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

Journals

Article Types

Countries / Regions

Search Results (60)

Search Parameters:
Keywords = raloxifene

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 28277 KiB  
Article
Enhancing Bone Repair: Impact of Raloxifene-Functionalized Cerabone® on Rat Calvarial Defects
by Laura Gabriela Macedo, Gabriel Mulinari-Santos, Natália Barbosa de Siqueira, Letícia Pitol-Palin, Ana Cláudia Ervolino da Silva, Paula Buzo Frigério, Paulo Roberto Botacin, Paulo Noronha Lisboa-Filho and Roberta Okamoto
J. Funct. Biomater. 2025, 16(2), 59; https://doi.org/10.3390/jfb16020059 - 11 Feb 2025
Viewed by 203
Abstract
Bone substitutes are commonly used in bone regeneration, and their functionalization with bioactive molecules can significantly enhance bone regeneration by directly influencing bone cells. This study aimed to evaluate the potential of raloxifene-functionalized Cerabone® (CB) for promoting bone repair and to highlight [...] Read more.
Bone substitutes are commonly used in bone regeneration, and their functionalization with bioactive molecules can significantly enhance bone regeneration by directly influencing bone cells. This study aimed to evaluate the potential of raloxifene-functionalized Cerabone® (CB) for promoting bone repair and to highlight the implications in bone regeneration. The effectiveness of Cerabone® functionalized with raloxifene via sonication or gel delivery in promoting bone repair in rat calvaria defects was assessed. Ninety-six male rats with critical-sized calvarial defects were divided into six treatment groups (n = 16): COAG (spontaneous blood clot), CB (Cerabone®), CBS (Cerabone® sonicated alone), CBRS (Cerabone® with raloxifene sonicated), CBG (Cerabone® with gel vehicle), and CBRG (Cerabone® with 20% raloxifene gel). After 14 and 28 days, samples were analyzed using microtomography, histomorphometry, immunohistochemistry, and fluorescence techniques. Quantitative data were statistically analyzed, comparing each group to the control CB group with significance set at p < 0.05. Micro-CT analysis demonstrated a significant increase in bone volume in the CBRS, CBRG, and CBS groups at 28 days compared to the CB group (p < 0.05). Specifically, the mean bone volume percentages for the CBRS, CBRG, CBS, and CB groups were 21.18%, 17.51%, 13.18%, and 7.8%, respectively. Histomorphometry showed increased new bone formation in the CBRS and CBRG groups at both 14 and 28 days. Fluorescence analysis revealed a significantly higher daily mineral apposition rate in the CBRS and CBRG groups at 28 days. These findings suggest that raloxifene-functionalized CB, delivered via sonication or gel, significantly enhances bone repair by improving bone volume and mineralization, highlighting its potential as an effective strategy for bone regeneration. Full article
(This article belongs to the Special Issue Biomaterials in Bone Reconstruction)
Show Figures

Figure 1

Figure 1
<p>Microcomputerized tomography analysis at the bone calvarial defect. Morphologic parameters with mean results and standard deviation were calculated and reported as follows: (<b>A</b>) bone volume per tissue volume (BV/TV); (<b>B</b>) trabecular thickness (Tb.Tb); (<b>C</b>) trabecular number (Tb.N.); (<b>D</b>) trabecular separation (Tb.Sp). The * indicate a significant statistical difference (<span class="html-italic">p</span> &lt; 0.05) in comparison to the CB group. Statistical tests: two-way ANOVA; Tukey post-test.</p>
Full article ">Figure 2
<p>The microtomographic reconstruction of bone repair for each group at 28 days is presented. The microtomography images are representative of all six groups: COAG, CB, CBG, CBRG, CBS, and CBRS, respectively. The microtomography was performed using CTvox software (SkyScan, Version 2.7).</p>
Full article ">Figure 3
<p>Histological images of bone repair at 14 days. The lower images of each group provide an overview in the sagittal plane, while the upper images present a detailed close-up of the tissue in the defect area for each group: COAG, CB, CBG, CBRG, CBS, and CBRS. Decalcified sections were prepared and stained with HE. NB indicates new bone formation, CT represents connective tissue, and CB denotes CB particles. The red dotted square in the lower images denotes the descriptive area from which the high-magnification areas in the upper images were derived. Scale bars represent 30 µm (upper images) and 1 mm (lower images). Original magnifications: 40× (upper images) and 4× (lower images).</p>
Full article ">Figure 4
<p>Histological images of bone repair at 28 days. The lower images of each group provide an overview in the sagittal plane, while the upper images present a detailed close-up of the tissue in the defect area for each group: COAG, CB, CBG, CBRG, CBS, and CBRS. Decalcified sections were prepared and stained with HE. NB indicates new bone formation, CT represents connective tissue, and CB denotes CB particles. The red dotted square in the lower images denotes the descriptive area from which the high-magnification areas in the upper images were derived. Scale bars represent 30 µm (upper images) and 1 mm (lower images). Original magnifications: 40× (upper images) and 4× (lower images).</p>
Full article ">Figure 5
<p>Column graphs of the histomorphometric parameters in the calvaria bone repair. Histomorphometric mean results and standard deviation of the new bone formed at 14 days (<b>A</b>) and at 28 days (<b>B</b>), following the percentage of biomaterial at 14 days (<b>C</b>) and at 28 days (<b>D</b>). The * indicates significant statistical difference in comparison to the CB group (<span class="html-italic">p</span> &lt; 0.05). Statistical tests: two-way ANOVA; Tukey post-test.</p>
Full article ">Figure 6
<p>Immunohistochemical staining of PECAM at 14 and 28 days for CB, CBG, CBRG, CBS, and CBRS. The red arrows indicate the positive imunostaining for PECAM. Scale bar = 100 µm. Original magnification: 20×.</p>
Full article ">Figure 7
<p>Immunohistochemical staining of RUNX-2 at 14 and 28 days for CB, CBG, CBRG, CBS, and CBRS. The red arrows indicate the positive imunostaining for RUNX-2. Scale bar = 100 µm. Original magnification: 20×.</p>
Full article ">Figure 8
<p>Immunohistochemical staining of osteopontin (OPN) at 14 and 28 days for CB, CBG, CBRG, CBS, and CBRS. The red arrows indicate the positive imunostaining for OPN. Scale bar = 100 µm. Original magnification: 20×.</p>
Full article ">Figure 9
<p>Fluorescent images showing bone mineralization at the edge (superior part) and the center of the bone defect (inferior part). Calcein (green) labels mature bone mineralization at 14 postoperative days, while alizarin (red) marks new bone mineralization at 42 postoperative days for COAG, CB, CBG, CBRG, CBS, and CBRS. The letter (S) indicates the superior part of the defect, while the letter (I) represents the inferior part of the defect. Original magnification: 20×.</p>
Full article ">Figure 10
<p>Column graphs showing bone mineralization marked by fluorochromes, with the mean and error bars. The calcein area is represented as green columns at 14 post-operative days, and the alizarin area as denoted as red columns at 42 post-operative days in μm<sup>2</sup> (<b>A</b>). Daily mineral apposition rate (MAR, μm/day) during calvarial bone repair (<b>B</b>). The asterisk (*) indicates a significant statistical difference compared to the CB group (<span class="html-italic">p</span> &lt; 0.05). Statistical analysis was performed using two-way ANOVA with Tukey’s post hoc test.</p>
Full article ">
16 pages, 4416 KiB  
Article
Raloxifene Protects Oxygen-Glucose-Deprived Astrocyte Cells Used to Mimic Hypoxic-Ischemic Brain Injury
by Nicolás Toro-Urrego, Juan P. Luaces, Tamara Kobiec, Lucas Udovin, Sofía Bordet, Matilde Otero-Losada and Francisco Capani
Int. J. Mol. Sci. 2024, 25(22), 12121; https://doi.org/10.3390/ijms252212121 - 12 Nov 2024
Viewed by 2577
Abstract
Perinatal asphyxia (PA) is a clinical condition characterized by oxygen supply suspension before, during, or immediately after birth, and it is an important risk factor for neurodevelopmental damage. Its estimated 1/1000 live births incidence in developed countries rises to 5–10-fold in developing countries. [...] Read more.
Perinatal asphyxia (PA) is a clinical condition characterized by oxygen supply suspension before, during, or immediately after birth, and it is an important risk factor for neurodevelopmental damage. Its estimated 1/1000 live births incidence in developed countries rises to 5–10-fold in developing countries. Schizophrenia, cerebral palsy, mental retardation, epilepsy, blindness, and others are among the highly disabling chronic pathologies associated with PA. However, so far, there is no effective therapy to neutralize or reduce PA-induced harm. Selective regulators of estrogen activity in tissues and selective estrogen receptor modulators like raloxifene have shown neuroprotective activity in different pathological scenarios. Their effect on PA is yet unknown. The purpose of this paper is to examine whether raloxifene showed neuroprotection in an oxygen–glucose deprivation/reoxygenation astrocyte cell model. To study this issue, T98G cells in culture were treated with a glucose-free DMEM medium and incubated at 37 °C in a hypoxia chamber with 1% O2 for 3, 6, 12, and 24 h. Cultures were supplemented with raloxifene 10, and 100 nM during both glucose and oxygen deprivation and reoxygenation periods. Raloxifene 100 nM and 10 nM improved cell survival—65.34% and 70.56%, respectively, compared with the control cell groups. Mitochondrial membrane potential was preserved by 58.9% 10 nM raloxifene and 81.57% 100 nM raloxifene cotreatment. Raloxifene co-treatment reduced superoxide production by 72.72% and peroxide production by 57%. Mitochondrial mass was preserved by 47.4%, 75.5%, and 89% in T98G cells exposed to 6-h oxygen–glucose deprivation followed by 3, 6, and 9 h of reoxygenation, respectively. Therefore, raloxifene improved cell survival and mitochondrial membrane potential and reduced lipid peroxidation and reactive oxygen species (ROS) production, suggesting a direct effect on mitochondria. In this study, raloxifene protected oxygen–glucose-deprived astrocyte cells, used to mimic hypoxic–ischemic brain injury. Two examiners performed the qualitative assessment in a double-blind fashion. Full article
(This article belongs to the Special Issue New Trends in Molecular Research of Aneurysm and Brain Injury)
Show Figures

