[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (437)

Search Parameters:
Keywords = anti-acetylcholinesterase

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 938 KiB  
Review
Mechanistic and Therapeutic Insights into Flavonoid-Based Inhibition of Acetylcholinesterase: Implications for Neurodegenerative Diseases
by Natalia Cichon, Weronika Grabowska, Leslaw Gorniak, Maksymilian Stela, Piotr Harmata, Michal Ceremuga and Michal Bijak
Nutrients 2025, 17(1), 78; https://doi.org/10.3390/nu17010078 (registering DOI) - 28 Dec 2024
Viewed by 340
Abstract
Flavonoids are naturally occurring polyphenolic compounds known for their extensive range of biological activities. This review focuses on the inhibitory effects of flavonoids on acetylcholinesterase (AChE) and their potential as therapeutic agents for cognitive dysfunction. AChE, a serine hydrolase that plays a crucial [...] Read more.
Flavonoids are naturally occurring polyphenolic compounds known for their extensive range of biological activities. This review focuses on the inhibitory effects of flavonoids on acetylcholinesterase (AChE) and their potential as therapeutic agents for cognitive dysfunction. AChE, a serine hydrolase that plays a crucial role in cholinergic neurotransmission, is a key target in the treatment of cognitive impairments due to its function in acetylcholine hydrolysis. Natural polyphenolic compounds, particularly flavonoids, have demonstrated significant inhibition of AChE, positioning them as promising alternatives or adjuncts in neuropharmacology. This study specifically examines flavonoids such as quercetin, apigenin, kaempferol, and naringenin, investigating their inhibitory efficacy, binding mechanisms, and additional neuroprotective properties, including their antioxidant and anti-inflammatory effects. In vitro, in vivo, and in silico analyses reveal that these flavonoids effectively interact with both the active and peripheral anionic sites of AChE, resulting in increased acetylcholine levels and the stabilization of cholinergic signaling. Their mechanisms of action extend beyond mere enzymatic inhibition, as they also exhibit antioxidant and anti-amyloidogenic properties, thereby offering a multifaceted approach to neuroprotection. Given these findings, flavonoids hold considerable therapeutic potential as modulators of AChE, with implications for enhancing cognitive function and treating neurodegenerative diseases. Future studies should prioritize the enhancement of flavonoid bioavailability, evaluate their efficacy in clinical settings, and explore their potential synergistic effects when combined with established therapies to fully harness their potential as neurotherapeutic agents. Full article
Show Figures

Figure 1

Figure 1
<p>Neuroprotective effects of flavonoids. Flavonoids exhibit neuroprotective properties through multiple mechanisms. (1) Antioxidant activity: Flavonoids activate the Nrf2 (nuclear factor erythroid 2-related factor 2) pathway, enhancing antioxidant response element (ARE) signaling. This activation promotes the expression of antioxidant enzymes (such as glutathione and superoxide dismutase), which safeguard neurons from reactive oxygen species (ROS). (2) β-amyloid interaction: Flavonoids bind to β-amyloid, stabilizing non-toxic conformations and inhibiting aggregation, thereby reducing neurotoxic effects. (3) Anti-inflammatory effects: Flavonoids inhibit the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and MAPK (mitogen-activated protein kinase) pathways, which are critical for the production of pro-inflammatory cytokines. This inhibition leads to decreased levels of TNF-α, IL-6, and other inflammatory cytokines, highlighting the role of flavonoids in modulating microglial activation and inflammation. Created in BioRender. Bijak, M. (2024) <a href="https://BioRender.com/w91a417" target="_blank">https://BioRender.com/w91a417</a> accessed on 27 December 2024.</p>
Full article ">Figure 2
<p>Chemical structures of flavonoids investigated for acetylcholinesterase inhibition and neuroprotective activity. Created in BioRender. Bijak, M. (2024) <a href="https://BioRender.com/o63e959" target="_blank">https://BioRender.com/o63e959</a> accessed on 27 December 2024.</p>
Full article ">
22 pages, 1658 KiB  
Article
Optimization of a Pressurized Extraction Process Based on a Ternary Solvent System for the Recovery of Neuroprotective Compounds from Eucalyptus marginata Leaves
by Soumaya Hasni, Hajer Riguene, Jose A. Mendiola, Elena Ibáñez, Lidia Montero, Gloria Domínguez-Rodríguez, Hanene Ghazghazi, Ghayth Rigane and Ridha Ben Salem
Int. J. Mol. Sci. 2025, 26(1), 94; https://doi.org/10.3390/ijms26010094 - 26 Dec 2024
Viewed by 177
Abstract
Green chemistry focuses on reducing the environmental impacts of chemicals through sustainable practices. Traditional methods for extracting bioactive compounds from Eucalyptus marginata leaves, such as hydro-distillation and organic solvent extraction, have limitations, including long extraction times, high energy consumption, and potential toxic solvent [...] Read more.
Green chemistry focuses on reducing the environmental impacts of chemicals through sustainable practices. Traditional methods for extracting bioactive compounds from Eucalyptus marginata leaves, such as hydro-distillation and organic solvent extraction, have limitations, including long extraction times, high energy consumption, and potential toxic solvent residues. This study explored the use of supercritical fluid extraction (SFE), pressurized liquid extraction (PLE), and gas-expanded liquid (GXL) processes to improve efficiency and selectivity. These techniques were combined in a single mixture design, where CO2 was used in the experiments carried out under SFE, while water and ethanol were used for the PLE and GXL experiments by varying the concentration of the solvents to cover all the extraction possibilities. The neuroprotective activity of the extracts was evaluated by measuring their antioxidant, anti-inflammatory, and acetylcholinesterase inhibition properties. The optimization resulted in a novel GXL extraction with an optimal ternary mixture of 27% CO2, 55% ethanol, and 18% water, with a high degree of desirability (R2 = 88.59%). Chromatographic analysis carried out by GC-MS and HPLC-ESI-MS/MS identified over 49 metabolites. The designed sustainable extraction process offers a promising approach for producing phenolic-rich plant extracts in industrial applications. Full article
(This article belongs to the Special Issue Neuroprotective Effects of Food Ingredients)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Cumulative yield curves for various solvent compositions. (<b>b</b>) Total extraction yields represented in a ternary phase diagram.</p>
Full article ">Figure 2
<p>GC-MS chromatogram of the optimal GXL extract from <span class="html-italic">Eucalyptus marginata</span> leaves, obtained with CO<sub>2</sub>–ethanol–water (27:55:18, <span class="html-italic">v</span>/<span class="html-italic">v</span>/<span class="html-italic">v</span>) (100 bar, T = 50 °C).</p>
Full article ">Figure 3
<p>HPLC-ESI-MS chromatogram of the optimal GXL extract from <span class="html-italic">Eucalyptus marginata</span> leaves, obtained with CO<sub>2</sub>–ethanol–water (27:55:18, <span class="html-italic">v</span>/<span class="html-italic">v</span>/<span class="html-italic">v</span>) (100 bar, 50 °C).</p>
Full article ">Figure 4
<p>Suggested schematic and chemical structure for the fragmentation of quercetin-7-O-rutinoside (peak 12) in the HPLC-ESI-MS analysis.</p>
Full article ">Figure 5
<p>Suggested schematic and chemical structure for the fragmentation of kaempferol-rutinoside in the HPLC-ESI-MS analysis.</p>
Full article ">
23 pages, 7865 KiB  
Article
Antioxidant, Anti-α-Glucosidase, Anti-Tyrosinase, and Anti-Acetylcholinesterase Components from Stem of Rhamnus formosana with Molecular Docking Study
by Chia-Hsuan Tsai, Ya-Lun Liou, Sin-Min Li, Hsiang-Ruei Liao and Jih-Jung Chen
Antioxidants 2025, 14(1), 8; https://doi.org/10.3390/antiox14010008 - 24 Dec 2024
Viewed by 508
Abstract
Rhamnus formosana is a creeping evergreen shrub endemic to Taiwan. In traditional medicine, Rhamnaceae plants are used as herbal remedies for conditions such as itching, difficulty urinating, and constipation. This study explores the inhibitory effects of various solvent extracts and bioactive components of [...] Read more.
Rhamnus formosana is a creeping evergreen shrub endemic to Taiwan. In traditional medicine, Rhamnaceae plants are used as herbal remedies for conditions such as itching, difficulty urinating, and constipation. This study explores the inhibitory effects of various solvent extracts and bioactive components of R. formosana on α-glucosidase, tyrosinase, acetylcholinesterase (AChE), and antioxidant activity. The 100 °C water extract exhibited strong antioxidant activity in DPPH, ABTS, superoxide, and FRAP assays. The methanol extract demonstrated the highest α-glucosidase inhibitory effect, while the ethanol extract displayed potent AChE inhibition and the acetone extract showed the most potential tyrosinase inhibitory activity among the extracts. Five main biocomponents were isolated and evaluated for their bioactivities. Among them, kaempferol (1) and quercetin (2) exhibited notable antioxidant activity in DPPH and ABTS assays. Particularly, kaempferol (1) performed the best α-glucosidase inhibitory effect, physcion (5) showed the strongest AChE inhibition, and quercetin (2) demonstrated the most potential for tyrosinase inhibitory activity. Further molecular docking studies revealed that there may be stronger binding mechanisms between bioactive components and target enzymes (including α-glucosidase, acetylcholinesterase, and tyrosinase) than the positive control. These findings suggest that bioactive extracts and compounds from the stems of R. formosana may have potential as natural antioxidant, anti-α-glucosidase, anti-AChE, and anti-tyrosinase drug candidates or dietary supplements for the management of oxidative stress-related conditions, including hyperglycemia, pigmentation disorders, and neurodegenerative diseases. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Extraction and isolation of active ingredients from <span class="html-italic">Rhamnus formosana</span>.</p>
Full article ">Figure 2
<p>Chemical structures of kaempferol (<b>1</b>), quercetin (<b>2</b>), emodin (<b>3</b>), chrysophanol (<b>4</b>), and physcion (<b>5</b>) from <span class="html-italic">Rhamnus formosana</span>.</p>
Full article ">Figure 3
<p>Interactions of kaempferol (<b>1</b>) with α-glucosidase active binding site.</p>
Full article ">Figure 4
<p>Interaction of quercetin (<b>2</b>) with α-glucosidase active binding site.</p>
Full article ">Figure 5
<p>Interaction of emodin (<b>3</b>) with α-glucosidase active binding site.</p>
Full article ">Figure 6
<p>Interaction of acarbose with α-glucosidase active binding site.</p>
Full article ">Figure 7
<p>Interactions of emodin (<b>3</b>) with AChE active binding site.</p>
Full article ">Figure 8
<p>Interactions of physcion (<b>5</b>) with AChE active binding site.</p>
Full article ">Figure 9
<p>Interactions of quercetin (<b>2</b>) with AChE active binding site.</p>
Full article ">Figure 10
<p>Interaction of chlorogenic acid with AChE active binding site.</p>
Full article ">Figure 11
<p>Interaction of kaempferol (<b>1</b>) with tyrosinase active binding site.</p>
Full article ">Figure 12
<p>Interaction of quercetin (<b>2</b>) with tyrosinase active binding site.</p>
Full article ">Figure 13
<p>Interaction of arbutin with tyrosinase active binding site.</p>
Full article ">
21 pages, 2955 KiB  
Article
Multivalent Neuroprotective Activity of Elettaria cardamomum (Cardamom) and Foeniculum vulgare (Fennel) in H2O2-Induced Oxidative Stress in SH-SY5Y Cells and Acellular Assays
by Himadri Sharma, Hyewon Yang, Niti Sharma and Seong Soo A. An
Pharmaceuticals 2025, 18(1), 2; https://doi.org/10.3390/ph18010002 - 24 Dec 2024
Viewed by 215
Abstract
Background: Elettaria cardamomum (Cardamom) and Foeniculum vulgare (Fennel) are well-known spices and are also used as natural mouth fresheners. This study was performed to evaluate their neuroprotective ability based on certain acellular and cellular assays. Methods: Hexane and ethyl acetate extracts were prepared [...] Read more.
Background: Elettaria cardamomum (Cardamom) and Foeniculum vulgare (Fennel) are well-known spices and are also used as natural mouth fresheners. This study was performed to evaluate their neuroprotective ability based on certain acellular and cellular assays. Methods: Hexane and ethyl acetate extracts were prepared using cardamom and fennel seeds. GC/MS was performed for the identification of important bioactive compounds. Cell-based assays were performed using SH-SY5Y cells. Hydrogen peroxide was used for the induction of oxidative stress, and evaluation was done based on neuroprotection, reduced reactive oxygen species, and restoration of mitochondrial membrane potential (MMP). Additionally, anti-Aβ fibrillization/oligomerization activities were also analyzed along with anti-acetylcholinesterase activity. Results: α-Terpinyl acetate and anethol were identified as major phytocompounds in cardamom and fennel, respectively. Cardamom extracts and α-terpinyl acetate were more potent acetylcholinesterase (AChE) inhibitors than fennel extracts and anethol [IC50 cardamom extracts, 130–150 μg/mL; α-terpinyl acetate, 61.87 μg/mL; anethol, 374.2 μg/mL; fennel extracts, >1 mg/mL] and showed mixed-type inhibition. Only the extracts displayed potent anti-Aβ fibrilization activity (>50%). Anethol showed potent anti-Aβ oligomerization activity (>50%), followed by α-terpinyl acetate and fennel-H (~36%). The neuroprotective potential of the spice extracts/phytochemicals was evaluated in SH-SY5Y cells by using H2O2-induced oxidative stress. Cardamom-EA displayed the best neuroprotection (0.01 to 30 μg/mL). No neuroprotection was observed by α-terpinyl acetate and anethol. Cardamom extracts and fennel-H restored the normal reactive oxygen species (ROS) levels at 30 µg/mL and 1 µg/mL, respectively. Conclusion: Overall, the extracts provided better neuroprotection than the pure compounds in cellular assays and displayed strong anti-Aβ fibrilization activity. Full article
(This article belongs to the Special Issue Neuropharmacology of Plant Extracts and Their Active Compounds)
Show Figures

