[go: up one dir, main page]

Next Issue
Volume 17, August
Previous Issue
Volume 17, June
 
 

Pharmaceuticals, Volume 17, Issue 7 (July 2024) – 158 articles

Cover Story (view full-size image): Lipid-lowering therapy (LLT) is a cornerstone of atherosclerotic cardiovascular disease prevention in women and men. However, in clinical practice, the use of potent LLT and low-density lipoprotein cholesterol goal attainment is lower in women than men. Sex differences occur in blood lipid levels, cardiovascular risk factors, and disease manifestations, which may be driven by sex-specific molecular pathways. To evaluate a potential need for sex-specific strategies for dyslipidemia management, this review describes the impact of sex on lipoprotein metabolism and lipid profiles, presents clinical trial and real-world data on LLT efficacy and safety in women, outlines female cardiovascular risk profiles, and discusses the diverse medical needs of women and men with dyslipidemia. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
17 pages, 2265 KiB  
Article
Sika Deer Velvet Antler Peptide Exerts Neuroprotective Effect in a Parkinson’s Disease Model via Regulating Oxidative Damage and Gut Microbiota
by Ying Liu, Hongyuan Li, Min Yang, Jia Guo, Zepeng Sun, Shuyue Wang, Ru Li, Xin Pang, Yumi Kim, Xiaohui Wang and Yinghua Peng
Pharmaceuticals 2024, 17(7), 972; https://doi.org/10.3390/ph17070972 - 22 Jul 2024
Viewed by 548
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder globally. Recognizing the potential of velvet antler in the nervous system, as shown in numerous studies, this research was aimed at evaluating the neuroprotective effects of Sika Deer velvet antler peptide (VAP), along [...] Read more.
Parkinson’s disease (PD) is the second most common neurodegenerative disorder globally. Recognizing the potential of velvet antler in the nervous system, as shown in numerous studies, this research was aimed at evaluating the neuroprotective effects of Sika Deer velvet antler peptide (VAP), along with the underlying mechanisms in neurotoxin-induced PD models. Initially, a peptidomic analysis of the VAP, which comprised 189 varieties of peptides, was conducted using LC-MS. Nine sequences were identified as significant using Proteome Discoverer 2.5 software. In a cellular model of PD, where PC12 cells are treated with the neurotoxin 1-methyl-4-phenylpyridinium (MPP+), the administration of the VAP reduced the cell damage and apoptosis induced by MPP+. This protective effect was associated with a decrease in oxidative stress. This protective mechanism was found to be mediated through the activation of the SIRT1-dependent Akt/Nrf2/HO-1-signaling pathway. In animal models, specifically in mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD, the administration of the VAP effectively reduced the dopaminergic neuron damage and reversed the neurobehavioral deficits. They also diminished microglia activation and apoptosis, all without any noticeable adverse effects. Additionally, the VAP was observed to beneficially alter the gut microbiota, as marked by an increase in the abundances of Prevotellaceae, Helicobacteraceae, and Prevotella. These findings suggest that VAP exerts its neuroprotective effect against neurodegeneration by inhibiting oxidative stress and modulating gut microbiota. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Peptidomics analysis of the VAP. (<b>A</b>) LC-MS TIC chromatographs of the VAP. (<b>B</b>) GO categorization of the sequences identified in the VAP. TIC, total ion current; GO, gene ontology.</p>
Full article ">Figure 2
<p>VAP protects PC12 cells from MPP<sup>+</sup> induced damage and apoptosis. (<b>A</b>) Post 24 h treatment with VAP and MPP<sup>+</sup>, PC12 cell viability was evaluated using the WST-1 assay. (<b>B</b>) LDH release was quantified in PC12 cells treated with VAP and MPP<sup>+</sup> for 24 h using an LDH assay kit. (<b>C</b>) After the 24 h treatment with VAP and MPP<sup>+</sup>, the mitochondrial membrane potential in PC12 cells was assessed using a JC-1 probe. Scale bar: 100 μm. (<b>D</b>) The expression levels of apoptosis-related proteins Bcl-2, Bax, and cleaved caspase-3 were determined in PC12 cells post-24 h treatment using Western blotting. β-actin served as the internal control. The relative expression of each protein was quantified based on normalization to GAPDH level and is shown on the right. (<b>E</b>) TUNEL staining (red) was conducted to detect the apoptosis in PC12 cells treated with VAP and MPP<sup>+</sup> for 24 h, with DAPI staining (blue) for nuclei. Scale bar: 100 μm. (<b>F</b>) Quantification of Bcl-2/Bax and cleaved caspase-3 expression. (<b>G</b>) Quantification of Δ<span class="html-italic">ψ</span>m was expressed as a ratio of JC-1 aggregate to monomer fluorescence intensity. (<b>H</b>) Quantification of TUNEL-positive cells. Data are presented as mean ± SEM (n = 3). Statistical significance is indicated as <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 versus the control group; * <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 versus the MPP<sup>+</sup>-treated group.</p>
Full article ">Figure 3
<p>VAP reduced the MPP<sup>+</sup>-induced oxidative stress in the PC12 cells. The PC12 cells were treated with the VAP and MPP<sup>+</sup> for 24 h. (<b>A</b>) The generation of ROS was measured using H<sub>2</sub>DCF-DA staining. Scale bar: 100 μm. (<b>B</b>) Quantitative analysis of the ROS production shown in (<b>A</b>). (<b>C</b>) The effect of the VAP on the MDA content was evaluated. The data are presented as the mean ± SEM (n = 3). Statistical significance is denoted as <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 versus the control group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus the MPP<sup>+</sup>-treated group.</p>
Full article ">Figure 4
<p>VAP protected PC12 cells against MPP<sup>+</sup> by activating the SIRT1-mediated Akt/Nrf2/HO-1 pathway. (<b>A</b>) PC12 cells were treated with VAP and MPP<sup>+</sup> for 24 h. SIRT1, p-Akt, Nrf2, and HO-1 were evaluated by Western blot analysis. β-actin was used as an internal control. (<b>B</b>) SIRT1 expression in PC12 cells treated with VAP and MPP<sup>+</sup> for 24 h was determined by immunocytochemistry. Scale bar: 100 μm. (<b>C</b>) Quantification of SIRT1, p-Akt, Nrf2, and HO-1 expression. (<b>D</b>) Quantification of SIRT1 fluorescence intensity. The values were presented as the mean ± SEM (n = 3). Statistical significance was indicated as follows: <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 versus the control group; * <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 versus the MPP<sup>+</sup>-treated group.</p>
Full article ">Figure 5
<p>VAP reduced the PD-related neurological damage in mice. (<b>A</b>) The timeline illustrating the construction of an MPTP-induced PD mouse model and the subsequent administration of the VAP. (<b>B</b>) Rotarod test. (<b>C</b>) Climbing pole test. (<b>D</b>) Immunohistochemical staining for TH and IBA-1 in the substantia nigra and α-synuclein (α-syn) in the striatum. The black arrows indicate α-syn-positive cells. Scale bar: 100 μm. (<b>E</b>) Protein gel blot analyses of TH, COX2, p-Akt, p-p38, p-Erk1/2, Bcl-2, Bax, and cleaved caspase3 protein in the striatum, with GAPDH used as the reference. (<b>F</b>,<b>G</b>) Quantitative analysis of TH, COX2, Bcl-2/Bax cleaved caspase3, p-Akt, p-p38, and p-Erk1/2. (<b>H</b>–<b>J</b>) Number of TH-, IBA-1-, and α-syn-positive cells determined using ImageJ. The values are presented as the mean ± SEM (n = 5). Statistical significance in all analyses is denoted as follows: <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 versus the control group; * <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 versus the MPTP-treated group.</p>
Full article ">Figure 6
<p>Hematoxylin and Eosin (H&amp;E) staining of the different organs (heart, liver, spleen, lung, and kidney) in mice treated with the VAP (30 mg/kg, n = 5). Scale bar: 100 μm.</p>
Full article ">Figure 7
<p>Hematoxylin and Eosin (H&amp;E) staining of the substantia nigra in three different treated groups of mice: the saline control, MPTP-treated group, and VAP (30 mg/kg) + MPTP-treated group. Scale bar: 100 μm.</p>
Full article ">Figure 8
<p>VAP improved the MPTP-induced gut microbiota dysbiosis in the mice. (<b>A</b>) Alpha diversity analysis of the gut microbiota showed no significant differences between the mice from the three different groups. (<b>B</b>,<b>C</b>) Barplots of the relative abundance of the three different groups at the family level (<b>B</b>) and genus level (<b>C</b>). (<b>D</b>) The relative abundances of f_<span class="html-italic">Prevotellaceae</span>, f_<span class="html-italic">Helicobacteraceae</span>, and g_<span class="html-italic">Prevotella</span> among the three different groups. * <span class="html-italic">p</span> &lt; 0.05 compared with the control group. (<b>E</b>) LEfSe analysis identified specific bacterial taxa that acted as biomarkers in each group. The cladogram highlighted the control-group-enriched taxa (red), MPTP-induced PD-group-enriched taxa (blue), and VAP-treated PD-group-enriched taxa (green). Only taxa with an LDA score greater than 2 are displayed, emphasizing the most significant differences in microbial composition between the groups.</p>
Full article ">Figure 9
<p>The possible pathway of PD protection induced by the VAP.</p>
Full article ">
22 pages, 2168 KiB  
Systematic Review
Strong Early Impact of Letrozole on Ovulation Induction Outperforms Clomiphene Citrate in Polycystic Ovary Syndrome
by Rita Zsuzsanna Vajna, András Mihály Géczi, Fanni Adél Meznerics, Nándor Ács, Péter Hegyi, Emma Zoé Feig, Péter Fehérvári, Szilvia Kiss-Dala, Szabolcs Várbíró, Judit Réka Hetthessy and Levente Sára
Pharmaceuticals 2024, 17(7), 971; https://doi.org/10.3390/ph17070971 - 22 Jul 2024
Viewed by 594
Abstract
Polycystic ovary syndrome is a common endocrine disorder, characterized by hyperandrogenism and/or chronic oligo/anovulation, which leads to infertility. The aim of this systematic review and meta-analysis was to explore the efficacy of letrozole compared with clomiphene citrate for ovulation induction in women with [...] Read more.
Polycystic ovary syndrome is a common endocrine disorder, characterized by hyperandrogenism and/or chronic oligo/anovulation, which leads to infertility. The aim of this systematic review and meta-analysis was to explore the efficacy of letrozole compared with clomiphene citrate for ovulation induction in women with polycystic ovarian syndrome. The study protocol has been registered with PROSPERO (registration number CRD42022376611). The literature search included randomized clinical trials. We conducted our systematic literature search across three medical databases: MEDLINE (via PubMed), Cochrane Library (CENTRAL), and Embase. The data synthesis employed a random effects model. Out of the 1994 articles screened, 25 studies fulfilled the inclusion criteria. The letrozole group exhibited a significant increase in endometrial thickness (mean difference = 1.70, confidence interval: 0.55–2.86; I2 = 97%, p-value = 0.008). The odds of ovulation (odds ratio = 1.8, confidence interval: 1.21–2.69; I2 = 51%, p-value = 0.010) and pregnancy (odds ratio = 1.96, confidence interval: 1.37–2.81; I2 = 32%, p-value = 0.002) were significantly higher. The resistance index of the subendometrial arteries showed a significant decrease (mean difference = −0.15, confidence interval: −0.27 to −0.04; I2 = 92%, p-value = 0.030). Women diagnosed with polycystic ovarian syndrome and treated with letrozole for ovulation induction had increased ovulation and pregnancy rates and increased endometrial thickness. The lower resistance index of subendometrial arteries can enhance intrauterine circulation, creating more favorable conditions for embryo implantation and development. Full article
(This article belongs to the Topic Research in Pharmacological Therapies)
Show Figures

