[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (37,338)

Search Parameters:
Keywords = death

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 3992 KiB  
Article
Encapsulation of Inositol Hexakisphosphate with Chitosan via Gelation to Facilitate Cellular Delivery and Programmed Cell Death in Human Breast Cancer Cells
by Ilham H. Kadhim, Adeolu S. Oluremi, Bijay P. Chhetri, Anindya Ghosh and Nawab Ali
Bioengineering 2024, 11(9), 931; https://doi.org/10.3390/bioengineering11090931 (registering DOI) - 17 Sep 2024
Abstract
Inositol hexakisphosphate (InsP6) is the most abundant inositol polyphosphate both in plant and animal cells. Exogenous InsP6 is known to inhibit cell proliferation and induce apoptosis in cancerous cells. However, cellular entry of exogenous InsP6 is hindered due to [...] Read more.
Inositol hexakisphosphate (InsP6) is the most abundant inositol polyphosphate both in plant and animal cells. Exogenous InsP6 is known to inhibit cell proliferation and induce apoptosis in cancerous cells. However, cellular entry of exogenous InsP6 is hindered due to the presence of highly negative charge on this molecule. Therefore, to enhance the cellular delivery of InsP6 in cancerous cells, InsP6 was encapsulated by chitosan (CS), a natural polysaccharide, via the ionic gelation method. Our hypothesis is that encapsulated InsP6 will enter the cell more efficiently to trigger its apoptotic effects. The incorporation of InsP6 into CS was optimized by varying the ratios of the two and confirmed by InsP6 analysis via polyacrylamide gel electrophoresis (PAGE) and atomic absorption spectrophotometry (AAS). The complex was further characterized by Scanning Electron Microscopy (SEM) and Fourier Transform Infrared Spectroscopy (FTIR) for physicochemical changes. The data indicated morphological changes and changes in the spectral properties of the complex upon encapsulation. The encapsulated InsP6 enters human breast cancer MCF-7 cells more efficiently than free InsP6 and triggers apoptosis via a mechanism involving the production of reactive oxygen species (ROS). This work has potential for developing cancer therapeutic applications utilizing natural compounds that are likely to overcome the severe toxic effects associated with synthetic chemotherapeutic drugs. Full article
(This article belongs to the Special Issue Advances in Hydrogels for Tissue Engineering Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic depiction of the concept of cellular entry of exogenously administered InsP<sub>6</sub> after encapsulation with chitosan by ionic gelation to shield off the negative charge. Note that the encapsulated InsP<sub>6</sub> enters the cell through cell membrane, whereas negatively charged free InsP<sub>6</sub> is unable to enter the cell membrane.</p>
Full article ">Figure 2
<p>Schematic illustration of the preparation of the CS:InsP<sub>6</sub> nanomaterial complex by ionic gelation. Chitosan dissolved in acetic acid (5.0 mg/mL) and InsP<sub>6</sub> dissolved in deionized water (5.0 mg/mL) were mixed in varying proportions and stirred for 30 min followed by pH adjustment. The CS:InsP<sub>6</sub> complex was purified by centrifugation and washing with ethanol and lyophilized to dry powder.</p>
Full article ">Figure 3
<p>Detection of InsP<sub>6</sub> contents in the CS:InsP<sub>6</sub> complex by PAGE. Optimization of InsP<sub>6</sub> incorporation in chitosan was carried out by varying the ratios of CS:InsP<sub>6</sub> (<b>B</b>). Standard InsP<sub>6</sub> with known concentrations were also run in parallel to establish the linearity of detection (<b>A</b>). Band densities were analyzed by image J software. (<b>B</b>) shows the amounts of InsP<sub>6</sub> detected in the samples with various ratios of CS:InsP<sub>6</sub> applied on the gel. The maximum amount of InsP<sub>6</sub> (0.49 μg) was detected in the sample with a CS:InsP<sub>6</sub> ratio of 2.5:1.0. This amount (0.49 μg), when calculated using the dilution factor of the samples loaded on the gel, provides a total incorporation of 49 ug InsP<sub>6</sub> per mg CS. Data shown are a representative of at least three independent experiments with similar results. The error bars are not shown as the data shown are from a single experiment repeated at least three times.</p>
Full article ">Figure 4
<p>SEM images of CS (<b>A</b>) and the CS:InsP<sub>6</sub> complex (<b>B</b>). Arrows point to the empty spaces in CS (<b>A</b>) that were perhaps filled by InsP<sub>6</sub> (<b>B</b>), showing morphological changes following encapsulation. The CS:InsP<sub>6</sub> complex at a ratio of 2.5:1.0 was used for SEM analysis in (<b>B</b>). Electron micrographs shown are representative images seen in replicate experiments with similar results.</p>
Full article ">Figure 5
<p>FTIR spectra of (<b>a</b>) InsP<sub>6</sub>, (<b>b</b>) CS, and (<b>c</b>) encapsulated complex with a CS:InsP<sub>6</sub> ratio of 2.5:1.0. Note that the spectral properties of the characteristic bands at specific wavenumbers in InsP<sub>6</sub> (<b>a</b>) and CS (<b>b</b>) are changed upon encapsulation (<b>c</b>).</p>
Full article ">Figure 6
<p>Cellular uptake of encapsulated InsP<sub>6</sub>. Band intensity was analyzed by image J software. (<b>A</b>) shows quantitative detection of InsP<sub>6</sub> in the CS:IsP<sub>6</sub> complex with a CS:InsP<sub>6</sub> ratio of 2.5:1.0. A volume of 5, 10, and 20 µL of the complex loaded on the gel gave 0.15, 0.36, and 0.64 µg InsP<sub>6</sub>, respectively, showing a concentration-dependent linear increase in the detection of InsP<sub>6</sub> in the complex. (<b>B</b>) shows a significant increase in InsP<sub>6</sub> uptake by MCF-7 cells using the encapsulated complex with a CS:InsP<sub>6</sub> ratio of 2.5:1.0 as compared to corresponding free InsP<sub>6</sub> and CS. Data shown are representative of experiments performed independently at least three times with similar results. Statistical analysis is not shown as the data are from a representative experiment.</p>
Full article ">Figure 7
<p>Dose- and time-dependent induction of cell viability in MCF-7 cells by encapsulated InsP<sub>6</sub> treatment. Cell viability was determined at 24 h (<b>A</b>), 48 h (<b>B</b>), and 72 h (<b>C</b>) by MTT assay using the given doses of free InsP<sub>6</sub> (1.0–4.0 µM) and equivalent amounts of the CS: InsP<sub>6</sub> complex that would give similar doses of free InsP<sub>6</sub>. Data are shown as means ± SD from three independent experiments. All experimental values were statistically compared with their respective controls to determine any significant differences. Only treatment with 4 µM encapsulated InsP<sub>6</sub> gave a significant difference as compared with 4 µM free InsP<sub>6</sub>. * <span class="html-italic">p</span> value ≤ 0.001 or ** <span class="html-italic">p</span> ≤ 0.0001 show significantly different values as compared to the respective controls.</p>
Full article ">Figure 8
<p>Effect of encapsulated InsP<sub>6</sub> on apoptosis. MCF-7 cells were incubated with 4 µM of encapsulated InsP<sub>6</sub> for 72 h to induce apoptosis. Etoposide (100 µM) was used as a positive control. (<b>A</b>) MCF-7 cells were stained with acridine orange/ethidium bromide and visualized under UV light using a fluorescent microscope. (<b>B</b>) The percentage of apoptosis was determined by counting 200–300 live (green) and/or dead (red) cells. Values shown are mean ±SD from three experiments, each performed in triplicate. ** <span class="html-italic">p</span> value ≤ 0.001 as compared to the control.</p>
Full article ">Figure 9
<p>Effect of encapsulated InsP<sub>6</sub> on ROS generation. MCF-7 cells were treated with 4 µM free InsP<sub>6</sub> equivalent of the encapsulated CS:InsP<sub>6</sub> complex for 72 h in a 96-well microplate. Etoposide (100 µM) was used as a positive control. Cells were then stained with 10 µM DCFH-DA and fluorescence intensity was recorded using a fluorescence microplate reader. Values shown are mean ± SD from three independent experiments, each performed in triplicate. * <span class="html-italic">p</span> value of ≤0.0001 was considered significantly different compared to the control.</p>
Full article ">Figure 10
<p>Determination of specificity of encapsulated InsP<sub>6</sub>-induced apoptosis by flow cytometry (<b>A</b>). Apoptosis was measured by using a commercially available Vybrant apoptosis assay kit #4. Live cells - are shown as green in lower left quadrant and apoptotic cells are shown as blue in <b>lower and upper right</b> quadrant. Necrotic cells give a red color which are expected to show up in upper left quadrant. The data shown are representative of an experiment repeated at least three times with similar results. (<b>B</b>) shows statistical analysis results of the flow cytometry data showing mean ± standard deviation (SD) from three independent experiments. The % apoptosis values were obtained by combining early and late apoptosis values from the lower and upper right quadrants, respectively. One-way ANOVA with multiple comparisons was used to determine values that were statistically significant. **** <span class="html-italic">p</span> &lt; 0.0001 was considered statistically significant values compared with their respective controls.</p>
Full article ">
19 pages, 3108 KiB  
Article
Phytochemical Characterization of Bilberries and Their Potential as a Functional Ingredient to Mitigate Ochratoxin A Toxicity in Cereal-Based Products
by Denisia Pașca, Massimo Frangiamone, Luciano Mangiapelo, Pilar Vila-Donat, Oana Mîrza, Ana-Maria Vlase, Doina Miere, Lorena Filip, Jordi Mañes, Felicia Loghin and Lara Manyes
Nutrients 2024, 16(18), 3137; https://doi.org/10.3390/nu16183137 (registering DOI) - 17 Sep 2024
Abstract
Mycotoxin contamination of cereals and cereal-based products is a serious problem for food safety. Antioxidant-rich ingredients such as bilberries (Vaccinium myrtillus L., VM) may mitigate their harmful effects. Firstly, total phenolic content, antioxidant activity, and analytical phytochemical composition (hydroxycinnamic and hydroxybenzoic acids, [...] Read more.