Figure 1

Figure 1
<p>The main phytocompound in (<b>A</b>) cardamom and (<b>B</b>) fennel extracts.</p>
Full article ">Figure 2
<p>IC<sub>50</sub> curves of cardamom and fennel extracts and their major phytocompounds (α-terpinyl acetate and anethol) with inhibitor control against AChE (<span class="html-italic">Electrophorus)</span>. The IC<sub>50</sub> values were computed using GraphPad Prism 10.2. All data are expressed as mean ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Lineweaver-Burk plots of AChE in the presence of 100 μg/mL and 200 μg/mL of (<b>A</b>) cardamom-H, (<b>B</b>) cardamom-EA, (<b>C</b>) α-terpinyl acetate, and (<b>D</b>) anethol. The graphs were plotted using GraphPad Prism 10.2. Abbreviations: V: Velocity of enzyme-catalyzed reaction; S: Substrate.</p>
Full article ">Figure 4
<p>Aβ oligomerization and fibrilization inhibition displayed by cardamom, fennel extracts, and pure compounds. (<b>A</b>) ThT anti-fibrilization assay. (<b>B</b>) Aβ oligomerization inhibition. All data were expressed as mean ± SD (<span class="html-italic">n</span> = 3). A significant difference * (<span class="html-italic">p</span> &lt; 0.05) and **** (<span class="html-italic">p</span> &lt; 0.0001) using the two-way ANOVA (<b>A</b>) and one-way ANOVA (<b>B</b>) followed by Dunnett’s post hoc test was observed in samples vs. negative control.</p>
Full article ">Figure 5
<p>Cytotoxicity assay of (<b>A</b>) cardamom-H, (<b>B</b>) cardamom-EA, (<b>C</b>) α-terpinyl acetate, (<b>D</b>) fennel-H, (<b>E</b>) fennel-EA, and (<b>F</b>) anethol on the SH-SY5Y cells. The cell viability was reported as the percentage of the control group (100%). All data were presented as mean ± SD (<span class="html-italic">n</span> = 3). No significant difference was observed using one-way ANOVA followed by Dunnett’s post hoc in the % of cell viability in the treated vs. the untreated control cells.</p>
Full article ">Figure 6
<p>Neuroprotective effects of various concentrations of (<b>A</b>) cardamom-H, (<b>B</b>) cardamom-EA, (<b>C</b>) α-terpinyl acetate, (<b>D</b>) fennel-H, (<b>E</b>) fennel-EA, and (<b>F</b>) anethol on H<sub>2</sub>O<sub>2</sub>-induced SH-SY5Y cells. The results indicate % cell viability vs. the control cells. All data were presented as mean ± SD (<span class="html-italic">n</span> = 3). Using one-way ANOVA followed by Dunnett’s test, a significant difference <sup>#</sup> (<span class="html-italic">p</span> &lt; 0.05), <sup>##</sup> (<span class="html-italic">p</span> &lt; 0.01), <sup>###</sup> (<span class="html-italic">p</span> &lt; 0.001), and ****<sup>/####</sup> (<span class="html-italic">p</span> &lt; 0.0001) was observed in the % of cell viability vs. untreated control cells (*) and H<sub>2</sub>O<sub>2</sub>-treated cells (<sup>#</sup>).</p>
Full article ">Figure 7
<p>Effects of various concentrations of (<b>A</b>) cardamom-H, (<b>B</b>) cardamom-EA, (<b>C</b>) fennel-H, and (<b>D</b>) fennel-EA on H<sub>2</sub>O<sub>2</sub>-induced ROS generation in neuroblastoma SH-SY5Y cells. The results indicate % cell viability vs. the control cells. All data are presented as mean ± SD (<span class="html-italic">n</span> = 3). Using one-way ANOVA followed by Dunnett’s test, a significant difference * (<span class="html-italic">p</span> &lt; 0.05), ** (<span class="html-italic">p</span> &lt; 0.01), ***<sup>/###</sup> (<span class="html-italic">p</span> &lt; 0.001), and ****<sup>/####</sup> (<span class="html-italic">p</span> &lt; 0.0001) was observed in the% of cell viability vs. untreated control cells (*) and H<sub>2</sub>O<sub>2</sub>-treated cells (<sup>#</sup>).</p>
Full article ">Figure 8
<p>Effects of various concentrations of (<b>A</b>) cardamom-H, (<b>B</b>) cardamom-EA, (<b>C</b>) fennel-H, and (<b>D</b>) fennel-EA on mitochondrial membrane potential (ΔΨm) in H<sub>2</sub>O<sub>2</sub>-induced SH-SY5Y cells. The results indicate % cell viability vs. the control cells. All data are presented as mean ± SD (<span class="html-italic">n</span> = 3). Using one-way ANOVA followed by Dunnett’s test, a significant difference <sup>#</sup> (<span class="html-italic">p</span> &lt; 0.05), **/<sup>##</sup> (<span class="html-italic">p</span> &lt; 0.01), ***/<sup>###</sup> (<span class="html-italic">p</span> &lt; 0.001), and ****<sup>/####</sup> (<span class="html-italic">p</span> &lt; 0.0001) was observed in the % of cell viability vs. untreated control cells (*) and H<sub>2</sub>O<sub>2</sub>-treated cells (<sup>#</sup>).</p>
Full article ">
19 pages, 1223 KiB  
Article
Anticholinesterase Activity and Bioactive Compound Profiling of Six Hop (Humulus lupulus L.) Varieties
by Bartłomiej Sagan, Bogusław Czerny, Anna Stasiłowicz-Krzemień, Piotr Szulc, Urszula Skomra, Tomasz M. Karpiński, Jolanta Lisiecka, Adam Kamiński, Aleksandra Kryszak, Oskar Zimak-Krótkopad and Judyta Cielecka-Piontek
Foods 2024, 13(24), 4155; https://doi.org/10.3390/foods13244155 - 22 Dec 2024
Viewed by 479
Abstract
Hops (Humulus lupulus L.) are widely recognized for their use in brewing, but they also possess significant pharmacological properties due to their rich bioactive compounds, with many varieties exhibiting diverse characteristics. This study investigates the chemical composition and biological activities of extracts [...] Read more.
Hops (Humulus lupulus L.) are widely recognized for their use in brewing, but they also possess significant pharmacological properties due to their rich bioactive compounds, with many varieties exhibiting diverse characteristics. This study investigates the chemical composition and biological activities of extracts from six hop varieties, focusing on quantifying xanthohumol and lupulone using High-Performance Liquid Chromatography (HPLC) and Total Phenolic Content (TPC) analysis. The hop varieties demonstrated significant variability in bioactive compound concentrations, with Aurora showing the highest xanthohumol (0.665 mg/g) and Zwiegniowski the highest lupulone (9.228 mg/g). TPC analysis revealed Aurora also had the highest phenolic content (22.47 mg GAE/g). Antioxidant activities were evaluated using DPPH, ABTS, CUPRAC, and FRAP assays, with Aurora and Oregon Fuggle displaying the most potent capacities. Aurora, in particular, showed the highest activity across multiple assays, including significant acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and tyrosinase inhibition, with IC50 values of 24.39 mg/mL, 20.38 mg/mL, and 9.37 mg/mL, respectively. The chelating activity was also assessed, with Apolon demonstrating the strongest metal ion binding capacity (IC50 = 1.04 mg/mL). Additionally, Aurora exhibited the most effective hyaluronidase inhibition (IC50 = 10.27 mg/mL), highlighting its potential for anti-inflammatory applications. The results underscore the influence of genetic and environmental factors on the bioactive compound profiles of hop varieties and their biological activity offering promising avenues for pharmaceutical and nutraceutical applications. However, further studies are needed to fully understand the potential interactions between hop cones components. Full article
(This article belongs to the Section Nutraceuticals, Functional Foods, and Novel Foods)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The chelating activity of hop flower extracts of different varieties is presented as IC<sub>50</sub> (mg/mL). Different letters (a–d) within the bars indicate statistical differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Inhibition of tyrosinase, by hop flower extracts of different varieties presented as IC<sub>50</sub> (mg/mL). Different letters (a–f) within the bars differ significantly (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Inhibition of hyaluronidase, by hop flower extracts of different varieties presented as IC<sub>50</sub> (mg/mL). Different letters (a–d) within the bars differ significantly (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Contributions of variables—xanthohumol (XAN), lupulone (LUP) content, Total Phenolic content (TPC), ABTS, DPPH, FRAP, CUPRAC, chelating (CHEL), acetylcholinesterase (AChE), butyrylcholinesterase (BChE), hyaluronidase (HYAL), tyrosinase (TYR) to PCs.</p>
Full article ">
22 pages, 6508 KiB  
Review
Alkaloids from Waltheria spp. (Malvaceae): Chemosystematic Aspects, Biosynthesis, Total Synthesis, and Biological Activities
by Raquel de M. Silva, Guilherme S. Caleffi and Fernando Cotinguiba
Int. J. Mol. Sci. 2024, 25(24), 13659; https://doi.org/10.3390/ijms252413659 - 20 Dec 2024
Viewed by 432
Abstract
Waltheria, a genus within the Malvaceae family, is abundantly distributed in tropical and subtropical areas worldwide. Many species of this genus are widely utilized in various ways, including chewing, in folk medicine, acting as an anti-inflammatory agent, and treating gastrointestinal disorders, rheumatism, [...] Read more.
Waltheria, a genus within the Malvaceae family, is abundantly distributed in tropical and subtropical areas worldwide. Many species of this genus are widely utilized in various ways, including chewing, in folk medicine, acting as an anti-inflammatory agent, and treating gastrointestinal disorders, rheumatism, and asthma, among other conditions. These applications are largely due to their secondary metabolites, primarily quinolone alkaloids and cyclopeptides. Several biological activities have been reported for Waltheria species, including antifungal, anticancer, trypanocidal, acetylcholinesterase inhibitory, potential anti-HIV, antinociceptive, analgesic, anti-inflammatory, antibacterial, antioxidant, and leishmanicidal activities. This review not only presents information on isolated alkaloids and their biological activities but also delves into biosynthetic, chemosystematic, medicinal chemistry, and total synthesis aspects. Additionally, the manuscript highlights other applications of alkaloids of the genus, such as a study on their herbicidal activity, which shows significant potential for agricultural use. Full article
Show Figures