Figure 1

Figure 1
<p>PRISMA [<a href="#B38-pharmaceuticals-17-00971" class="html-bibr">38</a>] flow diagram. The specifics of the search and selection process are presented in detail using a PRISMA [<a href="#B38-pharmaceuticals-17-00971" class="html-bibr">38</a>] flow diagram.</p>
Full article ">Figure 2
<p>Forest plots for the following outcomes: (<b>A</b>) endometrial thickness (ET) in all patients; (<b>B</b>) ovulation rate; (<b>C</b>) pregnancy rate in all patients. ET, ovulation rate, and pregnancy rate were significantly higher in the LE group compared to the CC group [<a href="#B1-pharmaceuticals-17-00971" class="html-bibr">1</a>,<a href="#B4-pharmaceuticals-17-00971" class="html-bibr">4</a>,<a href="#B5-pharmaceuticals-17-00971" class="html-bibr">5</a>,<a href="#B6-pharmaceuticals-17-00971" class="html-bibr">6</a>,<a href="#B15-pharmaceuticals-17-00971" class="html-bibr">15</a>,<a href="#B16-pharmaceuticals-17-00971" class="html-bibr">16</a>,<a href="#B19-pharmaceuticals-17-00971" class="html-bibr">19</a>,<a href="#B21-pharmaceuticals-17-00971" class="html-bibr">21</a>,<a href="#B22-pharmaceuticals-17-00971" class="html-bibr">22</a>,<a href="#B24-pharmaceuticals-17-00971" class="html-bibr">24</a>,<a href="#B25-pharmaceuticals-17-00971" class="html-bibr">25</a>,<a href="#B26-pharmaceuticals-17-00971" class="html-bibr">26</a>,<a href="#B27-pharmaceuticals-17-00971" class="html-bibr">27</a>,<a href="#B37-pharmaceuticals-17-00971" class="html-bibr">37</a>].</p>
Full article ">Figure 3
<p>Forest plots for subendometrial circulation: (<b>A</b>) resistance index (RI) of subendometrial arteries; (<b>B</b>) pulsatility index of subendometrial arteries. The RI of subendometrial arteries was significantly lower in the LE group compared to the CC group. The PI of subendometrial arteries was also lower in the LE group, although this difference did not reach statistical significance [<a href="#B1-pharmaceuticals-17-00971" class="html-bibr">1</a>,<a href="#B15-pharmaceuticals-17-00971" class="html-bibr">15</a>,<a href="#B19-pharmaceuticals-17-00971" class="html-bibr">19</a>].</p>
Full article ">
23 pages, 11722 KiB  
Article
A First Metabolite Analysis of Norfolk Island Pine Resin and Its Hepatoprotective Potential to Alleviate Methotrexate (MTX)-Induced Hepatic Injury
by Sherouk Hussein Sweilam, Dalia E. Ali, Ahmed M. Atwa, Ali M. Elgindy, Aya M. Mustafa, Manar M. Esmail, Mahmoud Abdelrahman Alkabbani, Mohamed Magdy Senna and Riham A. El-Shiekh
Pharmaceuticals 2024, 17(7), 970; https://doi.org/10.3390/ph17070970 - 22 Jul 2024
Cited by 1 | Viewed by 669
Abstract
Drug-induced liver injury (DILI) represents a significant clinical challenge characterized by hepatic dysfunction following exposure to diverse medications. Methotrexate (MTX) is a cornerstone in treating various cancers and autoimmune disorders. However, the clinical utility of MTX is overshadowed by its ability to induce [...] Read more.
Drug-induced liver injury (DILI) represents a significant clinical challenge characterized by hepatic dysfunction following exposure to diverse medications. Methotrexate (MTX) is a cornerstone in treating various cancers and autoimmune disorders. However, the clinical utility of MTX is overshadowed by its ability to induce hepatotoxicity. The current study aims to elucidate the hepatoprotective effect of the alcoholic extract of Egyptian Araucaria heterophylla resin (AHR) on MTX-induced liver injury in rats. AHR (100 and 200 mg/kg) significantly decreased hepatic markers (AST, ALT, and ALP), accompanied by an elevation in the antioxidant’s markers (SOD, HO-1, and NQO1). AHR extract also significantly inhibited the TGF-β/NF-κB signaling pathway as well as the downstream cascade (IL-6, JAK, STAT-3, and cyclin D). The extract significantly reduced the expression of VEGF and p38 with an elevation in the BCL2 levels, in addition to a significant decrease in the IL-1β and TNF-α levels, with a prominent effect at a high dose (200 mg/kg). Using LC-HRMS/MS analysis, a total of 43 metabolites were tentatively identified, and diterpenes were the major class. This study presents AHR as a promising hepatoprotective agent through inhibition of the TGF-β/NF-κB and JAK/STAT3 pathways, besides its antioxidant and anti-inflammatory effects. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Base peak chromatograms of negative (<b>A</b>) and positive (<b>B</b>) ionization modes of total methanolic extract of <span class="html-italic">Araucaria heterophylla</span> resin (AHR).</p>
Full article ">Figure 2
<p>Representation of the major bioactive compounds observed in the <span class="html-italic">Araucaria heterophylla</span> resin (AHR).</p>
Full article ">Figure 3
<p>Influence of AHR on hepatic injury biomarkers. (<b>A</b>) AST, (<b>B</b>) ALT, and (<b>C</b>) ALP. Results are displayed as mean +/− SD (number per group = 6 rats). a: significant vs. normal control group, b: significant vs. MTX group, c: significant vs. (SIL 100 mg + MTX) group, d: significant vs. (AHR 100 mg + MTX) group. Statistical analysis was conducted using ANOVA followed by Tukey’s post hoc test at a <span class="html-italic">p</span> value &lt; 0.05. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; ALT, alanine transaminase; AST, aspartate transaminase; ALP, alkaline phosphate.</p>
Full article ">Figure 4
<p>Influence of AHR on antioxidant markers. (<b>A</b>) SOD, (<b>B</b>) HO-1, and (<b>C</b>) NQO1. Results are displayed as the mean +/− SD (number per group = 6 rats). a: significant vs. control group, b: significant vs. MTX group, c: significant vs. (SIL 100 mg + MTX) group, d: significant vs. (AHR 100 mg + MTX) group. Statistical analysis was conducted using ANOVA followed by Tukey’s post hoc test at a <span class="html-italic">p</span> value &lt; 0.05. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; SOD, superoxide dismutases; HO-1, heme oxygenase-1; NQO1, NAD(P)H dehydrogenase [quinone] 1.</p>
Full article ">Figure 5
<p>Influence of AHR on tissue content of NF-κB, IL-1β, IL-6 and TNF-α. (<b>A</b>) NF-κB, (<b>B</b>) IL-1β, (<b>C</b>) IL-6, (<b>D</b>) TNF-α. Data are displayed as the mean +/− SD (number per group = 6 rats). a: significant vs. control group, b: significant vs. MTX group, c: significant vs. (SIL 100 mg + MTX) group, d: significant vs. (AHR 100 mg + MTX) group. Statistical analysis was conducted using ANOVA followed by Tukey’s post hoc test at a <span class="html-italic">p</span> value &lt; 0.05. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; NF-κB, nuclear factor-kappa B; IL-1β, interleukin-1β; IL-6, interleukin-6; TNF-α, tumor necrosis factor alpha.</p>
Full article ">Figure 6
<p>Influence of AHR on JAK, STAT3 and Cyclin D expression. (<b>A</b>) JAK, (<b>B</b>) STAT3, and (<b>C</b>) Cyclin D. Data are displayed as the mean +/− SD (number per group = 6 rats). a: significant vs. control group, b: significant vs. MTX group, c: significant vs. (SIL 100 mg + MTX) group, d: significant vs. (AHR 100 mg + MTX) group. Statistical analysis was conducted using ANOVA followed by Tukey’s post hoc test at a <span class="html-italic">p</span> value &lt; 0.05. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3.</p>
Full article ">Figure 7
<p>Effect of AHR on p38 and BCL2 expression. (<b>A</b>) p38, (<b>B</b>) BCL2. Data are displayed as the mean +/− SD (number per group = 6 rats). a: signficane vs. control group, b: significant vs. MTX group, c: significant vs. (SIL 100 mg + MTX) group. Statistical analysis was conducted using ANOVA followed by Tukey’s post hoc test at a <span class="html-italic">p</span> value &lt; 0.05. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; BCL2, B-cell lymphoma 2.</p>
Full article ">Figure 8
<p>Effect of AHR on MTX-induced histopathological alterations. (<b>A</b>–<b>E</b>) Photomicrographs representing staining of hepatocytes with H &amp; E (Scale bar 25 μm). (<b>A</b>) Control group, (<b>B</b>) MTX group, (<b>C</b>) SIL 100 mg + MTX-treated group, (<b>D</b>) AHR (100 mg)-treated group, and (<b>E</b>) AHR (200 mg)-treated group. Normal histological structure of hepatocytes (blue arrow), nuclear pyknosis in hepatocytes (black arrow), hepatic sinusoids engorged with blood (arrowhead), and congestion of the central vein (star). MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin.</p>
Full article ">Figure 9
<p>Effect of AHR on MTX-induced changes in TGF-β immunoreactivity. (<b>A</b>–<b>F</b>) Photomicrographs representing immunohistochemical analysis of TGF-β (Scale bar 25 μm). (<b>A</b>) Control group, (<b>B</b>) MTX group, (<b>C</b>) SIL 100 mg + MTX-treated group, (<b>D</b>) AHR (100 mg)-treated group, (<b>E</b>) AHR (200 mg)-treated group, and (<b>F</b>) % area of TGF-β immunoexpression. Data are displayed as the mean ± SD (number per group = 6 rats) using one-way ANOVA followed by Tukey’s post hoc test; <span class="html-italic">p</span> value &lt; 0.05. a vs. control group, b vs. MTX group, c vs. (SIL 100 mg + MTX) group, and d vs. (AHR 100 mg + MTX) group. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; TGF-β, transforming growth factor-beta.</p>
Full article ">Figure 10
<p>Effect of AHR on MTX-induced changes in VEGF immunoreactivity. (<b>A</b>–<b>F</b>) Photomicrographs representing immunohistochemical analysis of VEGF (scale bar: 25 μm). (<b>A</b>) Control group, (<b>B</b>) MTX group, (<b>C</b>) SIL 100 mg + MTX-treated group, (<b>D</b>) AHR (100 mg)-treated group, (<b>E</b>) AHR (200 mg)-treated group, and (<b>F</b>) % area of VEGF immunoexpression. Data are displayed as the mean ± SD (number per group = 6 rats) using one-way ANOVA followed by Tukey’s post hoc test; <span class="html-italic">p</span> value &lt; 0.05. a vs. control group, b vs. MTX group, c vs. (SIL 100 mg + MTX) group, and d vs. (AHR 100 mg + MTX) group. MTX, methotrexate; SIL, silymarin; AHR, <span class="html-italic">Araucaria heterophylla</span> resin; VEGF, vascular endothelial growth factor.</p>
Full article ">
17 pages, 7242 KiB  
Article
Exploring the Mechanism of Asiatic Acid against Atherosclerosis Based on Molecular Docking, Molecular Dynamics, and Experimental Verification
by Zhihao Wu, Luyin Yang, Rong Wang, Jie Yang, Pan Liang, Wei Ren and Hong Yu
Pharmaceuticals 2024, 17(7), 969; https://doi.org/10.3390/ph17070969 - 22 Jul 2024
Viewed by 565
Abstract
Asiatic acid (AA) is a pentacyclic triterpene derived from the traditional medicine Centella asiatica. It is known for its anti-inflammatory, antioxidant, and lipid-regulating properties. Though previous studies have suggested its potential therapeutic benefits for atherosclerosis, its pharmacological mechanism is unclear. The objective [...] Read more.
Asiatic acid (AA) is a pentacyclic triterpene derived from the traditional medicine Centella asiatica. It is known for its anti-inflammatory, antioxidant, and lipid-regulating properties. Though previous studies have suggested its potential therapeutic benefits for atherosclerosis, its pharmacological mechanism is unclear. The objective of this study was to investigate the molecular mechanism of AA in the treatment of atherosclerosis. Therefore, network pharmacology was employed to uncover the mechanism by which AA acts as an anti-atherosclerotic agent. Furthermore, molecular docking, molecular dynamics (MD) simulation, and in vitro experiments were performed to elucidate the mechanism of AA’s anti-atherosclerotic effects. Molecular docking analysis demonstrated a strong affinity between AA and PPARγ. Further MD simulations demonstrated the favorable stability of AA-PPARγ protein complexes. In vitro experiments demonstrated that AA can dose-dependently inhibit the expression of inflammatory factors induced by lipopolysaccharide (LPS) in RAW264.7 cells. This effect may be mediated through the PPARγ/NF-κB signaling pathway. This research underscores anti-inflammation as a crucial biological process in AA treatments for atherosclerosis, with PPARγ potentially serving as a key target. Full article
(This article belongs to the Special Issue The Mode of Action of Herbal Medicines and Natural Products)
Show Figures

Figure 1

Figure 1
<p>Network pharmacology analysis: (<b>A</b>) chemical structure of AA and (<b>B</b>) Venn diagram of potential targets.</p>
Full article ">Figure 2
<p>PPI network of common targets.</p>
Full article ">Figure 3
<p>Biological function enrichment analysis of AA treatment: (<b>A</b>) GO analysis of AA targets and (<b>B</b>) KEGG pathway analysis of AA targets.</p>
Full article ">Figure 4
<p>Molecular docking results of AA and core targets: (<b>A</b>) TNF-α, (<b>B</b>) IL-6, (<b>C</b>) PPARγ, (<b>D</b>) PTGS2, and (<b>E</b>) IL-1β.</p>
Full article ">Figure 5
<p>MD simulation: (<b>A</b>) RMSD curves, (<b>B</b>) RMSF curves, (<b>C</b>) Rg curves, (<b>D</b>) H-bonds plot, and (<b>E</b>) SASA plot.</p>
Full article ">Figure 6
<p>Plots of Gibbs FEL: (<b>A</b>) AA-PPARγ and (<b>B</b>) VSP-77-PPARγ.</p>
Full article ">Figure 7
<p>Plots of MM/GBSA binding energy: (<b>A</b>) AA-PPARγ and (<b>B</b>) VSP-77-PPARγ. VDWAALS, EEL, EGB, ESURF, GGAS, GSOLV, and TOTAL represent Van der Waals forces, electrostatic energy, polar solvation energy, nonpolar solvation energy, molecular mechanics terms, solvation energy terms, and average binding free energy, respectively.</p>
Full article ">Figure 8
<p>(<b>A</b>) Cell viability after treatment with AA. (<b>B</b>) Effect of AA on LPS-induced NO secretion. (<b>C</b>) Effect of AA on the morphology of cells. The results are presented as mean values ± standard error of the mean (SEM), with statistical significance indicated at a <span class="html-italic">p</span>-value below 0.05 (compared with the control group, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01; compared with LPS group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 9
<p>AA inhibited the LPS-induced generation of ROS in RAW264.7 cells: (<b>A</b>) fluorescence microscopy images and (<b>B</b>) intracellular relative ROS levels. The results are presented as mean values ± standard error of the mean (SEM), with statistical significance indicated at a <span class="html-italic">p</span>-value below 0.05 (compared with the control group, ## <span class="html-italic">p</span> &lt; 0.01; compared with LPS group, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 10
<p>Effect of AA on inflammatory factor expression: (<b>A</b>) Western blot results, (<b>B</b>) TNF-α, (<b>C</b>) IL-6, (<b>D</b>) COX-2, (<b>E</b>) iNOS, and (<b>F</b>) Arg-1. The results are presented as mean values ± standard error of the mean (SEM), with statistical significance indicated at a <span class="html-italic">p</span>-value below 0.05 (compared with the control group, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01; compared with LPS group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 11
<p>Effects of AA on NF-κB activation and PPARγ expression: (<b>A</b>) Western blot results, (<b>B</b>) NF-κB p65, p-p65 protein expression levels, and (<b>C</b>) PPARγ protein expression levels. The results are presented as mean values ± standard error of the mean (SEM), with statistical significance indicated at a <span class="html-italic">p</span>-value below 0.05 (compared with the control group, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01; compared with LPS group, * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 12
<p>AA regulates macrophage inflammation through the PPARγ/NF-κB signaling pathway: (<b>A</b>) Western blot results, (<b>B</b>) NF-κB p65, p-p65 protein expression levels, and (<b>C</b>) PPARγ protein expression levels. The results are presented as mean values ± standard error of the mean (SEM), with statistical significance indicated at a <span class="html-italic">p</span>-value below 0.05 (** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
23 pages, 5969 KiB  
Article
Parkia javanica Edible Pods Reveal Potential as an Anti-Diabetic Agent: UHPLC-QTOF-MS/MS-Based Chemical Profiling, In Silico, In Vitro, In Vivo, and Oxidative Stress Studies
by Alekhya Sarkar, Arjita Chakrabarti, Samhita Bhaumik, Bimal Debnath, Shiv Shankar Singh, Rajat Ghosh, Magdi E. A. Zaki, Sami A. Al-Hussain and Sudhan Debnath
Pharmaceuticals 2024, 17(7), 968; https://doi.org/10.3390/ph17070968 - 21 Jul 2024
Viewed by 832
Abstract
According to the World Health Organization, over 422 million people worldwide have diabetes, with the majority residing in low- and middle-income countries. Diabetes causes 1.5 million fatalities a year. The number of diabetes cases and its prevalence have progressively increased over the last [...] Read more.
According to the World Health Organization, over 422 million people worldwide have diabetes, with the majority residing in low- and middle-income countries. Diabetes causes 1.5 million fatalities a year. The number of diabetes cases and its prevalence have progressively increased over the last few decades. This study aims to determine the phytochemicals in the edible part of Perkia javanica, predict their α-glucosidase inhibitory potential, one of the promising targets for diabetes, and then carry out in vitro and in vivo studies. The phytochemicals present in the n-butanol fraction of the methanol extract of P. javanica pods were analyzed using UHPLC-QTOF-MS/MS (Ultra-High-Performance Liquid Chromatography-Quadrupole Time-of-Flight Mass Spectrometry). The UHPLC-QTOF analysis revealed the presence of 79 different compounds in the n-butanol fraction. Among these, six compounds demonstrated excellent binding affinities with α-glucosidase, surpassing the performance of two standard inhibitors, Miglitol and Voglibose. In vitro α-glucosidase inhibitory activities were assessed by the n-butanol fraction, followed by in vivo studies. According to the in vitro study, the inhibitory efficiency against α-glucosidase was determined to have an IC50 value of 261.9 µg/mL. The in vivo findings revealed a significant reduction in blood glucose levels in Swiss albino mice treated with the same extract, decreasing from 462.66 mg/dL to 228.66 mg/dL. Additionally, the extract significantly increased the activity of the enzymes catalase and superoxide dismutase (SOD) and decreased the amount of malondialdehyde (MDA) in the liver and kidney tissue. The predicted physicochemical parameters indicated that most of the compounds would be excreted from the body after inhibition in the small intestine without being absorbed. Considering the low cost and wide availability of raw materials, P. javanica pods can serve as a good food supplement that may help prevent type 2 diabetes management. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