Mycotoxin contamination of cereals and cereal-based products is a serious problem for food safety. Antioxidant-rich ingredients such as bilberries (Vaccinium myrtillus L., VM) may mitigate their harmful effects. Firstly, total phenolic content, antioxidant activity, and analytical phytochemical composition (hydroxycinnamic and hydroxybenzoic acids, flavanols, flavonols, and anthocyanins) were assessed in lyophilized wild bilberries from Romania. Secondly, this study evaluated bilberries’ effects on reducing ochratoxin A (OTA) bioaccessibility and cytotoxicity. An in vitro digestion model was developed and applied to four different types of bread: Control, VM (2%), OTA (15.89 ± 0.13 mg/kg), and OTA (16.79 ± 0.55 mg/kg)-VM (2%). The results indicated that VM decreased OTA bioaccessibility by 15% at the intestinal level. OTA-VM digests showed improved Caco-2 cell viability in comparison to OTA digests across different exposure times. Regarding the alterations in Jurkat cell line cell cycle phases and apoptosis/necrosis, significant increases in cell death were observed using OTA digests (11%), while VM addition demonstrated a protective effect (1%). Reactive oxygen species (ROS) analysis confirmed these findings, with OTA-VM digests showing significantly lower ROS levels compared to OTA digests, resulting in a 3.7-fold decrease. Thus, bilberries exhibit high potential as a functional ingredient, demonstrating protection in OTA mitigation effects. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of OTA.</p>
Full article ">Figure 2
<p>Gastric and intestinal OTA bioaccessibility (%) calculated after the in vitro simulated digestion (n = 3). Significant differences between OTA and OTA-VM are indicated as <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.001 (***). Bread with Ochratoxin A (OTA); bread with Ochratoxin A + <span class="html-italic">Vaccinium myrtillus</span> L. 2% (OTA-VM).</p>
Full article ">Figure 3
<p>Cell viability in differentiated Caco-2 cells after exposure to various dilutions of intestinal digests (3.2 μM OTA for no dilution) over five different time points. The data are presented as mean ± SD (n = 4). Significant differences between OTA and OTA-VM intestinal digests at the same dilution and exposure time are denoted as <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 (***). OTA: bread with wheat flour and barley contaminated with Ochratoxin A; OTA-VM: bread with wheat flour and barley flour contaminated with Ochratoxin A and <span class="html-italic">Vaccinium myrtillus</span> L. 2%.</p>
Full article ">Figure 4
<p>Effect of 1/10 intestinal bread digests (0.32 μM OTA) exposure for 7 days on Jurkat cells cycle phases (Sub G<sub>0</sub>/G<sub>1</sub>; G<sub>0</sub>/G<sub>1</sub>; S; G<sub>2</sub>/M). Data are presented as mean ± SD (n = 4) and significant differences between intestinal digests (VM, OTA, and OTA-VM) and control or OTA and OTA-VM are indicated as <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 (***). The acronyms for cell cycle phases are G for growth, S for DNA synthesis, and M for mitosis. C: wheat flour bread; VM: bread with wheat flour and <span class="html-italic">Vaccinium myrtillus</span> L. 2%; OTA: bread with wheat flour and barley flour contaminated with Ochratoxin A; OTA-VM: bread with wheat flour and barley flour contaminated with Ochratoxin A and <span class="html-italic">Vaccinium myrtillus</span> L. 2%.</p>
Full article ">Figure 5
<p>Effect of intestinal digests (0.32 μM OTA) exposure on Jurkat cells after 7 days on the apoptosis and necrosis pathway. Significant differences between intestinal digests (VM, OTA, and OTA-VM) and control or OTA and OTA-VM are indicated as <span class="html-italic">p</span> &lt; 0.05 (*); <span class="html-italic">p</span> &lt; 0.001 (***) and data graph bars represent the mean ± SD (n = 4). C: wheat flour bread; VM: bread with wheat flour and <span class="html-italic">Vaccinium myrtillus</span> L. 2%; OTA: bread with wheat flour and barley flour contaminated with Ochratoxin A; OTA-VM: bread with wheat flour and barley flour contaminated with Ochratoxin A and <span class="html-italic">Vaccinium myrtillus</span> L. 2%.</p>
Full article ">Figure 6
<p>Reactive oxygen species (ROS) measured by flow cytometry using Jurkat cells after 7 days of exposure. (<b>A</b>) Effect of intestinal digests (0.32 μM OTA) on ROS generation. Mean fluorescence intensity is expressed as relative units with ± SD (n = 4). (<b>B</b>) MitoSOX-based flow cytometry detection of mitochondrial ROS in Jurkat cells following exposure to intestinal digests (0.32 μM OTA). Data in the histogram are presented as mean ± SD (n = 4). Significant differences between intestinal digests (VM, OTA, and OTA-VM) and the control, or between OTA and OTA-VM, are indicated as <span class="html-italic">p</span> &lt; 0.001 (***). C: wheat flour bread; VM: bread with wheat flour and <span class="html-italic">Vaccinium myrtillus</span> L. 2%; OTA: bread with wheat flour contaminated with Ochratoxin A; OTA-VM: bread with wheat flour contaminated with Ochratoxin A and <span class="html-italic">Vaccinium myrtillus</span> L. 2%; TBHP: tert-Butyl hydroperoxide.</p>
Full article ">Figure 7
<p>Effect of intestinal digest (0.32 μM OTA) on mitochondrial mass using Jurkat cells after 7 days of exposure. Median fluorescence intensity (MFI) (n = 4) of MitoTracker dye measured after incubation of Jurkat cells with intestinal digests. Significant differences between intestinal digests (VM, OTA, and OTA-VM) and control or OTA and OTA-VM are indicated as <span class="html-italic">p</span> &lt; 0.001 (***). C: wheat flour bread; VM: bread with wheat flour and <span class="html-italic">Vaccinium myrtillus</span> L. 2%; OTA: bread with wheat flour contaminated with Ochratoxin A; OTA-VM: bread with wheat flour contaminated with Ochratoxin A and <span class="html-italic">Vaccinium myrtillus</span> L. 2%.</p>
Full article ">
17 pages, 7417 KiB  
Article
Anti-Tumor Immunity to Patient-Derived Breast Cancer Cells by Vaccination with Interferon-Alpha-Conditioned Dendritic Cells (IFN-DC)
by Caterina Lapenta, Stefano Maria Santini, Celeste Antonacci, Simona Donati, Serena Cecchetti, Patrizia Frittelli, Piera Catalano, Francesca Urbani, Iole Macchia, Massimo Spada, Sara Vitale, Zuleika Michelini, Domenico Cristiano Corsi, Ann Zeuner, Rosanna Dattilo and Manuela Tamburo De Bella
Vaccines 2024, 12(9), 1058; https://doi.org/10.3390/vaccines12091058 (registering DOI) - 17 Sep 2024
Abstract
Background: Breast cancer represents one of the leading causes of death among women. Surgery can be effective, but once breast cancer has metastasized, it becomes extremely difficult to treat. Conventional therapies are associated with substantial toxicity and poor efficacy due to tumor heterogeneity, [...] Read more.
