Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (98)

Search Parameters:
Keywords = tropomyosin receptor kinase B

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1659 KiB  
Article
7,8-DHF Modulates Aggressive Behavior in Sebastes schlegelii: Phenotype-Dependent Responses in Aggression-Dimorphic Individuals
by Shufei Xu, Xinna Ma, Yang Xiao, Tao Zhang, Chao Ma and Zhen Ma
Animals 2025, 15(10), 1463; https://doi.org/10.3390/ani15101463 - 19 May 2025
Viewed by 454
Abstract
Aggressive behavior is regulated by intricate neural circuits and molecular mechanisms, notably through the interaction of brain-derived neurotrophic factor (BDNF) with its receptor, tropomyosin receptor kinase B (TrkB), which influences neuroplasticity and related behavioral phenotypes. We investigate the role of the BDNF signaling [...] Read more.
Aggressive behavior is regulated by intricate neural circuits and molecular mechanisms, notably through the interaction of brain-derived neurotrophic factor (BDNF) with its receptor, tropomyosin receptor kinase B (TrkB), which influences neuroplasticity and related behavioral phenotypes. We investigate the role of the BDNF signaling pathway in fish aggression using juvenile black rockfish (Sebastes schlegelii), which exhibit distinct aggressive phenotypes. The TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) was administered intraperitoneally at doses of 1.25, 2.5, and 5 mg/kg to assess its effects on the behavioral characteristics of high-aggression (H-agg) and low-aggression (L-agg) phenotypes. Our findings indicate the following: (1) The effects of 7,8-DHF are dose-dependent, with 2.5 mg/kg identified as the effective threshold dose for H-agg individuals; (2) in the H-agg group, this dose significantly reduced locomotor acceleration, angular velocity, and activity frequency, while prolonging the first movement latency; (3) in the L-agg group, only angular velocity was significantly decreased with the 2.5 mg/kg treatment, with no significant changes observed in other behavioral parameters. This study provides the first evidence for differential behavioral responses to 7,8-DHF in S. schlegelii, demonstrating dose-dependent aggression suppression in H-agg phenotypes and threshold-specific responses in L-agg phenotypes. These insights into the neuro-molecular basis of fish aggression can guide phenotype-specific management in aquaculture, potentially improving stress management, reducing injuries and mortality, and boosting productivity. Full article
Show Figures

Figure 1

22 pages, 11258 KiB  
Article
High-Risk Neuroblastoma Stage 4 (NBS4): Developing a Medicinal Chemistry Multi-Target Drug Approach
by Amgad Gerges and Una Canning
Molecules 2025, 30(10), 2211; https://doi.org/10.3390/molecules30102211 - 19 May 2025
Viewed by 718
Abstract
Childhood neuroblastoma (NB) is a malignant tumour that is a member of a class of embryonic tumours that have their origins in sympathoadrenal progenitor cells. There are five stages in the clinical NB staging system: 1, 2A, 2B, 3, 4S, and 4. For [...] Read more.
Childhood neuroblastoma (NB) is a malignant tumour that is a member of a class of embryonic tumours that have their origins in sympathoadrenal progenitor cells. There are five stages in the clinical NB staging system: 1, 2A, 2B, 3, 4S, and 4. For those diagnosed with stage 4 neuroblastoma (NBS4), the treatment options are limited with a survival rate of between 40 and 50%. Since 1975, more than 15 targets have been identified in the search for a treatment for high-risk NBS4. This article is concerned with the search for a multi-target drug treatment for high-risk NBS4 and focuses on four possible treatment targets that research has identified as having a role in the development of NBS4 and includes the inhibitors Histone Deacetylase (HDAC), Bromodomain (BRD), Hedgehog (HH), and Tropomyosin Kinase (TRK). Computer-aided drug design and molecular modelling have greatly assisted drug discovery in medicinal chemistry. Computational methods such as molecular docking, homology modelling, molecular dynamics, and quantitative structure–activity relationships (QSAR) are frequently used as part of the process for finding new therapeutic drug targets. Relying on these techniques, the authors describe a medicinal chemistry strategy that successfully identified eight compounds (inhibitors) that were thought to be potential inhibitors for each of the four targets listed above. Results revealed that all four targets BRD, HDAC, HH and TRK receptors binding sites share similar amino acid sequencing that ranges from 80 to 100%, offering the possibility of further testing for multi-target drug use. Two additional targets were also tested as part of this work, Retinoic Acid (RA) and c-Src (Csk), which showed similarity (of the binding pocket) across their receptors of 80–100% but lower than 80% for the other four targets. The work for these two targets is the subject of a paper currently in progress. Full article
Show Figures

