Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,053)

Search Parameters:
Keywords = antiproliferative agents

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 3475 KiB  
Article
Gallic Acid Induces HeLa Cell Lines Apoptosis via the P53/Bax Signaling Pathway
by Umut Sarı, Fuat Zaman, İlhan Özdemir, Şamil Öztürk and Mehmet Cudi Tuncer
Biomedicines 2024, 12(11), 2632; https://doi.org/10.3390/biomedicines12112632 - 18 Nov 2024
Viewed by 401
Abstract
Background: Cervical cancer is a type of cancer that originates from the endometrium and is more common in developed countries and its incidence is increasing day by day in developing countries. The most commonly prescribed chemotherapeutic drugs limit their use due to serious [...] Read more.
Background: Cervical cancer is a type of cancer that originates from the endometrium and is more common in developed countries and its incidence is increasing day by day in developing countries. The most commonly prescribed chemotherapeutic drugs limit their use due to serious side effects and the development of drug resistance. For this reason, interest in new active ingredients obtained from natural products is increasing. This study aimed to reveal the apoptotic and antiproliferative effects of gallic acid and doxorubicin combination therapy against the HeLa cell line. Methods: We investigated the anti-cancer effects of doxorubicin and gallic acid in the human HeLa cervical cell line by using the MTT test, Nucblue staining for the identification of apoptotic cells due to nuclear condensation using fluorescent substance, and apoptotic markers P53 and Bax for the RT-PCR test. Results: The highest cytotoxic effect obtained in the study, the highest increase in apoptotic induction, and a significant difference in P53/Bax levels were seen in the gallic acid/doxorubicin combination. Additionally, it was determined that gallic acid exhibited an effective cytotoxic effect on HeLa and HaCat cells within 48 and 72 h of application. Conclusions: The obtained findings show that the gallic acid/doxorubicin combination applied to HeLa cells may be an alternative treatment against both the cytotoxic effect size and the side effects of the chemotherapy agent. Full article
(This article belongs to the Collection Feature Papers in Cell Biology and Pathology)
Show Figures

Figure 1

Figure 1
<p>The effect of DOX application on 9 different concentrations obtained by serial dilution in the concentration range of 10-1000 nM in HeLa cervix adenocarcinoma (<b>A</b>–<b>C</b>) and HaCaT human skin keratinocyte cell line (<b>D</b>–<b>F</b>) on compared to the vehicle group and the IC50 value of the chemotherapy agent (n = 6; data are mean ± standard deviation values, inhibition concentration (IC) values calculated by probit analysis). * Data are statistically significant compared to control, one-way ANOVA, Tukey HSD test, <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 2
<p>Effect of GA application on 9 different concentrations obtained by serial dilution between 10-1000 µM concentration range in HeLa cervix adenocarcinoma (<b>A</b>–<b>C</b>) and HaCaT human skin keratinocyte cell line (<b>D</b>–<b>F</b>) cell lines for 24, 48 and 72 hours on cell viability compared to the vehicle group and the IC50 value of GA (n = 6; data are mean ± standard deviation values, inhibition concentration (IC) values calculated by probit analysis). * Data are statistically significant compared to control, one-way ANOVA, Tukey HSD test, <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 3
<p>Cell morphology, nuclear structure, and apoptotic body formation (magnification: ×20) in HeLa cervical adenocarcinoma cell populations treated for 48 hours with vehicle control (<b>A</b>,<b>A1</b>), DOX IC50: 137.6 nM (<b>B</b>,<b>B1</b>), GA IC50: 239.2 μM (<b>C</b>,<b>C1</b>), and DOX IC50+GA IC50 (<b>D</b>,<b>D1</b>) (Arrow: apoptotic cell).</p>
Full article ">Figure 4
<p>H-scores were derived from semi-quantitative assessments of both staining intensity (scale 0–3) and the percentage of positive cells (0–100%) and, when multiplied, generated a score ranging from 0 to 300.</p>
Full article ">Figure 5
<p>Relative fold increases values of P53 and BAX gene expressions in HeLa cervical adenocarcinoma cell lines, DOX IC<sub>50</sub>: 137.6 nM, GA IC<sub>50</sub>: 239.2 μM, 48 h, after single and combined drug administration (data in multiple control with β-actin and GAPDH mRNA level). Method, n = 4 data mean ± SH), * means are statistically different, one-way ANOVA, Tukey HSD test, <span class="html-italic">p</span> values are given in the graph.</p>
Full article ">Figure 6
<p>PPI and interaction between various genes of cervical cancer.</p>
Full article ">Figure 7
<p>Enrichment analysis for the 530 common compound targets in cancer pathway.</p>
Full article ">Figure 8
<p>Enrichment analysis for the 331 common compound targets in human papilloma virüs infection and cervical cancer.</p>
Full article ">
15 pages, 5198 KiB  
Article
Evaluation of Antiproliferative Potentials Associated with the Volatile Compounds of Lantana camara Flowers: Selective In Vitro Activity
by Jennifer El Hajj, Louna Karam, Ali Jaber, Edmond Cheble, Elias Akoury, Philippe Hussein Kobeissy, José-Noel Ibrahim and Ali Yassin
Molecules 2024, 29(22), 5431; https://doi.org/10.3390/molecules29225431 - 18 Nov 2024
Viewed by 526
Abstract
Probing the chemical profiles and biological activities of medicinal plants is important for the discovery of new potent therapeutic products. Our study deciphers the chemical composition of the essential oils (EOs) obtained from three different flowers of Lantana camara and evaluates their antioxidant [...] Read more.
Probing the chemical profiles and biological activities of medicinal plants is important for the discovery of new potent therapeutic products. Our study deciphers the chemical composition of the essential oils (EOs) obtained from three different flowers of Lantana camara and evaluates their antioxidant and anticancer activities. This work represents the first study of EOs obtained from this plant and is based particularly on the difference in flower color. In addition, no other reports dealing specifically with the antitumor effects of such flower-derived EOs have been described in the literature. The collected flowers, white, pink, and orange, were extracted by hydrodistillation to yield EO1, EO2, and EO3 respectively. Gas chromatography–mass spectroscopy was primarily employed to identify the existing volatile compounds in the samples. Their antioxidant activities were screened through both DPPH (2,2-diphenyl-1-picrylhydrazyl) scavenging assays and FRAP (ferric-reducing antioxidant power) assays. The antiproliferative effects were evaluated on two distinct breast cancer cell lines, MCF-7 and MDA-MB-231, and compared to a normal human breast cell line, MCF-10A, using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium) assay. All EOs showed notable antioxidant potential attributed to the active phytochemical compounds, with results being supported by a positive correlation between such activity and the total phenolic and flavonoid content. The most eminent, EO1, revealed a selective dose-dependent antiproliferative effect in both breast cancer cell lines, thus reflecting its potent role as an anticancer agent. We suggest that this highly selective activity is associated with the presence of bicyclogermacrene and epi-bicyclosesquiphellandrene in its chemical composition. Full article
(This article belongs to the Special Issue Advances in Plant-Sourced Natural Compounds as Anticancer Agents)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The three flowers of <span class="html-italic">L. camara</span> (white, pink, and orange) as collected.</p>
Full article ">Figure 2
<p>(<b>A</b>–<b>C</b>): Main constituents of the EOs of <span class="html-italic">L. camara</span> obtained from GC-MS analysis. (<b>D</b>): Yields of extraction of the three samples. (<b>E</b>): Chemical structures of the different compounds.</p>
Full article ">Figure 3
<p>Results of IC50 of the three EOs as compared to ascorbic acid.</p>
Full article ">Figure 4
<p>In vitro cytotoxic effect of EO1, EO2, and EO3 on the MCF-10A, MCF-7, and MDA-MB231 cell lines. Representative graphs of the time-dependent cytotoxic effect of different concentrations of the oils on MCF-10A (<b>A</b>), MCF-7 (<b>B</b>), and MDA-MB-231 (<b>C</b>) cells. Dose-dependent inhibitory activity for EO1, EO2, and EO3 48 h post-treatment on MCF-7 (<b>D</b>) and MDA-MB-231 (<b>E</b>) is presented as a bar plot. Data are presented as mean ± SD (n = 3) with all treatments compared to each other by a two-way ANOVA test followed by the post hoc Tukey test for multiple comparison analysis; * <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, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Chemical structures of the possible antitumor-active phytochemicals in EO1.</p>
Full article ">
14 pages, 1407 KiB  
Article
Acid Sphingomyelinase Activation and ROS Generation Potentiate Antiproliferative Effects of Mitomycin in HCC
by Sirkka Buitkamp, Stephanie Schwalm, Katja Jakobi, Nerea Ferreiros, Christin Wünsche, Stefan Zeuzem, Erich Gulbins, Christoph Sarrazin, Josef Pfeilschifter and Georgios Grammatikos
Int. J. Mol. Sci. 2024, 25(22), 12175; https://doi.org/10.3390/ijms252212175 - 13 Nov 2024
Viewed by 322
Abstract
Sphingolipids play a major role in the regulation of hepatocellular apoptosis and proliferation. We have previously identified sphingolipid metabolites as biomarkers of chronic liver disease and hepatocellular carcinoma. Human hepatocellular carcinoma cell lines were transfected with a plasmid vector encoding for acid sphingomyelinase. [...] Read more.
Sphingolipids play a major role in the regulation of hepatocellular apoptosis and proliferation. We have previously identified sphingolipid metabolites as biomarkers of chronic liver disease and hepatocellular carcinoma. Human hepatocellular carcinoma cell lines were transfected with a plasmid vector encoding for acid sphingomyelinase. Overexpressing cells were subsequently treated with mitomycin and cell proliferation, acid sphingomyelinase activity, sphingolipid concentrations, and generation of reactive oxygen species were assessed. The stimulation of acid sphingomyelinase-overexpressing cell lines with mitomycin showed a significant activation of the enzyme (p < 0.001) followed by an accumulation of various ceramide species (p < 0.001) and reactive oxygen radicals (p < 0.001) as compared to control transfected cells. Consequently, a significant reduction in cell proliferation was observed in acid sphingomyelinase-overexpressing cells (p < 0.05) which could be diminished by the simultaneous application of antioxidant agents. Moreover, the application of mitomycin induced significant alterations in mRNA expression levels of ceramidases and sphingosine kinases (p < 0.05). Our data suggest that the overexpression of the acid sphingomyelinase in human hepatoma cell lines enhances the in vitro antiproliferative potential of mitomycin via accumulation of ceramide and reactive oxygen species. The selective activation of acid sphingomyelinase might offer a novel therapeutic approach in the treatment of hepatocellular carcinoma. Full article
Show Figures