Figure 1

Figure 1
<p>Degradation reaction of the alkaloids melicopine (<b>2</b>), melicopidine (<b>3</b>), and melicopicine (<b>4</b>), resulting in the formation of 1-methyl-4-quinolone (<b>5</b>). The corresponding acid, 1-methyl-4-quinolone-3-carboxylic acid (<b>1</b>), represents the first compound in the quinolone group (Adapted from Crow and Price, 1949 [<a href="#B14-ijms-25-13659" class="html-bibr">14</a>]).</p>
Full article ">Figure 2
<p>Examples of 4-quinolone alkaloids: phenyl-terminal open chain (waltherione G (<b>6</b>)), methyl-terminal open chain (antidesmone (<b>7</b>)), and cyclized (waltherione A (<b>8</b>)).</p>
Full article ">Figure 3
<p>4-quinolone alkaloids melochinone (<b>9</b>) and melovinone (<b>10</b>).</p>
Full article ">Figure 4
<p>Basic structure of cyclopeptide alkaloids (adapted from Gehm et al., 2022 [<a href="#B36-ijms-25-13659" class="html-bibr">36</a>]).</p>
Full article ">Figure 5
<p>Pandamine (<b>17</b>) isolated from <span class="html-italic">Panda oleosa</span> (Pandaceae).</p>
Full article ">Figure 6
<p>Lasiodine A (<b>38</b>) (adapted from Marchand et al., 1969 [<a href="#B46-ijms-25-13659" class="html-bibr">46</a>]).</p>
Full article ">Figure 7
<p><span class="html-italic">p</span>-phenylenedialanine (<b>42</b>) and its dihydrogenated counterpart (<b>43</b>) (adapted from Schmidt et al., 1985 [<a href="#B49-ijms-25-13659" class="html-bibr">49</a>]).</p>
Full article ">Figure 8
<p>Design strategy of the target compounds: <b>120</b> by the introduction of the ester group (<span class="html-italic">orange</span>) and pyrazole ring (<span class="html-italic">lilac</span>); compounds <b>121</b> and <b>122</b> by the introduction of an aromatic amide group (<span class="html-italic">green</span>); compounds <b>123</b> and <b>124</b> combining pyrazole ring (<span class="html-italic">lilac</span>) and piperazine-amide groups (<span class="html-italic">yellow</span>).</p>
Full article ">Figure 9
<p>Design strategy of target compounds <b>125</b>, <b>126</b>, and <b>127</b> by the introduction of the acethydrazide group (<span class="html-italic">pale pink</span>) and varying the presence of methyl in the quinoline ring.</p>
Full article ">Scheme 1
<p>Formation of the 4-quinolone nucleus (<b>23</b>) via anthranyl-CoA (<b>18</b>) and malonyl-CoA (<b>19</b>) (adapted from Dewick, 2009 [<a href="#B43-ijms-25-13659" class="html-bibr">43</a>]).</p>
Full article ">Scheme 2
<p>Melochinone biosynthesis proposal (adapted from Kapadia et al., 1975 [<a href="#B17-ijms-25-13659" class="html-bibr">17</a>]).</p>
Full article ">Scheme 3
<p>Antidesmone (<b>7</b>) biosynthesis (adapted from Bringmann et al., 2000 [<a href="#B19-ijms-25-13659" class="html-bibr">19</a>]).</p>
Full article ">Scheme 4
<p>Probable precursors of cyclopeptide alkaloids. Curved arrows (<span class="html-italic">red</span>) indicate probable formation of the macrocycle (adapted from Warnhoff, 1971; Bhat et al., 1987 [<a href="#B47-ijms-25-13659" class="html-bibr">47</a>,<a href="#B48-ijms-25-13659" class="html-bibr">48</a>]).</p>
Full article ">Scheme 5
<p>Kaufman and Larghi’s total synthesis of waltherione F (<b>44</b>).</p>
Full article ">Scheme 6
<p>Kaufman and Larghi’s total synthesis of melovinone (<b>10</b>).</p>
Full article ">Scheme 7
<p>Cox’s total synthesis of waltherione F (<b>44</b>).</p>
Full article ">Scheme 8
<p>Pabbaraja and Mehta’s total synthesis of waltherione F (<b>44</b>).</p>
Full article ">Scheme 9
<p>Nakagawa-Goto’s total synthesis of waltherione A (<b>8</b>).</p>
Full article ">
18 pages, 4432 KiB  
Review
Traditional Uses, Bioactive Compounds, and New Findings on Pharmacological, Nutritional, Cosmetic and Biotechnology Utility of Capsella bursa-pastoris
by Aleksandra Łukaszyk, Inga Kwiecień and Agnieszka Szopa
Nutrients 2024, 16(24), 4390; https://doi.org/10.3390/nu16244390 - 20 Dec 2024
Viewed by 554
Abstract
Capsella bursa-pastoris (L.) Medik. (shepherd’s purse) is a medicinal plant recently introduced to European Pharmacopoeia. The main active compounds responsible for the activity profile of the raw material are flavonoids, phenolic acids, amino acids, phytosterols, vitamins and bioelements. This species is known for [...] Read more.
Capsella bursa-pastoris (L.) Medik. (shepherd’s purse) is a medicinal plant recently introduced to European Pharmacopoeia. The main active compounds responsible for the activity profile of the raw material are flavonoids, phenolic acids, amino acids, phytosterols, vitamins and bioelements. This species is known for its properties supporting the functioning of the digestive system and antihemorrhagic properties in the ethnomedicine of Far Eastern countries. Modern research confirms these directions of activity. Additionally, the latest studies prove the anti-inflammatory, antioxidant, antibacterial, antifungal, acetylcholinesterase and anticancer properties and supportive action in the treatment of gynecological diseases. Shepherd’s purse herb also has a strong position as an edible plant due to the growing interest in this plant as “healthy food”. The protective, softening, antibacterial and antioxidant properties of sprout and herb extracts are useful in the production of modern cosmetics. Moreover, C. bursa-pastoris is valuable thanks to phytoremediation properties and the numerous practical uses in biotechnology for the creation of new resistant varieties of crop plants from the Brassicaceae family. Full article
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Capsella bursa-pastoris</span> plant (<b>a</b>); inflorescence shoot with cauline leaves (<b>b</b>); inflorescence shoot with flowers and heart-shaped siliques (<b>c</b>).</p>
Full article ">Figure 2
<p><span class="html-italic">Capsella bursa-pastoris</span> distribution map.</p>
Full article ">Figure 3
<p>Aglycone structures of flavonoids found in <span class="html-italic">Capsella bursa-pastoris</span> extracts: kaempferol (<b>a</b>), apigenin (<b>b</b>), sinensetin (<b>c</b>), quercetin (<b>d</b>), luteolin (<b>e</b>), acacetin (<b>f</b>), isorhamnetin (<b>g</b>), diosmetin (<b>h</b>), chryseriol (<b>i</b>), icaritin (<b>j</b>), licoflavonol (<b>k</b>) and tricin (<b>l</b>).</p>
Full article ">Figure 4
<p>Examples of phenolic glycosides reported in <span class="html-italic">Capsella bursa-pastoris</span>: lariciresinol-4′-<span class="html-italic">O-β</span>-D-glucoside (<b>a</b>); (+)-pinoresinol-<span class="html-italic">β</span>-D-glucoside (<b>b</b>); salidroside (<b>c</b>); <span class="html-italic">β</span>-hydroxy-propiovanillone-3-<span class="html-italic">O-β</span>-D-glucopyranoside (<b>d</b>).</p>
Full article ">Figure 5
<p>New metabolites recently isolated from the herb—capselic acid A (<b>a</b>) and seeds of <span class="html-italic">Capsella bursa-pastoris</span>—capselloside (<b>b</b>); bursapastoris A (<b>c</b>); bursapastoris B (<b>d</b>); 10-(methylsulfinyl)decanoic acid (<b>e</b>); 11-(methylsulfinyl)undecanoic acid (<b>f</b>).</p>
Full article ">
13 pages, 1984 KiB  
Article
A Novel Galantamine–Curcumin Hybrid Inhibits Butyrylcholinesterase: A Molecular Dynamics Study
by Evdokiya Salamanova, Mariyana Atanasova and Irini Doytchinova
Chemistry 2024, 6(6), 1645-1657; https://doi.org/10.3390/chemistry6060100 - 16 Dec 2024
Viewed by 430
Abstract
Cholinesterases are enzymes that break down the neurotransmitter acetylcholine in the nervous system. The two main types are acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). AChE inhibitors are used to treat Alzheimer’s disease by increasing acetylcholine levels. BChE activity increases in later stages of Alzheimer’s, [...] Read more.
Cholinesterases are enzymes that break down the neurotransmitter acetylcholine in the nervous system. The two main types are acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). AChE inhibitors are used to treat Alzheimer’s disease by increasing acetylcholine levels. BChE activity increases in later stages of Alzheimer’s, suggesting it might contribute to the disease. In previous experiments, it was found that a newly designed hybrid of galantamine (GAL) and curcumin (CCN) (compound 4b) decreases the activity of BChE in murine brain homogenates. Here, we explore this observation using molecular dynamics simulations. GAL and CCN were also studied for comparison. The structures of the complexes between the BChE and the ligands were predicted by molecular docking. Then, molecular dynamics simulations were performed to evaluate the stability of the complexes and the interactions between the ligands and the enzyme over a simulated time of 1 μs. All three ligands formed stable complexes with BChE. Compound 4b formed more hydrogen bonds and other interactions with BChE compared to GAL and CCN, suggesting a stronger binding affinity. The stronger binding of 4b to BChE might explain its superior anti-BChE activity observed in previous experiments. Full article
(This article belongs to the Section Theoretical and Computational Chemistry)
Show Figures