Graphical abstract

35 pages, 1992 KiB  
Review
BDNF Signaling and Pain Modulation
by Mariacristina Mazzitelli, Takaki Kiritoshi, Peyton Presto, Zachary Hurtado, Nico Antenucci, Guangchen Ji and Volker Neugebauer
Cells 2025, 14(7), 476; https://doi.org/10.3390/cells14070476 - 22 Mar 2025
Cited by 2 | Viewed by 1938
Abstract
Brain-derived neurotrophic factor (BDNF) is an important neuromodulator of nervous system functions and plays a key role in neuronal growth and survival, neurotransmission, and synaptic plasticity. The effects of BDNF are mainly mediated by the activation of tropomyosin receptor kinase B (TrkB), expressed [...] Read more.
Brain-derived neurotrophic factor (BDNF) is an important neuromodulator of nervous system functions and plays a key role in neuronal growth and survival, neurotransmission, and synaptic plasticity. The effects of BDNF are mainly mediated by the activation of tropomyosin receptor kinase B (TrkB), expressed in both the peripheral and central nervous system. BDNF has been implicated in several neuropsychiatric conditions such as schizophrenia and anxio-depressive disorders, as well as in pain states. This review summarizes the evidence for a critical role of BDNF throughout the pain system and describes contrasting findings of its pro- and anti-nociceptive effects. Different cellular sources of BDNF, its influence on neuroimmune signaling in pain conditions, and its effects in different cell types and regions are described. These and endogenous BDNF levels, downstream signaling mechanisms, route of administration, and approaches to manipulate BDNF functions could explain the bidirectional effects in pain plasticity and pain modulation. Finally, current knowledge gaps concerning BDNF signaling in pain are discussed, including sex- and pathway-specific differences. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Neuropathic Pain)
Show Figures

Graphical abstract

48 pages, 1545 KiB  
Review
Molecular Regulation and Therapeutic Applications of Brain-Derived Neurotrophic Factor–Tropomyosin-Related Kinase B Signaling in Major Depressive Disorder Though Its Interaction with Vascular Endothelial Growth Factor and N-Methyl-D-Aspartic Acid Receptors: A Narrative Review
by Yasunari Matsuzaka and Ryu Yashiro
Biologics 2025, 5(1), 7; https://doi.org/10.3390/biologics5010007 - 3 Mar 2025
Cited by 2 | Viewed by 2370
Abstract
The molecular regulation and therapeutic applications of brain-derived neurotrophic factor (BDNF)–tropomyosin-related kinase B (TrkB) signaling in major depressive disorder (MDD) through interaction with vascular endothelial growth factor (VEGF) and N-methyl-D-aspartic acid (NMDA) receptors show promise. While BDNF-TrkB signaling is implicated in antidepressant action, [...] Read more.
The molecular regulation and therapeutic applications of brain-derived neurotrophic factor (BDNF)–tropomyosin-related kinase B (TrkB) signaling in major depressive disorder (MDD) through interaction with vascular endothelial growth factor (VEGF) and N-methyl-D-aspartic acid (NMDA) receptors show promise. While BDNF-TrkB signaling is implicated in antidepressant action, the association between BDNFs and depression has not yielded conclusive results. Some studies show decreased BDNF levels in depression, while others indicate that increased BDNF expression in certain brain regions can induce depression susceptibility. The role of BDNFs varies across different brain regions, necessitating further study of individual mechanisms. This regional variability complicates the development of targeted therapies. The antidepressant-like and neurotrophic actions of BDNFs require VEGF signaling, but there is also a reciprocal interdependence, as VEGF actions are dependent on BDNFs. This complex relationship complicates the development of targeted therapies. Full article
Show Figures