Figure 1

Figure 1
<p>Raloxifene decreased OGD-induced cell death. (<b>A</b>) T98G cells were treated with different concentrations of raloxifene during 6 h of OGD and 3 h of reoxygenation, and cell viability was assessed by MTT assay. Data are represented as the mean ± SEM of four independent experiments. Control (101.99 ± 1.85); OGD/R (52.59 ± 2.02); OGD/R + 100 nM raloxifene (65.34 ± 2.03); OGD/R + 10 nM raloxifene (70.56 ± 2.36). Data were examined by analysis of variance, followed by the post hoc Dunnet’s test for between-group comparisons and Tukey’s test for multiple comparisons, * <span class="html-italic">p</span> &lt; 0.005. (<b>B</b>) Cell surface quantification with different concentrations of raloxifene during 6 h of OGD and 3 h of reoxygenation. Data are represented as the mean ± SEM of four independent experiments. Control (225.3 ± 13.01); OGD/R (278.7 ± 18.51); OGD/R + 100 nM raloxifene (318.2 ± 21.86); OGD/R + 10 nM raloxifene (277.1 ± 18.16). Data were examined by analysis of variance, followed by the post hoc Dunnet’s test for between-group comparisons and Tukey’s test for multiple comparisons, * <span class="html-italic">p</span> &lt; 0.005. (<b>C</b>–<b>F</b>) Raloxifene reduced morphological alterations induced by oxygen–glucose deprivation/reoxygenation. Representative microphotographs showing the morphology of cells exposed to (<b>C</b>) DMEM, (<b>D</b>) OGD/R, (<b>E</b>) OGD/R + Ral 100 nM, and (<b>F</b>) OGD/R + Ral 10 nM. Scale bar 50 µm.</p>
Full article ">Figure 2
<p>Raloxifene reduced superoxide production at 6 h of OGD and 3 h of reoxygenation. (<b>A</b>) Mean fluorescence values of dihydroethidium (DHE) intensity. (<b>B</b>–<b>E</b>) Representative fluorescence micrographs of dihydroethidium (DHE) staining in T98G cells exposed to (<b>B</b>) DMEM, (<b>C</b>) OGD/R, (<b>D</b>) OGD/R + Ral 100 nM with 6 h of OGD and 3 h of reoxygenation, and (<b>E</b>) OGD/R + Ral 10 nM with 6 h of OGD and 3 h of reoxygenation. *** <span class="html-italic">p</span> &lt; 0.0001. Scale bar 50 µm.</p>
Full article ">Figure 3
<p>Raloxifene reduced peroxide production at 6 h of OGD and 6 h of reoxygenation. The figure shows the representative fluorescence microphotographs of 2′,7′-Dichlorofluorescin Diacetate (DCFDA) staining of T98G cells exposed to (<b>A</b>) Control, (<b>B</b>) OGD/R, (<b>C</b>) OGD/R, OGD/R + Ral 100 nM with 6 h of OGD and 6 h of reoxygenation, (<b>D</b>) OGD/R, OGD/R + Ral 10 nM with 6 h of OGD and 6 h of reoxygenation, and (<b>E</b>) the mean fluorescence values of DCFDA intensity measured by flow cytometry. Data are represented as the mean ± SEM of five independent experiments. Control (55.51 ± 1.03); OGD/R (131.00 ± 4.01); OGD/R + 100 nM raloxifene (75.15 ± 6.60); OGD/R + 10 nM raloxifene (72.38 ± 7.82). Data were examined by analysis of variance, followed by the post hoc Dunnet’s test for between-group comparisons and Tukey’s test for multiple comparisons **** <span class="html-italic">p</span> &lt; 0.0001. Scale bar 50 µm.</p>
Full article ">Figure 4
<p>Raloxifene attenuated mitochondrial membrane potential loss at 6 h of OGD and 3 h of reoxygenation. (<b>A</b>) The figure shows the mean fluorescence values. (<b>B</b>–<b>E</b>) Representative fluores-cence micrographs of tetra-methyl rhodamine methyl ester (TMRM) staining in T98G cells exposed to (<b>B</b>) OGD/R, (<b>C</b>) DMEM, (<b>D</b>) OGD/R + Ral 100 nM with 6 h of OGD and 3 h of reoxygenation, and (<b>E</b>) OGD/R + Ral 10 nM with 6 h of OGD and 3 h of reoxygenation. *** <span class="html-italic">p</span> &lt; 0.0001. Scale bar 50 µm.</p>
Full article ">Figure 5
<p>Raloxifene preserved mitochondrial mass in T98G cells exposed to 6 h of OGD and 3 h of reoxygenation. The figure shows the mitochondrial mass in T98G cells exposed to 6 h of oxygen–glucose deprivation (OGD) to 3 h (<b>A</b>–<b>D</b>), 6 h (<b>E</b>–<b>H</b>), and 9 h (<b>I</b>–<b>L</b>) of reoxygenation. The representative microphotographs of acridine orange (NAO) fluorescence in T98G astrocytic cells exposed to (<b>A</b>) DMEM, (<b>B</b>) OGD/R, (<b>C</b>) OGD/R + Ral 100 nM with 3 h of reoxygenation, and (<b>D</b>) OGD/R + Ral 10 nM with 3 h of reoxygenation. (<b>M</b>) Mean fluorescence values of NAO intensity in this period of insult. Data are represented as the mean ± SEM of five independent experiments. Control (6671.00 ± 86.18); OGD/R (1903.00 ± 155.30); OGD/R + 100 nM raloxifene (2940.00 ± 142.90); OGD/R + 10 nM raloxifene (3163.00 ± 119.80). (<b>E</b>) DMEM, (<b>F</b>) OGD/R, (<b>G</b>) OGD/R + Ral 100 nM with 6 h of reoxygenation, and (<b>H</b>) OGD/R + Ral 10 nM with 6 h of reoxygenation. (<b>N</b>) Mean fluorescence values of NAO intensity in this period of insult. Data are represented as the mean ± SEM of five independent experiments. Control (416.7.00 ± 39.47); OGD/R (183.1 ± 17.70); OGD + 100 nM raloxifene (238.4 ± 26.43); OGD + 10 nM raloxifene (314.6 ± 27.45) (<b>I</b>) DMEM, (<b>J</b>) OGD/R, (<b>K</b>) OGD/R + Ral 100 nM with 9 h of reoxygenation, and (<b>L</b>) OGD/R + Ral 10 nM with 9 h of reoxygenation. (<b>O</b>) Mean fluorescence values of NAO intensity in this period of insult. Data are represented as the mean ± SEM of five independent experiments. Control (452.20 ± 22.28); OGD/R (330.42 ± 23.45); OGD/R + 100 nM raloxifene (404.71 ± 12.34); OGD/R + 10 nM raloxifene (374.64 ± 19.78). Data were examined by analysis of variance, followed by the post hoc Dunnet’s test for between-group comparisons and Tukey’s test for multiple comparisons, * <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001. Scale bar 50 µm.</p>
Full article ">
19 pages, 4984 KiB  
Article
Fabrication of an In Situ pH-Responsive Raloxifene-Loaded Invasome Hydrogel for Breast Cancer Management: In Vitro and In Vivo Evaluation
by Hanan O. Farouk, Marwa M. Nagib, Amr Gamal Fouad, Demiana M. Naguib, Sherif Faysal Abdelfattah Khalil, Amany Belal, Samar F. Miski, Nisreen Khalid Aref Albezrah, Shatha Hallal Al-Ziyadi, Gi-Hui Kim, Ahmed H. E. Hassan, Kyung-Tae Lee and Doaa S. Hamad
Pharmaceuticals 2024, 17(11), 1518; https://doi.org/10.3390/ph17111518 - 11 Nov 2024
Viewed by 1144
Abstract
Background/Objectives: Raloxifene (RLF) is a therapeutic option for invasive breast cancer because it blocks estrogen receptors selectively. Low solubility, limited targeting, first-pass action, and poor absorption are some of the challenges that make RLF in oral form less effective. This study aimed to [...] Read more.
Background/Objectives: Raloxifene (RLF) is a therapeutic option for invasive breast cancer because it blocks estrogen receptors selectively. Low solubility, limited targeting, first-pass action, and poor absorption are some of the challenges that make RLF in oral form less effective. This study aimed to create an intra-tumoral in situ pH-responsive formulation of RLF–invasome (IPHRLI) for breast cancer treatment, with the goals of sustaining RLF release, minimizing adverse effects, and enhancing solubility, bioavailability, targeting, and effectiveness. Methods: Numerous RLF–invasome formulations were optimized using design expert software (version 12.0.6.0, StatEase Inc., Minneapolis, MN, USA). Integrating an optimal formulation with an amalgam of chitosan and glyceryl monooleate resulted in the IPHRLI formulation. In vivo testing of the IPHRLI formulation was conducted utilizing the Ehrlich cancer model. Results: Requirements for an optimum RLF–invasome formulation were met by a mixture of phospholipids (2.46%), ethanol (2.84%), and cineole (0.5%). The IPHRLI formulation substantially sustained its release by 75.41% after 8 h relative to free RLF. The bioavailability of intra-tumoral IPHRLI was substantially raised by 4.07-fold compared to oral free RLF. Histopathological and tumor volume analyses of intra-tumoral IPHRLI confirmed its efficacy and targeting effect. Conclusions: the intra-tumoral administration of the IPHRLI formulation may provide a potential strategy for breast cancer management. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>,<b>C</b>) Correlation between observed and predicted values; (<b>B</b>,<b>D</b>) correlation between residual and run for vesicle size and entrapment efficiency.</p>
Full article ">Figure 2
<p>Three-dimensional charts showing the effects of factors on (<b>A</b>) vesicle size and (<b>B</b>) entrapment efficiency.</p>
Full article ">Figure 3
<p>(<b>A</b>) PDI; (<b>B</b>) zeta potential; (<b>C</b>) surface morphology, where photographs were taken at 4000× magnification and scale bar = 500 nm; and (<b>D</b>) DSC thermograms of optimum RLI formulation.</p>
Full article ">Figure 4
<p>(<b>A</b>) Viscosity of IPHRLI formulation compared to FRIPH formulation and optimum RLI formulation; (<b>B</b>) release profile of IPHRLI formulation compared to free RLF and optimum RLI formulation; (<b>C</b>) impact of different storage temperatures on IPHRLI formulation’s entrapment efficacy and vesicle size.</p>
Full article ">Figure 5
<p>(<b>A</b>) RLF plasma concentration–time profile between intra-tumor IPHRLI formulation and oral free RLF; (<b>B</b>) variation in tumor volume between control positive group and groups treated with intra-tumor IPHRLI formulation or oral free RLF; (<b>C</b>) variation in body weight between control positive group and groups treated with intra-tumor IPHRLI formulation or oral free RLF; (<b>D</b>) intra-tumor IPHRLI formulation’s RLF concentration in liver and tumor compared to oral free RLF. <sup>a</sup> Significance when compared to control positive group, with <span class="html-italic">p</span>-value &lt; 0.05; <sup>b</sup> Significance when compared to oral free RLF group, with <span class="html-italic">p</span>-value of &lt;0.05.</p>
Full article ">Figure 6
<p>Histopathology findings of tumor of (<b>A</b>) control negative group, (<b>B</b>) control positive group, (<b>C</b>) oral free RLF group, and (<b>D</b>) intra-tumor IPHRLI formulation group. Black arrow: hypercellular tumor; red arrow: infiltrating surrounding fat; blue arrow: necrosis area; violet arrow: cyst formation.</p>
Full article ">Figure 7
<p>(<b>A</b>) Kaplan–Meier survival curve of oral free RLF, intra-tumor IPHRLI formulation, and control positive group; (<b>B</b>) liver and tumor concentration of RLF for intra-tumor IPHRLI formulation and oral free RLF groups; (<b>C</b>) biochemical and hematological parameters of intra-tumor IPHRLI formulation group compared to control negative group.</p>
Full article ">
18 pages, 6510 KiB  
Article
Molecular Networking, Docking, and Biological Evaluation of Licarin A from Myristica fragrans as a Potential Cancer Chemopreventive Agent
by Peter J. Blanco Carcache, Ines Y. Castro-Dionicio, Nathan P. Mirtallo Ezzone, Eric D. Salinas-Arrellano, Joshua Bahar, Steven K. Clinton and A. Douglas Kinghorn
Molecules 2024, 29(20), 4919; https://doi.org/10.3390/molecules29204919 - 17 Oct 2024
Viewed by 1307
Abstract
Currently, clinically available cancer chemopreventive drug options are limited to mostly tamoxifen and its derivatives, such as raloxifene, and approved specifically for breast cancer. Thus, the availability of chemopreventive drug molecules for other types of malignant cancers would be desirable. In previous reports, [...] Read more.
Currently, clinically available cancer chemopreventive drug options are limited to mostly tamoxifen and its derivatives, such as raloxifene, and approved specifically for breast cancer. Thus, the availability of chemopreventive drug molecules for other types of malignant cancers would be desirable. In previous reports, the arils of Myristica fragrans (mace) have been found to exhibit cancer chemopreventive activity. Therefore, the purpose of the present study was to identify a natural product from this species with potential chemopreventive activity guided by chemoinformatic sample analysis via Global Natural Products Social (GNPS) molecular networking and molecular docking. The neolignan licarin A (1) was identified as a potential chemopreventive constituent, and subsequently submitted to several in vitro bioassays and a zebrafish toxicity evaluation. In this work, 1 afforded superior phosphoNF-κBp65 phosphorylation activity in DU-145 prostate cancer cells compared to isoliquiritigenin (2), which was used as a natural product chemopreventive control. Both 1 and 2 showed a longer-lasting reduction in cellular stress in a cell oxidative stress real-time dose–response assay than the positive control using Hepa1c1c7 mouse hepatoma cells. In addition, 1 displayed similar activities to 2, while also being less toxic to zebrafish (Danio rerio) than both this chalcone and the clinically used chemopreventive drug tamoxifen. Full article
Show Figures