Figure 1

Figure 1
<p>Structures of galantamine (GAL), curcumin (CCN), and their hybrid, <b>4b</b>.</p>
Full article ">Figure 2
<p>Superimposed conformations of the three complexes in the input conformation (blue) and in the most populated conformation (red). The enzyme is given in grey. The catalytic triad residues Ser195, Glu322, and His435 are given in green. (<b>Left</b>): BChE–GAL, (<b>middle</b>): BChE–CCN, (<b>right</b>): BChE–<b>4b</b>. The final conformation (cyan) is also given for <b>4b</b>. Ser195, Glu322, and His435 correspond to Ser198, Glu325, and His438 from human BChE (PDB ID: 6QAA).</p>
Full article ">Figure 3
<p>Superimposed representative frames of the most populated conformations of GAL (blue), CCN (red), and <b>4b</b> (green).</p>
Full article ">Figure 4
<p>RMSDs of the enzyme (<b>left panels</b>) and the ligands (<b>right panels</b>) in the studied complexes averaged over the three runs. The representative frame of the most populated conformation for each complex was used as a reference.</p>
Full article ">Figure 5
<p>Intermolecular hydrogen bonds between BChE and ligands GAL (<b>top</b>), CCN (<b>middle</b>), and <b>4b</b> (<b>bottom</b>) with lifetime ≥ 100 ns. The former atom corresponds to the H-bond acceptor and the latter to the H-bond donor. Atom numbering aligns with <a href="#chemistry-06-00100-f001" class="html-fig">Figure 1</a> for reference.</p>
Full article ">Figure 6
<p>Intermolecular interactions between BChE and ligands GAL (<b>top</b>), CCN (<b>middle</b>), and <b>4b</b> (<b>bottom</b>) detected in the most populated conformations of the complexes. Conventional hydrogen bonds (between heteroatoms) are given in dark green dashes, carbon hydrogen bonds—in light green, electrostatic attractions—in orange, π–π interactions—in dark purple, hydrophobic interactions—in light purple, and van der Waals interactions—without dashes.</p>
Full article ">Figure 6 Cont.
<p>Intermolecular interactions between BChE and ligands GAL (<b>top</b>), CCN (<b>middle</b>), and <b>4b</b> (<b>bottom</b>) detected in the most populated conformations of the complexes. Conventional hydrogen bonds (between heteroatoms) are given in dark green dashes, carbon hydrogen bonds—in light green, electrostatic attractions—in orange, π–π interactions—in dark purple, hydrophobic interactions—in light purple, and van der Waals interactions—without dashes.</p>
Full article ">
19 pages, 1992 KiB  
Article
Resveratrol-Loaded Pluronic Micelles Ameliorate Scopolamine-Induced Cognitive Dysfunction Targeting Acetylcholinesterase Activity and Programmed Cell Death
by Maria Lazarova, Miroslava Stefanova, Elina Tsvetanova, Almira Georgieva, Krasimira Tasheva, Lyubomira Radeva and Krassimira Yoncheva
Int. J. Mol. Sci. 2024, 25(23), 12777; https://doi.org/10.3390/ijms252312777 - 28 Nov 2024
Viewed by 622
Abstract
Numerous experimental studies suggest the potential for resveratrol (RVT) to be useful in the Alzheimer’s disease treatment, but its low bioavailability limits its application. This study aimed to assess the potential of resveratrol-loaded micelles as a neuronal delivery platform to protect rats from [...] Read more.
Numerous experimental studies suggest the potential for resveratrol (RVT) to be useful in the Alzheimer’s disease treatment, but its low bioavailability limits its application. This study aimed to assess the potential of resveratrol-loaded micelles as a neuronal delivery platform to protect rats from scopolamine-induced memory impairment. Resveratrol was incorporated into Pluronic micelles, and the effects of micellar (mRVT) and pure resveratrol (RVT) were compared in the model of scopolamine-induced dementia in male Wistar rats. Memory performance was assessed by a T maze test. The effect of the treatment on specific neurotransmitter levels and protein expression in the cortex and the hippocampus were evaluated biochemically. Our results revealed that the polymeric micelles were in nanoscale (approximately 33 nm) and reached 79% encapsulation efficiency. The treatment with mRVT demonstrated better spatial memory protective effect. The biochemical assays showed that mRVT in a dose of 10 mg/kg enhanced the effects of the pure drug in regard to noradrenalin neurotransmission and acetylcholinesterase inhibitory activity in the hippocampus. Furthermore, micellar resveratrol increased the cAMP-response element-binding protein expression in the cortex and hippocampus of rats as well as the Bcl2/BAX ratio, which indicated an anti-apoptotic effect in the experimental dementia model. In conclusion, our results indicated the potential of a micellar system loaded with resveratrol for neurodegenerative diseases treatment. Full article
Show Figures