Figure 1

14 pages, 4556 KiB  
Article
Ouabain Counteracts Retinal Ganglion Cell Death Through Modulation of BDNF and IL-1 Signaling Pathways
by Amanda Candida da Rocha Oliveira, Camila Saggioro Figueiredo, Ícaro Raony, Juliana Salles Von-Held-Ventura, Marcelo Gomes Granja, Thalita Mázala-de-Oliveira, Vinícius Henrique Pedrosa-Soares, Aline Araujo dos Santos and Elizabeth Giestal-de-Araujo
Brain Sci. 2025, 15(2), 123; https://doi.org/10.3390/brainsci15020123 - 26 Jan 2025
Viewed by 1060
Abstract
Background: Ouabain is a steroid hormone that binds to the sodium pump (Na+, K+-ATPase) at physiological (nanomolar) concentrations, activating different signaling pathways. This interaction has been shown to prevent the axotomy-induced death of retinal ganglion cells (RGCs), although the [...] Read more.
Background: Ouabain is a steroid hormone that binds to the sodium pump (Na+, K+-ATPase) at physiological (nanomolar) concentrations, activating different signaling pathways. This interaction has been shown to prevent the axotomy-induced death of retinal ganglion cells (RGCs), although the underlying mechanisms remain unclear. Objective: In this study, we investigated potential mechanisms by which ouabain promotes RGC survival using primary cultures of rat neural retina. Results: Our findings indicate that ouabain regulates brain-derived neurotrophic factor (BDNF) signaling in retinal cells via matrix metalloproteinase-9-mediated processing of proBDNF to mature BDNF (mBDNF) and by increasing the phosphorylation of the mBDNF receptor, tropomyosin-related receptor kinase B. Ouabain also enhances the maturation of interleukin (IL)-1β through the increased activation of caspase-1, which mediates the processing of proIL-1β into IL-1β, and transiently upregulates both IL-1 receptor and IL-1 receptor antagonist (IL-1Ra). Treatment using either IL-1β or IL-1Ra alone is sufficient to enhance RGC survival similarly to that achieved with ouabain. Finally, we further show that ouabain prevents RGC death through a complex signaling mechanism shared by BDNF and IL-1β, which includes the activation of the Src and protein kinase C pathways. Conclusions: Collectively, these results suggest that ouabain stimulates the maturation and signaling of both BDNF and IL-1β, which act as key mediators of RGC survival. Full article
(This article belongs to the Special Issue Retinal Neurochemistry and Development)
Show Figures

Figure 1

13 pages, 10525 KiB  
Article
Dexmedetomidine Improves Learning Functions in Male Rats Modeling Cognitive Impairment by Modulating the BDNF/TrkB/CREB Signaling Pathway
by Sinan Saral, Tolga Mercantepe, Atilla Topçu, Ali Koray Kaya and Aykut Öztürk
Life 2024, 14(12), 1672; https://doi.org/10.3390/life14121672 - 17 Dec 2024
Viewed by 979
Abstract
Dexmedetomidine (DEX) is a selective alpha-2 adrenergic receptor agonist with sedative and anxiolytic properties. Increasing evidence reports that DEX has a neuroprotective effect. In this study, we investigated the potential effects of DEX on learning and memory functions in rats with experimental cognitive [...] Read more.
Dexmedetomidine (DEX) is a selective alpha-2 adrenergic receptor agonist with sedative and anxiolytic properties. Increasing evidence reports that DEX has a neuroprotective effect. In this study, we investigated the potential effects of DEX on learning and memory functions in rats with experimental cognitive impairment. In the study, 21 adult male rats were used. The rats were divided into three groups, namely control, Scopolamine (SCOP) and SCOP + DEX. Cognitive impairment was induced with 1 mg/kg SCOP daily for 21 days. DEX was administered at a dose of 10 µg/kg between days 14 and 21 of the experiment. Following the injections, a spatial memory test was performed with a Morris Water Maze (MWM). At the end of the experiment, the hippocampus was dissected. The brain-derived neurotrophic factor (BDNF), acetylcholine (ACh) and acetylcholinesterase (AChE) levels were determined by ELISA. The tropomyosin receptor kinase B (TrkB) and Cyclic AMP-Response Element-Binding Protein (CREB) levels were measured by immunohistochemistry. DEX treatment improved the learning performance of rats compared to SCOP for 5 days. However, it did not significantly change memory performance. DEX increased the BDNF and ACh levels in the hippocampus while decreasing the AChE levels. Similarly, DEX treatment significantly increased CREB phosphorylation. No significant difference was observed between the TrkB receptor levels of the groups. This study demonstrated that the role of DEX in reducing SCOP-induced cognitive impairment is partially mediated by the increase in BDNF/TrkB/CREB signaling pathway activity. Full article
(This article belongs to the Special Issue Feature Paper in Physiology and Pathology: 2nd Edition)
Show Figures