Figure 1

Figure 1
<p>Structures of licarin A (<b>1</b>) and isoliquiritigenin (<b>2</b>).</p>
Full article ">Figure 2
<p>LC-MS/MS chemoinformatic analysis of all chromatographic fractions obtained from the ethyl acetate partition of <span class="html-italic">M. fragrans</span> revealed the presence of <b>1</b> in region A (see <a href="#app1-molecules-29-04919" class="html-app">Supplementary Figure S1</a>) of the molecular network. (Relevant data: GNPS library hit, Bronze: 0.81c.)</p>
Full article ">Figure 3
<p>Molecular docking of licarin A (<b>1</b>) based on binding energy and structural similarity. The binding pocket of <b>1</b> to NF-<span class="html-italic">κ</span>Bp65 is shown in a 3D perspective. The 2D perspective displays all binding interactions with relevant amino acids. (Relevant data: in silico binding affinity, 10.66 μM; binding energy, −6.78).</p>
Full article ">Figure 4
<p>Molecular docking for the SIRIUS CSI: FingerID structural predictions with NF-<span class="html-italic">κ</span>Bp65. (<b>A</b>) Structures of <b>3</b>–<b>6</b>. (<b>B</b>–<b>E</b>) Docking profiles for <b>3</b> (blue, <b>B</b>), <b>4</b> (gray, <b>C</b>), <b>5</b> (pink, <b>D</b>), and <b>6</b> (cyan, <b>E</b>) with NF-<span class="html-italic">κ</span>Bp65.</p>
Full article ">Figure 5
<p>Molecular docking for the GNPS library hits with NF-<span class="html-italic">κ</span>Bp65. (<b>A</b>) Structures of <b>7</b>–<b>10</b>. (<b>B</b>–<b>E</b>) Docking profiles for <b>7</b> (dark green, <b>B</b>), <b>8</b> (yellow, <b>C</b>), <b>9</b> (neon green, <b>D</b>), and <b>10</b> (blue, <b>E</b>) with NF-<span class="html-italic">κ</span>Bp65.</p>
Full article ">Figure 6
<p>(<b>A</b>) Crystal violet viability data for compounds <b>1</b> and <b>2</b>, and the rocaglamide positive control, with the DU-145 prostate cancer cell line. (<b>B</b>–<b>D</b>) Cell oxidative stress real-time dose–response assay profiles of <b>1</b>, <b>2</b>, and vitamin C, respectively, using Hepa1c1c7 mouse hepatoma cells.</p>
Full article ">Figure 7
<p>(<b>A</b>) Phosphorylated NF-<span class="html-italic">κ</span>Bp65 levels after treatment with 150 μM of <b>1</b>, <b>2</b>, and rocaglamide with the DU-145 prostate cancer cell line. (<b>B</b>–<b>D</b>) Individual phosphorylated NF-<span class="html-italic">κ</span>Bp65 levels after treatment with <b>1</b>, <b>2</b>, and rocaglamide, respectively, using the DU-145 prostate cancer cell line.</p>
Full article ">Figure 8
<p>Representative images of the observed morphological toxicity of tamoxifen, licarin A (<b>1</b>), and isoliquiritigenin (<b>2</b>) in a zebrafish model.</p>
Full article ">Figure 9
<p>Representative histograms showing the condition of zebrafish after separate exposure to tamoxifen, licarin A (<b>1</b>), and isoliquiritigenin (<b>2</b>).</p>
Full article ">
15 pages, 3699 KiB  
Article
Compared Antileishmanial Activity of Clomiphene and Tamoxifen
by Sergio Sifontes-Rodríguez, Alma Reyna Escalona-Montaño, Ricardo Mondragón Flores, Niurka Mollineda-Diogo, Lianet Monzote Fidalgo, Mónica Edith Mondragón-Castelán, Fedra Alardin-Gutiérrez, Lourdes Araceli López-Enzana, Daniel Andrés Sánchez-Almaraz, Ofelia Pérez-Olvera and María Magdalena Aguirre-García
Biomedicines 2024, 12(10), 2290; https://doi.org/10.3390/biomedicines12102290 - 9 Oct 2024
Viewed by 962
Abstract
Drug repositioning is an efficient strategy to search for new treatment alternatives that is especially valuable for neglected parasitic diseases such as leishmaniasis. Tamoxifen and raloxifene are selective estrogen receptor modulators (SERMs) that have shown antileishmanial activity. Clomiphene is a SERM structurally similar [...] Read more.
Drug repositioning is an efficient strategy to search for new treatment alternatives that is especially valuable for neglected parasitic diseases such as leishmaniasis. Tamoxifen and raloxifene are selective estrogen receptor modulators (SERMs) that have shown antileishmanial activity. Clomiphene is a SERM structurally similar to tamoxifen, whose antileishmanial potential is unknown. That is why the objective of the present work was to evaluate its antileishmanial activity in vitro and in vivo in comparison with tamoxifen. The inhibitory effect against promastigotes of L. amazonensis, L. major, and L. mexicana was evaluated for both compounds, as well as the cytotoxicity against mouse peritoneal macrophages, the growth inhibitory activity in intracellular amastigotes of L. mexicana, and the in vivo activity in mice experimentally infected with L. mexicana. Clomiphene was about twice as active as tamoxifen against both promastigotes and intracellular amastigotes, with IC50 values of 1.7–3.3 µM for clomiphene and 2.9–6.4 µM for tamoxifen against all three species of promastigotes and 2.8 ± 0.2 µM and 3.7 ± 0.3 µM, respectively, against L. mexicana amastigotes. Clomiphene structurally affected several parasite organelles in a concentration-dependent fashion, leading to the death of both promastigotes and intracellular amastigotes. Interestingly, the macrophage host cell did not appear damaged by any of the clomiphene concentrations tested. With oral administration at 20 mg/kg for 14 days, both compounds showed similar effects in terms of reducing the growth of the lesions, as well as the weight of the lesions and the parasite load at the end of the follow-up period. The results showed the potential of SERMs as antileishmanial drugs and support further testing of clomiphene and other compounds of this pharmacological group. Full article
(This article belongs to the Collection Feature Papers in Drug Discovery)
Show Figures

Figure 1

Figure 1
<p>Molecular structure of tamoxifen and clomiphene (downloaded from PubChemCompounds, <a href="https://pubchem.ncbi.nlm.nih.gov/" target="_blank">https://pubchem.ncbi.nlm.nih.gov/</a>, accessed on 15 July 2024).</p>
Full article ">Figure 2
<p>Effect of clomiphene on <span class="html-italic">L. mexicana</span> promastigotes treated with clomiphene. Images of scanning electron microscopy on the left side were obtained at low magnification, while on the right side are magnifications of the areas delimited by rectangular frames. *, deformed parasites; F, flagellum; VA, vesicular aggregates; v, vesicles, Scale bar= 500 nm.</p>
Full article ">Figure 3
<p>Effects of clomiphene on <span class="html-italic">L. mexicana</span> promastigotes visualized by transmission electron microscopy. The micrographs in the left column correspond to low magnification images to show the general structure of promastigotes. The flagellar pockets were framed and magnified to show the changes in these structures. In the right insert of the figure for 6 µM, the structure of a mitochondrion is shown, as well as its respective amplification (insert at bottom left). N, nucleus; FP, flagellar pocket; F, flagellum; m, mitochondrion; v, vesicles; kinetoplast (arrow), microtubules (arrowheads). Scale bar = 500 nm.</p>
Full article ">Figure 4
<p>Clomiphene did not affect the integrity of submembrane microtubules. Intact submembrane microtubules (arrowheads) were observed in promastigotes exposed to 6 µM clomiphene. N, nucleus; FP, flagellar pocket; DG, dense granule; v, vesicle. Scale bar = 500 nm.</p>
Full article ">Figure 5
<p>Structural changes induced by clomiphene on intracellular <span class="html-italic">L. mexicana</span> amastigotes. Electron transmission microscopy micrographs with red scale bars (subfigures denoted with “a”) correspond to low-magnification images to show the general structure of infected macrophages. Micrographs indicated by black scale bars (subfigures “b” and “c”) correspond to magnifications of framed zones. N, macrophage nucleus; n, amastigote nucleus; PV: parasitophorous vacuole; FP, flagellar pocket; star, intracellular parasites within parasitophorous vacuoles; red circles, enlarged parasitophorous vacuoles; red stars, destroyed parasites within the parasitophorous vacuole. Black scale bars = 2 µm; red scale bars= 1 µm.</p>
Full article ">Figure 6
<p>Effect of clomiphene and tamoxifen treatment on lesion growth. *: <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 (compared to the control group).</p>
Full article ">Figure 7
<p>Effect of treatment on lesion weight (<b>a</b>) and parasite load (<b>b</b>) one week after the end of treatment. *: <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 (compared to the control group).</p>
Full article ">
11 pages, 1814 KiB  
Article
Quantitative Structure–Property Relationship Analysis in Molecular Graphs of Some Anticancer Drugs with Temperature Indices Approach
by Xiaolong Shi, Ruiqi Cai, Jaber Ramezani Tousi and Ali Asghar Talebi
Mathematics 2024, 12(13), 1953; https://doi.org/10.3390/math12131953 - 24 Jun 2024
Cited by 2 | Viewed by 885
Abstract
The most important application of anticancer drugs in various forms (alkylating agents, hormones agents, and antimetabolites) is the treatment of malignant diseases. Topological indices are widely used in the field of chemical and medical sciences, especially in studying the chemical, biological, clinical, and [...] Read more.
The most important application of anticancer drugs in various forms (alkylating agents, hormones agents, and antimetabolites) is the treatment of malignant diseases. Topological indices are widely used in the field of chemical and medical sciences, especially in studying the chemical, biological, clinical, and therapeutic aspects of drugs. In this article, the temperature indices in anticancer drugs molecular graphs such as Carmustine, Convolutamine F, Raloxifene, Tambjamine K, and Pterocellin B were calculated and then analyzed based on physical and chemical properties. The analysis was performed by identifying the best regression models based on temperature indices for six physical and chemical features of anticancer drugs. The results indicated that temperature indices were essential topological indices that predict the properties of anticancer drugs, such as boiling point, flash point, enthalpy, molar refractivity, molar volume, and polarizability. It was also observed that the r value of the regression model was more than 0.6, and the p value was less than 0.05. Full article
Show Figures