Figure 1
<p>Post-docking 2D interactions of α-glucosidase (PDB ID: 5ZCC)-ligand (PJ_01, PJ_02, PJ_03, PJ_04, PJ_05, PJ_06, and control ligands).</p>
Full article ">Figure 2
<p>Post-docking protein–ligand 3D interactions of selected compounds (PJ_01, PJ_02, PJ_03, PJ_04, PJ_05, and PJ_06) and control ligands with their interacting distances. The hydrogen-bonding interactions were depicted in a yellow dotted line.</p>
Full article ">Figure 3
<p>In vitro inhibition of the n-butanol fraction in comparison to the standard inhibitor, Acarbose.</p>
Full article ">Figure 4
<p>Dose-dependent response curve of the n-butanol fraction and Acarbose.</p>
Full article ">Figure 5
<p>Effect of the n-butanol fraction on the blood glucose level of diabetic mice (CON—control; DB—diabetic mice; DBF—diabetic mice treated with pod extract; CONF—control treated with pod extract; and DBS—diabetes mice treated with standard drug acarbose).</p>
Full article ">Figure 6
<p>Effect of n-butanol fraction on oxidative stress in the liver and kidney of diabetic mice ((<b>A</b>): catalase activity, (<b>B</b>): superoxide dismutase, and (<b>C</b>): malondialdehyde). CON—control; CONF—control treated with plant fraction; DB—diabetic mice; DBS—diabetic mice treated with standard drug; and DBF—fraction-treated diabetic mice).</p>
Full article ">Figure 7
<p>Number of interactions exhibited by selected top six inhibitors (PJ_01, PJ_02, PJ_03, PJ_04, and PJ_06) with active site amino acid residues of α-glucosidase.</p>
Full article ">Figure 8
<p>Number of interactions exhibited by selected known α-glucosidase inhibitors Voglibose, Miglitol, and Acarbose with active site amino acid residues.</p>
Full article ">Figure 9
<p>Docked pose of compounds PJ_01, PJ_02, PJ_03, PJ_04, PJ_05, and PJ_06 (cyan color) superimposed on the docked pose of known inhibitor Acarbose (green color) in the active site of α-glucosidase.</p>
Full article ">Figure 10
<p>Docked pose of compounds PJ_01, PJ_02, PJ_03, PJ_04, PJ_05, and PJ_06 (cyan color) superimposed on the docked pose of known inhibitor Miglitol (green color) in the active site of α-glucosidase.</p>
Full article ">Figure 11
<p>Docked pose of compounds PJ_01, PJ_02, PJ_03, PJ_04, PJ_05, and PJ_06 (cyan color) superimposed on the docked pose of known inhibitor Voglibose (green color) in the active site of α-glucosidase.</p>
Full article ">Figure 12
<p>Photographic representation of the tree (<b>A</b>) and pods (<b>B</b>) of <span class="html-italic">P. javanica</span>.</p>
Full article ">
54 pages, 2198 KiB  
Review
Medicinal Plant Extracts against Cardiometabolic Risk Factors Associated with Obesity: Molecular Mechanisms and Therapeutic Targets
by Jorge Gutiérrez-Cuevas, Daniel López-Cifuentes, Ana Sandoval-Rodriguez, Jesús García-Bañuelos and Juan Armendariz-Borunda
Pharmaceuticals 2024, 17(7), 967; https://doi.org/10.3390/ph17070967 - 21 Jul 2024
Viewed by 949
Abstract
Obesity has increasingly become a worldwide epidemic, as demonstrated by epidemiological and clinical studies. Obesity may lead to the development of a broad spectrum of cardiovascular diseases (CVDs), such as coronary heart disease, hypertension, heart failure, cerebrovascular disease, atrial fibrillation, ventricular arrhythmias, and [...] Read more.
Obesity has increasingly become a worldwide epidemic, as demonstrated by epidemiological and clinical studies. Obesity may lead to the development of a broad spectrum of cardiovascular diseases (CVDs), such as coronary heart disease, hypertension, heart failure, cerebrovascular disease, atrial fibrillation, ventricular arrhythmias, and sudden cardiac death. In addition to hypertension, there are other cardiometabolic risk factors (CRFs) such as visceral adiposity, dyslipidemia, insulin resistance, diabetes, elevated levels of fibrinogen and C-reactive protein, and others, all of which increase the risk of CVD events. The mechanisms involved between obesity and CVD mainly include insulin resistance, oxidative stress, inflammation, and adipokine dysregulation, which cause maladaptive structural and functional alterations of the heart, particularly left-ventricular remodeling and diastolic dysfunction. Natural products of plants provide a diversity of nutrients and different bioactive compounds, including phenolics, flavonoids, terpenoids, carotenoids, anthocyanins, vitamins, minerals, fibers, and others, which possess a wide range of biological activities including antihypertensive, antilipidemic, antidiabetic, and other activities, thus conferring cardiometabolic benefits. In this review, we discuss the main therapeutic interventions using extracts from herbs and plants in preclinical and clinical trials with protective properties targeting CRFs. Molecular mechanisms and therapeutic targets of herb and plant extracts for the prevention and treatment of CRFs are also reviewed. Full article
(This article belongs to the Special Issue Anti-obesity and Anti-aging Natural Products)
Show Figures

Figure 1

Figure 1
<p>Overview of obesity. Obesity is associated with the development of cardiometabolic risk factors and cardiovascular diseases. However, healthy lifestyle and intake of plant extracts such as those described in this review (21 natural extracts), which have antioxidant and anti-inflammatory properties, can prevent these pathological conditions.</p>
Full article ">Figure 2
<p>Herb and plant extracts with inhibitory effects on adipogenesis, white adipose tissue accumulation, and cardiometabolic risk factors associated with obesity.</p>
Full article ">
23 pages, 6892 KiB  
Article
Enhancement of Cognitive Function by Andrographolide-Loaded Lactose β-Cyclodextrin Nanoparticles: Synthesis, Optimization, and Behavioural Assessment
by Debashish Paramanick, Kagithala Naga Rani, Vijay Kumar Singh, Parakh Basist, Rahmuddin Khan, Jameel H. Al-Tamimi, Omar M. Noman, Mansour N. Ibrahim and Abdulsalam Alhalmi
Pharmaceuticals 2024, 17(7), 966; https://doi.org/10.3390/ph17070966 - 21 Jul 2024
Viewed by 649
Abstract
This study investigates whether Andrographolide-loaded Lactose β-Cyclodextrin (ALN-βCD) nanoparticles enhance cognitive function, particularly spatial learning and memory. The successful conjugation of lactose to β-cyclodextrin was confirmed via 1H NMR spectroscopy, facilitating neuronal cell entry. The solvent evaporation method was used to create the [...] Read more.
This study investigates whether Andrographolide-loaded Lactose β-Cyclodextrin (ALN-βCD) nanoparticles enhance cognitive function, particularly spatial learning and memory. The successful conjugation of lactose to β-cyclodextrin was confirmed via 1H NMR spectroscopy, facilitating neuronal cell entry. The solvent evaporation method was used to create the nanoparticles, which were characterised for particle size, PDI, zeta potential, and drug release. The nanoparticles exhibited a size of 247.9 ± 3.2 nm, a PDI of 0.5 ± 0.02, and a zeta potential of 26.8 ± 2.5 mV. FTIR and TEM analyses, along with in vitro drug release and BBB permeability studies, confirmed their stability and efficacy. Behavioural tests, including the Elevated Plus Maze, Y-Maze, Object Recognition, and Locomotor Activity tests, demonstrated significant improvements in memory, motor coordination, and exploration time in the nanoparticle-treated groups. The group treated with ALN-βCD at a dose of 100 mg/kg/p.o. showed superior cognitive performance compared to the group receiving free andrographolides (AG). Biochemical assays indicated a significant reduction in acetylcholinesterase activity and lipid peroxidation, suggesting increased acetylcholine levels and reduced oxidative stress. Histopathological examination showed improved neuronal function without toxicity. The results showed significant improvements (p < 0.001) in memory and cognitive abilities in experimental animals, highlighting the potential of ALN-βCD nanoparticles as a non-invasive treatment for memory loss. These promising findings warrant further exploration through clinical trials. Full article
Show Figures

Figure 1

Figure 1
<p>Structure of andrographolides.</p>
Full article ">Figure 2
<p>NMR spectrum of (<b>A</b>) β-cyclodextrin and (<b>B</b>) lactose-appended β-cyclodextrin.</p>
Full article ">Figure 3
<p>Diagnostic plots of nanoparticles for particle size, entrapment efficacy (EE), and % drug release.</p>
Full article ">Figure 4
<p>Three-D response surface plots. (<b>A</b>–<b>I</b>) Comparative effects of surfactant conc., polymer conc., and sonication time on particle size (<b>A</b>–<b>C</b>), EE (<b>D</b>–<b>F</b>), and % drug release (<b>G</b>–<b>I</b>).</p>
Full article ">Figure 5
<p>In vitro drug release at pH 7.4 (<b>A</b>) and pH 5.4 (<b>B</b>).</p>
Full article ">Figure 6
<p>Percentage cell viability of prepared nanoparticles. *** <span class="html-italic">p</span> &lt; 0.001. ** <span class="html-italic">p</span> &lt; 0.1. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>In vitro permeability of prepared nanoparticles across the BBB. **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Cellular uptake of prepared nanoparticles. Free AG (<b>A</b>), ALN- βCD nanoparticles (<b>B</b>) and Lactose added β-Cyclodextrin (<b>C</b>).</p>
Full article ">Figure 9
<p>Effect of ALN-βCD nanoparticles using the Elevated Plus Maze model. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. ** <span class="html-italic">p</span> &lt; 0.1, # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 10
<p>Effect of ALN-βCD nanoparticles using the Y-maze model. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 11
<p>Effect of formulation on the latency time in the object recognition test. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 12
<p>Effect of formulation on exploration time in the object recognition test. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. ** <span class="html-italic">p</span> &lt; 0.1. # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 13
<p>Effect of formulation on % discrimination index in the object recognition test. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. vs. CP group. (Positive Control group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 14
<p>Effect of formulation on Motor coordination test. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 6), significantly different at *** <span class="html-italic">p</span> &lt; 0.001 when compared with the Control Negative (CN) group. # indicates <span class="html-italic">p</span> &lt; 0.01 when compared with the CP group. (Positive Control Group (CP), Scopolamine control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 15
<p>Effect of formulation on locomotor activity Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control Group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 16
<p>Effect of formulation on acetylcholinesterase level in animals. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control Group (CP), Scopolamine Control group (CN), Standard Group (ST), Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 17
<p>Effect of formulation on lipid peroxidation in animals. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 6), substantially different from the Control Negative (CN) group at *** <span class="html-italic">p</span> &lt; 0.001. # <span class="html-italic">p</span> &lt; 0.01 vs. CP group. (Positive Control Group (CP), Scopolamine control group (CN), Standard Group (ST) group, Free Andrographolides (T1), ALN-βCD nanoparticles (T2)).</p>
Full article ">Figure 18
<p>Histopathological analysis of mice brain (Scale bar of 50 μm).</p>
Full article ">
12 pages, 599 KiB  
Article
Close Cardiovascular Monitoring during the Early Stages of Treatment for Patients Receiving Immune Checkpoint Inhibitors
by Danielle Delombaerde, Christof Vulsteke, Nico Van de Veire, Delphine Vervloet, Veronique Moerman, Lynn Van Calster, Anne-Marie Willems, Lieselot Croes, Félix Gremonprez, Astrid De Meulenaere, Ximena Elzo Kraemer, Kristien Wouters, Marc Peeters, Hans Prenen and Johan De Sutter
Pharmaceuticals 2024, 17(7), 965; https://doi.org/10.3390/ph17070965 - 21 Jul 2024
Viewed by 535
Abstract
Background: There is an unmet medical need for the early detection of immune checkpoint inhibitor (ICI)-induced cardiovascular (CV) adverse events due to a lack of adequate biomarkers. This study aimed to provide insights on the incidence of troponin elevations and echocardiographic dynamics during [...] Read more.
Background: There is an unmet medical need for the early detection of immune checkpoint inhibitor (ICI)-induced cardiovascular (CV) adverse events due to a lack of adequate biomarkers. This study aimed to provide insights on the incidence of troponin elevations and echocardiographic dynamics during ICI treatment in cancer patients and their role as potential biomarkers for submyocardial damage. In addition, it is the first study to compare hs-TnT and hs-TnI in ICI-treated patients and to evaluate their interchangeability in the context of screening. Results: Among 59 patients, the mean patient age was 68 years, and 76% were men. Overall, 25% of patients received combination therapy. Although 10.6% [95% CI: 5.0–22.5] of the patients developed troponin elevations, none experienced a CV event. No significant changes were found in 3D left ventricular (LV) ejection fraction nor in global longitudinal strain f (56 ± 6% vs. 56 ± 6%, p = 0.903 and −17.8% [−18.5; −14.2] vs. −17.0% [−18.8; −15.1], p = 0.663) at 3 months. There were also no significant changes in diastolic function and right ventricular function. In addition, there was poor agreement between hs-TnT and hs-TnI. Methods: Here, we present a preliminary analysis of the first 59 patients included in our ongoing prospective clinical trial (NCT05699915) during the first three months of treatment. All patients underwent electrocardiography and echocardiography along with blood sampling at standardized time intervals. This study aimed to investigate the incidence of elevated hs-TnT levels within the first three months of ICI treatment. Elevations were defined as hs-TnT above the upper limit of normal (ULN) if the baseline value was normal, or 1.5 ≥ times baseline if the baseline value was above the ULN. Conclusions: Hs-TnT elevations occurred in 10.6% of the patients. However, no significant changes were found on 3D echocardiography, nor did any of the patients develop a CV event. There were also no changes found in NT-proBNP. The study is still ongoing, but these preliminary findings do not show a promising role for cardiac troponins nor for echocardiographic dynamics in the prediction of CV events during the early stages of ICI treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Cumulative incidence plot of troponin T elevation. The cumulative incidence of hs-TnT elevations was 10.6% [95% CI: 5.0; 22.5]. The dotted line represents the 95% CI.</p>
Full article ">
33 pages, 1718 KiB  
Review
Insights into Immune Exhaustion in Chronic Hepatitis B: A Review of Checkpoint Receptor Expression
by João Panão Costa, Armando de Carvalho, Artur Paiva and Olga Borges
Pharmaceuticals 2024, 17(7), 964; https://doi.org/10.3390/ph17070964 - 21 Jul 2024
Viewed by 946
Abstract
Hepatitis B, caused by the hepatitis B virus (HBV), often progresses to chronic infection, leading to severe complications, such as cirrhosis, liver failure, and hepatocellular carcinoma. Chronic HBV infection is characterized by a complex interplay between the virus and the host immune system, [...] Read more.
Hepatitis B, caused by the hepatitis B virus (HBV), often progresses to chronic infection, leading to severe complications, such as cirrhosis, liver failure, and hepatocellular carcinoma. Chronic HBV infection is characterized by a complex interplay between the virus and the host immune system, resulting in immune cell exhaustion, a phenomenon commonly observed in chronic viral infections and cancer. This state of exhaustion involves elevated levels of inhibitory molecules, cells, and cell surface receptors, as opposed to stimulatory counterparts. This review aims to elucidate the expression patterns of various co-inhibitory and co-stimulatory receptors on immune cells isolated from chronic hepatitis B (CHB) patients. By analyzing existing data, the review conducts comparisons between CHB patients and healthy adults, explores the differences between HBV-specific and total T cells in CHB patients, and examines variations between intrahepatic and peripheral immune cells in CHB patients. Understanding the mechanisms underlying immune exhaustion in CHB is crucial for developing novel immunotherapeutic approaches. This detailed analysis sheds light on the immune exhaustion observed in CHB and lays the groundwork for future combined immunotherapy strategies aimed at leveraging checkpoint receptors to restore immune function and improve clinical outcomes. Full article
(This article belongs to the Special Issue HIV and Viral Hepatitis: Prevention, Treatment and Coinfection)
Show Figures