Figure 1

20 pages, 3524 KiB  
Article
Ameliorating Effect of Fermented Perilla frutescens on Sleep Deprivation-Induced Cognitive Impairment Through Antioxidant and BDNF Signaling in Mice
by Chae-Ryeong Seo, Bo Kyung Lee, Hye Jin Jee, Jae Ryeong Yoo, Chul-Kyu Lee, Jin Wook Park and Yi-Sook Jung
Nutrients 2024, 16(23), 4224; https://doi.org/10.3390/nu16234224 - 6 Dec 2024
Viewed by 1463
Abstract
Background: Adequate sleep is essential for maintaining cognitive function, as evidenced by literature. Perilla frutescens var. acuta Kudo (PF) is a traditional medicinal herb reported to improve vascular cognitive impairment and induce sedation. However, the effects of PF on cognitive impairment caused by [...] Read more.
Background: Adequate sleep is essential for maintaining cognitive function, as evidenced by literature. Perilla frutescens var. acuta Kudo (PF) is a traditional medicinal herb reported to improve vascular cognitive impairment and induce sedation. However, the effects of PF on cognitive impairment caused by sleep deprivation (SD) have not yet been evaluated. This study aims to evaluate the effects of fermented PF (FPF) and its underlying mechanisms in a model of SD-induced cognitive impairment. Methods: Mice were subjected to SD to establish cognitive impairment, and FPF was administered once daily for 3 days. Cognitive performance was assessed using Y-maze and passive avoidance tests, followed by molecular mechanisms analyses. Results: FPF treatment improved SD-induced cognitive impairment, as evidenced by increased spontaneous alternation and extended latency time. Histological analysis revealed that SD impaired the hippocampus, and this impairment was alleviated by FPF treatment. FPF demonstrated antioxidant activity by increasing glutathione levels and decreasing malondialdehyde levels. Furthermore, the decreased levels of brain-derived neurotrophic factor (BDNF) observed in sleep-deprived mice were restored with FPF treatment. FPF also enhanced the phosphorylation of tropomyosin receptor kinase B, extracellular signal-regulated kinase, and cAMP response element-binding protein. Conclusions: These results indicate that FPF may have beneficial effects on SD-induced cognitive impairment by protecting against oxidative stress and increasing BDNF expression. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Graphical abstract

15 pages, 1123 KiB  
Article
Activation of BDNF–TrkB Signaling in Specific Structures of the Sheep Brain by Kynurenic Acid
by Katarzyna Roszkowicz-Ostrowska, Patrycja Młotkowska, Elżbieta Marciniak, Michał Szlis, Marcin Barszcz and Tomasz Misztal
Cells 2024, 13(23), 1928; https://doi.org/10.3390/cells13231928 - 21 Nov 2024
Viewed by 1232
Abstract
Fluctuations in kynurenic acid (KYNA) and brain-derived neurotrophic factor (BDNF) levels in the brain reflect its neurological status. The aim of the study was to investigate the effect of transiently elevated KYNA concentrations in the cerebroventricular circulation on the expression of BDNF and [...] Read more.
Fluctuations in kynurenic acid (KYNA) and brain-derived neurotrophic factor (BDNF) levels in the brain reflect its neurological status. The aim of the study was to investigate the effect of transiently elevated KYNA concentrations in the cerebroventricular circulation on the expression of BDNF and its high-affinity tropomyosin-related kinase receptor B (TrkB) in specific structures of the sheep brain. Intracerebroventricularly cannulated anestrous sheep were subjected to a series of four 30 min infusions of KYNA: 4 × 5 μg/60 μL/30 min (KYNA20, n = 6) and 4 × 25 μg/60 μL/30 min (KYNA100, n = 6) or a control infusion (n = 6), at 30 min intervals. Sections of the hippocampal CA3 field, amygdala (AMG), prefrontal cortex (PCx), and the hypothalamic medial-basal (MBH) and preoptic (POA) areas were dissected from the brain immediately after the experiment. The highest concentration of BDNF protein was found in the CA3 field (p < 0.001), which was 8-fold higher than in the AMG and 12-fold higher than that in the PCx (MBH and POA were not analyzed). The most pronounced BDNF mRNA expression was observed in the MBH, followed by the PCx, POA, AMG and CA3, while the highest abundance of TrkB mRNA was recorded in the AMG, followed by the MBH, PCx, CA3, and POA. KYNA increased (p < 0.05–p < 0.01) BDNF protein levels and the expression of its gene in the brain structures were examined, with the effect varying by dose and brain region. KYNA, particularly at the KYNA100 dose, also increased (p < 0.01) TrkB gene expression, except for the AMG, where the lower KYNA20 dose was more effective (p < 0.01). These findings suggest a positive relationship between KYNA levels in the cerebroventricular circulation and BDNF–TrkB expression in specific brain regions in a sheep model. This indicates that a transient increase in the CSF KYNA concentration can potentially restore BDNF production, for which deficiency underlies numerous neurological disorders. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