Figure 1

Figure 1
<p>Mitomycin C upregulates the expression and activity of ASM. mRNA-expression and enzyme activity after transfection with either an empty vector (pJK) or a plasmid encoding for ASM (<b>A</b>,<b>B</b>). The transfected Huh7.5 and HepG2 cells were treated for 48 h either with or without 3 µM mitomycin C (MMC) (<b>C</b>,<b>D</b>). The data are expressed in % of the basic expression level (<b>A</b>) or in % of untreated control (<b>C</b>). Given the specific activity of [<sup>14</sup>C] Sphingomyelin of 52 mCi/mmol, the activity of 100 cpm/μg/h corresponds to 1 pmol/μg/h specific activity of the acid sphingomyelinase. Shown are means +/− SD of three independent experiments (<span class="html-italic">n</span> = 3). *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05 indicate statistical significance when compared to the control values (<b>B</b>,<b>D</b>). The results are expressed in counts normalized on extracted protein (<b>B</b>) or in % of untreated control (<b>D</b>) and are means ± SD (<span class="html-italic">n</span> = 3, *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Mitomycin C causes enhanced cleavage of sphingomyeline and upregulates cellular levels of the ceramide species in ASM-overexpressing cells. The ceramide levels in ASM transfected (ASM) and control transfected (pJK) Huh7.5 after treatment with 3 µM mitomycin C (MMC) for 48 h. The data are expressed in % of the untreated control and are means ± SD from a total of three independent experiments (<span class="html-italic">n</span> = 3). *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05 are considered significantly different when compared to the control group.</p>
Full article ">Figure 3
<p>The effect of Mitomycin C on cell viability and ROS production is abrogated by antioxidants. The cells were either pretreated for 1 h with or without 25 µM imipramine prior to stimulation for 48 h either with or without 0.3 µM mitomycin C (MMC) (<b>A</b>). The data of cell counts are expressed as % of control and are means ± SD (<span class="html-italic">n</span> = 3). *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05 indicate statistical significance when compared to the control group. (<b>B</b>) The cells were incubated for 48 h either with or without 0.3 µM MMC C. The data are expressed in % of the untreated control and are means ± SD of at least three (<span class="html-italic">n</span> = 3) independent experiments. (<b>C</b>) The cells were treated for 48 h with either 0.3 µM MMC C (MMC), 3 mM N-acetyl-Cysteine (NAC), 100 µM Tiron, or combined. The results are expressed as % of control and are means ± SD (<span class="html-italic">n</span> = 3). *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05 are significantly different when compared to the control group.</p>
Full article ">Figure 4
<p>The effect of Mitomycin C on levels of sphingosine, S1P, and sphinganine-1-p. The ASM transfected (ASM) and control transfected (pJK) cells were treated for 48 h either with or without 3 µM mitomycin C (MMC). The results are expressed in % of the untreated control and are means ± SD (<span class="html-italic">n</span> = 3). *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05 indicate statistical significance when compared to the control group.</p>
Full article ">Figure 5
<p>The mRNA expression of various enzymes of the sphingolipid metabolism upon Mitomycin C treatment. The ASM transfected (ASM) and control transfected (pJK) Huh7.5 und HepG2 cells were incubated for 48 h either with or without 3 µM mitomycin C (MMC). The data were obtained by the ΔΔCt method as described in the methods section and are expressed in % of untreated control, shown are means ± SD (<span class="html-italic">n</span> = 3). *** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05 are considered significantly different when compared to the control group.</p>
Full article ">
24 pages, 3599 KiB  
Article
Complementary Treatment of Breast Cancer Cells with Different Metastatic Potential with Iscador Qu in the Presence of Clinically Approved Anticancer Drugs
by Ivan Iliev, Iana Tsoneva, Aleksandrina Nesheva, Galya Staneva, Bozhil Robev, Albena Momchilova and Biliana Nikolova
Curr. Issues Mol. Biol. 2024, 46(11), 12457-12480; https://doi.org/10.3390/cimb46110740 - 5 Nov 2024
Viewed by 582
Abstract
European mistletoe extract (Iscador Qu) has been studied for decades, but it has not ceased to arouse scientific interest. The purpose was to investigate the impact of Iscador Qu on the antiproliferative potential of 11 standard chemotherapeutic agents on two breast cancer cell [...] Read more.
European mistletoe extract (Iscador Qu) has been studied for decades, but it has not ceased to arouse scientific interest. The purpose was to investigate the impact of Iscador Qu on the antiproliferative potential of 11 standard chemotherapeutic agents on two breast cancer cell lines: MCF-7 low-metastatic and MDA-MB-231 high-metastatic and control cell lines (MCF-10A). MTT-dye reduction assay, FACS analysis, and PI staining were utilized. The most promising combinations acting against the MDA-MB-231 cell line were observed upon the simultaneous application of Iscador Qu (80 µg/mL) and Docetaxel, with 4-fold reduction in IC50. An antagonistic effect was found under treatment with Cisplatin and Iscador Qu (1.5-fold increase in IC50). The response of the low-metastatic breast cancer cell line MCF-7 to the tested combinations was different compared to the high-metastatic one. The most pronounced cytotoxic effect was found for the combination of Oxaliplatin and Iscador Qu (20 µg/mL) (5.2-fold IC50 reduction). An antagonistic effect for MCF-7 line was also observed when combinations with Olaparib and Tamoxifen were applied. This in vitro study offers new combinations between Iscador Qu and standard chemotherapeutic agents that hold great promise in establishing breast cancer therapeutic protocols compared to traditional monotherapies. Full article
Show Figures

Figure 1

Figure 1
<p>Antiproliferative activity of Docetaxel in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [nM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and * <span class="html-italic">p</span> &lt; 0.05; n = 6.</p>
Full article ">Figure 2
<p>Antiproliferative activity of Irinotecan in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [μM] from (<b>A</b>–<b>C</b>). * <span class="html-italic">p</span> &lt; 0.05; n = 6.</p>
Full article ">Figure 3
<p>Antiproliferative activity of Cisplatin in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [μM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001; n = 6.</p>
Full article ">Figure 4
<p>Antiproliferative activity of Oxaliplatin in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [µM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and * <span class="html-italic">p</span> &lt; 0.05; n = 6.</p>
Full article ">Figure 5
<p>Antiproliferative activity of 5-Fluorouracyl in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [µM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and ** <span class="html-italic">p</span> &lt; 0.01; n = 6.</p>
Full article ">Figure 6
<p>Antiproliferative activity of Doxorubicin in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and ** <span class="html-italic">p</span> &lt; 0.01; n = 6.</p>
Full article ">Figure 7
<p>Antiproliferative activity of Epirubicin in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and * <span class="html-italic">p</span> &lt; 0.05; n = 6.</p>
Full article ">Figure 8
<p>Antiproliferative activity of Methotrexate in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [nM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001; n = 6.</p>
Full article ">Figure 9
<p>Antiproliferative activity of Olaparib in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [µM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and ** <span class="html-italic">p</span> &lt; 0.01; n = 6.</p>
Full article ">Figure 10
<p>Antiproliferative activity of Ribociclib in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and * <span class="html-italic">p</span> &lt; 0.05; n = 6.</p>
Full article ">Figure 11
<p>Antiproliferative activity of Tamoxifen in combination with two concentrations of Iscador Qu against (<b>A</b>) MCF-10A, (<b>B</b>) MCF-7, and (<b>C</b>) MDA-MB-231. Mean IC<sub>50</sub> ± SD [µM] from (<b>A</b>–<b>C</b>). *** <span class="html-italic">p</span> &lt; 0.001 and * <span class="html-italic">p</span> &lt; 0.05; n = 6.</p>
Full article ">Scheme 1
<p>Chemical structure of all studied 11 anticancer drugs.</p>
Full article ">
13 pages, 597 KiB  
Review
Paclitaxel-Coated Versus Sirolimus-Coated Eluting Balloons for Percutaneous Coronary Interventions: Pharmacodynamic Properties, Clinical Evidence, and Future Perspectives
by Filippo Luca Gurgoglione, Mattia De Gregorio, Giorgio Benatti, Davide Donelli, Luigi Vignali, Emilia Solinas, Iacopo Tadonio, Andrea Denegri, Marco Covani, Gabriella Dallaglio, Bernardo Cortese and Giampaolo Niccoli
Future Pharmacol. 2024, 4(4), 775-787; https://doi.org/10.3390/futurepharmacol4040041 - 2 Nov 2024
Viewed by 1126
Abstract
Drug-coated balloons (DCBs) have emerged as an increasingly valuable option for the treatment of coronary artery disease (CAD). Percutaneous coronary intervention (PCI) with DCBs enables the localized delivery of antiproliferative drugs directly to the target coronary lesion, avoiding the need for permanent scaffold [...] Read more.
Drug-coated balloons (DCBs) have emerged as an increasingly valuable option for the treatment of coronary artery disease (CAD). Percutaneous coronary intervention (PCI) with DCBs enables the localized delivery of antiproliferative drugs directly to the target coronary lesion, avoiding the need for permanent scaffold implantation. Historically, paclitaxel-coated balloons (PCBs) have been the most used device in this context. Paclitaxel interferes with intracellular microtubule function, leading to cell cycle arrest. However, its cytotoxicity at a higher dosage and narrow therapeutic range has raised some safety concerns. To address these issues, sirolimus-coated balloons (SCBs) have been introduced as an alternative. Sirolimus acts as a cytostatic agent with potent anti-inflammatory and antiproliferative properties and is characterized by a wider therapeutic range, potentially offering a safer profile. Several experimental and clinical studies comparing the safety and efficacy of PCBs versus SCBs have yielded mixed results. Recently, a novel DCB (SirPlux Duo), which simultaneously releases both paclitaxel and sirolimus, has been tested in a porcine coronary model with promising results. In this review, we will elucidate the mechanisms of action of paclitaxel and sirolimus, examine contemporary preclinical and clinical evidence comparing PCB and SCB angioplasty, and discuss novel devices that may enhance the safety and efficacy of PCI with DCBs. Full article
Show Figures