Background: Breast cancer represents one of the leading causes of death among women. Surgery can be effective, but once breast cancer has metastasized, it becomes extremely difficult to treat. Conventional therapies are associated with substantial toxicity and poor efficacy due to tumor heterogeneity, treatment resistance and disease relapse. Moreover, immune checkpoint blockade appears to offer limited benefit in breast cancer. The poor tumor immunogenicity and the immunosuppressive tumor microenvironment result in scarce T-cell infiltration, leading to a low response rate. Thus, there is considerable interest in the development of improved active immunotherapies capable of sensitizing a patient’s immune system against tumor cells. Methods: We evaluated the in vitro anti-tumor activity of a personalized vaccine based on dendritic cells generated in the presence of interferon (IFN)-α and granulocyte-macrophage colony-stimulating factor (IFN-DC) and loaded with an oxidized lysate from autologous tumor cells expanded as 3D organoid culture maintaining faithful tumor antigenic profiles. Results: Our findings demonstrate that stimulation of breast cancer patients’ lymphocytes with autologous IFN-DC led to efficient Th1-biased response and the generation in vitro of potent cytotoxic activity toward the patients’ own tumor cells. Conclusions: This approach can be potentially applied in association with checkpoint blockade and chemotherapy in the design of new combinatorial therapies for breast cancer. Full article
(This article belongs to the Section Cancer Vaccines and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Phenotypic and functional analysis of peripheral blood lymphocytes (PBL) stimulated with HOCl-oxidized MCF-7 breast tumor cell lysate. (<b>a</b>) Dot-plot analysis of CD4+ and CD8+ cells as determined by flow cytometry in PBL stimulated with IFN-DC. PBL isolated from HLA-A2+ healthy blood donors were cultured with autologous IFN-DC (IFN-DC/PBL ratio of 1:4) pulsed with MCF-7 tumor cell lysate, as described in <a href="#sec2-vaccines-12-01058" class="html-sec">Section 2</a>. Representative results of three independent experiments are shown. (<b>b</b>) Cytokine production (IFN-γ, TNF-α and IL-10) in culture supernatants as evaluated by ELISA on days 7 and 14 of co-culture. (<b>c</b>) Degranulation assay as a surrogate evaluation of cytotoxic activity by detection of CD107a membrane expression and intracellular IFN-γ production in CD8+ and natural killer (NK) cells. Representative dot-plot analysis in electronically gated CD8+CD3+ and CD56+CD3- cells in PBL co-cultured with tumor cell-loaded IFN-DC. On day 21 of culture, PBL were restimulated with MCF-7 or K562 target cell lines for 4 h at 37 °C (E:T ratio of 2:1) (see <a href="#sec2-vaccines-12-01058" class="html-sec">Section 2</a>). Dot-plots show CD107a membrane exposure and IFN-γ expression in electronically gated CD8+CD3+ and CD56+CD3- lymphocytes in response to the indicated target cells. Results from one representative experiment out of four are shown. (<b>d</b>) Representative cytotoxicity assay against NK-sensitive K562 and MCF-7 target cell lines as evaluated by a Calcein-AM assay (see <a href="#sec2-vaccines-12-01058" class="html-sec">Section 2</a>) at different E:T ratios. Data are mean ± SD of a triplicate assay of PBL derived from an HLA-A2+ donor.</p>
Full article ">Figure 2
<p>Evaluation of tumor-growth inhibition by in vivo immunization of hu-PBL-NSG mice with IFN-DC loaded with HOCl-oxidized tumor cell lysate. (<b>a</b>) Vaccination schedule. Mice were reconstituted with HLA-A2+ PBL as soon as the implanted tumors became detectable by in vivo bioluminescence imaging (10–11 days). Humanized mice were then randomized into treatment and control groups. (<b>b</b>) Quantitative evaluation of tumor cell growth by bioluminescence analysis. Tumor burden was detected by in vivo non-invasive imaging of the firefly luciferase expressing MCF-7 cells after intraperitoneal luciferin injection. Tumor bioluminescence intensity was plotted in pseudocolor over black/white photographs and quantified as total flux in photons/seconds. Graph represents mean total flux of MCF-7 cell growth rate in hu-PBL-NSG mice immunized as described. The data are presented as mean ± SEM. The difference in tumor growth was highly statistically significant only at the last time point, day 47 (** <span class="html-italic">p</span> &lt; 0.01 by Mann–Whitney test). (<b>c</b>) Representative tumor burden images of the two groups (CTR vs. vaccine) at different time points by IVIS imaging system. (<b>d</b>) Evaluation of IFN-γ levels in mouse sera collected at the time of sacrifice.</p>
Full article ">Figure 3
<p>Isolation and characterization of patient-derived breast cancer organoids (PDBCOs). (<b>a</b>) Representative bright-field images of 5 PDBCOs used for the study, showing different structures: cohesive and discohesive organoids, dense and solid (PBR-13, PBR-14, PBR-16, PBR-17), cohesive organoids, cystic and grape-like (P-BR-22). Scale bar, 100 µm. (<b>b</b>) Comparative histological and immunohistochemical images of BC tissues and derived organoid lines. Shown are representative examples of H&amp;E staining and IHC of either HR or HER-2 status for PBR-17. Scale bar, 200 µm. (<b>c</b>) Stacked bar chart indicating the percentage of PDBCO lines found positive (grey) and negative (black) by IHC for the receptor expression grouped per original tumor receptor status. (<b>d</b>) Bar graph displaying proliferation rate percentage of PDBCOs and corresponding parental tumors as quantified by Ki67 immunohistochemical staining. (<b>e</b>) Representative dot-plot graphs showing CK14 and CK8-18 expression in PBR-13, PBR-16 and PBR-22 (<b>f</b>) CLSM analyses of PFA-fixed PDBCOs stained for CK8-18, SMA and E-cadherin (green); 4′-6-Diamidino-2-phenylindole (DAPI) was used to counterstain nuclei (light blue). Several (&gt;50 organoids) were observed for each condition and representative images are shown. Scale bars, 10 µm.</p>
Full article ">Figure 4
<p>Isolation and characterization of breast cancer patient-derived metastatic cells (PDMCs). (<b>a</b>) Representative phase-contrast images of PDMCs from ascitic fluid (MBR-1, MBR-2) or pleural effusion (MBR-3, MBR-4) cultured in serum-free conditions. Scale bar, 100 µm. (<b>b</b>) CLSM analyses of PFA-fixed PDMCs stained for E-cadherin, vimentin Taz and beta-catenin (green); DAPI was used to counterstain nuclei (light blue). Several fields were observed for each condition and representative images are shown. Scale bars, 10 µm. (<b>c</b>) Dot-plots showing luminal CK8-18 and myoepithelial CK14 expression in PDMC lines. (<b>d</b>) Bar chart reporting percentages of CD44<sup>high</sup>CD24<sup>−/low</sup> phenotype in MBR-1, MBR-2, MBR-3 and MBR-4 lines obtained from ascitic fluid or pleural effusion of breast cancer metastatic patients. Percentages, referring to CD44<sup>high</sup>CD24<sup>−/low</sup> positive cells, were determined by setting the gate on the isotype control from at least two independent FACS stainings.</p>
Full article ">Figure 5
<p>Characterization of PBL cultures from breast tumor patients stimulated with IFN-DC loaded with HOCl-oxidized autologous tumor cell lysate. (<b>a</b>). Representative phenotypical analysis of IFN-DC obtained from breast tumor patients. IFN-DC were differentiated from peripheral blood monocytes as described in <a href="#sec2-vaccines-12-01058" class="html-sec">Section 2</a>. Partially mature CD11c+ IFN-DC ex-pressed high levels of the costimulatory molecules CD80 and CD86, as well as variable levels of the maturation marker CD83. (<b>b</b>) IL-12 release in supernatants collected from IFN-DC cultures on day 3 of differentiation. (<b>c</b>) Representative phenotypic analysis of PBL isolated from breast tumor patients and cultured with autologous IFN-DC loaded with HOCl-oxidized tumor cell lysate for 14 days. (<b>d</b>) Evaluation of CD4, CD8 and NK cell percentages in PBL from breast cancer patients before (T0) and after 14 days of culture (T14) with autologous IFN-DC. (<b>e</b>) Flow cytometric analysis of CD4 and CD8 cell memory subsets of freshly purified PBL from breast cancer patients and after 14 days of culture with autologous IFN-DC loaded with HOCl-oxidized tumor cell lysate. (<b>f</b>) Cytokine release (IFN-γ, TNF-α and IL-10) in culture supernatants as evaluated by ELISA on day 14 of co-cultures.</p>
Full article ">Figure 6
<p>Antitumor activity of PBL from breast cancer patients co-cultured with IFN-DC loaded with HOCl-oxidized autologous tumor cell lysate. PBL isolated from nine patient blood donors were cultured with IFN-DC pulsed with HOCl-oxidized tumor cell lysate for 14 days. (<b>a</b>) Degranulation activity of expanded effectors cells tested against autologous breast cancer target cells as detected by flow cytometry; analysis of CD107a membrane expression and intracellular IFN-γ. (<b>b</b>) Representative degranulation assay as assessed by dot-plot analysis of ectopic CD107a and IFN-γ expression in CD8+ and CD3-CD56+ NK cells derived from three representative breast cancer patients in response to autologous tumor cells. (<b>c</b>) Cytotoxic assay of PBL culture from breast cancer patients after in vitro culture for 14 days, tested against autologous breast cancer cells. (<b>d</b>) Phenotypic analysis of CD8 cells from patient PBR-22 (HLA-A2+) stimulated with IFN-DC loaded with HOCl-oxidized autologous tumor cell lysate for 14 days. (<b>e</b>) Cytotoxic activity of PBL from PBR-22 (HLA-A2+) as compared to PBR-13 (HLA-A2–) as determined by cytotoxic assay towards HLA-A2+ MCF-7 target cells. (<b>f</b>) Degranulation activity as determined by dot plot analysis of CD107a and IFN-γ expression in CD8+ and CD3-CD56+ NK cells from patients PBR-22 and P-BR13 toward MCF-7 target cells.</p>
Full article ">
15 pages, 602 KiB  
Article
A Routine Coronary Angiography before Carotid Endarterectomy as an Example of Interdisciplinary Work: The Immediate Results of the Surgery
by Alexey N. Sumin, Anna V. Shcheglova, Olesya V. Adyakova, Darina N. Fedorova, Denis D. Yakovlev, Natalia A. Svinolupova, Svetlana V. Kabanova, Anastasia V. Malysheva, Marina Yu Karachenko, Vasily V. Kashtalap and Olga L. Barbarash
J. Clin. Med. 2024, 13(18), 5495; https://doi.org/10.3390/jcm13185495 (registering DOI) - 17 Sep 2024
Abstract
The aim: to evaluate the incidence of obstructive lesions of the coronary arteries during routine coronary angiography (CAG) before carotid endarterectomy (CEA) and the incidence of perioperative complications. Materials and Methods: We examined a continuous sample of 498 patients before CEA [...] Read more.