17 pages, 10721 KiB  
Article
Ethyl Acetate Fraction of Chestnut Honey Attenuates Scopolamine-Induced Cognitive Impairment in Mice and Glutamate-Induced Neurotoxicity in HT22 Cells
by Yun Hee Jeong, Wei Li, Hye Jin Yang, Se-Gun Kim, Hong Min Choi, Jang-Gi Choi and You-Chang Oh
Antioxidants 2024, 13(11), 1346; https://doi.org/10.3390/antiox13111346 - 2 Nov 2024
Cited by 1 | Viewed by 1607
Abstract
Chestnut honey has various benefits, such as antioxidative, anti-inflammatory, immunomodulatory, antibacterial, and antiviral effects. However, the effects of chestnut honey or the ethyl acetate fraction of chestnut honey (EACH) on neurodegenerative diseases and their related cognitive impairment and neurotoxicity have not yet been [...] Read more.
Chestnut honey has various benefits, such as antioxidative, anti-inflammatory, immunomodulatory, antibacterial, and antiviral effects. However, the effects of chestnut honey or the ethyl acetate fraction of chestnut honey (EACH) on neurodegenerative diseases and their related cognitive impairment and neurotoxicity have not yet been established. Therefore, in this study, we investigated the mitigating effect of the EACH on scopolamine (SCO)-injected cognitive decline in mice and glutamate-exposed neurotoxicity in HT22 cells. EACH administration significantly reversed SCO-induced cognitive decline in mice, as demonstrated through the Morris water maze and passive avoidance tests. The EACH treatment showed a significant alleviation effect by recovering more than 80% of the cell viability decrease induced by glutamate exposure in the HT22 neuronal cell model. Furthermore, the EACH significantly reduced reactive oxygen species accumulation, lactate dehydrogenase release, mitochondrial depolarization, and neuronal apoptosis. The EACH regulated the level of apoptosis-related proteins, induced the nuclear translocation of nuclear factor-E2-related factor 2 (Nrf-2) and the expression of related antioxidant proteins, and induced the phosphorylation of tropomyosin-related kinase receptor B (TrkB)/cAMP-calcium response element-binding protein (CREB) and the expression of brain-derived neurotrophic factor. These data indicate that the EACH can prevent neurons from oxidative damage and improve cognitive dysfunction by activating Nrf-2 and TrkB/CREB signaling pathways. Therefore, the EACH demonstrates potential therapeutic value in mitigating oxidative stress-induced neurotoxicity, cognitive decline, and related neurodegenerative diseases. Full article
(This article belongs to the Special Issue Role of Natural Antioxidants on Neuroprotection)
Show Figures