Figure 1

Figure 1
<p>Mechanisms of action between paclitaxel and sirolimus. Abbreviations. mTOR: mammalian target of rapamycin.</p>
Full article ">
12 pages, 1575 KiB  
Article
Arene Ruthenium Complexes Specifically Inducing Apoptosis in Breast Cancer Cells
by Adriana Grozav, Thomas Cheminel, Ancuta Jurj, Oana Zanoaga, Lajos Raduly, Cornelia Braicu, Ioana Berindan-Neagoe, Ovidiu Crisan, Luiza Gaina and Bruno Therrien
Inorganics 2024, 12(11), 287; https://doi.org/10.3390/inorganics12110287 - 2 Nov 2024
Viewed by 604
Abstract
Monocationic arene ruthenium complexes (RuL1RuL4) incorporating phenothiazinyl-hydrazinyl-thiazole ligands (L1L4) have been synthesized, characterized and evaluated as anticancer agents. Their cytotoxicity, antiproliferative activity and alteration of apoptotic gene expression were studied on [...] Read more.
Monocationic arene ruthenium complexes (RuL1RuL4) incorporating phenothiazinyl-hydrazinyl-thiazole ligands (L1L4) have been synthesized, characterized and evaluated as anticancer agents. Their cytotoxicity, antiproliferative activity and alteration of apoptotic gene expression were studied on three cancer cell lines, a double positive breast cancer cell line MCF-7 and two triple negative breast cancer cell lines Hs578T and MDA-MB-231. All arene ruthenium complexes were able to reduce the viability of the breast cancer cell lines, with the highest cytotoxicities being recorded for the [(p-cymene)RuL3Cl]+ (RuL3) complex on the MCF-7 (IC50 = 0.019 µM) and Hs578T cell lines (IC50 = 0.095 µM). In the double positive MCF-7 breast cancer cells, the complexes [(p-cymene)RuL1Cl]+ (RuL1) and [(p-cymene)RuL2Cl]+ (RuL2) significantly upregulated pro-apoptotic genes including BAK, FAS, NAIP, CASP8, TNF, XIAP and BAD, while downregulating TNFSF10. In the triple negative breast cancer cell line Hs578T, RuL1 reduced TNFSF-10 and significantly upregulated BAK, CASP8, XIAP, FADD and BAD, while complex RuL2 also increased BAK and CASP8 expression, but had limited effects on other genes. The triple negative MDA-MB-231 cancer cells treated with RuL1 upregulated NOD1 and downregulated p53, while RuL2 significantly downregulated p53, XIAP and TNFSF10, with minor changes in other genes. The significant alterations in the expression of key apoptotic genes suggest that such complexes have the potential to target cancer cells. Full article
(This article belongs to the Special Issue Noble Metals in Medicinal Inorganic Chemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Antiproliferative effects determined from MTT assays after 24 h incubation with <b>RuL1</b>–<b>RuL4</b> on NBC cells (MCF-7, MDA-MS-231, Hs578T) and normal cells (fR2). Log(conc, nM) = Log(concentration of complexes, nM) (mean ± SD, n = 6).</p>
Full article ">Figure 2
<p>(<b>A</b>) Fluorescence microscopy, showing nuclear features after DAPI staining on normal and NBC cell lines after 24 h incubation (40x magnification). Statistical analysis on various cell lines. (<b>B</b>) Normal cell line fR2. (<b>C</b>) Triple negative breast cancer cell line, Hs578T. (<b>D</b>) Double positive breast cancer cell line (data presented as mean ± SD; <span class="html-italic">p</span> * = 0.039 for compound 2, two-side <span class="html-italic">t</span>-test). (<b>E</b>) Triple negative breast cancer cell line, MDA-MB-231 (data presented as mean ± SD; <span class="html-italic">p</span> ** = 0.0047 for <b>RuL<sub>1</sub></b>, <span class="html-italic">p</span> *** = 0.0001 for <b>RuL<sub>2</sub></b>, two-side <span class="html-italic">t</span>-test).</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>) Fluorescence microscopy, showing nuclear features after DAPI staining on normal and NBC cell lines after 24 h incubation (40x magnification). Statistical analysis on various cell lines. (<b>B</b>) Normal cell line fR2. (<b>C</b>) Triple negative breast cancer cell line, Hs578T. (<b>D</b>) Double positive breast cancer cell line (data presented as mean ± SD; <span class="html-italic">p</span> * = 0.039 for compound 2, two-side <span class="html-italic">t</span>-test). (<b>E</b>) Triple negative breast cancer cell line, MDA-MB-231 (data presented as mean ± SD; <span class="html-italic">p</span> ** = 0.0047 for <b>RuL<sub>1</sub></b>, <span class="html-italic">p</span> *** = 0.0001 for <b>RuL<sub>2</sub></b>, two-side <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>Expression profile of selected genes in normal and breast cancer cell lines after incubation with <b>RuL<sub>1</sub></b> and <b>RuL<sub>2</sub></b> for 24 h. (<b>A</b>–<b>D</b>) The heatmap presents genes for breast cancer cell lines (color bars represent gene expression fold change: red color indicates the increased level and green indicates the decreased expression level in treated cells); (<b>A1</b>,<b>B1</b>,<b>C1</b>,<b>D1</b>) represents the STRING network [<a href="#B25-inorganics-12-00287" class="html-bibr">25</a>] for the genes with an altered expression level for genes of at least 1.25-fold increase or decrease with a <span class="html-italic">p</span>-value ≤ 0.05 as effect of <b>RuL<sub>1</sub></b> treatment; (<b>A2</b>,<b>B2</b>,<b>C2</b>,<b>D2</b>) represents the STRING network for the genes with an altered expression level considering the same cut-off values effect of the <b>RuL<sub>2</sub></b> treatment.</p>
Full article ">Scheme 1
<p>Synthesis of [(<span class="html-italic">p</span>-cymene)Ru<b>L</b>Cl]Cl (<b>RuL1</b>–<b>RuL4</b>) from [(<span class="html-italic">p</span>-cymene)RuCl<sub>2</sub>]<sub>2</sub> and the phenothiazinyl-hydrazinyl-thiazole ligands (<b>L1</b>–<b>L4</b>).</p>
Full article ">
20 pages, 4485 KiB  
Article
Synergistic Combination of Quercetin and Mafosfamide in Treatment of Bladder Cancer Cells
by Carmela Spagnuolo, Francesco Mautone, Anna Maria Iole Meola, Stefania Moccia, Giuseppe Di Lorenzo, Carlo Buonerba and Gian Luigi Russo
Molecules 2024, 29(21), 5176; https://doi.org/10.3390/molecules29215176 - 31 Oct 2024
Viewed by 939
Abstract
Bladder cancer, which has a rising incidence, is the 10th most common cancer. The transitional cell carcinoma histotype is aggressive and often current therapies are ineffective. We investigated the anti-proliferative effect of quercetin, a natural flavonoid, in combination with the alkylating agent mafosfamide [...] Read more.
Bladder cancer, which has a rising incidence, is the 10th most common cancer. The transitional cell carcinoma histotype is aggressive and often current therapies are ineffective. We investigated the anti-proliferative effect of quercetin, a natural flavonoid, in combination with the alkylating agent mafosfamide (MFA) on two human bladder cancer cell lines, namely RT112 and J82, representing the progression from low-grade to high-grade tumors, respectively. In both cell types, the combined treatment led to a synergic reduction in cell viability confirmed by a combination index of less than one, though different biological responses were noted. In J82 cells, MFA alone and, to a lesser extent, with quercetin caused cell cycle arrest in the G2/M phase, but only the combined treatment triggered apoptotic cell death. In contrast, in RT112 cells, quercetin induced autophagy, evidenced by the autophagosome formation and the increase in LC-3 lipidation. Interestingly, the synergistic effect was observed only when cells were pre-treated with MFA for 24 h before adding quercetin, not in the reverse order. This suggests that quercetin may help overcome MFA resistance to apoptosis. Although further studies are needed, investigating the combined effects of quercetin and MFA could help elucidate the mechanisms of drug resistance in bladder cancer treatment. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Quercetin enhances the cytotoxic effect of MFA in RT112 and J82 cells. RT112 (<b>a</b>) and J82 (<b>b</b>) cells were treated with mafosfamide (M), quercetin (Q), or their combination, as indicated. Cell viability was evaluated by crystal violet assay after 48 h of treatment. Line graphs represent the mean of n = 3 experiments performed in triplicate (s.d.). Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (#) with respect to untreated cells and <span class="html-italic">p</span> &lt; 0.05 (*) with respect to Q and MFA mono-treatments.</p>
Full article ">Figure 2
<p>Bliss synergy score evaluation. In surface plots, red area indicates combination was judged synergistic more than antagonistic; green indicates combination was judged antagonistic more than synergistic.</p>
Full article ">Figure 3