The aim: to evaluate the incidence of obstructive lesions of the coronary arteries during routine coronary angiography (CAG) before carotid endarterectomy (CEA) and the incidence of perioperative complications. Materials and Methods: We examined a continuous sample of 498 patients before CEA who underwent an invasive evaluation of the coronary bed during CAG. Depending on the hemodynamic significance of coronary artery lesions, the patients were divided into three groups: group I—obstructive coronary artery disease (≥70%) (n = 309, 62.0%); group II—non-obstructive lesions of the coronary arteries (<70%) (n = 118, 23.7%); group III—intact coronary arteries (n = 71, 14.3%). The groups were compared with each other according to the data of the preoperative examination (clinical and anamnestic parameters, laboratory data and results of echocardiography), as well as according to the immediate results of the operation. In the hospital period, adverse cardiovascular events were assessed: death, myocardial infarction (MI), stroke, arrhythmias, atrial fibrillation or flutter (AF/AFL) and combined endpoint. Results: The groups differed significantly in the presence of symptoms of angina pectoris, myocardial infarction and myocardial revascularization procedures in their medical history and in the presence of chronic ischemia of the lower extremities. However, in the group of intact coronary arteries, the symptoms of angina were in 14.1% of patients, and a history of myocardial infarction was in 12.7%. Myocardial revascularization before CEA or simultaneously with it was performed in 43.0% of patients. As a result, it was possible to reduce the number of perioperative cardiac complications (mortality 0.7%, perioperative myocardial infarction 1.96%). Conclusions: The high incidence of obstructive lesions in the coronary arteries in our patients and the minimum number of perioperative complications favor routine CAG before CEA. Full article
Show Figures

Figure 1

Figure 1
<p>Patient flowchart. Notes: CA—coronary artery.</p>
Full article ">Figure 2
<p>Managing patients with obstructive CAD and carotid artery disease (<span class="html-italic">n</span> = 309).</p>
Full article ">
24 pages, 3741 KiB  
Article
Protein Structure Inspired Discovery of a Novel Inducer of Anoikis in Human Melanoma
by Fangfang Qiao, Thomas Andrew Binkowski, Irene Broughan, Weining Chen, Amarnath Natarajan, Gary E. Schiltz, Karl A. Scheidt, Wayne F. Anderson and Raymond Bergan
Cancers 2024, 16(18), 3177; https://doi.org/10.3390/cancers16183177 (registering DOI) - 17 Sep 2024
Abstract
Drug discovery historically starts with an established function, either that of compounds or proteins. This can hamper discovery of novel therapeutics. As structure determines function, we hypothesized that unique 3D protein structures constitute primary data that can inform novel discovery. Using a computationally [...] Read more.
Drug discovery historically starts with an established function, either that of compounds or proteins. This can hamper discovery of novel therapeutics. As structure determines function, we hypothesized that unique 3D protein structures constitute primary data that can inform novel discovery. Using a computationally intensive physics-based analytical platform operating at supercomputing speeds, we probed a high-resolution protein X-ray crystallographic library developed by us. For each of the eight identified novel 3D structures, we analyzed binding of sixty million compounds. Top-ranking compounds were acquired and screened for efficacy against breast, prostate, colon, or lung cancer, and for toxicity on normal human bone marrow stem cells, both using eight-day colony formation assays. Effective and non-toxic compounds segregated to two pockets. One compound, Dxr2-017, exhibited selective anti-melanoma activity in the NCI-60 cell line screen. In eight-day assays, Dxr2-017 had an IC50 of 12 nM against melanoma cells, while concentrations over 2100-fold higher had minimal stem cell toxicity. Dxr2-017 induced anoikis, a unique form of programmed cell death in need of targeted therapeutics. Our findings demonstrate proof-of-concept that protein structures represent high-value primary data to support the discovery of novel acting therapeutics. This approach is widely applicable. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

Figure 1
<p>Computational pipeline schema for evaluating compound binding.</p>
Full article ">Figure 2
<p>Schema for identification and characterization of 3D protein structures with the potential to bind therapeutically active small molecules.</p>
Full article ">Figure 3
<p>Proteins that contain pockets structurally suited to binding drug-like small molecules. The surfaces of potential binding pockets are depicted, as are ribbon structures of proximal portions of the protein. The proteins are: 1-deoxy-D-xylulose 5-phosphate reductoisomerase (Dxr; Dxr1 (magenta) and Dxr2 (green) binding pockets), ß-ketoacyl acyl carrier protein reductase (FabG), 3-phosphoshikimate 1-carboxyvinyltransferase (EPSP synthase), dihydrofolate synthase (FolC; FolC1 (orange) and FolC2 (light green) binding pockets), hypoxanthine-guanine phosphoribosyltransferase (HGPRT), and glucose-1-phosphate thymidylyltransferase (TYLT).</p>
Full article ">Figure 4
<p>Structures of the acquired compounds.</p>
Full article ">Figure 5
<p>Effect of 5FU on normal bone marrow. (<b>A</b>) Effect of 5FU on eight- and fourteen-day human stem cell hematopoietic colony formation. (<b>B</b>) Effect of 5FU on eight-day colony formation for human stem cells and HT29 colon cancer cells. Data are the mean ± SEM of N = 4 and N = 2 replicates for stem and HT29 cells, respectively.</p>
Full article ">Figure 6
<p>Effect of compounds on cancer cell and bone marrow colony formation. The effect of denoted compounds on eight-day cancer cell colony formation (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>). The effect of denoted compounds on eight- and fourteen-day bone marrow colony formation (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>). Data are mean ± SD (N = 2 replicates), with similar findings in separate experiments (also N = 2 replicates).</p>
Full article ">Figure 7
<p>The predicted poses of bound Dxr2-017 and FolC2-001. The poses of FolC2-001 (<b>A</b>) and Dxr2-017 (<b>B</b>) bound to their respective FolC2 (yellow) and Dxr2 (green) binding surfaces are depicted, as are ribbon structures of proximal portions of the protein.</p>
Full article ">Figure 8
<p>Dxr2-017 inhibits melanoma cell growth through induction of anoikis. (<b>A</b>) Inhibition of melanoma cell growth. Cells were treated with different concentrations of Dxr2-017, and formation of colonies at eight days is depicted. Data are expressed as the percent of untreated control and are the mean ± SEM of three separate experiments. Each experiment was conducted with N = 3 replicates. (<b>B</b>) No effect on cell cycle progression. M14 and SK-MEL-5 cells were treated for eight days with 20 nM and 50 nM of Dxr2-017, respectively, and the phase of the cell cycle was determined by flow cytometry. Control cells were treated with DMSO vehicle only. Representative histograms are depicted. Graphical data are the mean ± SEM (N = 3). * Denotes <span class="html-italic">p</span>-value ≤ 0.05. (<b>C</b>,<b>D</b>) Transition to floating cells. Cells were treated for three days with different Dxr2-017 concentrations. (<b>C</b>) Depicted are representative light photomicrographs at 20X. The scale bar is 150 µm. Blue and green arrows denote adherent and floating cells, respectively. (<b>D</b>) Adherent and floating cells were quantified from captured images. Data are the mean ± SEM (N = 4), expressed as a percentage to total cells. * Denotes <span class="html-italic">p</span>-value ≤ 0.05 compared to respective floating or attached cells. (<b>E</b>) Cleaved caspase 3 is induced in floating cells. Cells were treated for 3 days, and cell lysate from only adherent cells, or from adherent and floating cells combined, was probed by Western blot for cleaved caspase 3. (<b>F</b>) Dxr2-017 decreases cadherin. Cell lysate from only adherent cells or from adherent and floating cells combined was probed by Western blot for pan-cadherin. All experiments were repeated at separate times, yielding similar results. Original western blots are presented in File S2.</p>
Full article ">
18 pages, 3363 KiB  
Article
Inhibition of Insulin-like Growth Factor 1 Receptor/Insulin Receptor Signaling by Small-Molecule Inhibitor BMS-754807 Leads to Improved Survival in Experimental Esophageal Adenocarcinoma
by Md Sazzad Hassan, Chloe Johnson, Saisantosh Ponna, Dimitri Scofield, Niranjan Awasthi and Urs von Holzen
Cancers 2024, 16(18), 3175; https://doi.org/10.3390/cancers16183175 (registering DOI) - 17 Sep 2024
Viewed by 124
Abstract
The insulin-like growth factor-1 (IGF-1) and insulin axes are upregulated in obesity and obesity-associated esophageal adenocarcinoma (EAC). Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a contemporary nanotechnology-based paclitaxel (PT) bound to human albumin, ensuring its solubility in water rather than a toxic solvent. Here, we [...] Read more.