Figure 1

Figure 1
<p>Molecular graph of Carmustine.</p>
Full article ">Figure 2
<p>Molecular of Raloxifene.</p>
Full article ">Figure 3
<p>Molecular of Tambjamine K.</p>
Full article ">Figure 4
<p>Molecular of Pterocellin B.</p>
Full article ">Figure 5
<p>Molecular of Convolutamine F.</p>
Full article ">Figure 6
<p>Physicochemical properties with topological indices.</p>
Full article ">Figure 6 Cont.
<p>Physicochemical properties with topological indices.</p>
Full article ">
18 pages, 1148 KiB  
Review
Beyond Psychotropic: Potential Repurposing of Fluoxetine toward Cancer Therapy
by Sultan F. Kadasah, Abdulaziz M. S. Alqahtani, Abdullah Alkhammash and Mohamed O. Radwan
Int. J. Mol. Sci. 2024, 25(12), 6314; https://doi.org/10.3390/ijms25126314 - 7 Jun 2024
Cited by 1 | Viewed by 2608
Abstract
Drug repurposing, rebranding an existing drug for a new therapeutic indication, is deemed a beneficial approach for a quick and cost-effective drug discovery process by skipping preclinical, Phase 1 trials and pharmacokinetic studies. Several psychotropic drugs, including selective serotonin reuptake inhibitors (SSRIs) and [...] Read more.
Drug repurposing, rebranding an existing drug for a new therapeutic indication, is deemed a beneficial approach for a quick and cost-effective drug discovery process by skipping preclinical, Phase 1 trials and pharmacokinetic studies. Several psychotropic drugs, including selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs), were studied for their potential application in different diseases, especially in cancer therapy. Fluoxetine (FLX) is one of the most prescribed psychotropic agents from the SSRIs class for the treatment of several neuropsychiatric disorders with a favorable safety profile. FLX exhibited different oncolytic effects via mechanisms distinct from its main serotonergic activity. Taking advantage of its ability to rapidly penetrate the blood–brain barrier, FLX could be particularly useful in brain tumors. This was proved by different in vitro and in vivo experiments using FLX as a monotherapy or combination with temozolomide (TMZ) or radiotherapy. In this review of the literature, we summarize the potential pleiotropic oncolytic roles of FLX against different cancers, highlighting the multifaceted activities of FLX and its ability to interrupt cancer proliferation via several molecular mechanisms and even surmount multidrug resistance (MDR). We elaborated on the successful synergistic combinations such as FXR/temozolomide and FXR/raloxifene for the treatment of glioblastoma and breast cancer, respectively. We showcased beneficial pharmaceutical trials to load FLX onto carriers to enhance its safety and efficacy on cancer cells. This is the first review article extensively summarizing all previous FLX repurposing studies for the management of cancer. Full article
(This article belongs to the Special Issue Techniques and Strategies in Drug Design and Discovery, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The number of scientific publications comprising scientific output linking repurposing studies with cancer over the period 2005–2023. Data were retrieved from the Web of Science database by using the keywords “repurposing” and “cancer”.</p>
Full article ">Figure 2
<p>Chemical structure of fluoxetine (FLX) and summary of different sensitive cancers to fluoxetine treatment.</p>
Full article ">Figure 3
<p>Similarity of fluoxetine (FLX) to a VEGFR2 inhibitor, CHEMBL437889; and a multitarget kinase inhibitor, CHEMBL3642580.</p>
Full article ">
18 pages, 23455 KiB  
Article
17β-Estradiol (E2) Activates Matrix Mineralization through Genomic/Nongenomic Pathways in MC3T3-E1 Cells
by Hiraku Suzuki, Yuki Fujiwara, Winda Ariyani, Izuki Amano, Sumiyasu Ishii, Ayane Kate Ninomiya, Seiichi Sato, Akinori Takaoka and Noriyuki Koibuchi
Int. J. Mol. Sci. 2024, 25(9), 4727; https://doi.org/10.3390/ijms25094727 - 26 Apr 2024
Viewed by 1423
Abstract
Estrogen plays an important role in osteoporosis prevention. We herein report the possible novel signaling pathway of 17β-estradiol (E2) in the matrix mineralization of MC3T3-E1, an osteoblast-like cell line. In the culture media-containing stripped serum, in which small lipophilic molecules such as steroid [...] Read more.
Estrogen plays an important role in osteoporosis prevention. We herein report the possible novel signaling pathway of 17β-estradiol (E2) in the matrix mineralization of MC3T3-E1, an osteoblast-like cell line. In the culture media-containing stripped serum, in which small lipophilic molecules such as steroid hormones including E2 were depleted, matrix mineralization was significantly reduced. However, the E2 treatment induced this. The E2 effects were suppressed by ICI182,780, the estrogen receptor (ER)α, and the ERβ antagonist, as well as their mRNA knockdown, whereas Raloxifene, an inhibitor of estrogen-induced transcription, and G15, a G-protein-coupled estrogen receptor (GPER) 1 inhibitor, had little or no effect. Furthermore, the E2-activated matrix mineralization was disrupted by PMA, a PKC activator, and SB202190, a p38 MAPK inhibitor, but not by wortmannin, a PI3K inhibitor. Matrix mineralization was also induced by the culture media from the E2-stimulated cell culture. This effect was hindered by PMA or heat treatment, but not by SB202190. These results indicate that E2 activates the p38 MAPK pathway via ERs independently from actions in the nucleus. Such activation may cause the secretion of certain signaling molecule(s), which inhibit the PKC pathway. Our study provides a novel pathway of E2 action that could be a therapeutic target to activate matrix mineralization under various diseases, including osteoporosis. Full article
(This article belongs to the Special Issue Steroids and Lipophilic Hormones, and Their Actions 3.0)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The scheme of matrix mineralization assays used in the present study.</p>
Full article ">Figure 2
<p>The effect of E2 treatment on the matrix mineralization of MC3T3-E1 cells. (<b>A</b>) A representative showing the matrix mineralization assay. Cells were cultured in the media with AA/βGP and cultured for 12 days. The indicated amount of E2 (1–100 nM) was added at Day 0 until Day 3. The cells were fixed and stained using Alizarin red S to determine the matrix mineralization level. The quantification results are shown in the right panel. (<b>B</b>) ALP assay. ALP activities in the culture medium on Day 12 of the cells being stimulated with/without E2 (1–100 nM) for 3 days (Day 0–Day 3). Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 versus control (no E2) using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 3
<p>Changes in the mRNA levels of the substance responsible for ossification via E2 treatment. Quantitative RT-PCR analysis for <span class="html-italic">Runx2</span>, <span class="html-italic">Alp</span> (<span class="html-italic">Alpl</span>), <span class="html-italic">Col1a,</span> and <span class="html-italic">Ocn</span> (<span class="html-italic">Bglap</span>) mRNA. mRNA was isolated from MC3T3-E1 cells on the indicated day. E2 (10 nM) was treated for 3 days (Day 0–Day 3). The relative mRNA levels of <span class="html-italic">Runx2</span>, <span class="html-italic">Alp</span> (<span class="html-italic">Alpl</span>), <span class="html-italic">Col1a</span>, and <span class="html-italic">Ocn</span> (<span class="html-italic">Bglap</span>) were normalized to the expression of <span class="html-italic">Rpl13a</span>. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. * <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, and **** <span class="html-italic">p</span> &lt; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 4
<p>The effect of ER or GPER antagonists on matrix mineralization. (<b>A</b>–<b>C</b>) MC3T3-E1 cells were treated with/without 10 nM E2 and 100 nM ICI182,780, 100 nM G15, or 100 nM Raloxifene for 3 days (Day 0–Day 3). The cells were fixed and stained with Alizarin red S on Day 12 to determine the matrix mineralization level. The quantification results are shown in the right panel. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 5
<p>The effects of ER antagonists on ALP activity in the culture media, <span class="html-italic">Ocn</span> mRNA, and p38 MAPK phosphorylation. (<b>A</b>) ALP assay. ALP activities in the culture medium on Day 12 of the cells treated with/without E2 (10 nM) for 3 days (Day 0–Day 3) in the presence of 100 nM ICI182,780 or 100 nM Raloxifene for 3 days (Day 0–Day 3). (<b>B</b>) The quantitative RT-PCR analysis of <span class="html-italic">Ocn</span> (<span class="html-italic">Bglap</span>). Cells were stimulated with/without E2 (10 nM) in the presence of 100 nM ICI182,780, 100 nM Raloxifene, or not on Day 0. mRNA was isolated from MC3T3-E1 cells on Day 12. Relative mRNA expression was normalized with the expression of <span class="html-italic">Rpl13a</span>. (<b>C</b>) The Western blot analysis of whole-cell lysates with antibodies for phosphor-p38 MAPK (p-p38 MAPK) or total p38 MAPK. MC3T3-E1 were stimulated with/without 10 nM E2 and 100 nM ICI182,780 on Day 0. Cells were collected 1 h after stimulation. The quantification results are shown in the right panel. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. * <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, and **** <span class="html-italic">p</span> &lt; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 6
<p>The knockdown of ERα or ERβ during matrix mineralization. (<b>A</b>) The quantitative RT-PCR analysis of <span class="html-italic">Esr1</span> (ERα) and <span class="html-italic">Esr2</span> (ERβ). MC3T3-E1 was treated with control siRNA (Control), si<span class="html-italic">Esr</span>1, or si<span class="html-italic">Esr</span>2. mRNA was isolated from the cells after 48 h of transfection. Relative mRNA expression was normalized to the expression of <span class="html-italic">Rpl13a</span>. (<b>B</b>) Matrix mineralization assays. The cells after 48 h of siRNA transfection were stimulated with/without 10 nM E2 for 3 days (Day 0–Day 3). The cells were fixed and stained with Alizarin red S on Day 12 to determine the matrix mineralization level. The quantification results are shown in the right panel. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. * <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; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 7
<p>E2 induced matrix mineralization through the inhibition of the PKC pathway and the activation of p38 MAPK pathway. (<b>A</b>) Cells were stimulated with/without 10 nM E2 for 3 days (Day 0–Day 3) and cultured with/without (<b>A</b>) 500 nM Wortmannin, (<b>B</b>) 150 nM PMA, and (<b>C</b>) 10 µM SB202190 for 3 days (Day 0–Day 3). The cells were fixed and stained with Alizarin red S on Day 12 to determine the matrix mineralization level. The quantification results are shown in the right panel. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 8