Figure 1

Figure 1
<p>HBV replication cycle after entering the hepatocyte. Image created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Mechanism and key factors leading to immune cell exhaustion in a CHB scenario. The increased HBV viral load and antigen level overwhelm the immune system, while cytokine imbalance, increased inhibitory receptors and ligands, decreased stimulatory receptors and ligands, increased regulatory T cells, decreased T helper activity, and decreased T-bet expression all contribute to T-cell exhaustion. Image created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>Outcomes of immune cell exhaustion in CHB patients. Immune cell exhaustion in CHB patients leads to decreased T cell proliferation, survival, and differentiation, as well as impaired cytokine responses and decreased cytotoxic activity. This exhaustion results in a lack of effective T cell responses against the virus, making it challenging for the immune system to effectively clear the infection.</p>
Full article ">Figure 4
<p>Depiction of co-stimulatory and co-inhibitory ligand–receptor interactions between APCs and T cells studied in CHB patients. The regulatory molecules and their ligands are depicted, highlighting the key interactions that influence T cell function. Image created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
26 pages, 1820 KiB  
Review
Galangin: A Promising Flavonoid for the Treatment of Rheumatoid Arthritis—Mechanisms, Evidence, and Therapeutic Potential
by Ghada Khawaja, Youmna El-Orfali, Aya Shoujaa and Sonia Abou Najem
Pharmaceuticals 2024, 17(7), 963; https://doi.org/10.3390/ph17070963 - 19 Jul 2024
Viewed by 766
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by progressive joint inflammation and damage. Oxidative stress plays a critical role in the onset and progression of RA, significantly contributing to the disease’s symptoms. The complex nature of RA and the role of [...] Read more.
Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by progressive joint inflammation and damage. Oxidative stress plays a critical role in the onset and progression of RA, significantly contributing to the disease’s symptoms. The complex nature of RA and the role of oxidative stress make it particularly challenging to treat effectively. This article presents a comprehensive review of RA’s development, progression, and the emergence of novel treatments, introducing Galangin (GAL), a natural flavonoid compound sourced from various plants, as a promising candidate. The bioactive properties of GAL, including its anti-inflammatory, antioxidant, and immunomodulatory effects, are discussed in detail. The review elucidates GAL’s mechanisms of action, focusing on its interactions with key targets such as inflammatory cytokines (e.g., TNF-α, IL-6), enzymes (e.g., SOD, MMPs), and signaling pathways (e.g., NF-κB, MAPK), which impact inflammatory responses, immune cell activation, and joint damage. The review also addresses the lack of comprehensive understanding of potential treatment options for RA, particularly in relation to the role of GAL as a therapeutic candidate. It highlights the need for further research and clinical studies to ascertain the effectiveness of GAL in RA treatment and to elucidate its mechanisms of action. Overall, this review provides valuable insights into the potential of GAL as a therapeutic option for RA, shedding light on its multifaceted pharmacological properties and mechanisms of action, while suggesting avenues for future research and clinical applications. Full article
(This article belongs to the Special Issue Bioactive Substances, Oxidative Stress, and Inflammation)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Overview of DMARDs used in RA treatment: mechanisms and effects.</p>
Full article ">Figure 2
<p>Chemical structure of GAL.</p>
Full article ">Figure 3
<p>Different biological activities and numerous health benefits of GAL.</p>
Full article ">
39 pages, 7143 KiB  
Review
Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development
by Federico Appetecchia, Emanuele Fabbrizi, Francesco Fiorentino, Sara Consalvi, Mariangela Biava, Giovanna Poce and Dante Rotili
Pharmaceuticals 2024, 17(7), 962; https://doi.org/10.3390/ph17070962 - 19 Jul 2024
Viewed by 702
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current [...] Read more.
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential. Full article
(This article belongs to the Special Issue Small Molecules as Antimicrobials 2022)
Show Figures

Figure 1

Figure 1
<p>The malaria parasite life cycle. <span class="html-italic">Plasmodium</span> sporozoites are injected into the human host’s dermis during an <span class="html-italic">Anopheles</span> mosquito blood meal before making their way to the liver. Hepatic schizogony begins when a sporozoite invades a hepatocyte, and the resultant merozoites (10<sup>4</sup>) enter the bloodstream to start the symptomatic ABS characterized by the presence of 10<sup>9</sup>–10<sup>11</sup> parasites in total. A small percentage of asexual parasites engage in gametocytogenesis, producing adult male and female gametocytes (10<sup>7</sup>–10<sup>9</sup> in total) in a development process that lasts 10–12 days. Roughly 103 gametocytes are transmitted to <span class="html-italic">Anopheles</span> mosquitoes following a blood meal. The midgut of the mosquito activates gametogenesis, which is followed by fertilization to form a diploid zygote, which, during meiosis, elongates into a tetraploid ookinete within ~24 h. Ookinetes develop in six morphologically distinct stages and progress to oocysts (~48 h) by penetrating the midgut wall. Each oocyst (1–5 in total) is attached to the basal lamina of the midgut and replicates its genome for the next 6–12 days to develop hundreds of sporozoites inside the cellular membrane (sporogony). The cycle is restarted when sporozoites develop and migrate to the salivary glands of the mosquito to infect another human host. Created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>.</p>
Full article ">Figure 2
<p>An overview of the most common transmission-blocking assays showing their targets in the <span class="html-italic">Plasmodium</span> life cycle. Viability assays are employed to assess the influence of potential drugs on gametocyte development. The DGFA is used to evaluate the ability of compounds to inhibit the production of gametes. The SMFA is employed to assess the transmission-blocking potential of drug candidates. In its indirect form, the SMFA informs on the effect of small molecules on <span class="html-italic">Plasmodium</span> gametocytogenesis, whereas in its direct version, it informs on the impact on gamete development into the oocyst. The ODA (* performed in <span class="html-italic">P. berghei</span>) enables the assessment of the effects of potential drugs on the early sporogonic development of parasites in the mosquito midgut. DGFA—dual gamete formation assay; ODA—ookinete development assay; SMFA—standard membrane feeding assay. Created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>.</p>
Full article ">Figure 3
<p>Structures and transmission-blocking activities of compounds <b>1</b>–<b>7</b> currently in clinical phases.</p>
Full article ">Figure 4
<p>Structures and biological activities of epigenetic inhibitors <b>20</b>–<b>25</b> identified by Coetzee et al. as transmission-blocking antiplasmodial agents.</p>
Full article ">Figure 5
<p>Structures and transmission-blocking activities of compounds <b>12a</b>–<b>c</b> and <b>26a</b>,<b>b</b> developed from HDAC inhibitors.</p>
Full article ">Figure 6
<p>Structures and transmission-blocking activities of KDM inhibitors <b>11a</b>, <b>27</b>, <b>28a</b>,<b>b</b> (<b>A</b>), and <b>29a</b>–<b>e</b> (<b>B</b>).</p>
Full article ">Figure 7
<p>Structures and transmission-blocking activities of kinase inhibitors <b>30</b>, <b>31</b>, <b>32a</b>,<b>b</b>, <b>33a</b>–<b>d</b>, and <b>34</b>.</p>
Full article ">Figure 8
<p>(<b>A</b>) The structure and transmission-blocking activity of the YRS inhibitor ML901 (<b>35</b>). (<b>B</b>) Structures, transmission-blocking activities, and SARs of the most relevant N-4HCS <b>36a</b>,<b>b</b>.</p>
Full article ">Figure 9
<p>Development, chemical structures, transmission-blocking activities, and mode of action of iPanAms <b>37a</b>–<b>e</b>.</p>
Full article ">Figure 10
<p>(<b>A</b>) Structures and transmission-blocking activities of compounds <b>38a</b>–<b>c</b> targeting microtubule assembly. (<b>B</b>) Structures and transmission-blocking activities of compounds inhibiting plasmepsins IX and X (<b>39</b>) and <span class="html-italic">Pf</span>20S proteasome (<b>40a</b>,<b>b</b>).</p>
Full article ">Figure 11
<p>Structures and transmission-blocking activities of compounds <b>41</b>–<b>45</b> (<b>A</b>) and NBDHEX (<b>46a</b>) and its metabolite NBDHEX-COOH (<b>46b</b>) (<b>B</b>).</p>
Full article ">Figure 12
<p>(<b>A</b>) Structures, SARs, and transmission-blocking activities of compounds <b>47a</b>–<b>c</b>. (<b>B</b>) Structures and transmission-blocking activities of compounds <b>48a</b>–<b>c</b>, <b>49</b>, and <b>50</b>.</p>
Full article ">Figure 13
<p>Structures and biological transmission-blocking activities of compounds <b>51</b>–<b>55</b>.</p>
Full article ">Figure 14
<p>(<b>A</b>) Atovaquone (<b>56</b>) structure and transmission-blocking activities. (<b>B</b>) Schematic of the atovaquone-coated-surface approach developed by Paton et al. [<a href="#B145-pharmaceuticals-17-00962" class="html-bibr">145</a>]. Created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>.</p>
Full article ">
10 pages, 1425 KiB  
Review
Trackins (Trk-Targeting Drugs): A Novel Therapy for Different Diseases
by George N. Chaldakov, Luigi Aloe, Stanislav G. Yanev, Marco Fiore, Anton B. Tonchev, Manlio Vinciguerra, Nikolai T. Evtimov, Peter Ghenev and Krikor Dikranian
Pharmaceuticals 2024, 17(7), 961; https://doi.org/10.3390/ph17070961 - 19 Jul 2024
Viewed by 722
Abstract
Many routes may lead to the transition from a healthy to a diseased phenotype. However, there are not so many routes to travel in the opposite direction; that is, therapy for different diseases. The following pressing question thus remains: what are the pathogenic [...] Read more.
Many routes may lead to the transition from a healthy to a diseased phenotype. However, there are not so many routes to travel in the opposite direction; that is, therapy for different diseases. The following pressing question thus remains: what are the pathogenic routes and how can be they counteracted for therapeutic purposes? Human cells contain >500 protein kinases and nearly 200 protein phosphatases, acting on thousands of proteins, including cell growth factors. We herein discuss neurotrophins with pathogenic or metabotrophic abilities, particularly brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), pro-NGF, neurotrophin-3 (NT-3), and their receptor Trk (tyrosine receptor kinase; pronounced “track”). Indeed, we introduced the word trackins, standing for Trk-targeting drugs, that play an agonistic or antagonistic role in the function of TrkBBDNF, TrkCNT−3, TrkANGF, and TrkApro-NGF receptors. Based on our own published results, supported by those of other authors, we aim to update and enlarge our trackins concept, focusing on (1) agonistic trackins as possible drugs for (1a) neurotrophin-deficiency cardiometabolic disorders (hypertension, atherosclerosis, type 2 diabetes mellitus, metabolic syndrome, obesity, diabetic erectile dysfunction and atrial fibrillation) and (1b) neurodegenerative diseases (Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis), and (2) antagonistic trackins, particularly TrkANGF inhibitors for prostate and breast cancer, pain, and arrhythmogenic right-ventricular dysplasia. Altogether, the druggability of TrkANGF, TrkApro-NGF, TrkBBDNF, and TrkCNT−3 receptors via trackins requires a further translational pursuit. This could provide rewards for our patients. Full article
(This article belongs to the Special Issue Synthetic Inhibitors of Nucleoside Monophosphate-Kinases)
Show Figures