The insulin-like growth factor-1 (IGF-1) and insulin axes are upregulated in obesity and obesity-associated esophageal adenocarcinoma (EAC). Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a contemporary nanotechnology-based paclitaxel (PT) bound to human albumin, ensuring its solubility in water rather than a toxic solvent. Here, we examined the benefits of inhibiting insulin-like growth factor-1 receptor/insulin receptor (IGF-1/IR) signaling and the enhancement of nab-paclitaxel effects by inclusion of the small-molecule inhibitor BMS-754807 using both in vitro and in vivo models of EAC. Using multiple EAC cell lines, BMS-754807 and nab-paclitaxel were evaluated as mono and combination therapies for in vitro effects on cell proliferation, cell death, and cell movement. We then analyzed the in vivo anticancer potency with survival improvement with BMS-754807 and nab-paclitaxel mono and combination therapies. BMS-754807 monotherapy suppressed in vitro cell proliferation and wound healing while increasing apoptosis. BMS-754807, when combined with nab-paclitaxel, enhanced those effects on the inhibition of cell proliferation, increment in cell apoptosis, and inhibition of wound healing. BMS-754807 with nab-paclitaxel produced substantially greater antitumor effects by increasing in vivo apoptosis, leading to increased mice survival compared to those of BMS-754807 or nab-paclitaxel monotherapy. Our outcomes support the use of BMS-754807, alone and in combination with nab-paclitaxel, as an efficient and innovative treatment choice for EAC. Full article
(This article belongs to the Special Issue Oesogastric Cancer: Treatment and Management)
Show Figures

Figure 1

Figure 1
<p>Phospho-IGF-1R/IR and total IGF-1R expression in human EC cell lines. Western blot experiment demonstrating phospho-IGF-1R/IR (95 kDa) and total IGF-1R (95 kda) expression in seven human EAC cell lines (FLO-1, ESO26, OE19, OE33, SK-GT-2, ESO51, and OACM5.1C) and two human ESCC cell lines (KYSE-270 and OE21). β-actin (42 kDa) served as a loading control. All these EC cell lines had IGF-1R and phospho-IGF-1R/IR expression, with the strongest phospho-IGF-1R/IR expression in FLO-1, OE19, and SK-GT-2. The uncropped blots are shown in the <a href="#app1-cancers-16-03175" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 2
<p>BMS-754807 only or in combination with a chemotherapy drug NPT induced cell proliferation inhibition in phospho-IGF-1R/IR-overexpressing EAC cells. (<b>A</b>–<b>C</b>) Dose-dependent effect of BMS-754807 on Flo-1, OE19, and SK-GT-2 cell proliferation inhibition. (<b>D</b>–<b>F</b>) BMS-754807 (BMS) showed enhancement of Flo-1, OE19, and SK-GT-2 cell proliferation inhibition when combined with the chemotherapy drug nab-paclitaxel (NPT). Flo-1, OE19, and SK-GT-2 EAC cells were seeded on 96-well plates and treated with (<b>A</b>–<b>C</b>) 0 nM to 10,000 nM concentrations of BMS-754807 alone and (<b>C</b>–<b>E</b>) a combination of 1 μM of BMS-754807 (BMS) and 5 µM of NPT simultaneously (NPT + BMS). After 72 h, the number of viable cells was analyzed by adding an equal amount of WST-1 reagent in each well, according to the manufacturer’s instructions. Results are the mean ± SDE of six values. * BMS-754807 additions significantly different from the control (0 nM). ** BMS + NPT additions significantly different from the single-agent addition (NPT or BMS). Results are representative of 3 separate experiments with similar results.</p>
Full article ">Figure 3
<p>BMS-754807 only or in combination with the chemotherapy drug NPT induced the expression of apoptosis-related proteins through the inhibition of the IGF signaling pathway in phospho-IGF-1R/IR-overexpressing EAC cells. A total of 60 to 70% confluent human EAC cells (<b>A</b>) Flo-1, (<b>B</b>) OE19, and (<b>C</b>) SK-GT-2 in culture were given 5 μM of nab-paclitaxel (NPT) and 10 μM of BMS-754807 (BMS), either as a single agent or in combination, for a continuous 16 h. Total cell lysates were subjected to Western blotting with antibodies to phospho-IGF-1R/IR (95 kDa), total IGF-1R (95 kDa), phospho AKT (pAKT) (60 kDa), AKT (60 kDa), cleaved PARP (Asp214) (89 kDa), cleaved caspase 3 (Asp 175) (19, 17 kDa), and β-actin (42 kDa). The uncropped blots are shown in the <a href="#app1-cancers-16-03175" class="html-app">Supplementary Materials</a>. The intensities of cleaved PARP (<b>D</b>–<b>F</b>) and cleaved caspase 3 (<b>G</b>–<b>I</b>) bands were quantified by densitometry and are represented in the bar graph, after normalizing values with β-actin. * NPT or BMS or BMS + NPT additions significantly different from the control (C). ** NPT + BMS additions significantly different from the single-agent addition (NPT or BMS) (N = 3).</p>
Full article ">Figure 4
<p>BMS-754807 alone or in combination with the chemotherapy drug NPT suppressed the migration of phospho-IGF-1R/IR-overexpressing Flo-1 EAC cells. (<b>A</b>) Near-confluent Flo-1 cells in culture were scratched with a pipette tip to make a linear gap and then 5 μM of nab-paclitaxel (NPT) and 10 μM of BMS-754807 (BMS) were added either as a single agent or in combination for a continuous 24, 48, 72, and 96 h (24 h, 48 h, 72 h, and 96 h). The microscopic photographs were taken at 0, 24, 48, 72, and 96 h, respectively (X10) (scale bar = 400 µm). Yellow lines indicate the borders of the scratched gap. (<b>B</b>) The gap area of Flo-1 cells was determined by Image J software to analyze the gap closure rate. All numbers were noted as the mean ± standard deviation (N = 5). * NPT or BMS-754807 (BMS) treatment significantly differed from the control (0 nM). ** NPT + BMS treatment significantly different from the single-agent therapy (NPT or BMS).</p>
Full article ">Figure 5
<p>Phospho-IGF-1R/IR-overexpressing OE19 EAC xenograft tumor size reduction by BMS-754807 (BMS) only and in combination with the chemotherapy drug nab-paclitaxel (NPT). High phospho-IGF-1R/IR-expressing OE19 cells (5 million per site) were inoculated under the skin of nude mice. When measurable tumor size was reached (usually 2 weeks after cell inoculation), mice received the vehicle (<b>C</b>) and BMS and NPT alone or in combination. (<b>A</b>,<b>B</b>) Relative tumor volume (RTV) was determined by dividing the tumor volume at any time point by the tumor volume at the start of therapy. (<b>A</b>) RTV variation was observed over a duration of 14 days from the start of the therapy. (<b>B</b>) RTV variation was observed at the end of the therapy under different therapy regimens. (<b>C</b>) Variation in net growth in tumor volume was observed at the end of the therapy under different therapy regimens. It was determined by deducting the tumor volume determined on the 1st treatment day from that on the last day. (<b>D</b>) Average tumor weight variation was observed at the end of the therapy under different therapy regimens. (<b>E</b>) Variation in mouse body weight was observed during the 14-day therapy time. Results are average values ± standard deviation from 5 mice in each group. * Significantly different from control (<b>C</b>). ** Significantly different from single-drug therapy. NS = not significant.</p>
Full article ">Figure 6
<p>Anti-proliferative and pro-apoptotic in vivo efficacy of BMS-754807 monotherapy and in combination with chemotherapy. Xenografts developed in immunodeficient nude mice with high phospho-IGF-1R/IR-expressing OE19 EAC cells received either BMS-754807 (BMS) or nab-paclitaxel (NPT) monotherapy or BMS plus NPT combination therapy for 14 consecutive days. At the conclusion of therapy, tumors were collected and processed for immunohistochemistry (IHC). (<b>A</b>) Cancer cell proliferation was determined within the tumor by immunostaining of the tissue microsections for Ki67 nuclear protein. Representative microscopic high-power field images of Ki67 brown staining are shown. (scale bar = 20 µm). (<b>B</b>) Brown-stained nuclei for Ki67-positive cells and blue-stained nuclei for the total number of cells within the tumor tissue microsections were counted in five different microscopic images of high-power fields, and data are summarized as a bar graph (proliferative index). In each high-power field image, the proliferative index was measured by the number of brown-stained Ki67-positive tumor nuclei as a percentage of the total number of nuclei. (<b>C</b>) Cancer cell apoptosis was determined within the tumor by immunostaining of tumor tissue microsections for cleaved caspase 3 cytoplasmic protein. Representative microscopic high-power field images of cleaved caspase 3 brown staining are shown. (scale bar = 20 µm). (<b>D</b>) Brown-stained cleaved caspase 3 and blue-stained tumor nuclei were counted in five different microscopic images of high-power fields, and data are summarized as a bar graph (apoptosis index). In each high-power field image, the apoptosis index was measured by the number of brown-stained cleaved caspase 3-positive cells as a percentage of the total number of nuclei. Results are shown as the mean ± standard deviation. * Significantly different from vehicle (control). ** Significantly different from single-agent therapy.</p>
Full article ">Figure 7
<p>Mice survival enhancement by BMS-754807 (BMS) only and in combination with chemotherapy. Kaplan–Meier survival curve for mice harboring phospho-IGF-1R/IR-overexpressing OE19 EAC intraperitoneal xenograft treated for 14 days with BMS-754807 (BMS) only and in combination with the chemotherapy drug nab-paclitaxel (NPT). The curve shows mice survival in days from the start of drug administration. Statistical grouping variation in the survival period was conducted by log-rank measurement (GraphPad Prism 7.0).</p>
Full article ">
15 pages, 4357 KiB  
Article
Dimethyl Fumarate Strongly Ameliorates Gray and White Matter Brain Injury and Modulates Glial Activation after Severe Hypoxia–Ischemia in Neonatal Rats
by Jon Ander Alart, Antonia Álvarez, Ana Catalan, Borja Herrero de la Parte and Daniel Alonso-Alconada
Antioxidants 2024, 13(9), 1122; https://doi.org/10.3390/antiox13091122 - 16 Sep 2024
Viewed by 209
Abstract
Neonatal hypoxia–ischemia is a major cause of infant death and disability. The only clinically accepted treatment is therapeutic hypothermia; however, cooling is less effective in the most severely encephalopathic infants. Here, we wanted to test the neuroprotective effect of the antioxidant dimethyl fumarate [...] Read more.