<p>The effect of the culture media obtained from E2 pre-exposed cells on matrix mineralization. (<b>A</b>) The scheme of matrix mineralization assays used in the experiments are shown in this figure. (<b>B</b>) Matrix mineralization assay. MC3T3-E1 cells were treated with/without 10 nM E2 at Day 0 for 6 h, 12 h, or 18 h, followed by the E2-free medium replacement and incubated for 18 h, 12 h, or 6 h, respectively. Then, cells directly exposed to E2 for 6 h, 12 h, or 18 h (i, ii or iii, respectively) were fixed on Day 12. Cells treated with the culture medium obtained from the 6 h, 12 h, or 18 h pre-exposed cell (i’, ii’, iii’) were also fixed on Day 12. Then, cells were stained with Alizarin red S to determine the matrix mineralization level. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) are representative of at least three independent experiments. **** <span class="html-italic">p</span> &lt; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">Figure 9
<p>The effect of PKC activation, p38 MAPK inhibition, and heat treatment on matrix mineralization. (<b>A</b>) The scheme of matrix mineralization assays used in the experiments is shown in this figure. (<b>B</b>,<b>C</b>) Matrix mineralization assay. The culture media were collected from the cells pre-exposed with/without 10 nM E2 on Day 0 for 18 h of stimulation. MC3T3-E1 was incubated with the media from the 18 h E2 pre-exposed culture for 72 h in the presence of PMA or SB202190. On Day 12, the cells were fixed and stained with Alizarin red S. The quantification of their results is shown in the right graph. (<b>D</b>) Matrix mineralization assay. The collected media were treated with/without 95 °C for 3 min and centrifuged with 14,000× <span class="html-italic">g</span>, for 3 min at 4 °C. MC3T3-E1 was incubated with this collected media or the heat-treated media from the 18 h E2 pre-exposed culture for 72 h. On Day 12, the cells were fixed and stained with Alizarin red S. The quantification results are shown in the right panel. The (<b>E</b>) quantitative RT-PCR analysis of <span class="html-italic">Ocn</span> (<span class="html-italic">Bglap</span>). Cells were treated in the same way as in (<b>D</b>). mRNA was isolated from MC3T3-E1 cells on Day 12. Relative mRNA expression was normalized to the expression of <span class="html-italic">Rpl13a</span>. Data presented as mean ± SD (<span class="html-italic">n</span> = 3) is representative of at least three independent experiments. ** <span class="html-italic">p</span> &lt; 0.01 and **** <span class="html-italic">p</span> &lt; 0.0001 using Bonferroni’s test. ns: not significant.</p>
Full article ">
2 pages, 149 KiB  
Retraction
RETRACTED: Aldawsari et al. Lipidic Nano-Sized Emulsomes Potentiates the Cytotoxic and Apoptotic Effects of Raloxifene Hydrochloride in MCF-7 Human Breast Cancer Cells: Factorial Analysis and In Vitro Anti-Tumor Activity Assessment. Pharmaceutics 2021, 13, 783
by Hibah M. Aldawsari, Osama A. A. Ahmed, Nabil A. Alhakamy, Thikryat Neamatallah, Usama A. Fahmy and Shaimaa M. Badr-Eldin
Pharmaceutics 2024, 16(2), 195; https://doi.org/10.3390/pharmaceutics16020195 - 30 Jan 2024
Cited by 1 | Viewed by 1206
Abstract
The journal retracts the article “Lipidic Nano-Sized Emulsomes Potentiates the Cytotoxic and Apoptotic Effects of Raloxifene Hydrochloride in MCF-7 Human Breast Cancer Cells: Factorial Analysis and In Vitro Anti-Tumor Activity Assessment” [...] Full article
46 pages, 25903 KiB  
Review
Lysine-Specific Demethylase 1 Inhibitors: A Comprehensive Review Utilizing Computer-Aided Drug Design Technologies
by Di Han, Jiarui Lu, Baoyi Fan, Wenfeng Lu, Yiwei Xue, Meiting Wang, Taigang Liu, Shaoli Cui, Qinghe Gao, Yingchao Duan and Yongtao Xu
Molecules 2024, 29(2), 550; https://doi.org/10.3390/molecules29020550 - 22 Jan 2024
Cited by 4 | Viewed by 3343
Abstract
Lysine-specific demethylase 1 (LSD1/KDM1A) has emerged as a promising therapeutic target for treating various cancers (such as breast cancer, liver cancer, etc.) and other diseases (blood diseases, cardiovascular diseases, etc.), owing to its observed overexpression, thereby presenting significant opportunities in drug development. Since [...] Read more.
Lysine-specific demethylase 1 (LSD1/KDM1A) has emerged as a promising therapeutic target for treating various cancers (such as breast cancer, liver cancer, etc.) and other diseases (blood diseases, cardiovascular diseases, etc.), owing to its observed overexpression, thereby presenting significant opportunities in drug development. Since its discovery in 2004, extensive research has been conducted on LSD1 inhibitors, with notable contributions from computational approaches. This review systematically summarizes LSD1 inhibitors investigated through computer-aided drug design (CADD) technologies since 2010, showcasing a diverse range of chemical scaffolds, including phenelzine derivatives, tranylcypromine (abbreviated as TCP or 2-PCPA) derivatives, nitrogen-containing heterocyclic (pyridine, pyrimidine, azole, thieno[3,2-b]pyrrole, indole, quinoline and benzoxazole) derivatives, natural products (including sanguinarine, phenolic compounds and resveratrol derivatives, flavonoids and other natural products) and others (including thiourea compounds, Fenoldopam and Raloxifene, (4-cyanophenyl)glycine derivatives, propargylamine and benzohydrazide derivatives and inhibitors discovered through AI techniques). Computational techniques, such as virtual screening, molecular docking and 3D-QSAR models, have played a pivotal role in elucidating the interactions between these inhibitors and LSD1. Moreover, the integration of cutting-edge technologies such as artificial intelligence holds promise in facilitating the discovery of novel LSD1 inhibitors. The comprehensive insights presented in this review aim to provide valuable information for advancing further research on LSD1 inhibitors. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The structure of LSD1 (PDB code: 6TUY) composed of three major domains, namely the N-terminal SWIRM domain (green), Tower domain (pink) and the C-terminal amino oxidase-like (AOL) domain (cyan).</p>
Full article ">Figure 2
<p>The structures of phenelzine derivatives. (<b>A</b>) Compound <b>1</b>; K<sub>i(inact)</sub> is the apparent maximum inactivation rate. (<b>B</b>) Compound <b>2</b> and the binding mode with LSD1; the key amino acids are illustrated, and orange dash lines represent the hydrogen bond interactions (Reprinted with permission from Ref. [<a href="#B40-molecules-29-00550" class="html-bibr">40</a>]. Copyright 2015 Taylor &amp; Francis). (<b>C</b>) Compound <b>3</b> and compound <b>4</b>; IC<sub>50</sub> is half maximal inhibitory concentration.</p>
Full article ">Figure 3
<p>The structures of tranylcypromine derivatives. (<b>A</b>) Compounds <b>5</b> and <b>6</b>; (<b>B</b>) the co-crystal structural comparison of MAO-B/compound <b>5</b> complex and LSD1/compound <b>5</b> complex (reprinted with permission from Ref. [<a href="#B50-molecules-29-00550" class="html-bibr">50</a>] Copyright 2010 American Chemical Society); (<b>C</b>) Compound <b>7</b> and the 2-PCPA benzene ring formed stable hydrophobic interactions with the surrounding residues (reprinted with permission from Ref. [<a href="#B50-molecules-29-00550" class="html-bibr">50</a>] Copyright 2010 American Chemical Society); (<b>D</b>) compound <b>8</b> and predicted binding model of compound <b>8</b> with LSD1 (reprinted with permission from Ref. [<a href="#B51-molecules-29-00550" class="html-bibr">51</a>]. Copyright 2017 Elsevier); (<b>E</b>) compound <b>9</b> and complex structure of LSD1 upon binding to compound <b>9</b> (reprinted with permission from Ref. [<a href="#B52-molecules-29-00550" class="html-bibr">52</a>]. Copyright 2022 American Chemical Society).</p>
Full article ">Figure 4
<p>The structures of pyridine derivatives. (<b>A</b>) Compound <b>10</b> (GSK-690) and compound <b>11</b>; (<b>B</b>) complex structure of LSD1 upon binding to compound <b>11</b> ((<b>left</b>), PDB code: 2V1D, reprinted with permission from Ref. [<a href="#B55-molecules-29-00550" class="html-bibr">55</a>]. Copyright 2016 American Chemical Society) and superposition of molecular docking result with the average structure during MD of compound <b>11</b> ((<b>right</b>), reprinted with permission from Ref. [<a href="#B56-molecules-29-00550" class="html-bibr">56</a>]. Copyright 2018 Taylor &amp; Francis); (<b>C</b>) compounds <b>12</b>–<b>16</b>.</p>
Full article ">Figure 5
<p>The structure of pyrimidine derivatives. (<b>A</b>) Compound <b>17</b> (<b>left</b>), the docking result ((<b>middle</b>), PDB code: 2H94) and the binding mode of compound <b>17</b> with LSD1 after MD simulation ((<b>right</b>), reprinted with permission from Ref. [<a href="#B64-molecules-29-00550" class="html-bibr">64</a>]. Copyright 2017 Elsevier); (<b>B</b>) compound <b>18</b> (Osimertinib) and compound <b>19,</b> and the binding mode of compound <b>19</b> with LSD1 (right, green dash lines represent the hydrogen bond interactions, yellow dash line is π–π stacking, reprinted with permission from Ref. [<a href="#B66-molecules-29-00550" class="html-bibr">66</a>]. Copyright 2022 Elsevier); (<b>C</b>) the structures of compound <b>20</b> and compound <b>21</b>; (<b>D</b>) the docking mode of compound <b>20</b> with LSD1 (adapted from Ref. [<a href="#B67-molecules-29-00550" class="html-bibr">67</a>]); (<b>E</b>) the co-crystal structure of compound <b>21</b> in complex with LSD1 (PDB code: 6W4K, reprinted from Ref. [<a href="#B67-molecules-29-00550" class="html-bibr">67</a>]).</p>
Full article ">Figure 6
<p>The structures of triazole derivatives (<b>A</b>) compounds <b>22</b> and <b>23</b>; (<b>B</b>) two conformations of triazole derivatives at the binding site (type A, the triazole moiety in close proximity to pocket 1, (<b>left</b>); type B, the triazole moiety in close proximity to pocket 2, (<b>right</b>), reprinted with permission from Ref. [<a href="#B73-molecules-29-00550" class="html-bibr">73</a>]. Copyright 2018 Royal Society of Chemistry); (<b>C</b>) compounds <b>24</b>–<b>27</b>.</p>
Full article ">Figure 7
<p>The structures of triazole derivatives. (<b>A</b>) Compounds <b>28</b> and <b>29</b>. (<b>B</b>) Complex structure of LSD1 upon binding to compound <b>28</b> (PDB code: 3ZMT, reprinted with permission from Ref. [<a href="#B79-molecules-29-00550" class="html-bibr">79</a>]. Copyright 2018 Royal Society of Chemistry); the key amino acids are illustrated. (<b>C</b>) Compounds <b>30</b> and <b>31</b>; (<b>D</b>) 2D diagram of the interaction between compound <b>30</b> and LSD1 (reprinted from Ref. [<a href="#B80-molecules-29-00550" class="html-bibr">80</a>]); (<b>E</b>) surface map for the compound <b>30</b> inside active site (reprinted from Ref. [<a href="#B80-molecules-29-00550" class="html-bibr">80</a>]).</p>
Full article ">Figure 8
<p>The structures of thiazole derivatives, compounds <b>32</b>–<b>36</b>.</p>
Full article ">Figure 9
<p>The structures of thiazole derivatives. (<b>A</b>) Compounds <b>37</b>–<b>42</b>; (<b>B</b>) compound <b>40</b> bound inside the active site of LSD1; yellow dotted line represents electrostatic interactions; pink dotted lines are π–π stacking (reprinted with permission from Ref. [<a href="#B82-molecules-29-00550" class="html-bibr">82</a>]. Copyright 2019 Elsevier).</p>
Full article ">Figure 10
<p>The structures of pyrazole derivatives. (<b>A</b>) Compounds <b>43</b> and <b>44</b>; (<b>B</b>) compound <b>45,</b> along with a 2D diagram depicting its interaction with LSD1 (reprinted with permission from Ref. [<a href="#B85-molecules-29-00550" class="html-bibr">85</a>]. Copyright 2019 Elsevier); (<b>C</b>) compounds <b>46</b> and <b>47</b>.</p>
Full article ">Figure 11
<p>The structures of thieno[3,2-b]pyrrole derivatives, compounds <b>48</b>–<b>52</b>.</p>
Full article ">Figure 12