<p>Quercetin and MFA’s impact on cell cycle in J82 cells. (<b>a</b>) J82 cells were stimulated with quercetin (Q; 10 μM) and mafosfamide (MFA 2.5 μg/mL) alone or in combination for 36 h and cell cycle analysis was performed using Modfit LT software. The average ± s.d. of the data obtained from n = 4 experiments, expressed as the number of cells (% events) present in the various phases of the cell cycle, is reported in the bar graph. Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (*) compared to CTRL. (<b>b</b>) Immunoblotting analysis of cyclin B and cyclin A expression in treated J82 cells. Blots are representative of one out of n = 3 experiments for cyclin B and n = 2 experiments for cyclin A, performed separately. Densitometric analysis is reported in the bar graph on the left and is expressed as the ratio between cyclins and α-tubulin band intensities means ± s.d. Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (*) compared to CTRL (C).</p>
Full article ">Figure 4
<p>Quercetin in association with MFA induces apoptosis in J82 cells. Cells were treated for 36 h with quercetin (Q 10 μM) and mafosfamide (MFA 2.5 μg/mL) alone or in combination. (<b>a</b>) Annexin-V positivity was measured by cytofluorimetric analysis. Dot plots are representative of one out of n = 4 experiments. The proteolytic activity of caspase-3 (<b>b</b>) and caspase-9 (<b>c</b>) (nmol AFC/min/mg protein) was measured after 24 h of treatment. (<b>d</b>) Immunoblot showing the PARP cleavage. The bar graphs represent means ± s.d. derived from n = 3 separate experiments performed in duplicate. Symbols indicate significance: # <span class="html-italic">p</span> &lt; 0.05 compared to CTRL, and <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.005 (**) compared to Q and MFA mono-treatments.</p>
Full article ">Figure 4 Cont.
<p>Quercetin in association with MFA induces apoptosis in J82 cells. Cells were treated for 36 h with quercetin (Q 10 μM) and mafosfamide (MFA 2.5 μg/mL) alone or in combination. (<b>a</b>) Annexin-V positivity was measured by cytofluorimetric analysis. Dot plots are representative of one out of n = 4 experiments. The proteolytic activity of caspase-3 (<b>b</b>) and caspase-9 (<b>c</b>) (nmol AFC/min/mg protein) was measured after 24 h of treatment. (<b>d</b>) Immunoblot showing the PARP cleavage. The bar graphs represent means ± s.d. derived from n = 3 separate experiments performed in duplicate. Symbols indicate significance: # <span class="html-italic">p</span> &lt; 0.05 compared to CTRL, and <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.005 (**) compared to Q and MFA mono-treatments.</p>
Full article ">Figure 5
<p>MFA and Q effect on the cell cycle in RT112 cells. Cells were stimulated with quercetin (Q 10 μM) and mafosfamide (MFA 2.5 μg/mL) alone or in combination for 36 h and cell cycle analysis was performed using Modfit LT software. The average ± s.d. of data obtained from n = 3 experiments, expressed as the number of cells (% events) present in the various phases of the cell cycle, was reported in the bar graph. Symbols indicate significance: * <span class="html-italic">p</span> &lt; 0.05 compared to CTRL.</p>
Full article ">Figure 6
<p>Quercetin induces autophagy in RT112 cells. (<b>a</b>) Representative images of autophagic vacuoles of cells untreated (left) and treated with 10 μM quercetin (Q) for 24 h visualized using a fluorescent microscopy and photographed in FITC/DAPI filters. (<b>b</b>) A representative histogram of CytoID flow cytometry acquisition of cells untreated (violet) and treated with 10 μM quercetin for 24 h (green line); the numbers below indicate means of n = 2 separate experiments ± s.d. (<b>c</b>) Immunoblotting analysis of LC3-I/LC3-II and (<b>d</b>) Beclin1 expression in RT112 cells treated with quercetin as indicated. Blots are representative of one out of n = 2 separate experiments performed. Densitometric analysis is reported in the bar graph on the left and is expressed as the ratio between LC3-II and the α-tubulin band intensities means ± s.d. Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (*) compared to CTRL.</p>
Full article ">Figure 6 Cont.
<p>Quercetin induces autophagy in RT112 cells. (<b>a</b>) Representative images of autophagic vacuoles of cells untreated (left) and treated with 10 μM quercetin (Q) for 24 h visualized using a fluorescent microscopy and photographed in FITC/DAPI filters. (<b>b</b>) A representative histogram of CytoID flow cytometry acquisition of cells untreated (violet) and treated with 10 μM quercetin for 24 h (green line); the numbers below indicate means of n = 2 separate experiments ± s.d. (<b>c</b>) Immunoblotting analysis of LC3-I/LC3-II and (<b>d</b>) Beclin1 expression in RT112 cells treated with quercetin as indicated. Blots are representative of one out of n = 2 separate experiments performed. Densitometric analysis is reported in the bar graph on the left and is expressed as the ratio between LC3-II and the α-tubulin band intensities means ± s.d. Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (*) compared to CTRL.</p>
Full article ">Figure 7
<p>Quercetin induces a protective form of autophagy in RT112 cells. Black bars indicate cells treated for 24 h with MFA, quercetin (Q), or their combination, as indicated; the experimental point represented by gray bars were pre-incubated for 1 h with the autophagic inhibitor chloroquine (CQ 20 μM). Cell viability was evaluated by crystal violet assay. Bar graphs represent the mean of n = 3 experiments performed in triplicate (± s.d.). Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (*) and <span class="html-italic">p</span> &lt; 0.005 (**) compared to respective treatment without CQ; <span class="html-italic">p</span> &lt; 0.05 (#) with respect to Q and MFA mono-treatments.</p>
Full article ">Figure 8
<p>Quercetin enhances sensitivity to MFA treatment in RT112 (<b>a</b>) and J82 cells (<b>b</b>). Cells were treated with mafosfamide (MFA 2.5 μg/mL), quercetin (Q; 10 μM), or their combination, as indicated. Cell viability was evaluated by crystal violet assay after 48 h. Bar graphs represent the mean of n = 2 experiments performed in triplicate (s.d.). Symbols indicate significance: <span class="html-italic">p</span> &lt; 0.05 (*) with respect to Q and MFA mono-treatments.</p>
Full article ">Figure 9
<p>Graphical summary of the different responses to quercetin and MFA treatment in J82 and RT112 cells. Created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>.</p>
Full article ">
16 pages, 1299 KiB  
Article
Synthesis of bis-Chalcones Based on Green Chemistry Strategies and Their Cytotoxicity Toward Human MeWo and A375 Melanoma Cell Lines
by Dorota Olender, Anna Pawełczyk, Anna Leśków, Katarzyna Sowa-Kasprzak, Lucjusz Zaprutko and Dorota Diakowska
Molecules 2024, 29(21), 5171; https://doi.org/10.3390/molecules29215171 - 31 Oct 2024
Viewed by 489
Abstract
Chalcone is an aromatic ketone that forms the central core of many important biological compounds. Chalcone derivatives show various biological activities, especially anti-inflammatory, antibacterial, antioxidant, and anticancer activities, and also inhibit melanoma cell growth. In this study, we synthesized chalcone compounds with bis [...] Read more.
Chalcone is an aromatic ketone that forms the central core of many important biological compounds. Chalcone derivatives show various biological activities, especially anti-inflammatory, antibacterial, antioxidant, and anticancer activities, and also inhibit melanoma cell growth. In this study, we synthesized chalcone compounds with bis-chalcone’s chemical structure under microwave (MW) and microwave–ultrasound (MW-US) conditions and compared them to chalcones produced using the classical synthesis method. All bis-chalcones were synthesized with terephthalaldehyde and an appropriate aromatic ketone as substrates in Claisen–Schmidt condensation. All the obtained compounds were tested regarding their roles as potential anticancer agents. The cytotoxic effect of the bis-chalcones against human MeWo and A375 melanoma cell lines was investigated through colorimetric MTT and SRB assays. The data were analyzed statistically. In the case of the synthesis of bis-chalcones, it was determined that the use of green conditions supported by the MW or MW-US factors led to an increase in the yield of the final products and a reduction in the reaction time compared to the classic method. The biological results showed the high cytotoxic effect of bis-chalcones. The present results show the compounds’ high antiproliferative and cytotoxic potential, especially for the two selected bis-chalcone derivatives (3b and 3c), in particular, at concentrations of 50 μM–200 μM at 24, 48 h, and 72 h of incubation. The use of MW and US for the synthesis of bis-chalcones significantly improved the process compared to the classical method. The derivatives containing two hydroxy and two methoxy groups were the most effective against the tested cancer cells. Full article
Show Figures