Neonatal hypoxia–ischemia is a major cause of infant death and disability. The only clinically accepted treatment is therapeutic hypothermia; however, cooling is less effective in the most severely encephalopathic infants. Here, we wanted to test the neuroprotective effect of the antioxidant dimethyl fumarate after severe hypoxia–ischemia in neonatal rats. We used a modified Rice–Vannucci model to generate severe hypoxic–ischemic brain damage in day 7 postnatal rats, which were randomized into four experimental groups: Sham, Sham + DMF, non-treated HI, and HI + DMF. We analyzed brain tissue loss, global and regional (cortex and hippocampus) neuropathological scores, white matter injury, and microglial and astroglial reactivity. Compared to non-treated HI animals, HI + DMF pups showed a reduced brain area loss (p = 0.0031), an improved neuropathological score (p = 0.0016), reduced white matter injuries by preserving myelin tracts (p < 0.001), and diminished astroglial (p < 0.001) and microglial (p < 0.01) activation. After severe hypoxia–ischemia in neonatal rats, DMF induced a strong neuroprotective response, reducing cerebral infarction, gray and white matter damage, and astroglial and microglial activation. Although further molecular studies are needed and its translation to human babies would need to evaluate the molecule in piglets or lambs, DMF may be a potential treatment against neonatal encephalopathy. Full article
11 pages, 1187 KiB  
Review
Carbon Monoxide: A Pleiotropic Redox Regulator of Life and Death
by Andrey Y. Abramov, Isabella Myers and Plamena R. Angelova
Antioxidants 2024, 13(9), 1121; https://doi.org/10.3390/antiox13091121 - 16 Sep 2024
Viewed by 164
Abstract
Despite recent technological progress, carbon monoxide poisoning is still one of the leading causes of domestic and industrial morbidity and mortality. The brain is particularly vulnerable to CO toxicity, and thus the majority of survivors develop delayed movement and cognitive complications. CO binds [...] Read more.
Despite recent technological progress, carbon monoxide poisoning is still one of the leading causes of domestic and industrial morbidity and mortality. The brain is particularly vulnerable to CO toxicity, and thus the majority of survivors develop delayed movement and cognitive complications. CO binds to haemoglobin in erythrocytes, preventing oxygen delivery to tissues, and additionally inhibits mitochondrial respiration. This renders the effect of CO to be closely related to hypoxia reperfusion injury. Oxygen deprivation, as well as CO poisoning and re-oxygenation, are shown to be able to activate the production of reactive oxygen species and to induce oxidative stress. Here, we review the role of reactive oxygen species production and oxidative stress in the mechanism of neuronal cell death induced by carbon monoxide and re-oxygenation. We discuss possible protective mechanisms used by brain cells with a specific focus on the inhibition of CO-induced ROS production and oxidative stress. Full article
Show Figures

Figure 1

Figure 1
<p><b>The role of carbon monoxide in physiology and pathology.</b> Left: at physiological conditions of the cell, CO is produced in the HO-1 and enables cell detoxification and signalling processes. Right: at pathologically acute high doses of CO-NOX, XO and mitochondria produce ROS with higher rates, which leads to lipid peroxidation and mPTP opening and, ultimately, to cell death. CO—carbon monoxide; ROS—reactive oxygen species; NOX—NADPH oxidases; HO-1—heme oxygenase; Nrf-2—nuclear factor erythroid 2-related factor 2; XO—xanthine oxidase; MAO—monoamine oxidase; NADPH—reduced nicotinamide adenine dinucleotide phosphate, NADP+—oxidised nicotinamide adenine dinucleotide phosphate; ATP—adenosine 3-phosphate; mPTP—mitochondrial permeability transition pore; MDA—malone dialdehyde; 8-OHdG-8-hydroxy-2′-deoxyguanosine; SOD—superoxide dismutase; CAT—catalase; GPx—glutathione peroxidase.</p>
Full article ">
23 pages, 11452 KiB  
Article
Exopolysaccharides from the Green Microalga Strain Coelastrella sp. BGV—Isolation, Characterization, and Assessment of Anticancer Potential
by Tanya Toshkova-Yotova, Inna Sulikovska, Vera Djeliova, Zdravka Petrova, Manol Ognyanov, Petko Denev, Reneta Toshkova and Ani Georgieva
Curr. Issues Mol. Biol. 2024, 46(9), 10312-10334; https://doi.org/10.3390/cimb46090614 (registering DOI) - 16 Sep 2024
Viewed by 179
Abstract
Algal metabolites have been extensively studied as potential anticancer therapeutics. Among them, polysaccharides have attracted much attention because of their beneficial biological effects and safety. In the present research, the chemical characteristics, antitumor, and proapoptotic activities of extracellular polysaccharides (EPS) isolated from a [...] Read more.
Algal metabolites have been extensively studied as potential anticancer therapeutics. Among them, polysaccharides have attracted much attention because of their beneficial biological effects and safety. In the present research, the chemical characteristics, antitumor, and proapoptotic activities of extracellular polysaccharides (EPS) isolated from a new Bulgarian strain of the green microalga Coelastrella sp. BGV were investigated. A fast and convenient method of precipitation with cold ethanol was used to isolate EPS from the culture medium. The chemical characteristics of the isolated EPS were examined by colorimetric and spectrophotometric analyses, HPSEC-RID and HPLC-UV chromatography, and FT-IR spectroscopy. The results showed that the isolated EPS sample consists of three carbohydrate fractions with different molecular weights (11.5 × 104 Da, 30.7 × 104 Da, and 72.4 × 104 Da, respectively) and contains 7.14 (w/w%) protein. HPLC-UV analysis revealed the presence of galactose and fucose. The total uronic acid content in the sample was 4.5 (w/w%). The IR-FT spectrum of EPS revealed the presence of various functional groups typical of a polysaccharide (or proteoglycan) composed primarily of neutral sugars. The anticancer potential of the obtained EPS was assessed using cell lines with cancerous and non-cancerous origins as in vitro experimental models. The results of the performed MTT assay showed that EPS reduced the viability of the cervical and mammary carcinoma cell lines HeLa and MCF-7, while the control non-cancer cell lines BALB/3T3 and HaCaT were less affected. The HeLa cell line showed the highest sensitivity to the effects of EPS and was therefore used for further studies of its anticancer potential. The ability of EPS to inhibit cancer cell migration was demonstrated by wound-healing (scratch) assay. The cell cycle FACS analysis indicated that the EPS treatment induced significant increases in the sub G1 cell population and decreases of the percentages of cells in the G1, S, and G2-M phases, compared to the control. The fluorescent microscopy studies performed using three different staining methods in combination with Annexin V-FITC flow cytometric analysis clearly demonstrate the ability of EPS to induce cancer cell death via the apoptosis pathway. Moreover, an altered pattern and intensity of the immunocytochemical staining for the apoptosis- and proliferation-related proteins p53, bcl2, and Ki67 was detected in EPS-treated HeLa cancer cells as compared to the untreated controls. The obtained results characterize the new local strain of green microalgae Coelastrella sp. BGV as a producer of EPS with selective antitumor activity and provide an opportunity for further studies of its pharmacological and biotechnological potential. Full article
(This article belongs to the Special Issue Phytochemicals in Cancer Chemoprevention and Treatment)
Show Figures

Figure 1

Figure 1
<p>An HPLC-UV chromatogram of PMP (1-phenyl-3-methyl-5-pyrazolone)—monosaccharide derivatives obtained from EPS from <span class="html-italic">Coelastrella</span> sp. BGV.</p>
Full article ">Figure 2
<p>HPSEC elution profile of EPS from <span class="html-italic">Coelastrella</span> sp. BGV. (<b>A</b>) exopolysaccharide; (<b>B</b>) pullulan standards. Molecular weights of the standards used—from left to right: 78.8 × 10<sup>4</sup>, 40.4 × 10<sup>4</sup>, 21.2 × 10<sup>4</sup>, 11.2 × 10<sup>4</sup>, 4.73 × 10<sup>4</sup>, 2.28 × 10<sup>4</sup>, 1.18 × 10<sup>4</sup>, 0.59 × 10<sup>4</sup> Da.</p>
Full article ">Figure 3
<p>FT-IR absorption spectrum of the EPS from <span class="html-italic">Coelastrella</span> sp. BGV over the range of 4000–400 cm<sup>−1</sup>.</p>
Full article ">Figure 4
<p>Effect of extracellular polysaccharide from Coelastrella sp. BGV on the viability of HeLa, MCF-7, HaCaT and BALB/3T3 cells assessed by an MTT test at 24 h and 48 h. Untreated cells and cultivated cells treated with the antitumor drug Doxorubicin (Dox; 2.5 μg/mL) were used as negative and positive controls, respectively. The data are expressed as the mean ± SD of five samples from each treatment group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 indicate significant differences compared to the negative control.</p>
Full article ">Figure 5
<p>Effect of Coelastrella sp. BGV EPS treatment on the migration of HeLa cervical carcinoma cells evaluated by wound-healing assay. (<b>Upper panel</b>) Light microscopy images of untreated cell cultures and cell cultures treated with EPS (250 μg/mL); (<b>Lower panel</b>) Quantification of the EPS effect on the migration potential of the cancer cells. Data are presented as Mean ± SD; *** <span class="html-italic">p</span> &lt; 0.001 compared to the untreated control.</p>
Full article ">Figure 6
<p>Morphological changes of the HeLa tumor cells cultured in the presence of EPS (500 μg/mL) for 24 h followed by AO/EB (<b>Upper row</b>), DAPI (<b>middle row),</b> and Annexin V (<b>Lower row</b>) staining. Fluorescence micrographs of: (<b>a</b>,<b>d</b>,<b>g</b>)—untreated HeLa cells; (<b>b</b>,<b>e</b>,<b>h</b>)—cells after incubation with 500 µg/mL of EPS; (<b>c</b>,<b>f</b>,<b>i</b>)—cells after incubation with 2.5 µg/mL Dox; Scale bar = 20 µm.</p>
Full article ">Figure 7
<p>Proapoptotic effect of <span class="html-italic">Coelastrella</span> sp. BGV EPS on HeLa cervical carcinoma cells as evaluated by FACS analysis. (<b>Upper panel</b>) Representative histogram of control untreated cells and cells treated with 500 µg/mL EPS for 24 h. (<b>Lower panel</b>) Bar graph showing the percentages of the live, early and late apoptotic, and necrotic cells. The data are expressed as mean ± SD from three independent experiments; *** <span class="html-italic">p</span> &lt; 0.001 indicates significant difference as compared to the negative control.</p>
Full article ">Figure 8
<p>Effect of Coelastrella sp BGV EPS treatment on the cell cycle progression of HeLa cervical carcinoma cells. (<b>Upper panel</b>) Representative histogram of control untreated cells and cells treated with 500 µg/mL EPS for 24 h. (<b>Lower panel</b>) Bar graph representing the distribution of the cells in the different cell cycle phases. The data are expressed as mean ± SD from three independent experiments; *** <span class="html-italic">p</span> &lt; 0.001 indicate significant difference as compared to the negative control.</p>
Full article ">Figure 9
<p>Immunocytochemical analysis of the EPS effects on the expression and intracellular localization of the p53, bcl2, and Ki67 proteins in HeLa carcinoma cells. (<b>a</b>,<b>d</b>,<b>g</b>) untreated control cells; (<b>b</b>,<b>e</b>,<b>h</b>) cells treated with EPS (500 μg/mL); (<b>c</b>,<b>f</b>,<b>i</b>) cells treated with Dox (2.5 μg/mL); (<b>a</b>–<b>c</b>) p53 immunostaining; (<b>d</b>–<b>f</b>) bcl2 immunostaining; (<b>g</b>–<b>i</b>) Ki67 immunostaining.</p>
Full article ">
35 pages, 2977 KiB  
Review
Gene Expression Regulation and the Signal Transduction of Programmed Cell Death
by Saqirile, Yuxin Deng, Kexin Li, Wenxin Yan, Ke Li and Changshan Wang
Curr. Issues Mol. Biol. 2024, 46(9), 10264-10298; https://doi.org/10.3390/cimb46090612 (registering DOI) - 16 Sep 2024
Viewed by 155
Abstract
Cell death is of great significance in maintaining tissue homeostasis and bodily functions. With considerable research coming to the fore, it has been found that programmed cell death presents in multiple modalities in the body, which is not only limited to apoptosis, but [...] Read more.