<p>The superposition of the docking structures (green) and MD average structures (cyan) of LSD1 with (<b>A</b>) compound <b>53</b>, (<b>B</b>) compound <b>54</b> and (<b>C</b>) compound <b>55</b>, respectively (reprinted from Ref. [<a href="#B88-molecules-29-00550" class="html-bibr">88</a>]).</p>
Full article ">Figure 13
<p>The structures of indole derivatives. (<b>A</b>) Compounds <b>56</b> and <b>57</b>; (<b>B</b>) complex structure of LSD1 upon binding to compound <b>57</b>; the key residues are labeled (reprinted with permission from Ref. [<a href="#B94-molecules-29-00550" class="html-bibr">94</a>]. Copyright 2018 Elsevier). (<b>C</b>) Compound <b>57</b> in the pocket cavity of LSD1 (reprinted with permission from Ref. [<a href="#B94-molecules-29-00550" class="html-bibr">94</a>]. Copyright 2018 Elsevier). (<b>D</b>) Compounds <b>58</b> and <b>59</b>; (<b>E</b>) compounds <b>60</b> and <b>61</b>; (<b>F</b>) complex structure of LSD1 upon binding to compound <b>61</b> (PDB code: 5YJB); green dash lines represent hydrogen bond interactions; yellow dash lines are π–π stacking (reprinted with permission from Ref. [<a href="#B96-molecules-29-00550" class="html-bibr">96</a>]. Copyright 2022 Elsevier).</p>
Full article ">Figure 14
<p>The structures and binding mode analysis of quinoline derivatives. (<b>A</b>) Binding mode of LSD1 with compound <b>62</b> (reprinted with permission from Ref. [<a href="#B97-molecules-29-00550" class="html-bibr">97</a>]. Copyright 2020 Elsevier); (<b>B</b>) binding mode of LSD1 with compound <b>63</b> (reprinted with permission from Ref. [<a href="#B97-molecules-29-00550" class="html-bibr">97</a>]. Copyright 2020 Elsevier); (<b>C</b>) compound <b>63</b> and compound <b>64</b>; (<b>D</b>) compound <b>62</b> and design and modification strategy of the target compound; (<b>E</b>) the proposed binding mode of LSD1 with compound <b>65</b> (reprinted with permission from Ref. [<a href="#B98-molecules-29-00550" class="html-bibr">98</a>]. Copyright 2021 John Wiley and Sons); (<b>F</b>) the proposed binding mode of LSD1 with compound <b>72</b> (reprinted with permission from Ref. [<a href="#B98-molecules-29-00550" class="html-bibr">98</a>]. Copyright 2021 John Wiley and Sons); (<b>G</b>) compounds <b>65</b>–<b>72</b>.</p>
Full article ">Figure 15
<p>(<b>A</b>) The structures of phenyloxazole derivatives compounds <b>73</b>–<b>75</b>; (<b>B</b>) binding orientation and (<b>C</b>) 2D diagram of interactions of compound <b>75</b> at the LSD1 binding site (reprinted with permission from Ref. [<a href="#B99-molecules-29-00550" class="html-bibr">99</a>]. Copyright 2013 Royal Society of Chemistry).</p>
Full article ">Figure 16
<p>(<b>A</b>) The structure of compound <b>76</b> (Sanguinarine) and Epiberberine; (<b>B</b>) predicted binding mode of compound <b>76</b> in the active site of LSD1 (PDB: 2V1D, reprinted from Ref. [<a href="#B100-molecules-29-00550" class="html-bibr">100</a>]); (<b>C</b>) overlap of the binding poses of compound <b>76</b>, Epiberberine and FAD (reprinted from Ref. [<a href="#B100-molecules-29-00550" class="html-bibr">100</a>]).</p>
Full article ">Figure 17
<p>(<b>A</b>) The structures of phenolic compounds of compounds <b>77</b>–<b>84</b>; (<b>B</b>) complex structure of LSD1 upon binding to compound <b>77</b> (PDB: 2IW5, reprinted from Ref. [<a href="#B110-molecules-29-00550" class="html-bibr">110</a>]).</p>
Full article ">Figure 18
<p>(<b>A</b>) The structures of resveratrol derivatives: compounds <b>85</b>–<b>87</b>; (<b>B</b>) complex structure of LSD1 upon binding to compound <b>85</b>; key amino acid residues and interactions are indicated (reprinted with permission from Ref. [<a href="#B113-molecules-29-00550" class="html-bibr">113</a>]. Copyright 2017 Elsevier); (<b>C</b>) Docking diagram of compound <b>87</b> with LSD1 (<b>left</b>) and 2D schematics of the protein–ligand interactions of compound <b>87</b> to LSD1 (<b>right</b>) (reprinted with permission from Ref. [<a href="#B114-molecules-29-00550" class="html-bibr">114</a>]. Copyright 2018 Elsevier).</p>
Full article ">Figure 19
<p>(<b>A</b>) 3D-QSAR contour maps visualize the effect of the introduced substituents on the biological activity (reprinted with permission from Ref. [<a href="#B102-molecules-29-00550" class="html-bibr">102</a>]); (<b>B</b>) structure–activity relationship (reprinted with permission from Ref. [<a href="#B102-molecules-29-00550" class="html-bibr">102</a>]); (<b>C</b>) the structures of resveratrol derivatives: compounds <b>88</b>–<b>93</b>.</p>
Full article ">Figure 20
<p>The structures and of flavonoids inhibitors. (<b>A</b>) Compounds <b>94</b> (Baicalin) and <b>95</b> (Wogonoside); (<b>B</b>) 3D docking model of compound <b>95</b> bound to LSD1(reprinted with permission from Ref. [<a href="#B123-molecules-29-00550" class="html-bibr">123</a>]. Copyright 2018 Elsevier); (<b>C</b>) 2D schematic of the docking model of compound <b>95</b> bound to LSD1(reprinted with permission from Ref. [<a href="#B123-molecules-29-00550" class="html-bibr">123</a>]. Copyright 2018 Elsevier); (<b>D</b>) compound <b>96</b> (IQ) and (<b>E</b>) 2D schematic of the docking model bound to LSD1(reprinted with permission from Ref. [<a href="#B124-molecules-29-00550" class="html-bibr">124</a>]. Copyright 2019 Elsevier).</p>
Full article ">Figure 21
<p>The structures of natural-product compounds <b>97</b>–<b>100</b>.</p>
Full article ">Figure 22
<p>The structures of thiourea compounds. (<b>A</b>) Compounds <b>101</b>–<b>104</b>; (<b>B</b>) computer-predicted binding mode of compounds <b>104</b> and <b>106</b> in the LSD1 binding site (<b>left</b>), molecular interactions between LSD1 and compound <b>104</b> (<b>right</b>) (reprinted with permission from Ref. [<a href="#B128-molecules-29-00550" class="html-bibr">128</a>]. Copyright 2015 Elsevier); (<b>C</b>) compounds <b>105</b> and <b>106</b>.</p>
Full article ">Figure 23
<p>(<b>A</b>) The structures of compounds <b>107</b> (Fenoldopam) and <b>108</b> (Raloxifene); molecular docking results of (<b>B</b>) compounds <b>107</b> (reprinted with permission from Ref. [<a href="#B130-molecules-29-00550" class="html-bibr">130</a>]. Copyright 2021 Elsevier) and (<b>C</b>) <b>108</b> bonding to LSD1; hydrogen bonds and their distances are shown (reprinted from Ref. [<a href="#B131-molecules-29-00550" class="html-bibr">131</a>]). (<b>D</b>) Compounds <b>107</b> (reprinted with permission from Ref. [<a href="#B130-molecules-29-00550" class="html-bibr">130</a>]. Copyright 2021 Elsevier) and (<b>E</b>) <b>108</b> were buried in a hydrophobic pocket of LSD1 (reprinted from Ref. [<a href="#B131-molecules-29-00550" class="html-bibr">131</a>]); (<b>F</b>) 2D diagram depicting their interaction (<b>up</b>, compounds <b>107</b> reprinted with permission from Ref. [<a href="#B130-molecules-29-00550" class="html-bibr">130</a>]. Copyright 2021 Elsevier and <b>108</b>, <b>down</b>, reprinted from Ref. [<a href="#B131-molecules-29-00550" class="html-bibr">131</a>]) with LSD1.</p>
Full article ">Figure 24
<p>The structures of (4-cyanophenyl)glycine derivatives. (<b>A</b>) Compounds <b>109</b> and <b>110</b>; (<b>B</b>) 2D diagram depicting interaction of compound <b>109</b> with LSD1 (reprinted with permission from Ref. [<a href="#B132-molecules-29-00550" class="html-bibr">132</a>]. Copyright 2017 American Chemical Society); (<b>C</b>) compounds <b>111</b>–<b>114</b>.</p>
Full article ">Figure 25
<p>The proposed binding modes of LSD1 with compounds (<b>A</b>) <b>111</b>, (<b>B</b>) <b>112</b>, (<b>C</b>) <b>113</b> and (<b>D</b>) <b>114</b>, respectively (reprinted with permission from Ref. [<a href="#B133-molecules-29-00550" class="html-bibr">133</a>]. Copyright 2019 Elsevier).</p>
Full article ">Figure 26
<p>(<b>A</b>) The structures of propargylamine derivatives: compounds <b>115</b>–<b>117</b>; complex structure of LSD1 upon binding to (<b>B</b>) compounds <b>115</b> (reprinted with permission from Ref. [<a href="#B135-molecules-29-00550" class="html-bibr">135</a>]. Copyright 2013 American Chemical Society) and (<b>C</b>) <b>116</b> (reprinted with permission from Ref. [<a href="#B135-molecules-29-00550" class="html-bibr">135</a>]. Copyright 2013 American Chemical Society).</p>
Full article ">Figure 27
<p>The structures of benzoylhydrazine derivatives. (<b>A</b>) Compounds <b>118</b>–<b>123</b>; (<b>B</b>) compounds <b>124</b>–<b>126</b>; (<b>C</b>) binding mode of compound <b>124</b> (reprinted with permission from Ref. [<a href="#B139-molecules-29-00550" class="html-bibr">139</a>]. Copyright 2016 Elsevier) and (<b>D</b>) compound <b>126</b> with LSD1; key residues are shown (reprinted with permission from Ref. [<a href="#B139-molecules-29-00550" class="html-bibr">139</a>]. Copyright 2016 Elsevier).</p>
Full article ">Figure 28
<p>The structures of LSD1 inhibitors discovered through artificial intelligence techniques. (<b>A</b>) Compounds <b>127</b>–<b>131</b> (with the predicted IC<sub>50</sub>); (<b>B</b>) compounds <b>132</b>–<b>136</b>.</p>
Full article ">
10 pages, 2146 KiB  
Article
The Interplay of Raloxifene and Sonochemical Bio-Oss in Early Maxillary Sinus Bone Regeneration: A Histological and Immunohistochemical Analysis in Rabbits
by Anderson Maikon de Souza Santos, Rodrigo dos Santos Pereira, Pietro Montemezzi, Rafael Coutinho Mello-Machado, Roberta Okamoto, Roberto Sacco, Paulo Noronha Lisboa-Filho, Michel Reis Messora, Carlos Fernando Mourão and Eduardo Hochuli-Vieira
Medicina 2023, 59(9), 1521; https://doi.org/10.3390/medicina59091521 - 23 Aug 2023
Cited by 1 | Viewed by 1783
Abstract
The study aimed to assess the efficacy of using Raloxifene with ultrasonic processing to enhance Bio-Oss®, a bone graft substitute, for maxillary sinus bone height reconstruction. A total of 24 rabbit maxillary sinuses were distributed into three groups, each receiving different [...] Read more.
The study aimed to assess the efficacy of using Raloxifene with ultrasonic processing to enhance Bio-Oss®, a bone graft substitute, for maxillary sinus bone height reconstruction. A total of 24 rabbit maxillary sinuses were distributed into three groups, each receiving different treatments: Bio-Oss® only, sonicated Bio-Oss, and sonicated Bio-Oss® with Raloxifene. Surgical procedures and subsequent histomorphometric and immunohistochemistry analyses were conducted to evaluate the bone formation, connective tissue, and remaining biomaterial, as well as the osteoblastic differentiation and maturation of collagen fibers. Results indicated that the sonicated Bio-Oss® and Bio-Oss® groups showed similar histological behavior and bone formation, but the Raloxifene group displayed inflammatory infiltrate, low bone formation, and disorganized connective tissue. The statistical analysis confirmed significant differences between the groups in terms of bone formation, connective tissue, and remaining biomaterial. In conclusion, the study found that while sonicated Bio-Oss® performed comparably to Bio-Oss® alone, the addition of Raloxifene led to an unexpected delay in bone repair. The findings stress the importance of histological evaluation for accurate bone repair assessment and the necessity for further investigation into the local application of Raloxifene. Future research may focus on optimizing bone substitutes with growth factors to improve bone repair. Full article
(This article belongs to the Special Issue Advances in Oral Surgery and Implant Dentistry)
Show Figures