Figure 1

Figure 1
<p>The results of the MTT assay on the viability of MeWo cells incubated with <span class="html-italic">bis</span>-chalcones <b>3a</b>–<b>3d</b> and <b>ChO</b> for (<b>A</b>) 24 h, (<b>B</b>) 48 h, and (<b>C</b>) 72 h. Descriptive data are presented as the mean ± SD. The results of the ANOVA analysis are described in figures (<b>A</b>–<b>C</b>). Statistically significant differences versus the control cells are marked with an asterisk (* <span class="html-italic">p</span> ≤ 0.0001). The horizontal bold line represents the viability of the control (0.2% DMSO-treated) cell.</p>
Full article ">Figure 2
<p>The results of the SRB assay on the viability of MeWo cells incubated with chalcones <b>3a</b>–<b>3d</b> and <b>ChO</b> for (<b>A</b>) 24 h, (<b>B</b>) 48 h, and (<b>C</b>) 72 h. Descriptive data are presented as the mean ± SD. The results of the ANOVA analysis are described in figures (<b>A</b>–<b>C</b>). Statistically significant differences versus the control cells are marked with an asterisk (* <span class="html-italic">p</span> ≤ 0.0001). The horizontal bold line represents the viability of the control (0.2% DMSO-treated) cells.</p>
Full article ">Figure 3
<p>The results of the MTT assay on the viability of A375 cells incubated with chalcones <b>3a</b>–<b>3d</b> and <b>ChO</b> for (<b>A</b>) 24 h, (<b>B</b>) 48 h, and (<b>C</b>) 72 h. Descriptive data are presented as the mean ± SD. The results of the ANOVA analysis are described in figures (<b>A</b>–<b>C</b>). Statistically significant differences versus the control cells are marked with an asterisk (* <span class="html-italic">p</span> ≤ 0.0001). The horizontal bold line represents the viability of the control (0.2% DMSO-treated) cells.</p>
Full article ">Figure 4
<p>The results of the SRB assay on the viability of A375 cells incubated with chalcones <b>3a</b>–<b>3d</b> and <b>ChO</b> for (<b>A</b>) 24 h, (<b>B</b>) 48 h, and (<b>C</b>) 72 h. Descriptive data are presented as the mean ± SD. The results of the ANOVA analysis are described in figures (<b>A</b>–<b>C</b>). Statistically significant differences versus the control cells are marked with an asterisk (* <span class="html-italic">p</span> ≤ 0.0001). The horizontal bold line represents the viability of the control (0.2% DMSO-treated) cells.</p>
Full article ">Scheme 1
<p>Synthesis of <span class="html-italic">bis</span>-chalcones <b>3a</b>–<b>3d</b> under the classical method, alternative microwave, and microwave–ultrasound-assisted synthesis conditions.</p>
Full article ">
26 pages, 2890 KiB  
Review
The Pivotal Role of One-Carbon Metabolism in Neoplastic Progression During the Aging Process
by Avisek Majumder, Shabana Bano and Kasturi Bala Nayak
Biomolecules 2024, 14(11), 1387; https://doi.org/10.3390/biom14111387 - 31 Oct 2024
Viewed by 773
Abstract
One-carbon (1C) metabolism is a complex network of metabolic reactions closely related to producing 1C units (as methyl groups) and utilizing them for different anabolic processes, including nucleotide synthesis, methylation, protein synthesis, and reductive metabolism. These pathways support the high proliferative rate of [...] Read more.
One-carbon (1C) metabolism is a complex network of metabolic reactions closely related to producing 1C units (as methyl groups) and utilizing them for different anabolic processes, including nucleotide synthesis, methylation, protein synthesis, and reductive metabolism. These pathways support the high proliferative rate of cancer cells. While drugs that target 1C metabolism (like methotrexate) have been used for cancer treatment, they often have significant side effects. Therefore, developing new drugs with minimal side effects is necessary for effective cancer treatment. Methionine, glycine, and serine are the main three precursors of 1C metabolism. One-carbon metabolism is vital not only for proliferative cells but also for non-proliferative cells in regulating energy homeostasis and the aging process. Understanding the potential role of 1C metabolism in aging is crucial for advancing our knowledge of neoplastic progression. This review provides a comprehensive understanding of the molecular complexities of 1C metabolism in the context of cancer and aging, paving the way for researchers to explore new avenues for developing advanced therapeutic interventions for cancer. Full article
(This article belongs to the Special Issue Homocysteine and H2S in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>This cartoon diagram illustrates the enzymatic reactions and compartmentalization of 1C metabolism. Cells utilize 1C units from methionine, serine, and glycine to produce various compounds that work as building blocks for the biosynthesis of nucleic acids and proteins, regulate methylation reactions, and help maintain a cellular redox status. Serine and glycine can enter cells from the outside or be synthesized de novo from the glycolysis intermediate, 3-phosphoglycerate (3-PG). Methionine and folate always come from diet, are carried over the methionine cycle, and can operate in both the cytoplasm and mitochondria (all abbreviations are given at the end).</p>
Full article ">Figure 2
<p>One-carbon (1C) metabolism through the methionine cycle and folate cycle and its utilization in other closely linked pathways (like polyamine synthesis and the transsulfuration pathway) (all abbreviations are given at the end).</p>
Full article ">Figure 3
<p>Schematic representation showing the inputs of 1C units from dietary sources and their processing and utilization in different biosynthesis processes as output. In this process, methionine, glucose, serine, and glycine can be used as inputs to carry over 1C metabolism. Serine can be obtained from the diet or produced from glucose via the de novo process. Folate from the diet is converted to THF, which accepts a 1C unit during the folate cycle. Then, serine is broken down into glycine, producing a 1C unit which combines with THF to form methylene-THF. Methionine from the diet can be used to produce SAM, which is subsequently used for methylation reactions and cellular antioxidant production. Different outputs from 1C metabolism also act as building blocks for the cellular biosynthesis of DNA, RNA, and protein.</p>
Full article ">Figure 4
<p>Regulation of redox homeostasis by glutathione (GSH). The enzyme glutathione peroxidase (GPx) uses glutathione (GSH) as a substrate to produce GSSG (oxidized glutathione) by utilizing the thiol (-SH) group of its cysteine residue to interact with reactive oxygen species (ROS) or electrophiles, whereas the enzyme glutathione reductase (GR) efficiently converts GSSG back to GSH with the help of NADPH, thereby preserving the antioxidant capacity of cells.</p>
Full article ">Figure 5
<p>Common transcription factors which are involved in 1C metabolism as well as cancer progression. This visual illustrates the enzymes involved in one-carbon metabolism and their regulating transcription factors, which also play a role in various stages of cancer progression. These transcription factors are organized into two main categories: serine synthesis, represented by different-colored star marks, and nucleotide synthesis, indicated by different-colored triangle marks. All the abbreviations are listed at the end of this article in the abbreviation section.</p>
Full article ">Figure 6
<p>Antiproliferative responses of damaged cells. This cartoon illustrates how damaged cells can become apoptotic, enter senescence, or continue replicating. If these antiproliferative responses are absent or fail, a cancerous lesion may be formed, further proliferating to form malignant cells.</p>
Full article ">Figure 7
<p>Schematic representation of the fate of cells undergoing senescence and apoptosis upon oncogenic insults. In response to various stressors, normal cells with pre-neoplastic lesions may undergo senescence or apoptosis with the final goal of removing the pre-neoplastic cells. However, in the absence of these antiproliferative responses, pre-neoplastic cells continue to grow and acquire additional oncogenic mutations. At this step, senescence can be reactivated, or it can progress toward malignant transformation.</p>
Full article ">
16 pages, 2076 KiB  
Article
Chemical Composition and Bioactivity Dataset Integration to Identify Antiproliferative Compounds in Phyllanthus Plants
by Luis Diaz, Taylor H. Díaz-Herrera and Ericsson Coy-Barrera
Pharmaceutics 2024, 16(11), 1381; https://doi.org/10.3390/pharmaceutics16111381 - 27 Oct 2024
Viewed by 609
Abstract
Background/Objectives: Phyllanthus species are renowned in traditional medicine for their diverse therapeutic properties, including potential anticancer activities. This study explored the antiproliferative potential of six Phyllanthus species by integrating chemical composition with bioactivity assays to identify key antiproliferative compounds. Methods: The integration of [...] Read more.
Background/Objectives: Phyllanthus species are renowned in traditional medicine for their diverse therapeutic properties, including potential anticancer activities. This study explored the antiproliferative potential of six Phyllanthus species by integrating chemical composition with bioactivity assays to identify key antiproliferative compounds. Methods: The integration of liquid chromatography–mass spectrometry (LC-MS)-based chemical composition data with antiproliferative activity against three cancer cell lines—PC-3 (prostate adenocarcinoma), SiHa (cervical carcinoma), and A549 (lung carcinoma)—as well as a normal mouse fibroblast line (L929) was performed by covariate analysis. These compounds were subsequently isolated and structurally characterized using spectroscopic methods. Results: Through covariate statistics, seven m/z features were found to be plausible active compounds, and after isolation, they were related to butyrolactone and arylnaphthalide lignans. Among the active isolates, an unreported compound, (+)-phyllanlathyrin 6, was discovered in the aerial part of Phyllanthus lathyroides. The isolated compounds exhibited moderate to good antiproliferative activity (IC50 < 20 µM) with selectivity to SiHa, validating the covariate-based identification approach. Conclusions: These findings highlight the potential of Phyllanthus species as sources of novel anticancer agents, with specific arylnaphthalide lignans showing promising cytotoxic effects that could be further developed into therapeutic leads. Additionally, this study underscores the value of combining advanced analytical techniques with bioactivity testing to uncover bioactive compounds from natural sources. The results contribute to the growing body of evidence supporting the therapeutic relevance of Phyllanthus species and provide a foundation for future drug development efforts targeting cancer treatment. Full article
Show Figures