Cell death is of great significance in maintaining tissue homeostasis and bodily functions. With considerable research coming to the fore, it has been found that programmed cell death presents in multiple modalities in the body, which is not only limited to apoptosis, but also can be divided into autophagy, pyroptosis, ferroptosis, mitotic catastrophe, entosis, netosis, and other ways. Different forms of programmed cell death have disparate or analogous characteristics with each other, and their occurrence is accompanied by multiple signal transduction and the role of a myriad of regulatory factors. In recent years, scholars across the world have carried out considerable in-depth research on programmed cell death, and new forms of cell death are being discovered continually. Concomitantly, the mechanisms of intricate signaling pathways and regulators have been discovered. More critically, cancer cells tend to choose distinct ways to evade cell death, and different tumors adapt to different manners of death. Therefore, targeting the cell death network has been regarded as an effective tumor treatment strategy for a long time. The objective of our paper is to review the signaling pathways and gene regulation in several typical types of programmed cell death and their correlation with cancer. Full article
14 pages, 266 KiB  
Article
Syndemic Connections: Overdose Death Crisis, Gender-Based Violence and COVID-19
by Ana M. Ning
Societies 2024, 14(9), 185; https://doi.org/10.3390/soc14090185 - 16 Sep 2024
Viewed by 210
Abstract
This article will use syndemic theory to identify and analyze overlapping health and social conditions, focusing specifically on how gender-based violence is systemically interconnected with contemporary public health issues. The overdose death crisis that continues to afflict Canadian populations is not an isolated [...] Read more.
This article will use syndemic theory to identify and analyze overlapping health and social conditions, focusing specifically on how gender-based violence is systemically interconnected with contemporary public health issues. The overdose death crisis that continues to afflict Canadian populations is not an isolated health issue. Across Canada, it is intertwined with mental health, HIV/AIDS, COVID-19 and structural violence—the chronic and systemic disadvantages affecting those living in poverty and oppressive circumstances. Opioid use is an often-avoidant coping strategy for many experiencing the effects of trauma, relentless fear, pain, ill health and social exclusion. In particular, Indigenous and non-Indigenous women’s experiences with opioid addiction are entangled with encounters with gender based-violence, poverty and chronic ailments within structurally imposed processes and stressors shaped by a history of colonialism, ruptured lifeways and Western ways of knowing and doing, leading to disproportionate harms and occurrences of illness. While biomedical models of comorbidity and mortality approach substance misuse, gender-based violence and major infectious diseases such as HIV/AIDS and COVID-19 as distinct yet compounding realities, this article argues that these conditions are synergistically interrelated via the critical/reflexive lens of syndemic frameworks. Through secondary research using academic, media and policy sources from the past decade in Canada, complemented by prior ethnographic research, the synergistic connections among opioid addiction, gender-based violence and the effects of the COVID pandemic on diverse women will be shown to be driven by socio-structural determinants of health including poverty, intergenerational trauma, the legacy of colonialism and Western optics. Together, they embody a contemporary Canadian syndemic necessitating coordinated responses. Full article
30 pages, 7247 KiB  
Review
Progress in Research of Nanotherapeutics for Overcoming Multidrug Resistance in Cancer
by Ayitila Maimaitijiang, Dongze He, Dingyang Li, Wenfang Li, Zhengding Su, Zhongxiong Fan and Jinyao Li
Int. J. Mol. Sci. 2024, 25(18), 9973; https://doi.org/10.3390/ijms25189973 (registering DOI) - 16 Sep 2024
Viewed by 193
Abstract
Chemotherapy has been widely applied in oncotherapy. However, the development of multidrug resistance (MDR) has diminished the effectiveness of anticancer drugs against tumor cells. Such resistance often results in tumor recurrence, metastasis, and patient death. Fortunately, nanoparticle-based drug delivery systems provide a promising [...] Read more.
Chemotherapy has been widely applied in oncotherapy. However, the development of multidrug resistance (MDR) has diminished the effectiveness of anticancer drugs against tumor cells. Such resistance often results in tumor recurrence, metastasis, and patient death. Fortunately, nanoparticle-based drug delivery systems provide a promising strategy by codelivery of multiple drugs and MDR reversal agents and the skillful, flexible, smart modification of drug targets. Such systems have demonstrated the ability to bypass the ABC transporter biological efflux mechanisms due to drug resistance. Hence, how to deliver drugs and exert potential antitumor effects have been successfully explored, applied, and developed. Furthermore, to overcome multidrug resistance, nanoparticle-based systems have been developed due to their good therapeutic effect, low side effects, and high tumor metastasis inhibition. In view of this, we systematically discuss the molecular mechanisms and therapeutic strategies of MDR from nanotherapeutics. Finally, we summarize intriguing ideas and future trends for further research in overcoming MDR. Full article
(This article belongs to the Special Issue The Application of Nanoparticles in Biomedicine)
Show Figures

Figure 1

Figure 1
<p>Mechanisms of anticancer drug resistance: efflux pump-mediated mechanisms of MDR and efflux pump-independent drug resistance mechanisms.</p>
Full article ">Figure 2
<p>ABC transporter superfamily partial members, and their substrates.</p>
Full article ">Figure 3
<p>Nanocarrier system with small interfering RNA. (<b>a</b>) The mesopore surface is modified with PEI to allow for the loading of P-gp modulator siRNA. (<b>b</b>) H-MSNs co-loaded with DOX and siRNA extravasate into tumor stroma across the blood vessel and angiogenic vasculature, and finally are endocytosed into cancer cells. (<b>c</b>) Mechanism scheme demonstrating the therapeutic functions of H-MSNs in suppressing MDR of cancer cells and enhancing chemotherapy efficiency [<a href="#B122-ijms-25-09973" class="html-bibr">122</a>]. (<b>d</b>) Top: Formation of the micelleplex between NSC–PLL–PA and siRNA was determined by the quenching method using EtBr. Bottom: In vitro drug release of Dox-micelle and siRNA in different media compared with Dox [<a href="#B121-ijms-25-09973" class="html-bibr">121</a>]. (<b>e</b>) P-gp levels were detected by RT-PCR (left) or Western blot (right) [<a href="#B121-ijms-25-09973" class="html-bibr">121</a>], * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with the controls (n = 3).</p>
Full article ">Figure 4
<p>Nitric oxide (NO) and its donor-redox responsive drug delivery system. (<b>A</b>) The formation of NapFFGEE-JSK/DOX supramolecular hydrogel by self-assembly, and further loading with DOX by electrostatic and hydrophobic interactions. (<b>B</b>) The synergistic antitumor mechanism of NapFFGEE-JSK/DOX supramolecular hydrogel for combating multidrug resistance [<a href="#B123-ijms-25-09973" class="html-bibr">123</a>]. (<b>C</b>) ROS cascade nanoplatform targeting regulation of P-glycoprotein and synergistic inducing of ferroptosis to reverse multidrug resistance in prostate cancer [<a href="#B124-ijms-25-09973" class="html-bibr">124</a>].</p>
Full article ">Figure 5
<p>Chip-based fabrication of rigid pH-sensitive micellar nanocomplex (RPN). (<b>A</b>) Schematic of the microfluidic chip and illustration of intracellular translocation of RPN and PM. (<b>B</b>) Chemical structure of diblock copolymer (PEG-b-PDPA) which could be activated in acidic endo-/lysosomes. (<b>C</b>) Simulations to show the structure of RPN and PM. (<b>D</b>) Hydrodynamic sizes of RPN with different mass ratios of PEG-b-PDPA/PLGA measured by DLS. (<b>E</b>) Hydrodynamic sizes of PM and RPN with the mass ratio of PEG-b-PDPA/PLGA of 9. (<b>F</b>) TEM images of RPN at pH 7.4, and dissociated RPN at pH 5.6 [<a href="#B139-ijms-25-09973" class="html-bibr">139</a>]. (<b>G</b>) Reversible magnetic nanogates drive drug release from magnetic mesoporous silica particles through DNA hybridization/dehybridization [<a href="#B141-ijms-25-09973" class="html-bibr">141</a>].</p>
Full article ">
27 pages, 35504 KiB  
Article
A Multi-Scale Analysis of the Extreme Precipitation in Southern Brazil in April/May 2024
by Michelle Simões Reboita, Enrique Vieira Mattos, Bruno César Capucin, Diego Oliveira de Souza and Glauber Willian de Souza Ferreira
Atmosphere 2024, 15(9), 1123; https://doi.org/10.3390/atmos15091123 - 16 Sep 2024
Viewed by 284
Abstract
Since 2020, southern Brazil’s Rio Grande do Sul (RS) State has been affected by extreme precipitation episodes caused by different atmospheric systems. However, the most extreme was registered between the end of April and the beginning of May 2024. This extreme precipitation caused [...] Read more.