Figure 1

38 pages, 3256 KiB  
Review
Harnessing Brain Plasticity: The Therapeutic Power of Repetitive Transcranial Magnetic Stimulation (rTMS) and Theta Burst Stimulation (TBS) in Neurotransmitter Modulation, Receptor Dynamics, and Neuroimaging for Neurological Innovations
by Minoo Sharbafshaaer, Giovanni Cirillo, Fabrizio Esposito, Gioacchino Tedeschi and Francesca Trojsi
Biomedicines 2024, 12(11), 2506; https://doi.org/10.3390/biomedicines12112506 - 1 Nov 2024
Cited by 11 | Viewed by 8782
Abstract
Transcranial magnetic stimulation (TMS) methods have become exciting techniques for altering brain activity and improving synaptic plasticity, earning recognition as valuable non-medicine treatments for a wide range of neurological disorders. Among these methods, repetitive TMS (rTMS) and theta-burst stimulation (TBS) show significant promise [...] Read more.
Transcranial magnetic stimulation (TMS) methods have become exciting techniques for altering brain activity and improving synaptic plasticity, earning recognition as valuable non-medicine treatments for a wide range of neurological disorders. Among these methods, repetitive TMS (rTMS) and theta-burst stimulation (TBS) show significant promise in improving outcomes for adults with complex neurological and neurodegenerative conditions, such as Alzheimer’s disease, stroke, Parkinson’s disease, etc. However, optimizing their effects remains a challenge due to variability in how patients respond and a limited understanding of how these techniques interact with crucial neurotransmitter systems. This narrative review explores the mechanisms of rTMS and TBS, which enhance neuroplasticity and functional improvement. We specifically focus on their effects on GABAergic and glutamatergic pathways and how they interact with key receptors like N-Methyl-D-Aspartate (NMDA) and AMPA receptors, which play essential roles in processes like long-term potentiation (LTP) and long-term depression (LTD). Additionally, we investigate how rTMS and TBS impact neuroplasticity and functional connectivity, particularly concerning brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase receptor type B (TrkB). Here, we highlight the significant potential of this research to expand our understanding of neuroplasticity and better treatment outcomes for patients. Through clarifying the neurobiology mechanisms behind rTMS and TBS with neuroimaging findings, we aim to develop more effective, personalized treatment plans that effectively address the challenges posed by neurological disorders and ultimately enhance the quality of neurorehabilitation services and provide future directions for patients’ care. Full article
Show Figures

Graphical abstract

26 pages, 2309 KiB  
Review
Natural Products and Their Neuroprotective Effects in Degenerative Brain Diseases: A Comprehensive Review
by Dong Wook Lim, Jung-Eun Lee, Changho Lee and Yun Tai Kim
Int. J. Mol. Sci. 2024, 25(20), 11223; https://doi.org/10.3390/ijms252011223 - 18 Oct 2024
Cited by 10 | Viewed by 8919
Abstract
As the global population ages, the incidence of neurodegenerative diseases such as Alzheimer’s and Parkinson’s is rapidly rising. These diseases present a significant public health challenge, as they severely impair cognitive and motor functions, ultimately leading to a substantial reduction in quality of [...] Read more.
As the global population ages, the incidence of neurodegenerative diseases such as Alzheimer’s and Parkinson’s is rapidly rising. These diseases present a significant public health challenge, as they severely impair cognitive and motor functions, ultimately leading to a substantial reduction in quality of life and placing a heavy burden on healthcare systems worldwide. Although several therapeutic agents have been developed to manage the symptoms of these diseases, their effectiveness is often limited, and there remains an urgent need for preventive strategies. Growing evidence indicates that bioactive compounds from natural products possess neuroprotective properties through antioxidant and anti-inflammatory effects, modulating key pathways such as phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) and brain-derived neurotrophic factor–tropomyosin receptor kinase B–cAMP response element-binding protein (BDNF-TrkB-CREB), which are crucial for neuronal survival. These compounds may also reduce amyloid-beta and tau pathology, as well as enhance cholinergic neurotransmission by inhibiting acetylcholinesterase activity. By targeting oxidative stress, neuroinflammation, and neurodegeneration, natural products offer a promising approach for both prevention and treatment. These findings suggest that natural products may be promising for preventing and treating neurodegenerative diseases. This review aims to explore the pathogenesis of neurodegenerative diseases, the limitations of current therapies, and the potential role of natural products as therapeutic agents. Full article
Show Figures