Figure 1

Figure 1
<p>LC-MS-based differentiation of <span class="html-italic">Phyllanthus</span> aerial part-derived extracts from six plants and five technical replicates per plant. <span class="html-italic">Pcb</span> = <span class="html-italic">P. caribaeus</span>; <span class="html-italic">Pcr</span> = <span class="html-italic">P. caroliniensis</span>; <span class="html-italic">Pl</span> = <span class="html-italic">P. lathyroides</span>; <span class="html-italic">Pm = P. madeirensis</span>; <span class="html-italic">Ps = P. salviifolius</span>; <span class="html-italic">Pu = P. urinaria</span>. (<b>a</b>) Heat map depicting the chemical distribution based on the intensity of the top 100 most contrasting <span class="html-italic">m</span>/<span class="html-italic">z</span> features detected across <span class="html-italic">Phyllanthus</span> aerial part-derived extracts. Each column represents the average levels (<span class="html-italic">n</span> = 5) per plant extract, and each colored cell corresponds to the autoscaled intensity of a detected <span class="html-italic">m/z</span> feature, with the color scale ranging from dark red (high intensity) to dark blue (low intensity). (<b>b</b>) Three-dimensional score plot derived from sparse partial least square discriminant analysis (sPLS-DA), showing the separation of samples based on the three first principal components. Plant extract grouping was used as a categorical variable, explaining 66.2% of the variance.</p>
Full article ">Figure 2
<p>Integration of chemical and bioactivity datasets (i.e., feature-based chemical composition and antiproliferative activity) for <span class="html-italic">Phyllanthus</span> aerial part extracts using single-<span class="html-italic">Y</span> orthogonal partial least squares (OPLS) analysis. The IC<sub>50</sub> values, represented as the continuous <span class="html-italic">Y</span>-variable, are depicted on a color scale (red = 250 µg/mL; aquamarine = 0 µg/mL). (<b>a</b>) Scores plot. (<b>b</b>) OPLS-derived <span class="html-italic">S</span>-plot. (<b>c</b>) OPLS-derived variable importance in the projection (VIP) plot.</p>
Full article ">Figure 3
<p>Structures of isolated compounds after statistical pattern recognition from <span class="html-italic">Phyllanthus</span> aerial parts. HMBC correlations in compound <b>6</b>.</p>
Full article ">
19 pages, 11956 KiB  
Article
Synthesis of New Pyrazolo[3,4-d]pyrimidine Derivatives: NMR Spectroscopic Characterization, X-Ray, Hirshfeld Surface Analysis, DFT, Molecular Docking, and Antiproliferative Activity Investigations
by Mohamed El Hafi, El Hassane Anouar, Sanae Lahmidi, Mohammed Boulhaoua, Mohammed Loubidi, Ashwag S. Alanazi, Insaf Filali, Mohamed Hefnawy, Lhoussaine El Ghayati, Joel T. Mague and El Mokhtar Essassi
Molecules 2024, 29(21), 5020; https://doi.org/10.3390/molecules29215020 - 24 Oct 2024
Viewed by 1099
Abstract
Four new pyrazolo[3,4-d]pyrimidines (P1P4) were successfully synthesized in good relative yields by reacting 3-methyl-1-phenyl-1H-pyrazolo[3,4-d]pyrimidin-4-ol with various alkylating agents (methyl iodide, propargyl bromide, and phenacyl bromide) at room temperature in DMF solvent, employing liquid–solid phase transfer [...] Read more.
Four new pyrazolo[3,4-d]pyrimidines (P1P4) were successfully synthesized in good relative yields by reacting 3-methyl-1-phenyl-1H-pyrazolo[3,4-d]pyrimidin-4-ol with various alkylating agents (methyl iodide, propargyl bromide, and phenacyl bromide) at room temperature in DMF solvent, employing liquid–solid phase transfer catalysis. The P1P4 structures were elucidated using NMR spectroscopy and X-ray diffraction. Intermolecular interactions in P1P4 were analyzed via Hirshfeld surface analysis and 2D fingerprint plots. Geometrical parameters were accurately modeled by DFT calculations using the B3LYP hybrid functional combined with a 6–311++G(d,p) basis set. The antiproliferative activity of P1P4 towards colorectal carcinoma (HCT 116), human hepatocellular carcinoma (HepG2), and human breast cancer (MCF-7) cell lines, along with one normal cell line (WI38) was investigated using the MTT assay and sunitinib as a reference. Compounds P1 and P2 exhibited antiproliferative activities comparable to the reference drug towards all tested cells, with an IC50 range of 22.7–40.75 µM. Both compounds also showed high selectivity indices and minimal cytotoxic effects on the normal cell line. Molecular docking revealed that the significant antiproliferative activity may attributed to the number and type of intermolecular hydrogen bonding established between pyrazolo[3,4-d]pyrimidines and DNA topoisomerase, a common target for various anticancer agents. Full article
(This article belongs to the Section Organic Chemistry)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of reported pyrazolo[3,4-<span class="html-italic">d</span>]pyrimidine derivatives.</p>
Full article ">Figure 2
<p>2D structures (<b>left</b>) and perspective views of <b>P1</b>–<b>P4</b> (<b>right</b>). The dashed line in <b>P3</b> represents the intramolecular hydrogen bond.</p>
Full article ">Figure 3
<p>A portion of one layer in <b>P1</b> projected onto <math display="inline"><semantics> <mrow> <mfenced separators="|"> <mrow> <mn>10</mn> <mover accent="true"> <mrow> <mn>3</mn> </mrow> <mo>¯</mo> </mover> </mrow> </mfenced> <mo>,</mo> </mrow> </semantics></math> with the b-axis horizontal and running from left to right. N—H···O hydrogen bonds and C—H···π(ring) interactions are depicted, respectively, by violet and green dashed lines. Non-interacting hydrogen atoms are omitted for clarity.</p>
Full article ">Figure 4
<p>Elevation view of the packing in <b>P2</b> seen parallel to (<math display="inline"><semantics> <mrow> <mn>10</mn> <mover accent="true"> <mrow> <mn>1</mn> </mrow> <mo>¯</mo> </mover> </mrow> </semantics></math>) with C—H···N hydrogen bonds and C—H···π(ring) interactions depicted, respectively, by light blue and green dashed lines.</p>
Full article ">Figure 5
<p>A portion of two chains in <b>P3</b> viewed along the <span class="html-italic">a</span>-axis direction with C—H···O and C—H···N hydrogen bonds depicted, respectively, by black and light blue dashed lines. The π-stacking interactions are depicted by orange dashed lines.</p>
Full article ">Figure 6
<p>A portion of one chain of <b>P4</b> view along the <span class="html-italic">b</span>-axis. Dashed lines C—H···O represent hydrogen intermolecular bonding.</p>
Full article ">Figure 7
<p>The optimized geometries of <b>P1</b>–<b>P4</b> (<b>left</b>) and their superposition with X-ray-generated ones (<b>right</b>, the green color corresponds to the optimized geometry, and the red color corresponds to the X-ray-generated structure).</p>
Full article ">Figure 8
<p>The d<sub>norm</sub> surfaces for viewing hydrogen bonding interactions in <b>P1</b>–<b>P4</b> crystals.</p>
Full article ">Figure 8 Cont.
<p>The d<sub>norm</sub> surfaces for viewing hydrogen bonding interactions in <b>P1</b>–<b>P4</b> crystals.</p>
Full article ">Figure 9
<p>2D fingerprints of the highest intercontacts in <b>P1</b>–<b>P4</b>.</p>
Full article ">Figure 9 Cont.
<p>2D fingerprints of the highest intercontacts in <b>P1</b>–<b>P4</b>.</p>
Full article ">Figure 10
<p>Bar representation of the in vitro antiproliferative activity of <b>P1</b>–<b>P4</b> and sunitinib against HCT 116, HepG2, and MCF-7 cancer cell lines, and one normal cell line WI38.</p>
Full article ">Figure 11
<p>2D binding affinities of <b>P1</b>–<b>P4</b> into the DNA topoisomerase binding site.</p>
Full article ">Figure 11 Cont.
<p>2D binding affinities of <b>P1</b>–<b>P4</b> into the DNA topoisomerase binding site.</p>
Full article ">Figure 12
<p>3D binding affinities of <b>P1</b>–<b>P4</b> into the DNA topoisomerase binding site.</p>
Full article ">Scheme 1
<p>Synthesis path of <b>P1</b>–<b>P4</b>.</p>
Full article ">Scheme 2
<p>Tautomerism of compound <b>P1</b>.</p>
Full article ">
11 pages, 3215 KiB  
Article
Napabucasin Inhibits Proliferation and Migration of Glioblastoma Cells (U87) by Regulating JAK2/STAT3 Signaling Pathway
by İlker Ünlü, İlhan Özdemir and Mehmet Cudi Tuncer
Medicina 2024, 60(10), 1715; https://doi.org/10.3390/medicina60101715 - 19 Oct 2024
Viewed by 763
Abstract
Background and Objectives: Napabucasin (NP) was discovered as a natural compound that suppresses cancer stemness by inhibiting the signal transducer and activator of the transcription 3 (STAT3) signaling pathway. In this study, the anti-proliferative and apoptotic effects of NP and the chemotherapy [...] Read more.
Background and Objectives: Napabucasin (NP) was discovered as a natural compound that suppresses cancer stemness by inhibiting the signal transducer and activator of the transcription 3 (STAT3) signaling pathway. In this study, the anti-proliferative and apoptotic effects of NP and the chemotherapy agent doxorubicin (DX), a natural compound, on glioblastoma cells (U87) were investigated. Materials and Methods: In this study, the effects of NP and DX on cell viability on the glioblastoma U87 cell line were determined by MTT test. Expressions of Jak2/Stat3 genes were examined by qRT-PCR. Apoptosis was evaluated by Hoescht 33258 staining. Moreover, NP, its antagonistic–synergistic effects and IC50 doses of the combined treatment of DX were determined. Results: Napabucacin and doxorubicin were found to inhibit glioblastoma U87 cell proliferation. It was determined that NP applied in the range of 0.3–1 µM and its combination with DX killed almost all of the glioblastoma cells in 48 h of application. Additionally, it was observed that Jak2/Stat3 expressions downregulated. Conclusions: These results show that NP suppresses the proliferation of glioblastoma cells. It was shown that the combination of NP and DX can prevent invasion of the U87 cell line due to its Jak2/Stat3 inhibitory effect. Since it can suppress Jak2/Stat3, an important cancer cell proliferation pathway in glioblastoma, the combination of NP and DX can be used as an alternative treatment agent. But no synergistic effect of NP and DX on the U87 cells of the glioblastoma cell line was observed. Full article
(This article belongs to the Section Genetics and Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Effect of NP and DX at different concentrations on the survival rate of U87 cells for 48 h (a: <span class="html-italic">p</span> &gt; 0.05, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001, *** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>% Cell viability determined by MTT test of application of 64 different doses of DX and NP in U87 cell lines with dosing square starting from 10 µM concentration for 48 h.</p>
Full article ">Figure 3
<p>NP and DX for 48 h to determine the antagonistic and synergistic effect. (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">Figure 4
<p>Contour view of the antagonistic or synergistic effect of NP and DX applied to U87 cell line.</p>
Full article ">Figure 5
<p>Vehicle control and U87 glioblastoma treated with NP IC<sub>50</sub>: 1 μM, DX IC<sub>50</sub>: 0.7 μM, NP + DX (1 + 0.7 µM) for 48 h. Cell morphology, nuclear structure and apoptotic body formation in cell populations (×20 magnification).</p>
Full article ">Figure 6
<p>Relative fold increase values of Jak2 gene expressions 48 h after single and combined application of NP and DX in U87 cell line (data were normalized with β-actin and GAPDH mRNA levels by multiple control method, n = 6, data mean ± SE), * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 7
<p>Relative fold increase values of STAT3 gene expressions 48 h after single and combined application of NP and DX in U87 cell line (data were normalized with β-actin and GAPDH mRNA levels by multiple control method, n = 6, data mean ± SE), * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 8
<p>Interaction between various genes of glioblastoma.</p>
Full article ">Figure 9
<p>Enrichment analysis of the 150 common compound targets. Ninety of these targets are directly related to cancer signaling pathways.</p>
Full article ">
23 pages, 6971 KiB  
Article
Antiproliferative Effects of Naja anchietae and Naja senegalensis Venom Peptides on Glioblastoma Cell Lines
by Yasmine Boughanmi, Caroline Berenguer-Daizé, Marielle Balzano, Hend Mosrati, Maxime Moulard, Pascal Mansuelle, Patrick Fourquet, Franck Torre, Harold de Pomyers, Didier Gigmes, Lhoucine Ouafik and Kamel Mabrouk