Figure 1

Figure 1
<p>Neurotrophins and their Trk receptors. Redrawn from [<a href="#B11-pharmaceuticals-17-00961" class="html-bibr">11</a>].</p>
Full article ">Figure 2
<p>Metabotrophic factors (MTF) and their Trk receptors on the crossroads of the pathogenesis of and therapy for cardiometabolic diseases (CMD) and neurometabolic diseases (NMD), particularly Alzheimer’s disease (AD). Credit Nikifor N. Chaldakov.</p>
Full article ">Figure 3
<p>Structure of the soul of plants, animals, and humans. In this vision, humans are unique in having all three types of souls symbiotically. Here, it is reasonable to quote Socrates—“Man is a soul that serves his body”—as a first conceptual step to envisage the soul-and-body interaction [<a href="#B92-pharmaceuticals-17-00961" class="html-bibr">92</a>].</p>
Full article ">
24 pages, 2676 KiB  
Review
Unlocking the Green Gold: Exploring the Cancer Treatment and the Other Therapeutic Potential of Fucoxanthin Derivatives from Microalgae
by Fatouma Mohamed Abdoul-Latif, Ayoub Ainane, Ibrahim Houmed Aboubaker, Ali Merito Ali, Houda Mohamed, Pannaga Pavan Jutur and Tarik Ainane
Pharmaceuticals 2024, 17(7), 960; https://doi.org/10.3390/ph17070960 - 18 Jul 2024
Cited by 1 | Viewed by 1015
Abstract
Fucoxanthin, a carotenoid widely studied in marine microalgae, is at the heart of scientific research because of its promising bioactive properties for human health. Its unique chemical structure and specific biosynthesis, characterized by complex enzymatic conversion in marine organisms, have been examined in [...] Read more.
Fucoxanthin, a carotenoid widely studied in marine microalgae, is at the heart of scientific research because of its promising bioactive properties for human health. Its unique chemical structure and specific biosynthesis, characterized by complex enzymatic conversion in marine organisms, have been examined in depth in this review. The antioxidant, anti-inflammatory, and anti-cancer activities of fucoxanthin have been rigorously supported by data from in vitro and in vivo experiments and early clinical trials. Additionally, this review explores emerging strategies to optimize the stability and efficacy of fucoxanthin, aiming to increase its solubility and bioavailability to enhance its therapeutic applications. However, despite these potential benefits, challenges persist, such as limited bioavailability and technological obstacles hindering its large-scale production. The medical exploitation of fucoxanthin thus requires an innovative approach and continuous optimization to overcome these barriers. Although further research is needed to refine its clinical use, fucoxanthin offers promising potential in the development of natural therapies aimed at improving human health. By integrating knowledge about its biosynthesis, mechanisms of action, and potential beneficial effects, future studies could open new perspectives in the treatment of cancer and other chronic diseases. Full article
(This article belongs to the Special Issue Network Pharmacology of Natural Products)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Fucoxanthin, Dinoxanthin, Peridinin, and Neoxanthin structures.</p>
Full article ">Figure 1 Cont.
<p>Fucoxanthin, Dinoxanthin, Peridinin, and Neoxanthin structures.</p>
Full article ">Figure 2
<p>Key characterized groups and bands of Fucoxanthin.</p>
Full article ">Figure 3
<p>The chemical structures of the two fucoxanthin derivatives: fucoxanthinol and amarouciaxanthin A.</p>
Full article ">Figure 4
<p>Model of the structure of fucoxanthin-chlorophyll proteins (FCP) in thylakoid membranes.</p>
Full article ">Figure 5
<p>Main steps of the fucoxanthin biosynthesis pathway in the majority of microalgae species.</p>
Full article ">Figure 5 Cont.
<p>Main steps of the fucoxanthin biosynthesis pathway in the majority of microalgae species.</p>
Full article ">Figure 6
<p>Recent antioxidant activities of fucoxanthin: in vitro and in vivo research. (created with <a href="http://www.map-this.com" target="_blank">www.map-this.com</a> accessed on 21 May 2024).</p>
Full article ">Figure 7
<p>Anti-inflammatory activities of fucoxanthin. (created with <a href="http://www.map-this.com" target="_blank">www.map-this.com</a> accessed on 22 February 2024).</p>
Full article ">
3 pages, 186 KiB  
Editorial
Vascular Endothelial Growth Factor (VEGF) and VEGF Receptor Inhibitors in Health and Disease
by Sylvain Broussy
Pharmaceuticals 2024, 17(7), 959; https://doi.org/10.3390/ph17070959 - 18 Jul 2024
Viewed by 383
Abstract
In this Special Issue of Pharmaceuticals, we present four reviews and seven original articles addressing recent aspects of research on Vascular Endothelial Growth Factors (VEGFs) and their receptors, from clinical practice to fundamental studies in new drug development [...] Full article
21 pages, 6336 KiB  
Article
Anti-Oxidative and Anti-Apoptotic Oligosaccharides from Pichia pastoris-Fermented Cress Polysaccharides Ameliorate Chromium-Induced Liver Toxicity
by Imdad Ullah Khan, Aqsa Aqsa, Yusra Jamil, Naveed Khan, Amjad Iqbal, Sajid Ali, Muhammad Hamayun, Abdulwahed Fahad Alrefaei, Turki Kh. Faraj, Bokyung Lee and Ayaz Ahmad
Pharmaceuticals 2024, 17(7), 958; https://doi.org/10.3390/ph17070958 - 18 Jul 2024
Viewed by 499
Abstract
Oxidative stress impairs the structure and function of the cell, leading to serious chronic diseases. Antioxidant-based therapeutic and nutritional interventions are usually employed for combating oxidative stress-related disorders, including apoptosis. Here, we investigated the hepatoprotective effect of oligosaccharides, produced through Pichia pastoris-mediated [...] Read more.
Oxidative stress impairs the structure and function of the cell, leading to serious chronic diseases. Antioxidant-based therapeutic and nutritional interventions are usually employed for combating oxidative stress-related disorders, including apoptosis. Here, we investigated the hepatoprotective effect of oligosaccharides, produced through Pichia pastoris-mediated fermentation of water-soluble polysaccharides isolated from Lepidium sativum (cress) seed mucilage, on chromium(VI)-induced oxidative stress and apoptosis in mice. Gel permeation chromatography (GPC), using Bio-Gel P-10 column, of the oligosaccharides product of fermentation revealed that P. pastoris effectively fermented polysaccharides as no long chain polysaccharides were observed. At 200 µg/mL, fractions DF73, DF53, DF72, and DF62 exhibited DPPH radical scavenging activity of 92.22 ± 2.69%, 90.35 ± 0.43%, 88.83 ± 3.36%, and 88.83 ± 3.36%, respectively. The antioxidant potential of the fermentation product was further confirmed through in vitro H2O2 radical scavenging assay. Among the screened samples, the highest H2O2 radical scavenging activity was displayed by DF73, which stabilized the free radicals by 88.83 ± 0.38%, followed by DF53 (86.48 ± 0.83%), DF62 (85.21 ± 6.66%), DF72 (79.9 4± 1.21%), and EPP (77.76 ± 0.53%). The oligosaccharide treatment significantly alleviated chromium-induced liver damage, as evident from the increase in weight gain, improved liver functions, and reduced histopathological alterations in the albino mice. A distinctly increased level of lipid peroxide (LPO) free radicals along with the endogenous hepatic enzymes were evident in chromium induced hepatotoxicity in mice. However, oligosaccharides treatment mitigated these effects by reducing the LPO production and increasing ALT, ALP, and AST levels, probably due to relieving the oxidative stress. DNA fragmentation assays illustrated that Cr(VI) exposure induced massive apoptosis in liver by damaging the DNA which was then remediated by oligosaccharides supplementation. Histopathological observations confirmed that the oligosaccharide treatment reverses the architectural changes in liver induced by chromium. These results suggest that oligosaccharides obtained from cress seed mucilage polysaccharides through P. pastoris fermentation ameliorate the oxidative stress and apoptosis and act as hepatoprotective agent against chromium-induced liver injury. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Growth phases of <span class="html-italic">P. pastoris</span> using cress seed mucilage polysaccharides as carbon source, where <span class="html-italic">Y</span>-axis shows <span class="html-italic">P. pastoris</span> count in logCFU/mL, <span class="html-italic">X</span>-axis shows time, and <span class="html-italic">Z</span>-axis shows temprature of the fermentation media.</p>
Full article ">Figure 2
<p>Total carbohydrate contents quantified in different oligosaccharide fractions were obtained through microbial bioprocessing of cress seed mucilage polysaccharides.</p>
Full article ">Figure 3
<p>Biochemical composition of different oligosaccharide fractions obtained through microbial bioprocessing of cress seed mucilage polysaccharides.</p>
Full article ">Figure 4
<p>FT-IR spectroscopic analysis of Df53, DF62, DF72 and DF73 fractions.</p>
Full article ">Figure 5
<p>Percent radical scavenging of the DPPH free radical at different concentrations, where EPP; ethanol precipitated polysaccharides, DF53–DF73; oligosaccharides fractions and AA: ascorbic acid as reference compound. Means denoted by different letters (a, b, c…) are significant from each other at <span class="html-italic">p</span> value of 0.05.</p>
Full article ">Figure 6
<p>Percent radical scavenging of the H<sub>2</sub>O<sub>2</sub> free radical at different concentrations, where EPP; ethanol precipitated polysaccharides, DF53–DF73; oligosaccharide fractions and Gallic acid: reference compound. Means denoted by different letters (a, b, c…) are significant from each other at <span class="html-italic">p</span> value of 0.05.</p>
Full article ">Figure 7
<p>Initial and final body weight along with absolute and relative liver weight of the mice exposed to 100 mg/kg of Cr(IV) and co-treated with 200 mg/kg of oligosaccharides fractions. Means denoted by different letters (a, b, c…) are significant from each other at <span class="html-italic">p</span> value of 0.05.</p>
Full article ">Figure 8
<p>Effect of oligosaccharides on liver ALT, MDA, ALP and AST level of chromium exposed mice. Where (<b>A</b>) is ALT level in U/L, (<b>B</b>) is MDA level in µM, (<b>C</b>) is ALP level in units/100 mL and (<b>D</b>) is AST level in U/L. Means denoted by different letters (a, b, c…) are significant from each other at P value of 0.05.</p>
Full article ">Figure 9
<p>Oligosaccharides protect the DNA damage in mice liver induced by Cr (VI) exposure, where M; DNA marker (1 Kb), C: Normal mice, Cr: mice received 100 mg/kg of Cr (VI) alone, 1–4: mice received 100 mg/kg of Cr (VI) along with 200 mg/kg of DF53-73 oligosaccharides and 5; mice received Cr (VI) along with ascorbic acid.</p>
Full article ">Figure 10
<p>Histology of the mice liver stained with H&amp;E stain, where (<b>A</b>); received 100 mg/kg of Cr(VI) only, (<b>B</b>,<b>C</b>); normal control, (<b>D</b>); received 100 mg/kg of Cr(VI) + 200 mg/kg of DF53, (<b>E</b>); received 100 mg/kg of Cr(VI) + 200 mg/kg of DF62, (<b>F</b>); received 100 mg/kg of Cr(VI) + 200 mg/kg of DF72, (<b>G</b>); received 100 mg/kg of Cr(VI) + 200 mg/kg of DF73 and (<b>H</b>); received 100 mg/kg of Cr(VI) + 100 mg/kg of ascorbic acid.</p>
Full article ">
23 pages, 1610 KiB  
Project Report
Introducing the NUATEI Consortium: A Mexican Research Program for the Identification of Natural and Synthetic Antimicrobial Compounds for Prevalent Infectious Diseases
by Julio César Carrero, Bertha Espinoza, Leonor Huerta, Mayra Silva-Miranda, Silvia-Laura Guzmán-Gutierrez, Alejandro Dorazco-González, Ricardo Reyes-Chilpa, Clara Espitia and Sergio Sánchez
Pharmaceuticals 2024, 17(7), 957; https://doi.org/10.3390/ph17070957 - 18 Jul 2024
Viewed by 697
Abstract
The need for new drugs to treat human infections is a global health concern. Diseases like tuberculosis, trypanosomiasis, amoebiasis, and AIDS remain significant problems, especially in developing countries like Mexico. Despite existing treatments, issues such as resistance and adverse effects drive the search [...] Read more.
The need for new drugs to treat human infections is a global health concern. Diseases like tuberculosis, trypanosomiasis, amoebiasis, and AIDS remain significant problems, especially in developing countries like Mexico. Despite existing treatments, issues such as resistance and adverse effects drive the search for new alternatives. Herein, we introduce the NUATEI research consortium, made up of experts from the Institute of Biomedical Research at UNAM, who identify and obtain natural and synthetic compounds and test their effects against human pathogens using in vitro and in vivo models. The consortium has evaluated hundreds of natural extracts and compounds against the pathogens causing tuberculosis, trypanosomiasis, amoebiasis, and AIDS, rendering promising results, including a patent with potential for preclinical studies. This paper presents the rationale behind the formation of this consortium, as well as its objectives and strategies, emphasizing the importance of natural and synthetic products as sources of antimicrobial compounds and the relevance of the diseases studied. Finally, we briefly describe the methods of the evaluation of the compounds in each biological model and the main achievements. The potential of the consortium to screen numerous compounds and identify new therapeutic agents is highlighted, demonstrating its significant contribution to addressing these infectious diseases. Full article
(This article belongs to the Special Issue Recent Advancements in the Development of Antiprotozoal Agents)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>General scheme for the evaluation of plant-derived products against infectious microorganisms in the NUATEI consortium. Image was made with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Workflow of the NUATEI consortium for the identification of new antimicrobial compounds. Imagen was made with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
18 pages, 2438 KiB  
Article
Novel Coumarin–Nucleobase Hybrids with Potential Anticancer Activity: Synthesis, In Vitro Cell-Based Evaluation, and Molecular Docking
by Maiara Correa de Moraes, Rafaele Frassini, Mariana Roesch-Ely, Favero Reisdorfer de Paula and Thiago Barcellos
Pharmaceuticals 2024, 17(7), 956; https://doi.org/10.3390/ph17070956 - 17 Jul 2024
Viewed by 451
Abstract
A new series of compounds planned by molecular hybridization of the nucleobases uracil and thymine, or the xanthine theobromine, with coumarins, and linked through 1,2,3-triazole heterocycles were evaluated for their in vitro anticancer activity against the human tumor cell lines: colon carcinoma (HCT116), [...] Read more.
A new series of compounds planned by molecular hybridization of the nucleobases uracil and thymine, or the xanthine theobromine, with coumarins, and linked through 1,2,3-triazole heterocycles were evaluated for their in vitro anticancer activity against the human tumor cell lines: colon carcinoma (HCT116), laryngeal tumor cells (Hep-2), and lung carcinoma cells (A549). The hybrid compound 9a exhibited better activity in the series, showing an IC50 of 24.19 ± 1.39 μM against the HCT116 cells, with a selectivity index (SI) of 6, when compared to the cytotoxicity against the non-tumor cell line HaCat. The in silico search for pharmacological targets was achieved through molecular docking studies on all active compounds, which suggested that the synthesized compounds possess a high affinity to the Topoisomerase 1–DNA complex, supporting their antitumor activity. The in silico toxicity prediction studies suggest that the compounds present a low risk of causing theoretical mutagenic and tumorigenic effects. These findings indicate that molecular hybridization from natural derivative molecules is an interesting approach to seek new antitumor candidates. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Representative examples of biologically active coumarin (<b>A</b>) and uracil-containing compounds (<b>B</b>) hybridized through the 1,2,3-triazole heterocycle.</p>
Full article ">Figure 2
<p>Synthesis of 4-(azidomethyl)coumarins <b>4a</b> and <b>4b</b>.</p>
Full article ">Figure 3
<p>Synthetic methodologies employed to prepare the <span class="html-italic">N1</span>-propargylated uracil <b>6a</b>, the <span class="html-italic">N1</span>-propargylated thymine <b>6b</b>, the <span class="html-italic">N1</span>,<span class="html-italic">N2</span>-dipropargylated uracil <b>6c</b>, and the propargylated theobromine <b>6d</b>.</p>
Full article ">Figure 4
<p>A Cu(I)-catalyzed [3+2]-cycloaddition reaction between the 4-(azidomethyl)coumarins <b>4a,b</b>, and the <span class="html-italic">N1</span>-propargylated uracil <b>6a</b>, <span class="html-italic">N1</span>-propargylated thymine <b>6b</b> (Scheme A), <span class="html-italic">N1</span>,<span class="html-italic">N2</span>-dipropargylated uracil <b>6c</b> (Scheme B), and the propargylated theobromine <b>6d</b> (Scheme C), leading to the corresponding target hybrid molecules.</p>
Full article ">Figure 5
<p>Predicted binding of most active in vitro compounds <b>9a,b</b> and <b>10a,b</b> poses (<b>A</b>) and <b>9a</b> pose (<b>B</b>) in active site of Topo 1-DNA complex (PDB code: 1K4S). Graphic visualization obtained using UCSF Chimera (v.1.10.1) [<a href="#B51-pharmaceuticals-17-00956" class="html-bibr">51</a>].</p>
Full article ">
14 pages, 681 KiB  
Article
Coffee with Cordyceps militaris and Hericium erinaceus Fruiting Bodies as a Source of Essential Bioactive Substances
by Katarzyna Kała, Małgorzata Cicha-Jeleń, Kamil Hnatyk, Agata Krakowska, Katarzyna Sułkowska-Ziaja, Agnieszka Szewczyk, Jan Lazur and Bożena Muszyńska
Pharmaceuticals 2024, 17(7), 955; https://doi.org/10.3390/ph17070955 - 17 Jul 2024
Viewed by 846
Abstract
Drinking coffee is a daily routine for many people. Supplement manufacturers have proposed adding powdered Cordyceps militaris, known for its ergogenic and immunostimulating properties, and Hericium erinaceus, known for its nerve growth factor (NGF)-stimulating properties, to coffee. The aim of this [...] Read more.
Drinking coffee is a daily routine for many people. Supplement manufacturers have proposed adding powdered Cordyceps militaris, known for its ergogenic and immunostimulating properties, and Hericium erinaceus, known for its nerve growth factor (NGF)-stimulating properties, to coffee. The aim of this work was to compare the bioactive substances in three types of coffee: machine-brewed, instant, and traditionally brewed, prepared with the addition of H. erinaceus and C. militaris fruiting bodies. The analysis of bioactive substances was performed using AAS and RP-HPLC methods. Among the control samples of coffee, traditionally brewed coffee was the best source of bioelements. Considering the mushroom species tested, the best additional source of Mg, Zn, Cu, Na, K, and Ca was C. militaris. A slightly higher Fe content was determined for H. erinaceus. With the addition of C. militaris, the amounts of 4-feruloylquinic acid (18.6 mg/200 mL) and 3,5-di-caffeoylquinic acid (3.76 mg/200 mL) also increased. In conclusion, the C. militaris species has been proven to be a better source of bioactive substances as a coffee additive in the daily diet. The combination of brewed coffee and the tested mushrooms seems to be the most beneficial in terms of health-promoting effects. Full article
(This article belongs to the Special Issue Natural Products Derived from Fungi and Their Biological Activities)
Show Figures