Figure 1

17 pages, 3711 KiB  
Article
A Combined Computational and Experimental Approach to Studying Tropomyosin Kinase Receptor B Binders for Potential Treatment of Neurodegenerative Diseases
by Duc D. Nguyen, Shomit Mansur, Lukasz Ciesla, Nora E. Gray, Shan Zhao and Yuping Bao
Molecules 2024, 29(17), 3992; https://doi.org/10.3390/molecules29173992 - 23 Aug 2024
Viewed by 1616
Abstract
Tropomyosin kinase receptor B (TrkB) has been explored as a therapeutic target for neurological and psychiatric disorders. However, the development of TrkB agonists was hindered by our poor understanding of the TrkB agonist binding location and affinity (both affect the regulation of disorder [...] Read more.
Tropomyosin kinase receptor B (TrkB) has been explored as a therapeutic target for neurological and psychiatric disorders. However, the development of TrkB agonists was hindered by our poor understanding of the TrkB agonist binding location and affinity (both affect the regulation of disorder types). This motivated us to develop a combined computational and experimental approach to study TrkB binders. First, we developed a docking method to simulate the binding affinity of TrkB and binders identified by our magnetic drug screening platform from Gotu kola extracts. The Fred Docking scores from the docking computation showed strong agreement with the experimental results. Subsequently, using this screening platform, we identified a list of compounds from the NIH clinical collection library and applied the same docking studies. From the Fred Docking scores, we selected two compounds for TrkB activation tests. Interestingly, the ability of the compounds to increase dendritic arborization in hippocampal neurons matched well with the computational results. Finally, we performed a detailed binding analysis of the top candidates and compared them with the best-characterized TrkB agonist, 7,8-dyhydroxyflavon. The screening platform directly identifies TrkB binders, and the computational approach allows for the quick selection of top candidates with potential biological activities based on the docking scores. Full article
(This article belongs to the Special Issue Featured Papers in Medicinal Chemistry II)
Show Figures

Graphical abstract

28 pages, 6931 KiB  
Review
BDNF-Regulated Modulation of Striatal Circuits and Implications for Parkinson’s Disease and Dystonia
by Daniel Wolf, Maurilyn Ayon-Olivas and Michael Sendtner
Biomedicines 2024, 12(8), 1761; https://doi.org/10.3390/biomedicines12081761 - 5 Aug 2024
Cited by 4 | Viewed by 2656
Abstract
Neurotrophins, particularly brain-derived neurotrophic factor (BDNF), act as key regulators of neuronal development, survival, and plasticity. BDNF is necessary for neuronal and functional maintenance in the striatum and the substantia nigra, both structures involved in the pathogenesis of Parkinson’s Disease (PD). Depletion of [...] Read more.
Neurotrophins, particularly brain-derived neurotrophic factor (BDNF), act as key regulators of neuronal development, survival, and plasticity. BDNF is necessary for neuronal and functional maintenance in the striatum and the substantia nigra, both structures involved in the pathogenesis of Parkinson’s Disease (PD). Depletion of BDNF leads to striatal degeneration and defects in the dendritic arborization of striatal neurons. Activation of tropomyosin receptor kinase B (TrkB) by BDNF is necessary for the induction of long-term potentiation (LTP), a form of synaptic plasticity, in the hippocampus and striatum. PD is characterized by the degeneration of nigrostriatal neurons and altered striatal plasticity has been implicated in the pathophysiology of PD motor symptoms, leading to imbalances in the basal ganglia motor pathways. Given its essential role in promoting neuronal survival and meditating synaptic plasticity in the motor system, BDNF might have an important impact on the pathophysiology of neurodegenerative diseases, such as PD. In this review, we focus on the role of BDNF in corticostriatal plasticity in movement disorders, including PD and dystonia. We discuss the mechanisms of how dopaminergic input modulates BDNF/TrkB signaling at corticostriatal synapses and the involvement of these mechanisms in neuronal function and synaptic plasticity. Evidence for alterations of BDNF and TrkB in PD patients and animal models are reviewed, and the potential of BDNF to act as a therapeutic agent is highlighted. Advancing our understanding of these mechanisms could pave the way toward innovative therapeutic strategies aiming at restoring neuroplasticity and enhancing motor function in these diseases. Full article
(This article belongs to the Special Issue Dopamine Signaling Pathway in Health and Disease—2nd Edition)
Show Figures