Toxins 2024, 16(10), 433; https://doi.org/10.3390/toxins16100433 - 10 Oct 2024
Viewed by 749
Abstract
This study explores the potential of natural bioactive peptides from animal venoms as targeted anti-cancer agents with reduced toxicity. Initially, we screened a broad collection of animal venoms for their antiproliferative activity against cancer cell lines. From this collection, we selected venoms from [...] Read more.
This study explores the potential of natural bioactive peptides from animal venoms as targeted anti-cancer agents with reduced toxicity. Initially, we screened a broad collection of animal venoms for their antiproliferative activity against cancer cell lines. From this collection, we selected venoms from Naja anchietae and Naja senegalensis due to their promising activity. Utilizing reverse- phase high-performance liquid chromatography (RP HPLC), mass spectrometry (MALDI-TOF MS and MALDI-TOF TOF MSMS), and Edman degradation sequencing, we isolated and characterized three peptides named CTNanc1, CTNanc2, and CTNanc3 from Naja anchietae, and three others named CTNsen1, CTNsen2, and CTNsen3 from Naja senegalensis, each with a molecular weight of around 7 kDa. These purified peptides demonstrated inhibition of U87 glioblastoma cell proliferation, but not of U251 and T98G cells, in cell viability assays. To assess the impact of these treatments on cell viability, apoptosis, and necrosis, flow cytometry assays were conducted on U87 cells at 72 h. The results showed a decrease in cell viability and an increase in dead cells, suggesting that the treatments not only promote apoptosis, but may also lead to increased necrosis or late-stage apoptosis as the exposure time increases. These findings suggest that these peptides could be developed as leads for cancer therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Preparative HPLC chromatogram of <span class="html-italic">Naja anchietae</span> venom using gradient 0–70% for 70 min at 214 nm a C18 100 Å column. Fractions were collected at intervals of every 0.1 min, corresponding to a flow rate of 6 mL per minute. The region containing the active peptides is highlighted with a red square in the figure. These peptides were eluted between 40 and 52 min, which corresponds to an elution percentage of approximately 30%.</p>
Full article ">Figure 2
<p>Analytical HPLC chromatogram of the isolated peptides from <span class="html-italic">Naja anchietae</span> at 214 nm on a C18 100 Å column.</p>
Full article ">Figure 3
<p>Mass spectrum of CTNanc3 using MALDI-ToF.</p>
Full article ">Figure 4
<p>Preparative HPLC chromatogram of <span class="html-italic">Naja anchietae</span> venom using gradient 20–60% for 120 min at 214 nm on a C18 300 Å column. Fractions were collected at intervals of every 0.1 min, corresponding to a flow rate of 6 mL per minute. The region containing the active peptides is highlighted with a red square in the figure. These peptides were eluted between 80 and 120 min, which corresponds to an elution percentage of approximately 37%.</p>
Full article ">Figure 5
<p>Preparative HPLC chromatogram of <span class="html-italic">Naja senegalensis</span> venom using gradient 20–60% for 120 min at 214 nm on a C18 300 Å column. Fractions were collected at intervals of every 0.1 min, corresponding to a flow rate of 6 mL per minute. The region containing the active peptides is highlighted with a red square in the figure. These peptides were eluted between 90 and 120 min, which corresponds to an elution percentage of approximately 40%.</p>
Full article ">Figure 6
<p>Analytical HPLC chromatogram of the isolated peptides from <span class="html-italic">Naja senegalensis</span> at 214 nm on a C18 100 Å column.</p>
Full article ">Figure 7
<p>Mass spectrum of peptides CTNsen1 (Aa), CTNsen2 (Ab), CTNsen3 (Ac), CTNanc1 (Bd), CTNanc2 (Be), and CTNanc3 (Bf) using MALDI-ToF.</p>
Full article ">Figure 8
<p>Comparative analysis of CTNsen3, CTNanc3, and Cytotoxin 2 from <span class="html-italic">Naja nivea</span> sequences. The differences are in bold and cysteines forming disulfide bridges are highlighted in yellow.</p>
Full article ">Figure 9
<p>Antiproliferative activity of CTNsen1 (<b>a</b>), CTNsen2 (<b>b</b>), CTNsen3 (<b>c</b>), CTNanc1 (<b>d</b>), CTNanc2 (<b>e</b>), and CTNanc3 (<b>f</b>) from <span class="html-italic">Naja senegalensis</span> and <span class="html-italic">Naja anchietae</span>, respectively. These results were obtained using an MTT assay on U87 cells after 72 h of treatment with a series of increasing concentrations (1 × 10<sup>−</sup>⁷, 0.5 × 10<sup>−6</sup>, 1 × 10<sup>−</sup>⁶, 0.5 × 10<sup>−5</sup>, 1 × 10<sup>−</sup>⁵, 0.5 × 10<sup>−4</sup>, and 1 × 10<sup>−</sup>⁴ Molar) of peptides. The OD was measured at 570 nm.</p>
Full article ">Figure 10
<p>Antiproliferative activity of CTNsen1 (<b>a</b>), CTNsen2 (<b>b</b>), CTNsen3 (<b>c</b>), CTNanc1 (<b>d</b>), CTNanc2 (<b>e</b>), and CTNanc3 (<b>f</b>) from <span class="html-italic">Naja senegalensis</span> and <span class="html-italic">Naja anchietae</span> respectively. These results were obtained using an MTT assay on U251 cells after 72 h of treatment with a series of increasing concentrations (1 × 10<sup>−</sup>⁷, 0.5 × 10<sup>−6</sup>, 1 × 10<sup>−</sup>⁶, 0.5 × 10<sup>−5</sup>, 1 × 10<sup>−</sup>⁵, 0.5 × 10<sup>−4</sup>, and 1 × 10<sup>−4</sup> Molar) of peptides. The OD was measured at 570 nm.</p>
Full article ">Figure 11
<p>Antiproliferative activity of CTNsen1 (<b>a</b>), CTNsen2 (<b>b</b>), CTNsen3 (<b>c</b>), CTNanc1 (<b>d</b>), CTNanc2 (<b>e</b>), and CTNanc3 (<b>f</b>) from <span class="html-italic">Naja senegalensis</span> and <span class="html-italic">Naja anchietae</span>, respectively. These results were obtained using an MTT assay on T98G cells after 72 h of treatment with a series of increasing concentrations (1 × 10<sup>−</sup>⁷, 0.5 × 10<sup>−6</sup>, 1 × 10<sup>−</sup>⁶, 0.5 × 10<sup>−5</sup>, 1 × 10<sup>−</sup>⁵, 0.5 × 10<sup>−4</sup>, and 1 × 10<sup>−</sup>⁴ Molar) of peptides. The OD was measured at 570 nm.</p>
Full article ">Figure 12
<p>Cytotoxic activity of CTNsen1 (<b>a</b>), CTNsen2 (<b>b</b>), CTNsen3 (<b>c</b>), CTNanc1 (<b>d</b>), CTNanc2 (<b>e</b>), and CTNanc3 (<b>f</b>) from <span class="html-italic">Naja senegalensis</span> and <span class="html-italic">Naja anchietae</span>, respectively. These results were obtained using an MTT assay on HUVEC cells after 72 h of treatment with a series of increasing concentrations (1 × 10<sup>−</sup>⁷, 0.5 × 10<sup>−6</sup>, 1 × 10<sup>−</sup>⁶, 0.5 × 10<sup>−5</sup>, 1 × 10<sup>−</sup>⁵, 0.5 × 10<sup>−4</sup>, and 1 × 10<sup>−</sup>⁴ Molar) of peptides. The OD was measured at 570 nm.</p>
Full article ">Figure 13
<p>Antiproliferative activity of reduced and alkylated CTNsen1 (<b>a</b>) and CTNanc1 (<b>b</b>). These results were obtained using an MTT assay on U87 cells after 72 h of treatment with a series of increasing concentrations (1 × 10<sup>−</sup>⁷, 0.5 × 10<sup>−6</sup>, 1 × 10<sup>−</sup>⁶, 0.5 × 10<sup>−5</sup>, 1 × 10<sup>−</sup>⁵, 0.5 × 10<sup>−4</sup>, and 1 × 10<sup>−</sup>⁴ Molar) of peptides. The OD was measured at 570 nm.</p>
Full article ">Figure 14
<p>Flow cytometry gating strategy: representative example using untreated U87 cells. Doublets and debris are excluded (<b>A</b>); the first gating strategy identifies living cells (PI-negative and Annexin V-negative), apoptotic cells (Annexin V-positive and PI-negative), and dead cells (Annexin V-positive and PI-positive) (<b>B</b>); the second gating assesses the intensity of Annexin V fluorescence and the percentage of Annexin V-positive cells, Annexin V-negative and positive cell subsets are colored in pink and orange, respectively (<b>C</b>).</p>
Full article ">Figure 15
<p>Dot plots analysis of U87 cell responses to cytotoxin peptides over 72 h by flow cytometry. Cells were labeled with Annexin-V and Propidium iodide (PI). Cells that did not undergo treatment served as the negative control, Annexin V-negative and positive cell subsets are colored in pink and orange, respectively. Annexin V-negative and positive cell subsets are colored in pink and orange, respectively on histograms.</p>
Full article ">Figure A1
<p>Antiproliferative activity of different animal venoms. These results were obtained using an MTT assay on MCF7 cells after 72 h of treatment with 3 and 15 µg/mL. The OD was measured at 570 nm after 72 h of treatment. The abbreviations include Na: <span class="html-italic">Naja anchietae</span>; Vg: <span class="html-italic">Viridovipera gumprechti</span>; Vk: <span class="html-italic">Vipera kaznakovi</span>; Tv: <span class="html-italic">Trimeresurus vogeli</span>; Tt: <span class="html-italic">Trimeresurus trigonocephalus</span>; Ach: <span class="html-italic">Atheris chlorechis</span>; Ath ni: <span class="html-italic">Atheris nitschei nitschei</span>; Nox: <span class="html-italic">Naja oxiana</span>; Np: <span class="html-italic">Naja philippinensis</span>; At sq: <span class="html-italic">Atheris squamigera</span>; Nhl: <span class="html-italic">Naja haje legionis</span>; Por Oph: <span class="html-italic">Porthidium ophryomegas;</span> Ph: <span class="html-italic">Parias hageni</span>; Ns: <span class="html-italic">Naja senegalensis</span>.</p>
Full article ">Figure A2
<p>Antiproliferative activity of CTNsen1, CTNsen2, CTNsen3, CTNanc1, CTNanc2, and CTNanc3 from <span class="html-italic">Naja senegalensis</span> (<b>b</b>) and <span class="html-italic">Naja anchietae</span> (<b>a</b>), respectively. These results were obtained using an MTT assay on MCF7 cells after 72 h of treatment at 4 µg and 20 µg of peptide. Na and Ns here were used as a positive control at two concentrations (3 µg and 15 µg). The OD was measured at 570 nm.</p>
Full article ">
19 pages, 3182 KiB  
Article
Deferasirox’s Anti-Chemoresistance and Anti-Metastatic Effect on Non-Small Cell Lung Carcinoma
by Yamixa Delgado, Anamaris Torres-Sanchez, Daraishka Perez, Grace Torres, Sthephanie Estrada, Natalia Ortiz Alvelo, Jaisy Vega, Laurie Santos, Aracelis Torres, Bismark A. Madera and Yancy Ferrer-Acosta
Biomedicines 2024, 12(10), 2272; https://doi.org/10.3390/biomedicines12102272 - 7 Oct 2024
Viewed by 1342
Abstract
Clinically approved iron chelators, originally designed to address iron overload disorders, have emerged as potential anticancer agents. Deferasirox (Def), a tridentate iron chelator, has demonstrated antiproliferative effects in cancer. Background/Objectives: This study aims to elucidate the mechanism of action of Def and [...] Read more.
Clinically approved iron chelators, originally designed to address iron overload disorders, have emerged as potential anticancer agents. Deferasirox (Def), a tridentate iron chelator, has demonstrated antiproliferative effects in cancer. Background/Objectives: This study aims to elucidate the mechanism of action of Def and its impact on non-small cell lung carcinoma (NSCLC). Methods: NSCLC A549 cells were treated with Def to assess cytotoxicity, the effect on nuclear and mitochondrial pathways, and iron-containing proteins and genes to evaluate anti-metastasis and chemoresistance. A lung carcinoma mouse model was used for in vivo studies. Results: Our findings revealed that Def induced cytotoxicity, effectively chelated intracellular iron, and triggered apoptosis through the increase in phosphatidylserine externalization and caspase 3 activity. Additionally, Def caused G0/G1 cell cycle arrest by downregulating the ribonucleotide reductase catalytic subunit. Furthermore, Def perturbed mitochondrial function by promoting the production of reactive oxygen species and the inhibition of glutathione as a measurement of ferroptosis activation. Def demonstrated inhibitory effects on cell migration in scratch assays, which was supported by the upregulation of n-myc downstream-regulated gene 1 and downregulation of the epidermal growth factor receptor protein. Also, Def downregulated one of the main markers of chemoresistance, the ABCB1 gene. In vivo experiments using a lung carcinoma mouse model showed that Def treatment did not affect the animal’s body weight and showed a significant decrease in tumor growth. Conclusions: This investigation lays the groundwork for unraveling Def action’s molecular targets and mechanisms in lung carcinoma, particularly within iron-related pathways, pointing out its anti-metastasis and anti-chemoresistance effect. Full article
Show Figures