Figure 1

Figure 1
<p>Comparative evaluation of the efficacy of three distinct biomaterials (BO, BS, and BR) in the process of bone formation. The chart provides a quantified representation of the bone formation (<b>A</b>), connective tissue (<b>B</b>), and biomaterial remaining (<b>C</b>) percentages for each group. Considering the significance * <span class="html-italic">p</span> &lt; 0.05 and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Slides of the biological responses across three biomaterial groups: BO, BS, and BR. (<b>A1</b>,<b>B1</b>,<b>C1</b>) lines show the histological evaluation (arrows mean new bone formed; * means connective tissue; and # means biomaterial remaining). (<b>A2</b>,<b>B2</b>,<b>C2</b>) lines show the results from immunohistochemistry demonstrating varying levels of Runx 2 staining in the groups evaluated (red arrows indicating positive immunostaining for Runx 2 cells; and (<b>A3</b>,<b>B3</b>,<b>C3</b>) line showing the collagen fiber maturation analysis).</p>
Full article ">
19 pages, 341 KiB  
Review
Chemoprevention and Lifestyle Modifications for Risk Reduction in Sporadic and Hereditary Breast Cancer
by Eliza Del Fiol Manna, Davide Serrano, Gaetano Aurilio, Bernardo Bonanni and Matteo Lazzeroni
Healthcare 2023, 11(16), 2360; https://doi.org/10.3390/healthcare11162360 - 21 Aug 2023
Cited by 3 | Viewed by 2483
Abstract
Female breast cancer is the most commonly diagnosed malignancy worldwide. Risk assessment helps to identify women at increased risk of breast cancer and allows the adoption of a comprehensive approach to reducing breast cancer incidence through personalized interventions, including lifestyle modification, chemoprevention, intensified [...] Read more.
Female breast cancer is the most commonly diagnosed malignancy worldwide. Risk assessment helps to identify women at increased risk of breast cancer and allows the adoption of a comprehensive approach to reducing breast cancer incidence through personalized interventions, including lifestyle modification, chemoprevention, intensified surveillance with breast imaging, genetic counseling, and testing. Primary prevention means acting on modifiable risk factors to reduce breast cancer occurrence. Chemoprevention with tamoxifen, raloxifene, anastrozole, and exemestane has already shown benefits in decreasing breast cancer incidence in women at an increased risk for breast cancer. For healthy women carrying BRCA 1 or BRCA 2 pathogenic/likely pathogenic (P/LP) germline variants, the efficacy of chemoprevention is still controversial. Adopting chemoprevention strategies and the choice among agents should depend on the safety profile and risk–benefit ratio. Unfortunately, the uptake of these agents has been low. Lifestyle modifications can reduce breast cancer incidence, and the recommendations for BRCA 1 or BRCA 2 P/LP germline variant carriers are comparable to the general population. This review summarizes the most recent evidence regarding the efficacy of chemoprevention and lifestyle interventions in women with sporadic and hereditary breast cancer. Full article
(This article belongs to the Special Issue Breast Cancer Prevention in Healthcare: A Comprehensive Overview)
20 pages, 4500 KiB  
Article
Formulation and Evaluation of a Self-Microemulsifying Drug Delivery System of Raloxifene with Improved Solubility and Oral Bioavailability
by Muhammad Mohsin Ansari, Dang-Khoa Vo, Ho-Ik Choi, Jeong-Su Ryu, Yumi Bae, Nadeem Irfan Bukhari, Alam Zeb, Jin-Ki Kim and Han-Joo Maeng
Pharmaceutics 2023, 15(8), 2073; https://doi.org/10.3390/pharmaceutics15082073 - 2 Aug 2023
Cited by 7 | Viewed by 3030
Abstract
Poor aqueous solubility and dissolution limit the oral bioavailability of Biopharmaceutics Classification System (BCS) class II drugs. In this study, we aimed to improve the aqueous solubility and oral bioavailability of raloxifene hydrochloride (RLX), a BCS class II drug, using a self-microemulsifying drug [...] Read more.
Poor aqueous solubility and dissolution limit the oral bioavailability of Biopharmaceutics Classification System (BCS) class II drugs. In this study, we aimed to improve the aqueous solubility and oral bioavailability of raloxifene hydrochloride (RLX), a BCS class II drug, using a self-microemulsifying drug delivery system (SMEDDS). Based on the solubilities of RLX, Capryol 90, Tween 80/Labrasol ALF, and polyethylene glycol 400 (PEG-400) were selected as the oil, surfactant mixture, and cosurfactant, respectively. Pseudo-ternary phase diagrams were constructed to determine the optimal composition (Capryol 90/Tween 80/Labrasol ALF/PEG-400 in 150/478.1/159.4/212.5 volume ratio) for RLX-SMEDDS with a small droplet size (147.1 nm) and stable microemulsification (PDI: 0.227). Differential scanning calorimetry and powder X-ray diffraction of lyophilized RLX-SMEDDS revealed the loss of crystallinity, suggesting a molecularly dissolved or amorphous state of RLX in the SMEDDS formulation. Moreover, RLX-SMEDDS exhibited significantly higher saturation solubility and dissolution rate in water, simulated gastric fluid (pH 1.2), and simulated intestinal fluid (pH 6.8) than RLX powder. Additionally, oral administration of RLX-SMEDDS to female rats resulted in 1.94- and 1.80-fold higher area under the curve and maximum plasma concentration, respectively, than the RLX dispersion. Collectively, our findings suggest SMEDDS is a promising oral formulation to enhance the therapeutic efficacy of RLX. Full article
(This article belongs to the Special Issue Micro- and Nano-Emulsions as Drug Delivery Systems)
Show Figures

Figure 1

Figure 1
<p>Pseudo-ternary phase diagrams of the self-microemulsifying drug delivery system (SMEDDS) prepared using Caproyl-90 as an oil, Tween 80/Labrasol ALF as a surfactant mixture, and polyethylene glycol (PEG)-400 as a cosurfactant. Different surfactant to cosurfactant ratios (Km) of 3:1 (<b>A</b>), 2:1 (<b>B</b>), 1:1 (<b>C</b>), 1:2 (<b>D</b>), and 1:3 (<b>E</b>) were investigated. Gray regions in pseudo-ternary phase diagrams indicate the microemulsion phase.</p>
Full article ">Figure 2
<p>Droplet size distribution curve of the optimized formulation of liquid RLX-incorporated SMEDDS (RLX-SMEDDS) measured using the dynamic light scattering (DLS) technique after 100-times dilution with double distilled water.</p>
Full article ">Figure 3
<p>Scanning electron microscopy (SEM) micrographs of RLX (<b>A</b>), mannitol (<b>B</b>), and lyophilized RLX-SMEDDS (<b>C</b>).</p>
Full article ">Figure 4
<p>Differential scanning calorimetry (DSC) thermograms (<b>A</b>) and powder X-ray diffraction (PXRD) patterns (<b>B</b>) of RLX (a), mannitol (b), physical mixture (c), and lyophilized RLX-SMEDDS (d).</p>
Full article ">Figure 5
<p>Fourier-transform infrared (FTI)R spectra (<b>A</b>) of RLX powder (a), mannitol (b), physical mixture (c), and lyophilized RLX-SMEDDS (d), and chemical structures of RLX (<b>B</b>) and mannitol (<b>C</b>).</p>
Full article ">Figure 6
<p>Saturation solubilities of RLX and lyophilized RLX-SMEDDS in water, simulated gastric fluid (SGF; pH 1.2), and simulated intestinal fluid (SIF; pH 6.8). Data are represented as the mean ± S.D. (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 7
<p>Dissolution profiles of RLX and lyophilized RLX-SMEDDS in water (<b>A</b>), simulated gastric fluid (pH 1.2) (<b>B</b>), and simulated intestinal fluid (pH 6.8) (<b>C</b>). Data are represented as the mean ± S.D. (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 8
<p>Average RLX plasma concentration–time profiles after the oral administration of RLX dispersion and RLX-SMEDDS to female rats at a dose equivalent to 10 mg/kg. Data are represented as the mean ± S.D. (<span class="html-italic">n</span> = 4).</p>
Full article ">
19 pages, 3612 KiB  
Article
Neuroprotective Potential of Raloxifene via G-Protein-Coupled Estrogen Receptors in Aβ-Oligomer-Induced Neuronal Injury
by Tetsuhito Nohara, Mayumi Tsuji, Tatsunori Oguchi, Yutaro Momma, Hideaki Ohashi, Miki Nagata, Naohito Ito, Ken Yamamoto, Hidetomo Murakami and Yuji Kiuchi
Biomedicines 2023, 11(8), 2135; https://doi.org/10.3390/biomedicines11082135 - 28 Jul 2023
Cited by 6 | Viewed by 2122
Abstract
Amyloid-β (Aβ) is one of the causes of Alzheimer’s disease (AD), damaging nerve membranes and inducing neurotoxicity. AD is more prevalent in female patients than in male patients, and women are more susceptible to developing AD due to the decline in estrogen levels [...] Read more.
Amyloid-β (Aβ) is one of the causes of Alzheimer’s disease (AD), damaging nerve membranes and inducing neurotoxicity. AD is more prevalent in female patients than in male patients, and women are more susceptible to developing AD due to the decline in estrogen levels around menopause. Raloxifene, a selective estrogen receptor modulator, exhibits protective effects by activating the transmembrane G-protein-coupled estrogen receptor (GPER). Additionally, raloxifene prevents mild cognitive impairment and restores cognition. However, the influence of raloxifene via GPER on highly toxic Aβ-oligomers (Aβo)-induced neurotoxicity remains uncertain. In this study, we investigated the GPER-mediated neuroprotective effects of raloxifene against the neurotoxicity caused by Aβo-induced cytotoxicity. The impact of raloxifene on Aβo-induced cell damage was evaluated using measures such as cell viability, production of reactive oxygen species (ROS) and mitochondrial ROS, peroxidation of cell-membrane phospholipids, and changes in intracellular calcium ion concentration ([Ca2+]i) levels. Raloxifene hindered Aβo-induced oxidative stress and reduced excessive [Ca2+]i, resulting in improved cell viability. Furthermore, these effects of raloxifene were inhibited with pretreatment with a GPER antagonist. Our findings suggest that raloxifene safeguards against Aβo-induced neurotoxicity by modifying oxidative parameters and maintaining [Ca2+]i homeostasis. Raloxifene may prove effective in preventing and inhibiting the progression of AD. Full article
(This article belongs to the Special Issue Advanced Diagnostic and Treatment Methods in Alzheimer's Disease)
Show Figures