Figure 1

23 pages, 6915 KiB  
Review
Pyrazolo[1,5-a]pyrimidine as a Prominent Framework for Tropomyosin Receptor Kinase (Trk) Inhibitors—Synthetic Strategies and SAR Insights
by Amol T. Mahajan, Shivani, Ashok Kumar Datusalia, Carmine Coluccini, Paolo Coghi and Sandeep Chaudhary
Molecules 2024, 29(15), 3560; https://doi.org/10.3390/molecules29153560 - 29 Jul 2024
Cited by 2 | Viewed by 3775
Abstract
Tropomyosin receptor kinases (Trks) are transmembrane receptor tyrosine kinases named TrkA, TrkB, and TrkC and encoded by the NTRK1, NTRK2, and NTRK3 genes, respectively. These kinases have attracted significant attention and represent a promising therapeutic target for solid tumor treatment due to their [...] Read more.
Tropomyosin receptor kinases (Trks) are transmembrane receptor tyrosine kinases named TrkA, TrkB, and TrkC and encoded by the NTRK1, NTRK2, and NTRK3 genes, respectively. These kinases have attracted significant attention and represent a promising therapeutic target for solid tumor treatment due to their vital role in cellular signaling pathways. First-generation TRK inhibitors, i.e., Larotrectinib sulfate and Entrectinib, received clinical approval in 2018 and 2019, respectively. However, the use of these inhibitors was significantly limited because of the development of resistance due to mutations. Fortunately, the second-generation Trk inhibitor Repotrectinib (TPX-0005) was approved by the FDA in November 2023, while Selitrectinib (Loxo-195) has provided an effective solution to this issue. Another macrocycle-based analog, along with many other TRK inhibitors, is currently in clinical trials. Two of the three marketed drugs for NTRK fusion cancers feature a pyrazolo[1,5-a] pyrimidine nucleus, prompting medicinal chemists to develop numerous novel pyrazolopyrimidine-based molecules to enhance clinical applications. This article focuses on a comprehensive review of chronological synthetic developments and the structure–activity relationships (SAR) of pyrazolo[1,5-a]pyrimidine derivatives as Trk inhibitors. This article will also provide comprehensive knowledge and future directions to the researchers working in the field of medicinal chemistry by facilitating the structural modification of pyrazolo [1,5-a]pyrimidine derivatives to synthesize more effective novel chemotherapeutics as TRK inhibitors. Full article
Show Figures

Figure 1

27 pages, 3498 KiB  
Article
Molecular Adaptations of BDNF/NT-4 Neurotrophic and Muscarinic Pathways in Ageing Neuromuscular Synapses
by Marta Balanyà-Segura, Aleksandra Polishchuk, Laia Just-Borràs, Víctor Cilleros-Mañé, Carolina Silvera, Anna Ardévol, Marta Tomàs, Maria A. Lanuza, Erica Hurtado and Josep Tomàs
Int. J. Mol. Sci. 2024, 25(15), 8018; https://doi.org/10.3390/ijms25158018 - 23 Jul 2024
Cited by 3 | Viewed by 1574
Abstract
Age-related conditions, such as sarcopenia, cause physical disabilities for an increasing section of society. At the neuromuscular junction, the postsynaptic-derived neurotrophic factors brain-derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) have neuroprotective functions and contribute to the correct regulation of the exocytotic machinery. [...] Read more.
Age-related conditions, such as sarcopenia, cause physical disabilities for an increasing section of society. At the neuromuscular junction, the postsynaptic-derived neurotrophic factors brain-derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) have neuroprotective functions and contribute to the correct regulation of the exocytotic machinery. Similarly, presynaptic muscarinic signalling plays a fundamental modulatory function in this synapse. However, whether or not these signalling pathways are compromised in ageing neuromuscular system has not yet been analysed. The present study analyses, through Western blotting, the differences in expression and activation of the main key proteins of the BDNF/NT-4 and muscarinic pathways related to neurotransmission in young versus ageing Extensor digitorum longus (EDL) rat muscles. The main results show an imbalance in several sections of these pathways: (i) a change in the stoichiometry of BDNF/NT-4, (ii) an imbalance of Tropomyosin-related kinase B receptor (TrkB)-FL/TrkB-T1 and neurotrophic receptor p 75 (p75NTR), (iii) no changes in the cytosol/membrane distribution of phosphorylated downstream protein kinase C (PKC)βI and PKCε, (iv) a reduction in the M2-subtype muscarinic receptor and P/Q-subtype voltage-gated calcium channel, (v) an imbalance of phosphorylated mammalian uncoordinated-18-1 (Munc18-1) (S313) and synaptosomal-associated protein 25 (SNAP-25) (S187), and (vi) normal levels of molecules related to the management of acetylcholine (Ach). Based on this descriptive analysis, we hypothesise that these pathways can be adjusted to ensure neurotransmission rather than undergoing negative alterations caused by ageing. However, further studies are needed to assess this hypothetical suggestion. Our results contribute to the understanding of some previously described neuromuscular functional age-related impairments. Strategies to promote these signalling pathways could improve the neuromuscular physiology and quality of life of older people. Full article
(This article belongs to the Special Issue Molecular and Neuromuscular Mechanisms in Skeletal Muscle Aging)
Show Figures

Figure 1

Back to TopTop