Figure 1

Figure 1
<p>Iron chelation by Def. Two molecules of Def bind an iron (Fe<sup>+3</sup>) atom to form an octahedral structure. (Ball and stick model color code: terracotta-iron, red-oxygen, dark gray-carbon, blue-nitrogen, white-hydrogen). Adapted from Wikipedia and available at <a href="https://en.wikipedia.org/wiki/Deferasirox#" target="_blank">https://en.wikipedia.org/wiki/Deferasirox#</a> (accessed on 8 March 2024). All structured data images from Wikipedia are available under the Creative Commons CC0 License (No copyright).</p>
Full article ">Figure 2
<p>Def’s cytotoxicity. (<b>A</b>) MTS viability assay. Def was tested in three different lung cell lines: A549, MRC5 CAF, and NL20 cells. Data showed the average of eight measurements (mean ± SD) of at least three independent experiments. (<b>B</b>) Early apoptosis activation by the phosphatidylserine (PS) externalization (green fluorescence). The upper images show the bright field. The NucBlue DAPI (blue) and Annexin V (green) fluorescence channels are overlaid in the bottom images. All images were taken at 40× magnification. Green fluorescence (525 nm) of Annexin V was statistically quantified ((<b>B</b>), down). Def was analyzed using confocal in duplicate in two independent experiments. The <span class="html-italic">p</span>-value thresholds for statistical significance were set as follows: **** for <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Nuclear-related mechanistic pathways. (<b>A</b>) Cell cycle arrest assay. Dot plot histograms (<b>left</b>) and bar graph with statistics (<b>right</b>). Def was analyzed in duplicate in two independent experiments. (<b>B</b>) DNA damage assay. Dot plot histograms (<b>left</b>) and bar graph with statistics (<b>right</b>). Def was analyzed in duplicate in two independent experiments The <span class="html-italic">p</span>-value thresholds for statistical significance were set as follows: **** for <span class="html-italic">p</span> &lt; 0.0001, *** for <span class="html-italic">p</span> ranging from 0.0001 to 0.001 and ** for <span class="html-italic">p</span> between 0.01 and 0.001.</p>
Full article ">Figure 4
<p>Mitochondrial-related mechanistic pathways. (<b>A</b>) Mitochondrial depolarization assay. This assay was performed in triplicates in two independent experiments. Dot plot histograms (<b>left</b>) and bar graph with statistics (<b>right</b>). (<b>B</b>) Caspase 3 activation. Data showed the average of four measurements (mean ± SD) of at least three independent experiments. The <span class="html-italic">p</span>-value thresholds for statistical significance were set as follows: **** for <span class="html-italic">p</span> &lt; 0.0001, and * for <span class="html-italic">p</span> from 0.05 to 0.01.</p>
Full article ">Figure 5
<p>Iron-related processes. (<b>A</b>) Iron binding capacity assay. Treated and untreated cells were supplemented with a 200 μM iron citrate for this assay. Data showed the average of four measurements (mean ± SD) of at least three independent experiments. (<b>B</b>) RT-qPCR for <span class="html-italic">RR</span> mRNA gene expression. Data showed the average of three measurements (mean ± SD) of at least two independent experiments. (<b>C</b>) Total ROS production assay. H<sub>2</sub>O<sub>2</sub> was used as a positive control. Data showed the average of four measurements (mean ± SD) of at least three independent experiments. (<b>D</b>) % GSH production. Iron citrate was used as a positive control. Data showed the average of four measurements (mean ± SD) of at least two independent experiments. The <span class="html-italic">p</span>-value thresholds for statistical significance were set as follows: **** for <span class="html-italic">p</span> &lt; 0.0001 and *** for <span class="html-italic">p</span> ranging from 0.0001 to 0.001.</p>
Full article ">Figure 6
<p>Migration and resistance-related processes. (<b>A</b>) Scratch Migration Assay (<b>left</b>) and statistics of the migration (<b>right</b>). Data showed the average of two measurements (mean ± SD) of at least three independent experiments. Blue lines show the measured area to determine migration. (<b>B</b>) mRNA Gene Expression of A549 cells treated with Def for 24 h using qPCR for <span class="html-italic">ABCB1</span> and <span class="html-italic">NDRG1</span> genes. Data showed the average of three measurements (mean ± SD) of at least two independent experiments. (<b>C</b>) EGFR expression and activation. Total Expression = Inactivated + Activated. Dot plot histograms (<b>left</b>) and bar graph with statistics (<b>right</b>). This assay was performed in triplicates in two independent experiments. The <span class="html-italic">p</span>-value thresholds for statistical significance were set as follows: **** for <span class="html-italic">p</span> &lt; 0.0001 and *** for <span class="html-italic">p</span> ranging from 0.0001 to 0.001.</p>
Full article ">Figure 7
<p>In vivo assays. (<b>A</b>) Average mouse weight of vehicle versus Def-treated mice was monitored on days 3, 6, 8, 10, and 12. (<b>B</b>) Tumor volume using a concentration range (20–90 mg/kg) of Def. (<b>C</b>) Tumor growth of vehicle vs. Def treated mice (from 20 to 90 mg/kg) after 12 days. Vehicle (n = 4) and Def-treated mice (n = 4). The <span class="html-italic">p</span>-value thresholds for statistical significance were set as * for <span class="html-italic">p</span> from 0.05 to 0.01.</p>
Full article ">
18 pages, 6524 KiB  
Review
Arylpiperazine Derivatives and Cancer: A New Challenge in Medicinal Chemistry
by Giorgia Andreozzi, Angela Corvino, Beatrice Severino, Elisa Magli, Elisa Perissutti, Francesco Frecentese, Vincenzo Santagada, Giuseppe Caliendo and Ferdinando Fiorino
Pharmaceuticals 2024, 17(10), 1320; https://doi.org/10.3390/ph17101320 - 2 Oct 2024
Viewed by 693
Abstract
Background: In recent decades, there has been a startling rise in the number of cancer patients worldwide, which has led to an amazing upsurge in the development of novel anticancer treatment candidates. On a positive note, arylpiperazines have garnered attention in cancer research [...] Read more.
Background: In recent decades, there has been a startling rise in the number of cancer patients worldwide, which has led to an amazing upsurge in the development of novel anticancer treatment candidates. On a positive note, arylpiperazines have garnered attention in cancer research due to their potential as scaffolds for developing anticancer agents. These compounds exhibit a diverse array of biological activities, including cytotoxic effects against cancer cells. Indeed, one of the key advantages of arylpiperazines lies in their ability to interact with various molecular targets implicated in cancer pathogenesis. Aim: Here, we focus on the chemical structures of several arylpiperazine derivatives, highlighting their anti-proliferative activity in different tumor cell lines. The modular structure, diverse biological activities, and potential for combination therapies of arylpiperazine compounds make them valuable candidates for further preclinical and clinical investigations in the fight against cancer. Conclusion: This review, providing a careful analysis of different arylpiperazines and their biological applications, allows researchers to refine the chemical structures to improve potency, selectivity, and pharmacokinetic properties, thus advancing their therapeutic potential in oncology. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>General structure of <span class="html-italic">N</span>-arylpiperazine and pharmacophoric model of 5-HT<sub>1A</sub> agonist.</p>
Full article ">Figure 2
<p>Signaling pathways of 5-HT<sub>1A</sub> receptor [<a href="#B11-pharmaceuticals-17-01320" class="html-bibr">11</a>].</p>
Full article ">Figure 3
<p>Chemical structure of5HT<sub>1A</sub> antagonists.</p>
Full article ">Figure 4
<p>Structure of SDZ 216–525 (<b>2</b>).</p>
Full article ">Figure 5
<p>Structure of BP554 (<b>3</b>).</p>
Full article ">Figure 6
<p>Structure of Naftopidil (<b>4</b>).</p>
Full article ">Figure 7
<p>Structures of Terazosin (<b>5</b>) and Prazosin (<b>6</b>).</p>
Full article ">Figure 8
<p>Structure of Doxazosin (<b>7</b>).</p>
Full article ">Figure 9
<p>General structure of arylpiperazine derivatives. Ar indicates variously substituted phenyl or benzyl piperazine.</p>
Full article ">Figure 10
<p>Structure of androgen antagonist YM-92088 (<b>8</b>).</p>
Full article ">Figure 11
<p>Compound <b>10</b> inhibited cell viability in prostate cell lines PC-3 and DU145. All cells were exposed to escalating concentrations of arylpiperazine derivatives respectively for 24 h, and the cell viability was detected by CCK-8 assay [<a href="#B27-pharmaceuticals-17-01320" class="html-bibr">27</a>].</p>
Full article ">Figure 12
<p>Chemical structures of oxicams derivatives acting on colorectal adenocarcinoma cell lines.</p>
Full article ">Figure 13
<p>Chemical structure of C2 (<b>18</b>).</p>
Full article ">Figure 14
<p>Chemical structure of SER-79 (<b>19</b>) and SER-68 (<b>20</b>).</p>
Full article ">Figure 15
<p>Chemical structure of compound <b>30</b> and its combination effect with different cytotoxic drugs on the growth of MDA-MB-231 cancer cells [<a href="#B52-pharmaceuticals-17-01320" class="html-bibr">52</a>]. The drugs were used at the following doses: (a,b): 2 and 3 nM paclitaxel; (c,d): 5 and 10 nM doxorubicin; (e,f): 100 and 200 nM gemcitabine; (g,h) 3 and 5 μM fluorouracil; (i,j): 300 and 500 μM cisplatin.</p>
Full article ">Figure 16
<p>Chemical structures of SMO inhibitors.</p>
Full article ">Figure 17
<p>Chemical structure of SGI-7079 (<b>33</b>).</p>
Full article ">Figure 18
<p>Potent anticancer drugs bearing <span class="html-italic">N</span>-arylpiperazine moieties.</p>
Full article ">
Back to TopTop