Figure 1

Figure 1
<p>Structures of raloxifene and 17β-estradiol.</p>
Full article ">Figure 2
<p>Separation and collection of Aβ molecular species with HPLC.</p>
Full article ">Figure 3
<p>Effects of raloxifene and estradiol on Aβ<sub>1–42</sub> (<b>A</b>,<b>B</b>) and Aβ<sub>1–40</sub> (<b>C</b>,<b>D</b>) aggregation kinetics. Aggregation kinetics of Aβ<sub>1–40</sub> and Aβ<sub>1–42</sub> peptides measured using thioflavin T fluorescence and shown as 100% of the fluorescence intensity at the start of the measurement. (<b>A</b>) Aβ<sub>1−42</sub> (25 μM) with 0.1% DMSO or raloxifene (1, 5, 10, and 20 μM). (<b>B</b>) Aβ<sub>1−42</sub> (25 μM) with 0.1% DMSO or estradiol (1, 5, 10, and 20 μM). (<b>C</b>) Aβ<sub>1−40</sub> (25 μM) with 0.1% DMSO or raloxifene (1, 5, 10, and 20 μM). (<b>D</b>) Aβ<sub>1−40</sub> (25 μM) with 0.1% DMSO or estradiol (1, 5, 10, and 20 μM). The <span class="html-italic">p</span>-values in ANOVA were &lt;0.001. Each value represents the mean ± standard error of the mean (SEM) (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 4
<p>Changes in cell viability in response to Aβo-stimulated SH-SY5Y cells. Viability of Aβo-stimulated SH-SY5Y cells was assessed using the MTT assay. (<b>A</b>) SH-SY5Y cells were exposed to Aβo (0.5, 1, 5, and 10 μM) for 3 h. #: <span class="html-italic">p</span> &lt; 0.0001 for control versus Aβo-exposed cells (<span class="html-italic">n</span> = 8, Dunnett’s). (<b>B</b>) SH-SY5Y cells were exposed to Aβo (5 μM) and treated with Aβo + raloxifene (0.5, 1, and 5 μM) or Aβo + estradiol (0.5, 1, and 5 μM) for 3 h. (<b>C</b>,<b>D</b>) SH-SY5Y cells were pretreated with GPER antagonist (G-15) and nuclear estrogen receptor antagonist (fulvestrant) and treated with Aβo + raloxifene or Aβo + estradiol. +: inclusion of 5 μM Aβo, raloxifene, estradiol; −: non-inclusion. The <span class="html-italic">p</span>-values in ANOVA were &lt;0.001. Results are expressed as means + SEMs of 10 individually treated samples. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.0001 vs. 5 μM Aβo-exposed cells (<span class="html-italic">n</span> = 10, Tukey’s).</p>
Full article ">Figure 5
<p>Effect of raloxifene and estradiol on cytotoxicity in Aβo-stimulated SH-SY5Y cells. Cytotoxicity of Aβo-stimulated SH-SY5Y cells was stained with ethidium homodimer-1. (<b>A</b>) SH-SY5Y cells were exposed to Aβo (5 μM) and treated with Aβo + raloxifene or Aβo + estradiol (5 μM) for 3 h. In the absence of 5 μM Aβo, the cytotoxicity values of control, 5 μM raloxifene-treated, 5 μM estradiol-treated cells were 5.46 ± 0.6, 5.57± 0.79, and 7.15 ± 0.91%, respectively (no significant differences, <span class="html-italic">n</span> = 10, Tukey’s). The <span class="html-italic">p</span>-values in ANOVA were &lt;0.001. Measurements were expressed as the means of 10 individually treated samples + SEM. *: <span class="html-italic">p</span> &lt; 0.0005 vs. Aβo. (<b>B</b>–<b>I</b>) Cytotoxicity of Aβo-stimulated SH-SY5Y cells stained with calcein-AM/ethidium homodimer-1 was observed using phase-contrast microscopy (<b>B</b>–<b>E</b>) and fluorescence microscopy (<b>F</b>–<b>I</b>). Cytotoxicity of Aβo-stimulated SH-SY5Y cells stained only with ethidium homodimer-1 is shown as gray-scale images (<b>J</b>–<b>M</b>). Scale bars represent 100 μm.</p>
Full article ">Figure 6
<p>Effect of raloxifene and estradiol on ROS production in Aβo-stimulated SH-SY5Y cells. (<b>A</b>) ROS production in SH-SY5Y cells exposed to Aβo was evaluated with CM-H2DCFDA. ROS production in SH-SY5Y cells treated with 5 μM Aβo and 5 μM raloxifene or 5 μM estradiol for 3 h after 5 μM G-15 pretreatment for 30 min was examined. The <span class="html-italic">p</span>-values in ANOVA were &lt;0.0001. Measurements were expressed as the means + SEMs of 10 individually treated samples. In the absence of 5 μM Aβo, ROS levels of 5 μM raloxifene-treated and 5 μM estradiol-treated cells were 17.79 ± 1.89 and 19.04 ± 1.36 fluorescence intensity × 10<sup>5</sup>/μg protein, respectively (no significant differences, <span class="html-italic">n</span> = 10, Tukey’s). *: <span class="html-italic">p</span> &lt; 0.01 vs. 5 μM Aβo. (<b>B</b>–<b>I</b>) ROS production was observed using phase-contrast microscopy (<b>B</b>–<b>E</b>) and fluorescence microscopy (<b>F</b>–<b>I</b>). (<b>J</b>–<b>Q</b>) ROS production under G-15 pretreatment was observed using phase-contrast microscopy (<b>J</b>–<b>M</b>) and fluorescence microscopy (<b>N</b>–<b>Q</b>). Scale bars represent 100 μm.</p>
Full article ">Figure 7
<p>Effect of raloxifene and estradiol on mitochondrial ROS production in SH-SY5Y cells exposed to Aβo. (<b>A</b>) SH-SY5Y cells treated with 5 µM Aβo and 5 µM raloxifene or 5 µM estradiol for 3 h were examined. Moreover, cells were pretreated with 5 µM G-15 for 30 min and treated with Aβo + raloxifene or Aβo + estradiol for 3 h. (<b>B</b>) SH-SY5Y cells treated with 5 µM Aβo and 5 µM raloxifene or 5 µM estradiol for 24 h were examined. Moreover, cells were pretreated with 5 µM G-15 for 30 min and treated with Aβo + raloxifene or Aβo + estradiol for 24 h. The <span class="html-italic">p</span>-values in ANOVA were &lt;0.0001. Measurements were expressed as means + SEMs of 8 individually treated samples. In the absence of 5 μM Aβo, mitochondrial ROS levels of 5 μM raloxifene-treated and 5 μM estradiol-treated cells were 7.67 ± 2.85 and 8.93 ± 4.12 fluorescence intensity × 10<sup>5</sup>/μg protein, respectively (no significant differences, <span class="html-italic">n</span> = 10, Tukey’s). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001 vs. 5 μM Aβo.</p>
Full article ">Figure 8
<p>Effect of raloxifene and estradiol on membrane phospholipid peroxidation levels in Aβo-exposed SH-SY5Y cells. (<b>A</b>) Membrane phospholipid peroxidation level in Aβo-stimulated SH-SY5Y cells was evaluated with DPPP. SH-SY5Y cells exposed to Aβo (5 μM) and treated with Aβo + 5 μM raloxifene or Aβo + 5 μM estradiol for 30 min were examined. (<b>B</b>) Membrane phospholipid peroxidation levels in Aβo-stimulated SH-SY5Y cells were evaluated using DPPP. SH-SY5Y cells exposed to Aβo (5 μM) and treated with Aβo + 5 μM raloxifene or Aβo + 5 μM estradiol for 3 h were examined. Measurements were expressed as the means + SEMs of 8 individually treated samples. In the absence of 5 μM Aβo, membrane phospholipid peroxidation levels of 5 μM raloxifene-treated and 5 μM estradiol-treated cells were 18.82 ± 0.86 and 19.00 ± 1.64 fluorescence intensity × 10<sup>5</sup>/μg protein, respectively (no significant differences, <span class="html-italic">n</span> = 8, Tukey’s). The <span class="html-italic">p</span>-values in ANOVA were &lt;0.0001. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001 vs. 5 μM Aβo (<span class="html-italic">n</span> = 8, Tukey’s). (<b>C</b>–<b>J</b>) membrane phospholipid peroxidation was also observed using phase-contrast microscopy (<b>C</b>–<b>F</b>) and fluorescence microscopy (<b>G</b>–<b>J</b>). Scale bars represent 100 μm. The fluorescence intensity values are shown as gray-scale images.</p>
Full article ">Figure 9
<p>Effect of raloxifene and estradiol on intracellular ionized calcium concentration ([Ca<sup>2+</sup>]<sub>i</sub>) in SH−SY5Y cells. (<b>A</b>) The change in [Ca<sup>2+</sup>]<sub>i</sub> in SH-SY5Y cells after the addition of Aβo was measured using fluorescence intensity for 300 sec. SH-SY5Y cells were exposed to 5 μM Aβo in Ca<sup>2+</sup>-containing or Ca<sup>2+</sup>-less buffer. Furthermore, SH-SY5Y cells were exposed to 5 μM Aβo after calcium-channel antagonist (10 μM nicardipine) or an NMDA receptor blocker (10 μM MK801) pretreatment for 10 min. (<b>B</b>) SH-SY5Y cells were supplemented with raloxifene or estradiol after G-15 pretreatment or no pretreatment. (<b>C</b>) SH-SY5Y cells treated with raloxifene were exposed to 5 μM Aβo after G-15 pretreatment or no pretreatment. (<b>D</b>) SH-SY5Y cells treated with estradiol were exposed to 5 μM Aβo after G-15 pretreatment or no pretreatment. The time of administration is indicated by an arrow. Fluorescence intensity was expressed as 100% of the value at the onset.</p>
Full article ">Figure 10
<p>Schematic of the neurotoxic mechanism of Aβo and inhibition by raloxifene.</p>
Full article ">
11 pages, 2187 KiB  
Article
Hemostatic Effects of Raloxifene in Ovariectomized Rats
by Denys Alva-Chavarría, Maribel Soto-Núñez, Edgar Flores-Soto and Ruth Jaimez
Life 2023, 13(7), 1612; https://doi.org/10.3390/life13071612 - 23 Jul 2023
Cited by 1 | Viewed by 1578
Abstract
This study aimed to explore the effects of raloxifene (Rx) and estradiol (E2) on prothrombin time (PT), partial thromboplastin time (APTT), coagulation factors (VII, X, XI), and fibrinogen concentrations in rats. Female rats were ovariectomized 11 days prior to starting the [...] Read more.
This study aimed to explore the effects of raloxifene (Rx) and estradiol (E2) on prothrombin time (PT), partial thromboplastin time (APTT), coagulation factors (VII, X, XI), and fibrinogen concentrations in rats. Female rats were ovariectomized 11 days prior to starting the treatment. Afterward, they received Rx or E2 (1, 10, 100, and 1000 µg/kg) or propylene glycol (0.3 mL; vehicle, V) subcutaneously for 3 consecutive days. Plasma was collected to measure the hemostatic parameters. Rx significantly increased PT (8%, at 1000 µg/kg; p < 0.05) and APTT at all doses evaluated (32, 70, 67, 30%; p < 0.05, respectively). Rx (1, 10, 100, and 1000 µg/kg) decreased the activity of factor VII by −20, −40, −37, and −17% (p < 0.05), respectively, and E2 increased it by 9, 34, 52, and 29%. Rx reduced factor X activity at 10 and 100 µg/kg doses (−30, and −30% p < 0.05), and E2 showed an increment of 24% with 1000 µg/kg dose only. Additionally, Rx (1, 10, 100 µg/kg) diminished FXI activity (−71, −62, −66; p < 0.05), E2 (1 and 10 µg/kg) in −60 and −38, respectively (p < 0.05), and Rx (1000 µg/kg) produced an increment of 29% (p < 0.05) in fibrinogen concentration, but not E2. Our findings suggest that raloxifene has a protective effect on hemostasis in rats. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

Figure 1
<p>Raloxifene (⧫) and estradiol (•) on prothrombin (<b>A</b>) and thromboplastin (<b>B</b>) time. Each point represents the mean ± SEM of 15 determinations. * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle (o) Dunnett’s test.</p>
Full article ">Figure 2
<p>Raloxifene (⧫) and estradiol (•) on the activity of coagulation factors VII (<b>A</b>) and X (<b>B</b>). Each point represents the percentage ± SEM of 15 determinations. * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle (o) Dunnett´s test.</p>
Full article ">Figure 3
<p>Raloxifene (⧫) and estradiol (•) on the activity of coagulation factor XI (<b>A</b>) and fibrinogen concentrations (<b>B</b>). Each point represents the percentage ± SEM of 15 determinations. * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle (o) Dunnett´s test.</p>
Full article ">Figure 4
<p>Raloxifene and estradiol have contrasting effects on hemostasis and thrombosis due to their interactions with estrogen receptors and their impact on various components of the coagulation system. The orange arrows represent the effect of raloxifene while the green arrows represent the effect of estradiol. Raloxifene showed an increased-on PT, APTT, and fibrinogen and produced a diminution in factor VII, X, and XI activity. E2 induced an increment of factor VII and X activity and lowered factor XI, whereas it did not show any effect on PT and APTT.</p>
Full article ">
Back to TopTop