Figure 1

Figure 1
<p>Example chromatogram of neochlorogenic acid (<b>1</b>), chlorogenic acid (<b>2</b>), caffeine (<b>3</b>), 4-feruloylquinic acid (<b>4</b>), isochlorogenic acid (<b>5</b>), and 3,5-di-caffeoylquinic acid (<b>6</b>) in the brewed coffee with <span class="html-italic">Cordyceps militaris</span>.</p>
Full article ">Figure 2
<p>Structural formulas of bioactive substances characteristic of coffee (caffeine, chlorogenic acid, neochlorogenic acid) and mushroom material (lovastatin, ergosterol).</p>
Full article ">
1 pages, 144 KiB  
Correction
Correction: Singh et al. Nanotechnology-Aided Advancement in Combating the Cancer Metastasis. Pharmaceuticals 2023, 16, 899
by Arun Kumar Singh, Rishabha Malviya, Bhupendra Prajapati, Sudarshan Singh, Deepika Yadav and Arvind Kumar
Pharmaceuticals 2024, 17(7), 954; https://doi.org/10.3390/ph17070954 - 17 Jul 2024
Viewed by 325
Abstract
In the original publication [...] Full article
32 pages, 1631 KiB  
Review
Dilemmas in Elderly Diabetes and Clinical Practice Involving Traditional Chinese Medicine
by Chongxiang Xue, Ying Chen, Yuntian Bi, Xiaofei Yang, Keyu Chen, Cheng Tang, Xiaolin Tong, Linhua Zhao and Han Wang
Pharmaceuticals 2024, 17(7), 953; https://doi.org/10.3390/ph17070953 - 16 Jul 2024
Viewed by 662
Abstract
Diabetes is a widespread chronic disease that occurs mainly in the elderly population. Due to the difference in pathophysiology between elderly and young patients, the current clinical practice to treat elderly patients with anti-diabetes medications still faces some challenges and dilemmas, such as [...] Read more.
Diabetes is a widespread chronic disease that occurs mainly in the elderly population. Due to the difference in pathophysiology between elderly and young patients, the current clinical practice to treat elderly patients with anti-diabetes medications still faces some challenges and dilemmas, such as the urgent need for early diagnosis and prevention, and an imbalance between restricted dietary intake and the risk of undernutrition. Traditional Chinese medicine (TCM) offers various treatment regimens that are actively utilized in the field of diabetes management. Through multiple targets and multiple pathways, TCM formulas, medicinal herbs, and active natural products enhance the efficacy of diabetes prevention and diabetes control measures, simplify complex medication management, and improve common symptoms and common diabetic complications in elderly people. Historically, natural products have played a key role in material composition analysis of TCM and mechanism interpretation to enable drug discovery. However, there have been few conclusions on this topic. This review summarizes the development of TCM for the prevention and management of diabetes in elderly people, existing evidence-based clinical practices, and prospects for future development. Full article
(This article belongs to the Special Issue Therapeutic Effects of Natural Products and Their Clinical Research)
Show Figures

Figure 1

Figure 1
<p>Dilemma in elderly diabetes and the role of TCM and natural products in elderly diabetes. The person used to represent elderly diabetes in this figure is Fu Du, a prominent Chinese poet in the Tang dynasty, who is said to have suffered from diabetes near the end of his life. Yin (black background) and Yang (white background) are two halves, representing the role of traditional Chinese medicine in maintaining body balance. The dilemma mentioned and the related solution are connected by a balance board.</p>
Full article ">Figure 2
<p>Example of TCM and highly potent chemical compounds used for elderly diabetes and their chemical structure.</p>
Full article ">
21 pages, 5275 KiB  
Article
Computational Design of Novel Tau-Tubulin Kinase 1 Inhibitors for Neurodegenerative Diseases
by Shahzaib Ahamad, Iqbal Taliy Junaid and Dinesh Gupta
Pharmaceuticals 2024, 17(7), 952; https://doi.org/10.3390/ph17070952 - 16 Jul 2024
Viewed by 603
Abstract
The tau-tubulin kinase 1 (TTBK1) protein is a casein kinase 1 superfamily member located at chromosome 6p21.1. It is expressed explicitly in the brain, particularly in the cytoplasm of cortical and hippocampal neurons. TTBK1 has been implicated in the phosphorylation and aggregation of [...] Read more.
The tau-tubulin kinase 1 (TTBK1) protein is a casein kinase 1 superfamily member located at chromosome 6p21.1. It is expressed explicitly in the brain, particularly in the cytoplasm of cortical and hippocampal neurons. TTBK1 has been implicated in the phosphorylation and aggregation of tau in Alzheimer’s disease (AD). Considering its significance in AD, TTBK1 has emerged as a promising target for AD treatment. In the present study, we identified novel TTBK1 inhibitors using various computational techniques. We performed a virtual screening-based docking study followed by E-pharmacophore modeling, cavity-based pharmacophore, and ligand design techniques and found ZINC000095101333, LD7, LD55, and LD75 to be potential novel TTBK1 lead inhibitors. The docking results were complemented by Molecular Mechanics/Generalized Born Surface Area (MMGBSA) calculations. The molecular dynamics (MD) simulation studies at a 500 ns scale were carried out to monitor the behavior of the protein toward the identified ligands. Pharmacological and ADME/T studies were carried out to check the drug-likeness of the compounds. In summary, we identified a new series of compounds that could effectively bind the TTBK1 receptor. The newly designed compounds are promising candidates for developing therapeutics targeting TTBK1 for AD. Full article
(This article belongs to the Special Issue Structural and Other Proteomics Approaches in Drug Discovery)
Show Figures