Figure 1

Figure 1
<p>In vitro dissolution of encapsulated (mRVT) and pure resveratrol (RVT) in a phosphate buffer with pH = 7.4.</p>
Full article ">Figure 2
<p>Effect of mRVT (5 and 10 mg/kg) and RVT (5 and 10 mg/kg) on rewarded spontaneous alternation behavior of rats before all treatments (<b>A</b>) and after 11 days’ scopolamine treatment (<b>B</b>) evaluated by the T maze test. Mean values ± SEM (<span class="html-italic">n</span> = 12 animals per group). Statistical analysis involved one-way analysis of variance (ANOVA), followed by Tukey’s multiple comparison test. Significance vs. saline-treated group: ### <span class="html-italic">p</span> &lt; 0.001; significance vs. Sco-treated group: *** <span class="html-italic">p</span> &lt; 0.001; significance between RVT and mRVT-treated groups: ƒ <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Effect of pure (RVT) and micellar resveratrol (mRVT) at concentrations of 5 and 10 mg/kg on locomotion (<b>A</b>), anxiety, and exploratory activity (<b>B</b>) of rats with scopolamine-induced memory impairment in the hole-board test. Mean values ± SEM (<span class="html-italic">n</span> = 12 animals per group). Statistical analysis involved one-way analysis of variance (ANOVA), followed by Tukey’s multiple comparison test. Significance vs. saline-treated group: # <span class="html-italic">p</span> &lt; 0.05; significance vs. Sco-treated group: * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001; significance between RVT and mRVT-treated groups: ƒƒƒ <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Effect of pure (RVT) and micellar resveratrol (mRVT) at 5 and 10 mg/kg concentrations on AChE activity and ACh content in cortex (<b>A</b>,<b>C</b>) and hippocampus (<b>B</b>,<b>D</b>) of rats with scopolamine-induced memory impairment. Mean values ± SEM (<span class="html-italic">n</span> = 6 animals per group). Statistical analysis involved one-way analysis of variance (ANOVA), followed by Tukey’s multiple comparison test. Significance vs. saline-treated group: # <span class="html-italic">p</span> &lt; 0.05; ## <span class="html-italic">p</span> &lt; 0.01 significance vs. Sco-treated group: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; significance between RVT and mRVT-treated groups: ƒ <span class="html-italic">p</span> &lt; 0.05, ƒƒ <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Effect of pure (RVT) and micellar resveratrol (mRVT) at 5 and 10 mg/kg concentrations on DA, NA, and Sero content in cortex (<b>A</b>–<b>C</b>) and hippocampus (<b>D</b>–<b>F</b>) of rats with scopolamine-induced memory impairment. Mean values ± SEM (<span class="html-italic">n</span> = 6 animals per group). Statistical analysis involved one-way analysis of variance (ANOVA), followed by Tukey’s multiple comparison test. Significance vs. saline-treated group: ### <span class="html-italic">p</span> &lt; 0.001; significance vs. Sco-treated group: ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; significance between RVT/mRVT-treated groups: ƒ <span class="html-italic">p</span> &lt; 0.05; ƒƒ <span class="html-italic">p</span> &lt; 0.01; ƒƒƒ <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Effect of pure (RVT) and micellar resveratrol (mRVT) (5 and 10 mg/kg) on BDNF and pCREB protein expression in cortex (<b>A</b>,<b>C</b>) and hippocampus (<b>B</b>,<b>D</b>) of rats with Sco-induced memory deficit. Each column represents mean ± S.E.M. of 6 animals. Data analysis was performed using one-way ANOVA followed by Tukey’s multiple comparison test. Significance versus saline-treated group: ### <span class="html-italic">p</span> &lt; 0.001; significance versus Sco-treated group: *** <span class="html-italic">p</span> &lt; 0.001; significance between RVT and mRVT-treated groups: ƒƒƒ <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Effect of pure (RVT) and micellar (mRVT) resveratrol (5 and 10 mg/kg) on Bcl2 and BAX protein expression and Bcl2/BAX ratio in cortex (<b>A</b>–<b>C</b>) and hippocampus (<b>D</b>–<b>F</b>) of rats with Sco-induced memory deficit. Each column represents mean ± S.E.M. of 6 animals. Data analysis was performed using one-way ANOVA followed by Tukey’s multiple com-parison test. Significance versus saline-treated group: # <span class="html-italic">p</span> &lt; 0.05, ### <span class="html-italic">p</span> &lt; 0.001; significance versus Sco-treated group: * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001; significance between RVT and mRVT-treated groups: ƒƒ <span class="html-italic">p</span> &lt; 0.01, ƒƒƒ <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Timeline of the experiment.</p>
Full article ">
19 pages, 3888 KiB  
Article
Natural Deep Eutectic Solvents Combined with Supercritical Carbon Dioxide for the Extraction of Curcuminoids from Turmeric
by Anna Stasiłowicz-Krzemień, Julia Wójcik, Anna Gościniak, Marcin Szymański, Piotr Szulc, Krzysztof Górecki and Judyta Cielecka-Piontek
Pharmaceuticals 2024, 17(12), 1596; https://doi.org/10.3390/ph17121596 - 27 Nov 2024
Viewed by 628
Abstract
Background: Curcuminoids, the bioactive compounds found in turmeric, exhibit potent antioxidant, anti-inflammatory, and neuroprotective properties. This study aims to enhance the extraction of curcuminoids from turmeric using environmentally friendly solvents supercritical CO2 (scCO2) combined with natural deep eutectic solvents (NADESs) [...] Read more.
Background: Curcuminoids, the bioactive compounds found in turmeric, exhibit potent antioxidant, anti-inflammatory, and neuroprotective properties. This study aims to enhance the extraction of curcuminoids from turmeric using environmentally friendly solvents supercritical CO2 (scCO2) combined with natural deep eutectic solvents (NADESs) in one process, and to evaluate the resulting biological activity. Methods: A Box–Behnken statistical design was applied to optimize scCO2 extraction conditions—pressure, CO2 volume, and temperature—to maximize curcuminoid yield. Next, the menthol and lactic acid NADESs were selected, and these two solvents were combined into a single turmeric extraction process. The biological activity of the resulting extract was evaluated using antioxidant assays (ferric reducing antioxidant power and 2,2-diphenyl-1-picrylhydrazyl) and enzyme inhibition assays (acetylcholinesterase, butyrylcholinesterase, and tyrosinase). Toxicity assessments were conducted on the aquatic invertebrates Daphnia pulex, Artemia sp., and Chironomus aprilinus. Results: The most effective extraction was achieved using a menthol–lactic acid NADES as a cosolvent, integrated at a 1:20 ratio of plant material to NADESs while in combination with scCO2. The optimized scCO2–NADES extraction resulted in a high curcuminoid yield (33.35 mg/g), outperforming scCO2 extraction (234.3 μg/g), NADESs ultrasound-assisted extraction (30.50 mg/g), and alcohol-based solvents (22.95–26.42 mg/g). In biological assays, the extract demonstrated significant antioxidant activity and effective inhibition of enzymes (acetylcholinesterase, butyrylcholinesterase, and tyrosinase). Toxicity studies showed a concentration-dependent response, with EC50 for Chironomus aprilinus at the level of 0.098 μL/mL and Daphnia pulex exhibiting high sensitivity to the extract. Conclusions: This study highlights the potential of combining NADESs and scCO2 extraction in one process, demonstrating the effectiveness of scCO2–NADES extraction in maximizing curcuminoid yield and enhancing bioactivity. Full article
(This article belongs to the Special Issue Natural Products in Health Promotion and Disease Prevention 2024)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The chromatogram of curcuminoids present in turmeric extracts.</p>
Full article ">Figure 2
<p>Pareto chart of standardized effects of Box–Behnken experimental analysis for curcuminoid content in the extracts.</p>
Full article ">Figure 3
<p>Response surface curve illustrating the effect of pressure to temperature (<b>a</b>), pressure to CO<sub>2</sub> volume (<b>b</b>), and CO<sub>2</sub> volume to temperature (<b>c</b>) on curcuminoid content.</p>
Full article ">Figure 4
<p>Curcuminoid content in the extracts was obtained using various ratios of plant material to eutectic solvent (<span class="html-italic">m</span>/<span class="html-italic">v</span>). Different letters (a–f) within the bars indicate statistical differences between curcuminoid content in the extracts (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>The antioxidant potential of the extract in DPPH (<b>A</b>) and FRAP (<b>B</b>) assays with references (curcumin, trolox). Different letters (a–c) within the bars indicate statistical differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>The potential of the extract to inhibit acetylcholinesterase (<b>A</b>), butyrylcholinesterase (<b>B</b>), and tyrosinase (<b>C</b>) with references (curcumin, galantamine, azelaic acid). Different letters (a–c) within the bars indicate statistical differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>Percentage mortality of test organisms after 24 h of incubation with CUR–scCO<sub>2</sub>–NADES_1:20 (10 µL/mL).</p>
Full article ">Figure 8
<p>Graphs of the dependence of the number of dead organisms of <span class="html-italic">Daphnia p.</span> (<b>A</b>) and <span class="html-italic">Chironomus a.</span> (<b>B</b>) on the incubation time.</p>
Full article ">Figure 9
<p>Petri dishes with test organisms—preliminary studies.</p>
Full article ">
38 pages, 2898 KiB  
Article
Therapeutic Options in Alzheimer’s Disease: From Classic Acetylcholinesterase Inhibitors to Multi-Target Drugs with Pleiotropic Activity
by Ramón Cacabelos, Olaia Martínez-Iglesias, Natalia Cacabelos, Iván Carrera, Lola Corzo and Vinogran Naidoo
Life 2024, 14(12), 1555; https://doi.org/10.3390/life14121555 - 26 Nov 2024
Viewed by 921
Abstract
Alzheimer’s disease (AD) is a complex/multifactorial brain disorder involving hundreds of defective genes, epigenetic aberrations, cerebrovascular alterations, and environmental risk factors. The onset of the neurodegenerative process is triggered decades before the first symptoms appear, probably due to a combination of genomic and [...] Read more.
Alzheimer’s disease (AD) is a complex/multifactorial brain disorder involving hundreds of defective genes, epigenetic aberrations, cerebrovascular alterations, and environmental risk factors. The onset of the neurodegenerative process is triggered decades before the first symptoms appear, probably due to a combination of genomic and epigenetic phenomena. Therefore, the primary objective of any effective treatment is to intercept the disease process in its presymptomatic phases. Since the approval of acetylcholinesterase inhibitors (Tacrine, Donepezil, Rivastigmine, Galantamine) and Memantine, between 1993 and 2003, no new drug was approved by the FDA until the advent of immunotherapy with Aducanumab in 2021 and Lecanemab in 2023. Over the past decade, more than 10,000 new compounds with potential action on some pathogenic components of AD have been tested. The limitations of these anti-AD treatments have stimulated the search for multi-target (MT) drugs. In recent years, more than 1000 drugs with potential MT function have been studied in AD models. MT drugs aim to address the complex and multifactorial nature of the disease. This approach has the potential to offer more comprehensive benefits than single-target therapies, which may be limited in their effectiveness due to the intricate pathology of AD. A strategy still unexplored is the combination of epigenetic drugs with MT agents. Another option could be biotechnological products with pleiotropic action, among which nosustrophine-like compounds could represent an attractive, although not definitive, example. Full article
(This article belongs to the Special Issue New Trends in Pharmaceutical Science: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Confluent pathogenetic cascades and risk factors contributing to the phenotypic definition of Alzheimer’s disease.</p>
Full article ">Figure 2
<p>Correlation analysis between number of mutant AD genes per patient and age at diagnosis.</p>
Full article ">Figure 3
<p>Effect of Nosustrophine on neuropathological hallmarks in transgenic AD mouse models. Comparative photomicrographs of 3xTg-AD mouse brain sections from 3- and 9-month-old mice, representing early and late stages of AD neuropathology, stained with anti-Aβ (<b>A</b>,<b>B</b>), anti-TH, and anti-Cox2 antibodies (<b>C</b>,<b>D</b>). Images of the cortical areas demonstrate how 3xTg-AD mice treated with Nosustrophine (<b>B</b>) exhibit a significant reduction in Aβ plaques compared to control (untreated) mice (<b>A</b>). Treatment with Nosustrophine (<b>D</b>) also significantly attenuates the inflammatory reaction (COX-2) present in dopaminergic neurons (TH) of the midbrain of untreated transgenic animals (<b>C</b>). Scale bar: 100 μm.</p>
Full article ">Figure 4
<p>Regulation of AD-related gene expression by Nosustrophine (NST) in the hippocampus of 3- and 9-month-old wild type (WT) and AD-3xTg mice.</p>
Full article ">Figure 5
<p>Nosustrophine (NST) regulation of inflammation-related gene expression in the hippocampus of 3- and 9-month-old wild type (WT) and AD-3xTg mice.</p>
Full article ">Figure 6
<p>Nosustrophine regulation of DNA methylation, <span class="html-italic">DNMT3a</span> expression, SIRT activity, and <span class="html-italic">SIRT1</span> expression in the hippocampus of 3- and 9-month-old wild type and AD-3xTg mice.</p>
Full article ">Figure 7
<p>Effect of Nosustrophine on HDAC activity and <span class="html-italic">HDAC3</span> expression in the hippocampus of 3- and 9-month-old wild type and AD-3xTg mice.</p>
Full article ">
20 pages, 4640 KiB  
Article
In Vivo and Computational Studies on Sitagliptin’s Neuroprotective Role in Type 2 Diabetes Mellitus: Implications for Alzheimer’s Disease
by Vasudevan Mani and Minhajul Arfeen
Brain Sci. 2024, 14(12), 1191; https://doi.org/10.3390/brainsci14121191 - 26 Nov 2024
Viewed by 653
Abstract
Background/Objectives: Diabetes mellitus (DM), a widespread endocrine disorder characterized by chronic hyperglycemia, can cause nerve damage and increase the risk of neurodegenerative diseases such as Alzheimer’s disease (AD). Effective blood glucose management is essential, and sitagliptin (SITG), a dipeptidyl peptidase-4 (DPP-4) [...] Read more.
Background/Objectives: Diabetes mellitus (DM), a widespread endocrine disorder characterized by chronic hyperglycemia, can cause nerve damage and increase the risk of neurodegenerative diseases such as Alzheimer’s disease (AD). Effective blood glucose management is essential, and sitagliptin (SITG), a dipeptidyl peptidase-4 (DPP-4) inhibitor, may offer neuroprotective benefits in type 2 diabetes mellitus (T2DM). Methods: T2DM was induced in rats using nicotinamide (NICO) and streptozotocin (STZ), and biomarkers of AD and DM-linked enzymes, inflammation, oxidative stress, and apoptosis were evaluated in the brain. Computational studies supported the in vivo findings. Results: SITG significantly reduced the brain enzyme levels of acetylcholinesterase (AChE), beta-secretase-1 (BACE-1), DPP-4, and glycogen synthase kinase-3β (GSK-3β) in T2DM-induced rats. It also reduced inflammation by lowering cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and nuclear factor-κB (NF-κB). Additionally, SITG improved oxidative stress markers by reducing malondialdehyde (MDA) and enhancing glutathione (GSH). It increased anti-apoptotic B-cell lymphoma protein-2 (Bcl-2) while reducing pro-apoptotic markers such as Bcl-2-associated X (BAX) and Caspace-3. SITG also lowered blood glucose levels and improved plasma insulin levels. To explore potential molecular level mechanisms, docking was performed on AChE, COX-2, GSK-3β, BACE-1, and Caspace-3. The potential binding affinity of SITG for the above-mentioned target enzymes were 10.8, 8.0, 9.7, 7.7, and 7.9 kcal/mol, respectively, comparable to co-crystallized ligands. Further binding mode analysis of the lowest energy conformation revealed interactions with the critical residues. Conclusions: These findings highlight SITG’s neuroprotective molecular targets in T2DM-associated neurodegeneration and its potential as a therapeutic approach for AD, warranting further clinical investigations. Full article
Show Figures

Figure 1

Figure 1
<p>The timeline of the drug treatment and the experiment schedule.</p>
Full article ">Figure 2
<p>Effect of diabetes and sitagliptin on body weight in rats over a 30-day treatment period (<span class="html-italic">n</span> = 6). Data are presented as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. Day-1 in Control; ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. Day 1 in SITG10; <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. Day 1 in T2DM + SITG30.</p>
Full article ">Figure 3
<p>Effect of sitagliptin on blood glucose levels in diabetes-induced rats (<span class="html-italic">n</span> = 6). Data are presented as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. Day 1 in T2DM + SITG10; ### <span class="html-italic">p</span> &lt; 0.001 vs. Day 1 in T2DM + SITG30.</p>
Full article ">Figure 4
<p>Effect of sitagliptin on plasma insulin levels in diabetes-induced rats (<span class="html-italic">n</span> = 6). Data are presented as mean ± SEM. *** <span class="html-italic">p</span> &lt; 0.001 vs. Control; ### <span class="html-italic">p</span> &lt; 0.001 vs. T2DM.</p>
Full article ">Figure 5
<p>Effect of sitagliptin on enzyme activity in the brains of diabetes-induced rats (<span class="html-italic">n</span> = 6): (<b>A</b>) <span class="html-italic">AChE</span>, (<b>B</b>) <span class="html-italic">BACE-1</span>, (<b>C</b>) <span class="html-italic">DPP-4</span>, and (<b>D</b>) <span class="html-italic">GSK-3β</span>. Data are presented as mean ± SEM. *** <span class="html-italic">p</span> &lt; 0.001 vs. Control; ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. T2DM.</p>
Full article ">Figure 6
<p>Effect of sitagliptin on inflammatory markers in the brains of diabetes-induced rats (<span class="html-italic">n</span> = 6): (<b>A</b>) <span class="html-italic">COX-2</span>, (<b>B</b>) PGE2, (<b>C</b>) TNF-α, and (<b>D</b>) NF-κB. Data are presented as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 vs. Control; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. T2DM.</p>
Full article ">Figure 7
<p>Effect of sitagliptin on oxidative and antioxidant markers in the brains of diabetes-induced rats (<span class="html-italic">n</span> = 6): (<b>A</b>) MDA, (<b>B</b>) GSH, and (<b>C</b>) Catalase. Data are presented as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. Control; # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. T2DM; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. T2DM + SITG10.</p>
Full article ">Figure 8
<p>Effect of sitagliptin on apoptotic proteins in the brains of diabetes-induced rats (<span class="html-italic">n</span> = 6): (<b>A</b>) Bcl-2, (<b>B</b>) BAX, and (<b>C</b>) Caspace-3. Data are presented as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. Control; # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, and ### <span class="html-italic">p</span> &lt; 0.001 vs. T2DM.</p>
Full article ">Figure 9
<p>Binding mode of SITG in the active site of <span class="html-italic">AChE</span>, <span class="html-italic">COX-2</span>, <span class="html-italic">GSK-3β</span>, <span class="html-italic">BACE-1</span>, and Caspace-3.</p>
Full article ">
14 pages, 3265 KiB  
Article
Chemical, In Cellulo, and In Silico Characterization of the Aminocholine Analogs of VG
by Stavroula Kostoudi, Nikolaos Iatridis, Dimitra Hadjipavlou-Litina, Eleni Pontiki and Georgios Pampalakis
Int. J. Mol. Sci. 2024, 25(23), 12656; https://doi.org/10.3390/ijms252312656 - 25 Nov 2024
Viewed by 498
Abstract
V-type nerve agents are exceedingly toxic chemical warfare agents that irreversibly inhibit acetylcholinesterase (AChE), leading to acetylcholine accumulation in synapses and the disruption of neurotransmission. VG or O.O-diethyl S-(diethylamino)ethyl phosphorothiolate was the first compound of this class that was synthesized. The selenocholines (-Se-), [...] Read more.
V-type nerve agents are exceedingly toxic chemical warfare agents that irreversibly inhibit acetylcholinesterase (AChE), leading to acetylcholine accumulation in synapses and the disruption of neurotransmission. VG or O.O-diethyl S-(diethylamino)ethyl phosphorothiolate was the first compound of this class that was synthesized. The selenocholines (-Se-), cholines (-O-), and methylene-cholines (-CH2-) analogs of V-agents have been synthesized and their anti-AChE activities reported. Nevertheless, the aminocholine derivatives have not been pursued. Here, we have designed and synthesized a series of phosphorylated aminocholines analogs of VG that were characterized by NMR spectroscopy (H1, C13, P31, and TOCSY). Their pharmacological properties were analyzed in silico, while their toxicological properties were in vitro investigated using the SH-SY5Y cellular model. Despite the drug likeness of the new compounds, these fail to inhibit AChE in vitro and in cellulo. This may be partially explained by the fact that aminocholine is not a good leaving group compared to thiocholine. Remarkably, one of the compounds (P4) was found to even increase the activity of AChE. These compounds may serve as new nerve agent mimics that are safer alternatives for testing countermeasures. Importantly, P4 may act as a lead compound for developing a new class of alternative nerve agent pretreatments that are safer from pyridostigmine. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of VX, VG (<b>left</b>), the synthesized compounds (<b>P1</b>–<b>P4</b>) (<b>right</b>).</p>
Full article ">Figure 2
<p>TOCSY-NMR spectrum of compound <b>P1</b>.</p>
Full article ">Figure 3
<p>Best docking pose of compounds <b>P2</b> (cyan), <b>P4</b> (purple), and VG (light blue) with the 3D model of eeAChE generated based on the 4EY6 human structure deposited in the PDB. The one hydrogen bond is illustrated with dashed grey lines. The heteroatoms are colored differently: nitrogen (blue), oxygen (red), sulfur (yellow) and phosphorus (orange).</p>
Full article ">Figure 4
<p>Activity of eeAChE in the presence of compounds <b>P1</b>–<b>P4</b>. None of the compounds inhibited the activity of eeAChE. On the contrary, stimulation of eeAChE activity was observed mainly for compounds <b>P1</b> and <b>P4</b>. All compounds were tested at 100 μM.</p>
Full article ">Figure 5
<p>SHSY-5Y after 24 h incubation with inhibitors <b>P1</b>–<b>P4</b> at 50 μM (40× magnification). No alterations in the morphology of SHSY-5Y cells were observed.</p>
Full article ">Figure 6
<p>The <b>P1</b>–<b>P4</b> compounds did not exhibit cytotoxicity in SH-SY5Y cells. The results shown are from an MTT assay carried out 24 h after the incubation of cells with the <b>P1</b>–<b>P4</b> compounds at a concentration of 50 μM. The results are shown ± standard deviation.</p>
Full article ">Figure 7
<p>AchE (dark bars) and BChE (light gray bars) inhibition by compounds <b>P1</b>–<b>P4</b>. The compounds fail to inhibit AChE but, interestingly, compounds <b>P1</b> and <b>P4</b> inhibit BChE at 50 μM. Especially, compound <b>P4</b> shows approximately 60% inhibition. Furthermore, all compounds appear to stimulate the activity of AChE (negative values of inhibition). The results are shown ± standard deviation.</p>
Full article ">Figure 8
<p>Differentiation of SH-SY5Y cells. The SH-SY5Y cells are shown on day 1 (<b>a</b>) and day 7 (<b>b</b>) of differentiation with 10 μM retinoic acid. The development of neurites is easily detectable.</p>
Full article ">Figure 9
<p>Inhibition of AChE and BChE by <b>P1</b>–<b>P4</b> in differentiated SH-SY5Y cells. The results are shown ± standard deviation.</p>
Full article ">Scheme 1
<p>Chemical synthesis of the compounds (<b>P1</b>–<b>P4</b>).</p>
Full article ">
20 pages, 1506 KiB  
Article
Essential Oil Composition and Anti-Cholinesterase Properties of Cryptomeria japonica Foliage Harvested in São Miguel Island (Azores) in Two Different Seasons
by Tânia Rodrigues, Ana Lima, Tanner Wortham, Filipe Arruda, Alexandre Janeiro, José Baptista and Elisabete Lima
Plants 2024, 13(23), 3277; https://doi.org/10.3390/plants13233277 - 22 Nov 2024
Viewed by 502
Abstract
The Azorean Cryptomeria japonica forest operations and wood industry generate considerable foliage biomass residues that are used for local essential oil (EO) production. However, research on seasonal variation of C. japonica EO remains scarce. In this study, the EOs from fresh Azorean C. [...] Read more.
The Azorean Cryptomeria japonica forest operations and wood industry generate considerable foliage biomass residues that are used for local essential oil (EO) production. However, research on seasonal variation of C. japonica EO remains scarce. In this study, the EOs from fresh Azorean C. japonica foliage (Az–CJF) collected in autumn (Aut) and spring (Spr) were obtained via hydrodistillation and investigated for their physical properties, yield, chemical composition, and bioactivities. Both EOs presented a strong odor, a yellowish color, a density around 0.9 g·mL−1, and similar yields (approximately 1% v/w, dry matter). Nevertheless, the GC–MS analyses showed a decrease in monoterpene hydrocarbons (MH) and an increase in oxygenated sesquiterpenes (OS) contents in Spr–EO compared with Aut–EO (16% vs. 35% for MH and 45% vs. 31% for OS, respectively). In addition, the predominant components were kaur-16-ene (23%) for Spr–EO and phyllocladene (19%) for Aut–EO, revealing that both EOs were rich in diterpene hydrocarbons (29% vs. 26%). Concerning its toxicity against brine shrimp, a low mortality (0–38%) was observed at a concentration range of 100–180 μg·mL−1. Regarding the anti-cholinesterase properties, both EOs were inactive against acetylcholinesterase but showed anti-butyrylcholinesterase activity superior to (–)-α-pinene, a major compound of Az–CJF EO (IC50 values: 84, 148, and 648 μg·mL−1 for Spr–EO, Aut–EO, and α-pinene, respectively). Overall, the results indicate the potential benefit of both seasonal EOs in Alzheimer’s disease treatment. In conclusion, this study demonstrated that season strongly influences the Az–CJF EO quantitative composition and thus its bioactivity, aiding in the selection of the most high-quality raw materials for use in Azorean C. japonica EO aromatherapy industry. Full article
Show Figures

Figure 1

Figure 1
<p>Azorean <span class="html-italic">Cryptomeria japonica</span>: (<b>A</b>) woodland; (<b>B</b>) current distribution (shown in red) across the three island groups of the Azores archipelago: western (Flores and Corvo), central (Faial, Pico, São Jorge, Graciosa, and Terceira), and eastern (São Miguel and Santa Maria) [<a href="#B22-plants-13-03277" class="html-bibr">22</a>]; and (<b>C</b>) fresh foliage, the plant part utilized in this study, after removal of attached cones.</p>
Full article ">Figure 2
<p>Means of the air temperature and precipitation levels by month in São Miguel Island, Azores, from September 2022 to August 2023 [<a href="#B50-plants-13-03277" class="html-bibr">50</a>].</p>
Full article ">Figure 3
<p>Total ion current (TIC) chromatogram on a ZB–5MSPlus capillary column of the essential oil extracted via hydrodistillation from fresh Azorean <span class="html-italic">Cryptomeria japonica</span> foliage collected in one site in São Miguel Island (<b>A</b>) during autumn; (<b>B</b>) during spring. Legend: 1—α-Pinene; 2—Sabinene; 3—Limonene; 4—Elemol; 5—γ-Eudesmol; 6—β + α-Eudesmol; 7—Phyllocladene; 8—Kaur-16-ene.</p>
Full article ">Figure 4
<p>Toxicity of the essential oil (EO) extracted via hydrodistillation from fresh Azorean <span class="html-italic">Cryptomeria japonica</span> foliage collected in autumn (Aut) and spring (Spr) in one site in São Miguel Island. There are no statistically significant differences between samples within the used concentration range.</p>
Full article ">
19 pages, 2323 KiB  
Article
Effect of Different Extraction Techniques on Phenolic Profile and Phytochemical Potential of Gymnema inodorum Leaf Extract
by Muhammad Hassnain Haideri, Titi Phanjaroen, Wiritphon Khiaolaongam, Thanarat Boonchalaem, Jiraporn Laoung-on, Supakit Chaipoot, Surat Hongsibsong, Kongsak Boonyapranai and Sakaewan Ounjaijean
Molecules 2024, 29(22), 5475; https://doi.org/10.3390/molecules29225475 - 20 Nov 2024
Viewed by 699
Abstract
The therapeutic potential of plant extracts has attracted significant interest, especially regarding indigenous species with health-promoting properties. Gymnema inodorum, native to Northern Thailand, is recognized for its rich phytochemical profile; however, the impact of various extraction techniques on its phenolic composition and [...] Read more.
The therapeutic potential of plant extracts has attracted significant interest, especially regarding indigenous species with health-promoting properties. Gymnema inodorum, native to Northern Thailand, is recognized for its rich phytochemical profile; however, the impact of various extraction techniques on its phenolic composition and bioactivity remains underexplored. Optimizing extraction methods is essential to enhance the pharmacological efficacy of this plant’s bioactive compounds. This study investigated the influence of four extraction methods—ethanol maceration, ethanol reflux, aqueous decoction, and microwave-assisted extraction—on the bioactive profile of G. inodorum leaves, with a focus on the phenolic content and biological activities. Antioxidant activities were evaluated using DPPH, ABTS, and FRAP assays, while the total phenolic and flavonoid contents were quantified by colorimetric methods. High-Performance Liquid Chromatography (HPLC) quantified gymnemic acid and key phenolic compounds. Among the methods, ethanol reflux yielded the highest antioxidant activities (DPPH and ABTS scavenging), with a total phenolic content of 82.54 mg GAE/g and flavonoid content of 31.90 mg QE/g. HPLC analysis identified sinapic acid, myricetin, and p-hydroxybenzoic acid as major phenolics. Furthermore, the ethanol reflux extract displayed potent anti-diabetic activity, with IC50 values of 13.36 mg/mL for α-amylase and 7.39 mg/mL for α-glucosidase, as well as strong anti-inflammatory activity (IC50 of 1.6 mg/mL) and acetylcholinesterase inhibition (IC50 of 1.2 mg/mL). These findings suggest that ethanol reflux extraction is a highly effective method for producing bioactive-rich G. inodorum extracts, with substantial pharmacological potential for developing herbal remedies and nutraceuticals, particularly in enhancing therapeutic approaches for diabetes and other health-related conditions. Full article
(This article belongs to the Special Issue Natural Bioactive Compounds from Traditional Asian Plants)
Show Figures

Figure 1

Figure 1
<p>HPLC Chromatograms of phenolic standards and various <span class="html-italic">G. inodorum</span> leaf extracts. Compounds were identified based on their retention times and characteristic UV absorbance spectra compared to those of authentic standards. Peak are indicated as follows: (1) gallic acid, (2) Theobromine, (3) Protocatechuic acid, (4) p-Hydroxybenzoic acid, (5) catechin, (6) chlorogenic acid, (7) Caffeine, (8) Vanillic acid, (9) caffeic acid, (10) Syringic acid, (11) epicatechin, (12) Vanillin, (13) p-Coumaric acid, (14) ferulic acid, (15) sinapic acid, (16) rutin, (17) myricetin, (18) quercetin, and (19) Trans-cinnamic acid. Detection was performed at 280 nm.</p>
Full article ">Figure 2
<p>Antioxidant activities of <span class="html-italic">G. inodorum</span> leaf extract obtained from various extraction methods determined by different antioxidant assays: ABTS radical scavenging activity, DPPH radical scavenging activity, and Ferric-reducing antioxidant power (FRAP). The statistical significance was determined using one-way ANOVA, followed by Duncan’s multiple range test at <span class="html-italic">p</span> &lt; 0.05 and represented by lowercase letters (a, b, or c). The variables with the same letters are not statistically significant. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Pearson’s correlation coefficients (<span class="html-italic">r</span>) between the phytochemical contents, antioxidants, and anti-diabetic, anti-acetylcholinesterase, and anti-inflammatory activities. The color scale represents the strength and direction of the correlations, with blue indicating positive correlations and red indicating negative correlations. * correlation (<span class="html-italic">r</span>) is significant at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Cell viability of <span class="html-italic">G. inodorum</span> extracts in RAW 264.7 cells. Data are presented as the percentage of viable cells relative to the untreated control. The ethanolic reflux extract showed dose-dependent cytotoxicity, while others maintained cell viability above 80% at higher concentrations. Data represents the mean ± SD of three independent studies (<span class="html-italic">n</span> = 3).</p>
Full article ">
Back to TopTop