Since 2020, southern Brazil’s Rio Grande do Sul (RS) State has been affected by extreme precipitation episodes caused by different atmospheric systems. However, the most extreme was registered between the end of April and the beginning of May 2024. This extreme precipitation caused floods in most parts of the state, affecting 2,398,255 people and leading to 183 deaths and 27 missing persons. Due to the severity of this episode, we need to understand its drivers. In this context, the main objective of this study is a multi-scale analysis of the extreme precipitation between 26 April and 5 May, i.e., an analysis of the large-scale patterns of the atmosphere, a description of the synoptic environment, and an analysis of the mesoscale viewpoint (cloud-top features and lightning). Data from different sources (reanalysis, satellite, radar, and pluviometers) were used in this study, and different methods were applied. The National Center for Monitoring and Early Warning of Natural Disasters (CEMADEN) registered accumulated rainfall above 400 mm between 26 April and 5 May using 27 pluviometers located in the central-northern part of RS. The monthly volumes reached 667 mm and 803 mm, respectively, for April and May 2024, against a climatological average of 151 mm and 137 mm for these months. The maximum precipitation recorded was 300 mm in a single day on 30 April 2024. From a large-scale point of view, an anomalous heat source in the western Indian Ocean triggered a Rossby wave that contributed to a barotropic anticyclonic anomalous circulation over mid-southeastern Brazil. While the precipitant systems were inhibited over this region (the synoptic view), the anomalous stronger subtropical jet southward of the anticyclonic circulation caused uplift over RS State and, consequently, conditions leading to mesoscale convective system (MCS) development. In addition, the low-level jet east of the Andes transported warm and moist air to southern Brazil, which also interacted with two cold fronts that reached RS during the 10-day period, helping to establish the precipitation. Severe deep MCSs (with a cloud-top temperature lower than −80 °C) were responsible for a high lightning rate (above 10 flashes km−2 in 10 days) and accumulated precipitation (above 600 mm in 10 days), as observed by satellite measurements. This high volume of rainfall caused an increase in soil moisture, which exceeded a volume fraction of 0.55, making water infiltration into the soil difficult and, consequently, favoring flood occurrence. Full article
18 pages, 1415 KiB  
Review
New Insights into Mitochondria in Health and Diseases
by Ya Li, Huhu Zhang, Chunjuan Yu, Xiaolei Dong, Fanghao Yang, Mengjun Wang, Ziyuan Wen, Mohan Su, Bing Li and Lina Yang
Int. J. Mol. Sci. 2024, 25(18), 9975; https://doi.org/10.3390/ijms25189975 (registering DOI) - 16 Sep 2024
Viewed by 387
Abstract
Mitochondria are a unique type of semi-autonomous organelle within the cell that carry out essential functions crucial for the cell’s survival and well-being. They are the location where eukaryotic cells carry out energy metabolism. Aside from producing the majority of ATP through oxidative [...] Read more.
Mitochondria are a unique type of semi-autonomous organelle within the cell that carry out essential functions crucial for the cell’s survival and well-being. They are the location where eukaryotic cells carry out energy metabolism. Aside from producing the majority of ATP through oxidative phosphorylation, which provides essential energy for cellular functions, mitochondria also participate in other metabolic processes within the cell, such as the electron transport chain, citric acid cycle, and β-oxidation of fatty acids. Furthermore, mitochondria regulate the production and elimination of ROS, the synthesis of nucleotides and amino acids, the balance of calcium ions, and the process of cell death. Therefore, it is widely accepted that mitochondrial dysfunction is a factor that causes or contributes to the development and advancement of various diseases. These include common systemic diseases, such as aging, diabetes, Parkinson’s disease, and cancer, as well as rare metabolic disorders, like Kearns–Sayre syndrome, Leigh disease, and mitochondrial myopathy. This overview outlines the various mechanisms by which mitochondria are involved in numerous illnesses and cellular physiological activities. Additionally, it provides new discoveries regarding the involvement of mitochondria in both disorders and the maintenance of good health. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Mitochondrial structure and the process by which NADH + H<sup>+</sup> enters the mitochondria via specific shuttling in the inner mitochondrial membrane. The process of producing ATP is intricate. For instance, the metabolism of glucose involves the cytoplasmic glycolysis process, the mitochondrial matrix TCA cycle, and oxidative phosphorylation accompanied by the production of ATP. Of these, the pyruvate transporter facilitates the entry of pyruvate generated during glycolysis into the mitochondria, but the mechanism of NADH + H+ entering the mitochondria is more intricate: 1. NADH + H<sup>+</sup> in the cytoplasm is treated by malate dehydrogenase to make oxaloacetic acid (OAA) accept 2 H and become malic acid (MAL). 2. Malic acid enters mitochondria via transport carriers in the inner membrane.3. Under the action of malic acid entering mitochondria, NAD<sup>+</sup> is used as acceptor to form oxaloacetic acid and NADH + H<sup>+</sup>. 4. Oxaloacetic acid and glutamic acid are transformed into aspartic acid and alpha-ketoglutaric acid by the interaction of glutamic acid with glutamic acid through glutamic oxaloacetic acid transaminase. 5. Aspartate (Asp) and α-ketoglutaric acid enter the cytoplasm with the help of mitochondrial transport carriers. 6. Glutamate (Glu) consumed in the mitochondria is supplemented by the exchange of glutamate in cellular fluid and outgoing aspartic acid through the reverse glutamate–aspartic acid transport carrier.</p>
Full article ">Figure 2
<p>Mitophagy mediated by the PINK1–Parkin pathway during aging. PINK1 accumulates on the outer membrane of the mitochondria under depolarization or stress, and autophosphorylation activates it. In addition, the active PINK1 draws the cytoplasmic Parkin protein to the mitochondria and triggers Parkin’s E3 ubiquitin ligase activity by phosphorylating ubiquitin, which polyubiquitinates the protein found in the mitochondrial membrane. Specifically, signals for the identification of autophagy receptors are provided by ubiquitin chains connected by K63, and autophagy receptor proteins, like p62, OPTN, and NDP52, are attracted to mitochondria modified by ubiquitination to facilitate selective autophagy.</p>
Full article ">Figure 3
<p>Complex regulatory networks in Parkinson’s disease. Parkinson’s disease has a complex etiology that includes both genetic and environmental influences. The three that will most likely impact mitochondrial function, cause abnormalities in mitochondrial electron transfer and oxidative phosphorylation, produce reactive oxygen species (ROS), and ultimately result in nerve cell death are oxidative stress, SNCA mutation, and LRRK2 mutation.</p>
Full article ">
19 pages, 5619 KiB  
Article
Paraquat Poisoning: Insights from Autopsy, Histology, and Liquid Chromatography with Tandem Mass Spectrometry in Multidisciplinary Forensic Toxicology Practice
by Issarapong Nuwongsa, Tanyarat Markmee, Nareerat Pholpo, Manoch Chockjamsai, Tawachai Monum, Yutti Amornlertwatana and Preechaya Tajai
Toxics 2024, 12(9), 675; https://doi.org/10.3390/toxics12090675 - 16 Sep 2024
Viewed by 421
Abstract
The herbicide paraquat (PQ) is responsible for a significant number of fatalities resulting from self-poisoning. Nevertheless, only a limited number of comprehensive studies focusing on fatal PQ poisoning, which include examination of autopsy findings, histopathology, and quantitative analysis of post-mortem samples, have been [...] Read more.
The herbicide paraquat (PQ) is responsible for a significant number of fatalities resulting from self-poisoning. Nevertheless, only a limited number of comprehensive studies focusing on fatal PQ poisoning, which include examination of autopsy findings, histopathology, and quantitative analysis of post-mortem samples, have been published. This study aimed to evaluate autopsy findings, histopathology, and quantitative analysis of PQ in post-mortem human serum samples using liquid chromatography-tandem mass spectrometry (LC-MS/MS), a simple, sensitive, and specific method. Autopsies were performed on all deaths due to PQ poisoning, and serum samples were sent to the toxicology laboratory for chemical analysis. The method was successfully applied to seven human serum samples, and the results indicate its reliability for detecting PQ. The study reports fatal serum PQ levels ranging from 0.5 to 372.0 µg/mL. The comprehensive data presented in this study can be useful for further research and practical applications. Full article
(This article belongs to the Section Agrochemicals and Food Toxicology)
Back to TopTop