Figure 1

Figure 1
<p>E-pharmacophore models for co-crystal ligands 4BTK-DTQ, 4BTM-F8E, 4NFN-2KC, JXX-VP7, and 7JXY-VSY and their features (<b>A</b>–<b>E</b>). The graphs were generated using the Schrödinger molecular modeling software version 12.8.117 [<a href="#B21-pharmaceuticals-17-00952" class="html-bibr">21</a>].</p>
Full article ">Figure 2
<p>Two-dimensional interaction plots of the co-crystal ligand 2KC and top three compounds shortlisted viz. ZINC000095101333, ZINC000009936617, and ZINC001209984530 (<b>A</b>–<b>D</b>, respectively).</p>
Full article ">Figure 3
<p>The TTBK1 active site (blue circular region). The ligand design determines the location of H-bond donors and acceptors (<b>A</b>), and after the generation of the newly designed molecules, the blue color is less apparent (<b>B</b>). The plots were generated using the Ligand Designer module of Schrödinger software [<a href="#B21-pharmaceuticals-17-00952" class="html-bibr">21</a>].</p>
Full article ">Figure 4
<p>Docking (2D) plots of the top five newly designed compounds LD7, LD10, LD51, LD55, and LD75 (<b>A</b>–<b>E</b>, respectively). The H-bonds are represented in the pink arrow.</p>
Full article ">Figure 5
<p>Plots depicting the RMSD (<b>A</b>), RMSF (<b>B</b>), Rg (<b>C</b>), and SASA (<b>D</b>) of the native TTBK1 protein and complexes. The results of the MD simulation were visualized using the GROMACS 5.18.3 software package [<a href="#B22-pharmaceuticals-17-00952" class="html-bibr">22</a>].</p>
Full article ">Figure 6
<p>Plots of hydrogen bonds between TTBK1 protein residues and selected ligands over a 500 ns MD simulation were analyzed using GROMACS, providing detailed data for each frame.</p>
Full article ">Figure 7
<p>The Gibbs energy landscape (FEL) plot was estimated during a 500 ns MD simulation. The graphs were generated with the help of GROMACS software [<a href="#B22-pharmaceuticals-17-00952" class="html-bibr">22</a>].</p>
Full article ">Figure 8
<p>HOMO, LUMO orbitals, and the electrostatic potential map of shortlisted compounds, namely, 2KC, ZINC000095101333, LD7, LD55, and LD75 (<b>A</b>–<b>E</b>, respectively). The calculations performed in the gas phase were executed using the Spartan20 package with the B3LYP/6-31G* method, and the graphs were generated with the assistance of Spartan20 (Version 20.1.2) software [<a href="#B27-pharmaceuticals-17-00952" class="html-bibr">27</a>].</p>
Full article ">Figure 9
<p>The workflow of this study utilized ZINC15’s 6.6 million active compounds and involved virtual screening. The top-scoring molecules were further utilized for pharmacophore analysis, MD simulations, and DFT calculations.</p>
Full article ">Chart 1
<p>Chemical structure of the inhibitors reported in the literature [<a href="#B13-pharmaceuticals-17-00952" class="html-bibr">13</a>,<a href="#B15-pharmaceuticals-17-00952" class="html-bibr">15</a>,<a href="#B16-pharmaceuticals-17-00952" class="html-bibr">16</a>].</p>
Full article ">
13 pages, 4416 KiB  
Article
Cancer Cell Secreted Legumain Promotes Gastric Cancer Resistance to Anti-PD-1 Immunotherapy by Enhancing Macrophage M2 Polarization
by Xu Pei, Shi-Long Zhang, Bai-Quan Qiu, Peng-Fei Zhang, Tian-Shu Liu and Yan Wang
Pharmaceuticals 2024, 17(7), 951; https://doi.org/10.3390/ph17070951 - 16 Jul 2024
Viewed by 525
Abstract
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in [...] Read more.
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in modulating the tumor immune microenvironment and the biological effect on the immune evasion of gastric cancer remains unclear. In this study, we found that forced expression of sLGMN in gastric cancer serum correlates with increased M2 macrophage infiltration in GC tissues and predicted resistance to anti-PD-1 immunotherapy. Mechanistically, gastric cancer cells secrete LGMN via binding to cell surface Integrin αvβ3, then activate Integrin αvβ3/PI3K (Phosphatidylinositol-4,5-bisphosphate3-kinase)/AKT (serine/threonine kinase)/mTORC2 (mammalian target of rapamycin complex 2) signaling, promote metabolic reprogramming, and polarize macrophages from the M1 to the M2 phenotype. Either blocking LGMN, Integrin αv, or knocking out Integrin αv expression and abolishing the LGMN/Integrin αvβ3 interaction significantly inhibits metabolic reprogramming and polarizes macrophages from the M1 to the M2 phenotype. This study reveals a critical molecular crosstalk between gastric cancer cells and macrophages through the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis in promoting gastric cancer immune evasion and resistance to anti-PD-1 immunotherapy, indicating that the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis may act as a promising therapeutic target. Full article
(This article belongs to the Special Issue Small Molecules in Cancer Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Relationship between LGMN expression levels and M2 macrophage infiltration in gastric cancer. (<b>A</b>) Analysis of TCGA database suggests a correlation between LGMN expression levels in gastric cancer and M2 macrophage polarization. (<b>B</b>) Differential serum LGMN expression in patients sensitive and resistant to anti-PD-1 therapy. (<b>C</b>,<b>D</b>) Immunohistochemical detection of M1 or M2 macrophage infiltration in gastric cancer tissues, and their correlation with serum LGMN expression levels in gastric cancer patients (bar 100 μm). * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>LGMN induces polarization of macrophages from M1 to M2 phenotype. (<b>A</b>) Co-culture of THP-1-derived M1 macrophages with ASG-L and BGC823-L cells followed by flow cytometry to detect expression of M2 macrophage markers CD163 and CD206, and the polarization of M1 macrophages towards M2 macrophages can be blocked by LGMN-neutralizing antibodies. (<b>B</b>) Incubation of THP-1-derived M1 macrophages with rh-LGMN protein for 5 days followed by flow cytometry to detect expression of M2 macrophage markers CD163 and CD206. (<b>C</b>) Stimulation of healthy human PBMCs with IFN-γ followed by flow cytometry to detect expression of CD80 and CD86. (<b>D</b>) Induction of M1 macrophages from PBMCs by culture with rh-LGMN, followed by flow cytometry to detect expression of CD206 and CD163.</p>
Full article ">Figure 3
<p>LGMN induces polarization of macrophages from M1 to M2 phenotype by forming a complex with Integrin αvβ3. (<b>A</b>) Western blot analysis of the effect of rh-LGMN, rh-LGMN + Integrin αv antibody, and rh-LGMN + Integrin β3 antibody on mTORC2 activity in M1 macrophages. (<b>B</b>) Flow cytometry analysis of the effect of rh-LGMN, rh-LGMN + Integrin αv antibody, and rh-LGMN + Integrin β3 antibody on the polarization of macrophages from M1 to M2 phenotype. (<b>C</b>) Establishment of M1 macrophage cells derived from THP-1 cells with knockdown of Integrin β3 using lentivirus. (<b>D</b>) Flow cytometry analysis of the effect of knocking down Integrin β3 expression on LGMN-mediated polarize macrophages from M1 to M2 phenotype. (<b>E</b>) Stimulation of M1 macrophages derived from PBMCs with rh-LGMN protein for 48 h followed by Western blot analysis to detect changes in mTORC2 signaling pathway activity. (<b>F</b>) Stimulation of M1 macrophages derived from healthy human PBMCs with rh-LGMN for 48 h followed by flow cytometry to detect changes in CD36 expression.</p>
Full article ">Figure 4
<p>mTOR inhibitor blocks LGMN-induced polarization of macrophages from M1 to M2 phenotype. (<b>A</b>) Flow cytometry analysis of the effect of AZD2014 on LGMN-mediated polarization of macrophages from M1 to M2 phenotype. (<b>B</b>) Western blot analysis of the effect of AZD2014 on upregulation of mTORC2 signaling pathway activity by rh-LGMN in M1 macrophages. (<b>C</b>) Western blot analysis of the effect of rh-LGMN on mTORC1 signaling pathway activity in M1 macrophages.</p>
Full article ">Figure 5
<p>rh-LGMN protein promotes glycolysis and fatty acid oxidation in M1 macrophages derived from healthy human PBMCs. (<b>A</b>) ECAR assay. (<b>B</b>) Fatty acid oxidation assay. (<b>C</b>) Oxygen consumption assay. (<b>D</b>) Fatty acid uptake assay. (<b>E</b>,<b>F</b>) Low-dose mTOR inhibitor AZD2014 inhibits rh-LGMN-induced glycolysis and oxidative phosphorylation in M1 macrophages. (<b>G</b>,<b>H</b>) Blocking antibodies against Integrin αv but not Integrin β3 inhibits rh-LGMN protein-induced glycolysis (<b>G</b>) and oxygen consumption (<b>H</b>) in M1 macrophages derived from healthy human PBMCs. * <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 ((<b>G</b>,<b>H</b>): LGMN vs. LGMN + Integrin αv).</p>
Full article ">Figure 6
<p>sLGMN induces resistance of gastric cancer to anti-PD-1 therapy. (<b>A</b>) Relationship between serum LGMN levels and PFS of gastric cancer to anti-PD-1 therapy. (<b>B</b>) Establishment of 615 mouse gastric cancer model using MCF cells, followed by different therapies to observe the relationship between sLGMN and sensitivity of gastric cancer to PD-1 mAb. (<b>C</b>) Statistical analysis of tumor volume after different treatments in 615 mouse MFC cell gastric cancer model. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; NS, not significant.</p>
Full article ">
17 pages, 2901 KiB  
Review
Roles of Two-Dimensional Materials in Antibiofilm Applications: Recent Developments and Prospects
by Lei Xin, Hongkun Zhao, Min Peng and Yuanjie Zhu
Pharmaceuticals 2024, 17(7), 950; https://doi.org/10.3390/ph17070950 - 16 Jul 2024
Viewed by 497
Abstract
Biofilm-associated infections pose a significant challenge in healthcare, constituting 80% of bacterial infections and often leading to persistent, chronic conditions. Conventional antibiotics struggle with efficacy against these infections due to the high tolerance and resistance induced by bacterial biofilm barriers. Two-dimensional nanomaterials, such [...] Read more.
Biofilm-associated infections pose a significant challenge in healthcare, constituting 80% of bacterial infections and often leading to persistent, chronic conditions. Conventional antibiotics struggle with efficacy against these infections due to the high tolerance and resistance induced by bacterial biofilm barriers. Two-dimensional nanomaterials, such as those from the graphene family, boron nitride, molybdenum disulfide (MoS2), MXene, and black phosphorus, hold immense potential for combating biofilms. These nanomaterial-based antimicrobial strategies are novel tools that show promise in overcoming resistant bacteria and stubborn biofilms, with the ability to circumvent existing drug resistance mechanisms. This review comprehensively summarizes recent developments in two-dimensional nanomaterials, as both therapeutics and nanocarriers for precision antibiotic delivery, with a specific focus on nanoplatforms coupled with photothermal/photodynamic therapy in the elimination of bacteria and penetrating and/or ablating biofilm. This review offers important insight into recent advances and current limitations of current antibacterial nanotherapeutic approaches, together with a discussion on future developments in the field, for the overall benefit of public health. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration for the preparation of multifunctional MoS<sub>2</sub>/ICG/Ag nanocomposites for the photothermally activated triple-mode synergistic antibacterial therapy. Copyright 2022, Elsevier.</p>
Full article ">Figure 2
<p>Schematic illustration of the preparation and application of multiple stimuli-responsive nanozyme-based cryogel. (<b>A</b>) The preparation of CMCSG, MSPA and C/N/MPA; (<b>B</b>) Mechanism of multiple stimuli-responsive antibacterial and self-adaptive wound management. Copyright 2023, Wiley-VCH.</p>
Full article ">Figure 3
<p>Schematic describing (<b>a</b>) the design and synthetic process of DNase-I@V<sub>2</sub>C and (<b>b</b>) the neutrophil immune function conversion (NIFC) mechanism of DNase-I@V<sub>2</sub>C in combating diabetic-related biofilm infections. IRBJIs, implant-related biofilm joint infections; BWIs, biofilm wound infections. Copyright 2023, Wiley-VCH.</p>
Full article ">Figure 4
<p>Schematic illustration of (<b>A</b>) the preparation of monolayer V<sub>2</sub>N and (<b>B</b>) 1064 nm laser irradiation enhanced dual-enzyme-like catalytic activities of V<sub>2</sub>N for promoting the healing of S. aureus-infected abscesses <span class="html-italic">in vivo</span>. Copyright 2023, Elsevier.</p>
Full article ">Figure 5
<p>Synthesis and targeting of Ce6@Man-BPN to macrophages and demonstration of the inactivation of intracellular bacteria by combined photothermal and photodynamic therapy. Copyright 2023, Elsevier.</p>
Full article ">Figure 6
<p>Preparation of the hydrogels: (<b>a</b>) schematic illustration of OCS, APBA-G-OCS and NPs@gel-2; (<b>b</b>) injectable and sprayable process of NPs@gel-2; (<b>c</b>) schematic diagram of the hydrogel that promotes healing of infected diabetic wounds by combined chemo-photothermal antimicrobial and cytokines modulation. Copyright 2023, Elsevier.</p>
Full article ">Scheme 1
<p>Two-dimensional nanomaterials for antibiofilm.</p>
Full article ">
38 pages, 7933 KiB  
Review
Why Is Wnt/β-Catenin Not Yet Targeted in Routine Cancer Care?
by Auriane de Pellegars-Malhortie, Laurence Picque Lasorsa, Thibault Mazard, Fabien Granier and Corinne Prévostel
Pharmaceuticals 2024, 17(7), 949; https://doi.org/10.3390/ph17070949 - 16 Jul 2024
Viewed by 952
Abstract
Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the [...] Read more.
Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach. Full article
(This article belongs to the Special Issue Wnt Signaling in Cancer: New Advances)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Mutation rates in key players of the canonical Wnt/B-catenin pathway in different cancer types (NIH GDC Data Portal release 40.0-March 2024): green (&lt;20%), orange/red (&gt;20%).</p>
Full article ">Figure 2
<p>Flowchart for using Wnt/β-catenin inhibitors as anti-cancer treatments on the basis of the data presented in <a href="#pharmaceuticals-17-00949-f001" class="html-fig">Figure 1</a> and <a href="#pharmaceuticals-17-00949-sch001" class="html-scheme">Scheme 1</a>, <a href="#pharmaceuticals-17-00949-sch002" class="html-scheme">Scheme 2</a>, <a href="#pharmaceuticals-17-00949-sch003" class="html-scheme">Scheme 3</a> and <a href="#pharmaceuticals-17-00949-sch004" class="html-scheme">Scheme 4</a>.</p>
Full article ">Scheme 1
<p>Clinical trials (<a href="https://clinicaltrials.gov/" target="_blank">https://clinicaltrials.gov/</a>) using antibodies as Wnt-dependent inhibitors (WDi). API: active pharmaceutical ingredient; SD: stable disease; PR: partial response; CR: complete response [<a href="#B91-pharmaceuticals-17-00949" class="html-bibr">91</a>,<a href="#B92-pharmaceuticals-17-00949" class="html-bibr">92</a>,<a href="#B93-pharmaceuticals-17-00949" class="html-bibr">93</a>,<a href="#B97-pharmaceuticals-17-00949" class="html-bibr">97</a>,<a href="#B98-pharmaceuticals-17-00949" class="html-bibr">98</a>,<a href="#B99-pharmaceuticals-17-00949" class="html-bibr">99</a>,<a href="#B100-pharmaceuticals-17-00949" class="html-bibr">100</a>,<a href="#B101-pharmaceuticals-17-00949" class="html-bibr">101</a>,<a href="#B104-pharmaceuticals-17-00949" class="html-bibr">104</a>,<a href="#B108-pharmaceuticals-17-00949" class="html-bibr">108</a>,<a href="#B109-pharmaceuticals-17-00949" class="html-bibr">109</a>,<a href="#B112-pharmaceuticals-17-00949" class="html-bibr">112</a>,<a href="#B119-pharmaceuticals-17-00949" class="html-bibr">119</a>].</p>
Full article ">Scheme 2
<p>Clinical trials (<a href="https://clinicaltrials.gov/" target="_blank">https://clinicaltrials.gov/</a>) using small molecules inhibitors (SMi) as Wnt-dependent inhibitors (WDi). API: active pharmaceutical ingredient; SD: stable disease; PR: partial response; CR: complete response [<a href="#B135-pharmaceuticals-17-00949" class="html-bibr">135</a>,<a href="#B136-pharmaceuticals-17-00949" class="html-bibr">136</a>,<a href="#B137-pharmaceuticals-17-00949" class="html-bibr">137</a>,<a href="#B138-pharmaceuticals-17-00949" class="html-bibr">138</a>,<a href="#B140-pharmaceuticals-17-00949" class="html-bibr">140</a>,<a href="#B147-pharmaceuticals-17-00949" class="html-bibr">147</a>,<a href="#B148-pharmaceuticals-17-00949" class="html-bibr">148</a>,<a href="#B151-pharmaceuticals-17-00949" class="html-bibr">151</a>,<a href="#B152-pharmaceuticals-17-00949" class="html-bibr">152</a>,<a href="#B153-pharmaceuticals-17-00949" class="html-bibr">153</a>,<a href="#B154-pharmaceuticals-17-00949" class="html-bibr">154</a>,<a href="#B155-pharmaceuticals-17-00949" class="html-bibr">155</a>,<a href="#B156-pharmaceuticals-17-00949" class="html-bibr">156</a>,<a href="#B157-pharmaceuticals-17-00949" class="html-bibr">157</a>,<a href="#B158-pharmaceuticals-17-00949" class="html-bibr">158</a>,<a href="#B159-pharmaceuticals-17-00949" class="html-bibr">159</a>,<a href="#B160-pharmaceuticals-17-00949" class="html-bibr">160</a>].</p>
Full article ">Scheme 3
<p>Clinical trials (<a href="https://clinicaltrials.gov/" target="_blank">https://clinicaltrials.gov/</a>) using small molecules inhibitors (SMi) as Wnt-independent inhibitors (WIi) preventing β-catenin stabilization. API: active pharmaceutical ingredient; SD: stable disease; PR: partial response; CR: complete response [<a href="#B166-pharmaceuticals-17-00949" class="html-bibr">166</a>,<a href="#B169-pharmaceuticals-17-00949" class="html-bibr">169</a>,<a href="#B175-pharmaceuticals-17-00949" class="html-bibr">175</a>,<a href="#B178-pharmaceuticals-17-00949" class="html-bibr">178</a>,<a href="#B184-pharmaceuticals-17-00949" class="html-bibr">184</a>,<a href="#B193-pharmaceuticals-17-00949" class="html-bibr">193</a>,<a href="#B195-pharmaceuticals-17-00949" class="html-bibr">195</a>,<a href="#B196-pharmaceuticals-17-00949" class="html-bibr">196</a>,<a href="#B197-pharmaceuticals-17-00949" class="html-bibr">197</a>,<a href="#B198-pharmaceuticals-17-00949" class="html-bibr">198</a>,<a href="#B199-pharmaceuticals-17-00949" class="html-bibr">199</a>,<a href="#B200-pharmaceuticals-17-00949" class="html-bibr">200</a>,<a href="#B201-pharmaceuticals-17-00949" class="html-bibr">201</a>,<a href="#B202-pharmaceuticals-17-00949" class="html-bibr">202</a>,<a href="#B203-pharmaceuticals-17-00949" class="html-bibr">203</a>,<a href="#B204-pharmaceuticals-17-00949" class="html-bibr">204</a>,<a href="#B205-pharmaceuticals-17-00949" class="html-bibr">205</a>,<a href="#B206-pharmaceuticals-17-00949" class="html-bibr">206</a>,<a href="#B207-pharmaceuticals-17-00949" class="html-bibr">207</a>,<a href="#B208-pharmaceuticals-17-00949" class="html-bibr">208</a>,<a href="#B209-pharmaceuticals-17-00949" class="html-bibr">209</a>,<a href="#B210-pharmaceuticals-17-00949" class="html-bibr">210</a>,<a href="#B211-pharmaceuticals-17-00949" class="html-bibr">211</a>,<a href="#B220-pharmaceuticals-17-00949" class="html-bibr">220</a>,<a href="#B221-pharmaceuticals-17-00949" class="html-bibr">221</a>].</p>
Full article ">Scheme 3 Cont.
<p>Clinical trials (<a href="https://clinicaltrials.gov/" target="_blank">https://clinicaltrials.gov/</a>) using small molecules inhibitors (SMi) as Wnt-independent inhibitors (WIi) preventing β-catenin stabilization. API: active pharmaceutical ingredient; SD: stable disease; PR: partial response; CR: complete response [<a href="#B166-pharmaceuticals-17-00949" class="html-bibr">166</a>,<a href="#B169-pharmaceuticals-17-00949" class="html-bibr">169</a>,<a href="#B175-pharmaceuticals-17-00949" class="html-bibr">175</a>,<a href="#B178-pharmaceuticals-17-00949" class="html-bibr">178</a>,<a href="#B184-pharmaceuticals-17-00949" class="html-bibr">184</a>,<a href="#B193-pharmaceuticals-17-00949" class="html-bibr">193</a>,<a href="#B195-pharmaceuticals-17-00949" class="html-bibr">195</a>,<a href="#B196-pharmaceuticals-17-00949" class="html-bibr">196</a>,<a href="#B197-pharmaceuticals-17-00949" class="html-bibr">197</a>,<a href="#B198-pharmaceuticals-17-00949" class="html-bibr">198</a>,<a href="#B199-pharmaceuticals-17-00949" class="html-bibr">199</a>,<a href="#B200-pharmaceuticals-17-00949" class="html-bibr">200</a>,<a href="#B201-pharmaceuticals-17-00949" class="html-bibr">201</a>,<a href="#B202-pharmaceuticals-17-00949" class="html-bibr">202</a>,<a href="#B203-pharmaceuticals-17-00949" class="html-bibr">203</a>,<a href="#B204-pharmaceuticals-17-00949" class="html-bibr">204</a>,<a href="#B205-pharmaceuticals-17-00949" class="html-bibr">205</a>,<a href="#B206-pharmaceuticals-17-00949" class="html-bibr">206</a>,<a href="#B207-pharmaceuticals-17-00949" class="html-bibr">207</a>,<a href="#B208-pharmaceuticals-17-00949" class="html-bibr">208</a>,<a href="#B209-pharmaceuticals-17-00949" class="html-bibr">209</a>,<a href="#B210-pharmaceuticals-17-00949" class="html-bibr">210</a>,<a href="#B211-pharmaceuticals-17-00949" class="html-bibr">211</a>,<a href="#B220-pharmaceuticals-17-00949" class="html-bibr">220</a>,<a href="#B221-pharmaceuticals-17-00949" class="html-bibr">221</a>].</p>
Full article ">Scheme 4
<p>Clinical trials (<a href="https://clinicaltrials.gov/" target="_blank">https://clinicaltrials.gov/</a>) using small molecules inhibitors (SMi) as Wnt-independent inhibitors (WIi) preventing β-catenin transcriptional activity. API: active pharmaceutical ingredient; SD: stable disease; PR: partial response; CR: complete response [<a href="#B231-pharmaceuticals-17-00949" class="html-bibr">231</a>,<a href="#B232-pharmaceuticals-17-00949" class="html-bibr">232</a>,<a href="#B233-pharmaceuticals-17-00949" class="html-bibr">233</a>,<a href="#B234-pharmaceuticals-17-00949" class="html-bibr">234</a>,<a href="#B235-pharmaceuticals-17-00949" class="html-bibr">235</a>,<a href="#B238-pharmaceuticals-17-00949" class="html-bibr">238</a>,<a href="#B239-pharmaceuticals-17-00949" class="html-bibr">239</a>,<a href="#B246-pharmaceuticals-17-00949" class="html-bibr">246</a>].</p>
Full article ">Scheme 5
<p>Antibody-drug conjugates (ADC) and peptide-drug conjugates (PDC) targeting the Wnt/β-catenin signaling. MMAE: Monomethyl Auristatin E; Aur0101: Auristatin Microtubule Inhibitor; PNU159682: DNA damaging topoisomerase-inhibiting anthracycline; (ABD)-PA: (Albumin Binding Domain)- Pseudomonas endotoxin A; ABD; DMSA: Streptomyces Duocarmycin [<a href="#B276-pharmaceuticals-17-00949" class="html-bibr">276</a>,<a href="#B277-pharmaceuticals-17-00949" class="html-bibr">277</a>,<a href="#B278-pharmaceuticals-17-00949" class="html-bibr">278</a>,<a href="#B281-pharmaceuticals-17-00949" class="html-bibr">281</a>,<a href="#B283-pharmaceuticals-17-00949" class="html-bibr">283</a>,<a href="#B284-pharmaceuticals-17-00949" class="html-bibr">284</a>,<a href="#B285-pharmaceuticals-17-00949" class="html-bibr">285</a>,<a href="#B286-pharmaceuticals-17-00949" class="html-bibr">286</a>].</p>
Full article ">
43 pages, 3516 KiB  
Systematic Review
Ethnomedicinal Usage, Phytochemistry and Pharmacological Potential of Solanum surattense Burm. f.
by Kamrul Hasan, Shabnam Sabiha, Nurul Islam, João F. Pinto and Olga Silva
Pharmaceuticals 2024, 17(7), 948; https://doi.org/10.3390/ph17070948 - 15 Jul 2024
Viewed by 708
Abstract
Solanum surattense Burm. f. is a significant member of the Solanaceae family, and the Solanum genus is renowned for its traditional medicinal uses and bioactive potential. This systematic review adheres to PRISMA methodology, analyzing scientific publications between 1753 and 2023 from B-on, Google [...] Read more.
Solanum surattense Burm. f. is a significant member of the Solanaceae family, and the Solanum genus is renowned for its traditional medicinal uses and bioactive potential. This systematic review adheres to PRISMA methodology, analyzing scientific publications between 1753 and 2023 from B-on, Google Scholar, PubMed, Science Direct, and Web of Science, aiming to provide comprehensive and updated information on the distribution, ethnomedicinal uses, chemical constituents, and pharmacological activities of S. surattense, highlighting its potential as a source of herbal drugs. Ethnomedicinally, this species is important to treat skin diseases, piles complications, and toothache. The fruit was found to be the most used part of this plant (25%), together with the whole plant (22%) used to treat different ailments, and its decoction was found to be the most preferable mode of herbal drug preparation. A total of 338 metabolites of various chemical classes were isolated from S. surattense, including 137 (40.53%) terpenoids, 56 (16.56%) phenol derivatives, and 52 (15.38%) lipids. Mixtures of different parts of this plant in water–ethanol have shown in vitro and/or in vivo antioxidant, anti-inflammatory, antimicrobial, anti-tumoral, hepatoprotective, and larvicidal activities. Among the metabolites, 51 were identified and biologically tested, presenting antioxidant, anti-inflammatory, and antitumoral as the most reported activities. Clinical trials in humans made with the whole plant extract showed its efficacy as an anti-asthmatic agent. Mostly steroidal alkaloids and triterpenoids, such as solamargine, solanidine, solasodine, solasonine, tomatidine, xanthosaponin A–B, dioscin, lupeol, and stigmasterol are biologically the most active metabolites with high potency that reflects the new and high potential of this species as a novel source of herbal medicines. More experimental studies and a deeper understanding of this plant must be conducted to ensure its use as a source of raw materials for pharmaceutical use. Full article
(This article belongs to the Special Issue Feature Reviews in Natural Products)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>General picture of <span class="html-italic">S. surattense</span>.</p>
Full article ">Figure 2
<p>Screening of published data based on the PRISMA methodology.</p>
Full article ">Figure 3
<p>Ethnomedicinal uses of different parts of the <span class="html-italic">S. surattense</span> plant. Abbreviation: Wp—whole plant; L—leaf; F—fruit; S—seed; Fl—flower; R—root.</p>
Full article ">Figure 4
<p>Examples of phenolic compounds isolated from different parts of <span class="html-italic">S. surattense</span>.</p>
Full article ">Figure 5
<p>Examples of steroidal alkaloids isolated from different parts of <span class="html-italic">S. surattense</span>.</p>
Full article ">Figure 6
<p>Triterpenoids isolated from different parts of <span class="html-italic">S. surattense</span>.</p>
Full article ">Figure 7
<p>Some fatty acids isolated from different parts of <span class="html-italic">S. surattense</span>.</p>
Full article ">Figure 8
<p>Different biological activities (%) of <span class="html-italic">S. surattense</span>.</p>
Full article ">Figure 9
<p>Percentage of different plant parts of <span class="html-italic">S. surattense</span> used in different biological activities. Abbreviation: F—fruit; L—leaf; Wp—whole plant; Ap—aerial part; R—root; Stb—stem bark; Fl—flower; S—seed; B—bark; St—stem.</p>
Full article ">
1 pages, 144 KiB  
Correction
Correction: Heruye et al. Current Trends in the Pharmacotherapy of Cataracts. Pharmaceuticals 2020, 13, 15
by Segewkal H. Heruye, Leonce N. Maffofou Nkenyi, Neetu U. Singh, Dariush Yalzadeh, Kalu K. Ngele, Ya-Fatou Njie-Mbye, Sunny E. Ohia and Catherine A. Opere
Pharmaceuticals 2024, 17(7), 947; https://doi.org/10.3390/ph17070947 - 15 Jul 2024
Viewed by 290
Abstract
In the original publication [...] Full article
23 pages, 2166 KiB  
Article
Unraveling the Bioactive Potential of Camellia japonica Edible Flowers: Profiling Antioxidant Substances and In Vitro Bioactivity Assessment
by Antia G. Pereira, Maria Fraga-Corral, Aurora Silva, Maria Fatima Barroso, Clara Grosso, Maria Carpena, Pascual Garcia-Perez, Rosa Perez-Gregorio, Lucia Cassani, Jesus Simal-Gandara and Miguel A. Prieto
Pharmaceuticals 2024, 17(7), 946; https://doi.org/10.3390/ph17070946 - 15 Jul 2024
Viewed by 804
Abstract
In recent years, the search for novel natural-based ingredients by food and related industries has sparked extensive research aimed at discovering new sources of functional molecules. Camellia japonica, traditionally known as an ornamental plant, has gained attention due to its diverse array [...] Read more.
In recent years, the search for novel natural-based ingredients by food and related industries has sparked extensive research aimed at discovering new sources of functional molecules. Camellia japonica, traditionally known as an ornamental plant, has gained attention due to its diverse array of bioactive compounds with potential industrial applications. Although C. japonica flowers are edible, their phytochemical profile has not been thoroughly investigated. In this study, a phenolic profile screening through an HPLC–ESI-QQQ-MS/MS approach was applied to C. japonica flower extracts, revealing a total of 36 compounds, including anthocyanins, curcuminoids, dihydrochalcones, dihydroflavonols, flavonols, flavones, hydroxybenzoic acids, hydroxycinnamic acids, isoflavonoids, stilbenes, and tyrosols. Following extract profiling, their bioactivity was assessed by means of in vitro antioxidant, antimicrobial, cytotoxic, and neuroprotective activities. The results showed a multifaceted high correlation of phenolic compounds with all the tested bioactivities according to Pearson’s correlation analysis, unraveling the potential of C. japonica flowers as promising sources of nutraceuticals. Overall, these findings provide insight into the valorization of C. japonica flowers from different unexplored cultivars thus diversifying their industrial outcome. Full article
(This article belongs to the Special Issue Advances in Mass Spectrometry Metrology in Pharmaceutical Sciences)
Show Figures

Figure 1

Figure 1
<p>Results of crocin assay. (<b>A</b>) Raw experimental signal for the Elegans Variegated cultivarcultivar, with the experimental data fitted to Equation (2) to determine IC<sub>50</sub> values. (<b>B</b>) Compilation of IC<sub>50</sub> values for all cultivars, providing a comparative view of crocin assay outcomes across the different <span class="html-italic">C. japonica</span> flower cultivars.</p>
Full article ">Figure 2
<p>Graphs of in vitro antioxidant studies. Column 1 illustrates hydroxyl radical scavenging activity, column 2 showcases H<sub>2</sub>O<sub>2</sub> scavenging activity, column 3 demonstrates superoxide radical scavenging activity, and column 4 displays nitric oxide scavenging activity. Symbols represent experimental data, while lines depict model values. The bar graphs in the final row present the IC<sub>50</sub> values (mg/mL) for each assay across all the cultivars examined in this study.</p>
Full article ">Figure 3
<p>Pearson’s correlation heatmap between the different biological activities of <span class="html-italic">C. japonica</span> L. flower extracts (antioxidant activity, by means of DPPH, ABTS, CBA, SRSA, OHSA, NOSA, H<sub>2</sub>O<sub>2</sub> methods; cytotoxic activity towards Vero, AGS, HepG2, A549 cell lines; anti-microbial activity against <span class="html-italic">E. coli</span>, <span class="html-italic">S. epidermidis</span>, <span class="html-italic">S. aureus</span>, <span class="html-italic">P. aeruginosa</span>, <span class="html-italic">S. enteritidis</span>, and <span class="html-italic">B. cereus</span>; and neuroprotective activity).</p>
Full article ">
27 pages, 4126 KiB  
Review
Advances in Nanomedicine for Precision Insulin Delivery
by Alfredo Caturano, Roberto Nilo, Davide Nilo, Vincenzo Russo, Erica Santonastaso, Raffaele Galiero, Luca Rinaldi, Marcellino Monda, Celestino Sardu, Raffaele Marfella and Ferdinando Carlo Sasso
Pharmaceuticals 2024, 17(7), 945; https://doi.org/10.3390/ph17070945 - 15 Jul 2024
Viewed by 1925
Abstract
Diabetes mellitus, which comprises a group of metabolic disorders affecting carbohydrate metabolism, is characterized by improper glucose utilization and excessive production, leading to hyperglycemia. The global prevalence of diabetes is rising, with projections indicating it will affect 783.2 million people by 2045. Insulin [...] Read more.
Diabetes mellitus, which comprises a group of metabolic disorders affecting carbohydrate metabolism, is characterized by improper glucose utilization and excessive production, leading to hyperglycemia. The global prevalence of diabetes is rising, with projections indicating it will affect 783.2 million people by 2045. Insulin treatment is crucial, especially for type 1 diabetes, due to the lack of β-cell function. Intensive insulin therapy, involving multiple daily injections or continuous subcutaneous insulin infusion, has proven effective in reducing microvascular complications but poses a higher risk of severe hypoglycemia. Recent advancements in insulin formulations and delivery methods, such as ultra-rapid-acting analogs and inhaled insulin, offer potential benefits in terms of reducing hypoglycemia and improving glycemic control. However, the traditional subcutaneous injection method has drawbacks, including patient compliance issues and associated complications. Nanomedicine presents innovative solutions to these challenges, offering promising avenues for overcoming current drug limitations, enhancing cellular uptake, and improving pharmacokinetics and pharmacodynamics. Various nanocarriers, including liposomes, chitosan, and PLGA, provide protection against enzymatic degradation, improving drug stability and controlled release. These nanocarriers offer unique advantages, ranging from enhanced bioavailability and sustained release to specific targeting capabilities. While oral insulin delivery is being explored for better patient adherence and cost-effectiveness, other nanomedicine-based methods also show promise in improving delivery efficiency and patient outcomes. Safety concerns, including potential toxicity and immunogenicity issues, must be addressed, with the FDA providing guidance for the safe development of nanotechnology-based products. Future directions in nanomedicine will focus on creating next-generation nanocarriers with precise targeting, real-time monitoring, and stimuli-responsive features to optimize diabetes treatment outcomes and patient safety. This review delves into the current state of nanomedicine for insulin delivery, examining various types of nanocarriers and their mechanisms of action, and discussing the challenges and future directions in developing safe and effective nanomedicine-based therapies for diabetes management. Full article
(This article belongs to the Special Issue Advancements in Cardiovascular and Antidiabetic Drug Therapy)
Show Figures

Figure 1

Figure 1
<p>Barriers in the gastrointestinal tract to oral insulin delivery and potential pathways.</p>
Full article ">Figure 2
<p>Mechanisms of the gastrointestinal barrier: gap junctions and intracellular pathways for oral insulin delivery.</p>
Full article ">Figure 3
<p>Structural overview of nanocarriers used for insulin delivery.</p>
Full article ">
20 pages, 933 KiB  
Systematic Review
Impact of Anti-CD38 Monoclonal Antibody Therapy on CD34+ Hematopoietic Stem Cell Mobilization, Collection, and Engraftment in Multiple Myeloma Patients—A Systematic Review
by Flavia Bigi, Enrica Manzato, Simona Barbato, Marco Talarico, Michele Puppi, Simone Masci, Ilaria Sacchetti, Roberta Restuccia, Miriam Iezza, Ilaria Rizzello, Chiara Sartor, Katia Mancuso, Lucia Pantani, Paola Tacchetti, Michele Cavo and Elena Zamagni
Pharmaceuticals 2024, 17(7), 944; https://doi.org/10.3390/ph17070944 - 15 Jul 2024
Viewed by 842
Abstract
This systematic review examines the available clinical data on CD34+ cell mobilization, collection, and engraftment in multiple myeloma patients treated with the anti-CD38 monoclonal antibodies daratumumab and isatuximab in clinical trials and in real life. Twenty-six clinical reports were published between 2019 and [...] Read more.
This systematic review examines the available clinical data on CD34+ cell mobilization, collection, and engraftment in multiple myeloma patients treated with the anti-CD38 monoclonal antibodies daratumumab and isatuximab in clinical trials and in real life. Twenty-six clinical reports were published between 2019 and February 2024. Most studies documented lower circulating CD34+ cells after mobilization compared to controls, leading to higher plerixafor requirements. Although collection yields were significantly lower in approximately half of the studies, the collection target was achieved in similar proportions of daratumumab- and isatuximab-treated and nontreated patients, and access to autologous stem cell transplant (ASCT) was comparable. This could be explained by the retained efficacy of plerixafor in anti-CD38 monoclonal antibody-treated patients, while no chemotherapy-based or sparing mobilization protocol proved superior. Half of the studies reported slower hematopoietic reconstitution after ASCT in daratumumab- and isatuximab-treated patients, without an excess of infectious complications. While no direct effect on stem cells was observed in vitro, emerging evidence suggests possible dysregulation of CD34+ cell adhesion after daratumumab treatment. Overall, anti-CD38 monoclonal antibodies appear to interfere with CD34+ cell mobilization, without consistently leading to significant clinical consequences. Further research is needed to elucidate the underlying mechanisms and define optimal mobilization strategies in this patient population. Full article
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram.</p>
Full article ">Figure 2
<p>Visual summary of outcomes: Studies with and without a significant impact of daratumumab or isatuximab induction on CD34+ cell-related outcomes. Abbreviations: ASCT: autologous stem cell transplant, MoAb: monoclonal antibody, pts: patients.</p>
Full article ">
19 pages, 11576 KiB  
Article
In Vitro Biological Evaluation of an Alginate-Based Hydrogel Loaded with Rifampicin for Wound Care
by Tudor Bibire, Radu Dănilă, Cătălina Natalia Yilmaz, Liliana Verestiuc, Isabella Nacu, Ramona Gabriela Ursu and Cristina Mihaela Ghiciuc
Pharmaceuticals 2024, 17(7), 943; https://doi.org/10.3390/ph17070943 - 14 Jul 2024
Viewed by 793
Abstract
We report a biocompatible hydrogel dressing based on sodium alginate-grafted poly(N-vinylcaprolactam) prepared by encapsulation of Rifampicin as an antimicrobial drug and stabilizing the matrix through the repeated freeze–thawing method. The hydrogel structure and polymer-drug compatibility were confirmed by FTIR, and a series of [...] Read more.
We report a biocompatible hydrogel dressing based on sodium alginate-grafted poly(N-vinylcaprolactam) prepared by encapsulation of Rifampicin as an antimicrobial drug and stabilizing the matrix through the repeated freeze–thawing method. The hydrogel structure and polymer-drug compatibility were confirmed by FTIR, and a series of hydrogen-bond-based interactions between alginate and Rifampicin were identified. A concentration of 0.69% Rifampicin was found in the polymeric matrix using HPLC analysis and spectrophotometric UV–Vis methods. The hydrogel’s morphology was evaluated by scanning electron microscopy, and various sizes and shapes of pores, ranging from almost spherical geometries to irregular ones, with a smooth surface of the pore walls and high interconnectivity in the presence of the drug, were identified. The hydrogels are bioadhesive, and the adhesion strength increased after Rifampicin was encapsulated into the polymeric matrix, which suggests that these compositions are suitable for wound dressings. Antimicrobial activity against S. aureus and MRSA, with an increased effect in the presence of the drug, was also found in the newly prepared hydrogels. In vitro biological evaluation demonstrated the cytocompatibility of the hydrogels and their ability to stimulate cell multiplication and mutual cell communication. The in vitro scratch assay demonstrated the drug-loaded alginate-grafted poly(N-vinylcaprolactam) hydrogel’s ability to stimulate cell migration and wound closure. All of these results suggest that the prepared hydrogels can be used as antimicrobial materials for wound healing and care applications. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Description of the preparation steps of the hydrogel based on sodium alginate grafted with poly(N-vinylcaprolactam) and loaded with Rifampicin via solution mixing and freeze–thawing processes.</p>
Full article ">Figure 2
<p>FTIR spectra: H -Alg-PNVCL, H-Alg-PNVCL-Rif, and Rifampicin.</p>
Full article ">Figure 3
<p>SEM data and porosity distribution of H-Alg-PNVCL and H-Alg-PNVCL-Rif hydrogels.</p>
Full article ">Figure 4
<p>HPLC chromatogram of the Rifampicin-loaded hydrogel and the calibration curve for drug release.</p>
Full article ">Figure 5
<p>Detachment force and the work of adhesion for H-Alg-PNVCL and H-Alg-PNVCL-Rif hydrogels (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>MTT cell viability of NHDF cells for (<b>A</b>) Rifampicin solutions and (<b>B</b>) H-Alg-PNVCL and H-Alg-PNVCL-Rif hydrogels (n = 3; * <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).</p>
Full article ">Figure 7
<p>Viable NHDF cells and fixed cells, respectively, after 72 h of cell culturing with H-Alg-PNVCl and H-Alg-PNVCL-Rif hydrogels (<b>A</b>) and Rifampicin (<b>B</b>).</p>
Full article ">Figure 8
<p>In vitro evaluation of NHDF migration in the presence of different concentrations of Rifampicin (NHDF cell line stained with Calcein AM at various times).</p>
Full article ">Figure 9
<p>In vitro evaluation of NHDF migration in the presence of H-Alg-PNVCL hydrogel and H-Alg-PNVCL-Rif hydrogel (NHDF cell line stained with Calcein AM at various times).</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop