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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (383)

Search Parameters:
Keywords = chelation therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
8 pages, 525 KiB  
Article
Evaluation of Solubility and Complexation Ability of Vanillic, Syringic and Gallic Acids Towards Aluminum Cation
by Donatella Aiello, Tiziana Marino, Anna Napoli, Emilia Furia and Pierluigi Plastina
Inorganics 2025, 13(1), 2; https://doi.org/10.3390/inorganics13010002 - 25 Dec 2024
Viewed by 218
Abstract
Chelation therapy is currently successfully applied to reduce the aluminum burden and its neurodegenerative consequences. In view of a possible application to aluminum chelation therapy, here we have studied the complexation of hydroxybenzoic acids, namely, vanillic, syringic and gallic acids, towards aluminum ion [...] Read more.
Chelation therapy is currently successfully applied to reduce the aluminum burden and its neurodegenerative consequences. In view of a possible application to aluminum chelation therapy, here we have studied the complexation of hydroxybenzoic acids, namely, vanillic, syringic and gallic acids, towards aluminum ion at physiologically relevant conditions as regards temperature (37 °C) and ionic strength (i.e., 0.16 M NaCl). The solubility values and the protonation constants of the hydroxybenzoic acids were primarily assessed to estimate the competition of these acids towards aluminum and H+ ions. Then, potentiometric titrations were carried out, and the speciation analysis indicated a pH-dependent complexation occurring at a 1:1 hydroxybenzoic acid-to-aluminum ratio for vanillic and syringic, and 1:1, 2:1 and 3:1 ligand-to-Al(III) ratios for gallic. Gallic acid forms more stable complexes with Al(III) ion than vanillic and syringic acids and could therefore represent a good candidate for being used as sequestering agents for Al(III) ion. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of hydroxybenzoic acids.</p>
Full article ">Figure 2
<p>UV–Vis spectra of the free ligand 0.1 mM (violet line) and of the complexes formed by phenolic acids and AlCl<sub>3</sub> 0.1 mM (red line): (<b>a</b>) vanillic acid; (<b>b</b>) syringic acid; (<b>c</b>) gallic acid.</p>
Full article ">
23 pages, 2443 KiB  
Article
Neuroprotective Potential of Indole-Based Compounds: A Biochemical Study on Antioxidant Properties and Amyloid Disaggregation in Neuroblastoma Cells
by Tania Ciaglia, Maria Rosaria Miranda, Simone Di Micco, Mariapia Vietri, Gerardina Smaldone, Simona Musella, Veronica Di Sarno, Giulia Auriemma, Carla Sardo, Ornella Moltedo, Giacomo Pepe, Giuseppe Bifulco, Carmine Ostacolo, Pietro Campiglia, Michele Manfra, Vincenzo Vestuto and Alessia Bertamino
Antioxidants 2024, 13(12), 1585; https://doi.org/10.3390/antiox13121585 - 23 Dec 2024
Viewed by 421
Abstract
Based on the established neuroprotective properties of indole-based compounds and their significant potential as multi-targeted therapeutic agents, a series of synthetic indole–phenolic compounds was evaluated as multifunctional neuroprotectors. Each compound demonstrated metal-chelating properties, particularly in sequestering copper ions, with quantitative analysis revealing approximately [...] Read more.
Based on the established neuroprotective properties of indole-based compounds and their significant potential as multi-targeted therapeutic agents, a series of synthetic indole–phenolic compounds was evaluated as multifunctional neuroprotectors. Each compound demonstrated metal-chelating properties, particularly in sequestering copper ions, with quantitative analysis revealing approximately 40% chelating activity across all the compounds. In cellular models, these hybrid compounds exhibited strong antioxidant and cytoprotective effects, countering reactive oxygen species (ROS) generated by the Aβ(25–35) peptide and its oxidative byproduct, hydrogen peroxide, as demonstrated by quantitative analysis showing on average a 25% increase in cell viability and a reduction in ROS levels to basal states. Further analysis using thioflavin T fluorescence assays, circular dichroism, and computational studies indicated that the synthesized derivatives effectively promoted the self-disaggregation of the Aβ(25–35) fragment. Taken together, these findings suggest a unique profile of neuroprotective actions for indole–phenolic derivatives, combining chelating, antioxidant, and anti-aggregation properties, which position them as promising compounds for the development of multifunctional agents in Alzheimer’s disease therapy. The methods used provide reliable in vitro data, although further in vivo validation and assessment of blood–brain barrier penetration are needed to confirm therapeutic efficacy and safety. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Scheme of the synthesized molecules.</p>
Full article ">Figure 2
<p>(<b>A</b>) UV spectra (in the range of 280 to 400 nm) of compounds (30 µM) alone and in the presence of 40 µM FeSO<sub>4</sub>, FeCl<sub>3</sub>, and CuSO<sub>4</sub>. (<b>B</b>) Copper-chelating quantitative analyses of compounds (30 µM). EDTA (1 mM) was used as the positive control. Results are shown as mean ± standard deviation (SD) from three independent experiments. **, *** denote <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001 vs. Ctrl.</p>
Full article ">Figure 3
<p>Neuroprotective activity of compounds. (<b>A</b>) SH-SY5Y cells were exposed to compounds at a concentration of 30 µM. Neuroprotective effects of compounds against (<b>B</b>) H<sub>2</sub>O<sub>2</sub>-induced (500 μM) cytotoxicity, (<b>C</b>) H<sub>2</sub>O<sub>2</sub>-induced (500 μM) ROS production, and (<b>D</b>) Aβ(25–35)-induced (40 μM) cytotoxicity. The 2′,7′-dichlorofluorescin diacetate (DCFH-DA) assay was conducted to reveal ROS production. The changes in viability were determined by calculating the percentage of viable cells in treated cultures relative to untreated controls. The results are presented as the mean ± standard deviation (SD) from three independent experiments. *, **, and *** denote, respectively, <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01, and <span class="html-italic">p</span> &lt; 0.001 vs. Ctrl; <sup>##</sup>, and <sup>###</sup> denote, respectively, <span class="html-italic">p</span> &lt; 0.01, and <span class="html-italic">p</span> &lt; 0.001 vs. H<sub>2</sub>O<sub>2</sub> or Aβ(25–35).</p>
Full article ">Figure 4
<p>Disaggregating properties of compounds. In-cell ThT assay was performed for both fluorescence microscopy (<b>A</b>) and spectrophotometry (<b>B</b>). Scale bar: 100 μm. (<span class="html-italic">N</span> ≥ 10). Cells were observed at 20× magnification. (<b>C</b>) ThT shows a direct disaggregating effect of compounds against Aβ. Data are shown as mean ± SD of three different experiments performed in triplicate. *** denotes <span class="html-italic">p</span> &lt; 0.001 vs. Ctrl; <sup>##</sup>, and <sup>###</sup> denote, respectively, <span class="html-italic">p</span> &lt; 0.01, and <span class="html-italic">p</span> &lt; 0.001 vs. H<sub>2</sub>O<sub>2</sub> or Aβ(25–35).</p>
Full article ">Figure 5
<p>(<b>A</b>,<b>B</b>) CD curves and secondary structure analysis of the Aβ(25−35) peptide were performed using the CONTIN algorithm after 24 h of aggregation. Aβ(25–35) 40 μM was used as a positive control. A different colour has been selected for Aβ(25–35) and each compound in presence Aβ(25–35), as indicated in the legend.</p>
Full article ">Figure 6
<p>Three-dimensional model of the interactions of <b>12</b> (<b>A</b>), <b>13</b> (<b>B</b>), <b>14</b> (<b>C</b>), <b>20</b> (<b>D</b>), <b>21</b> (<b>E</b>), and <b>22</b> (<b>F</b>) with Aβ(25–35). The biological target is depicted by a ribbon colored by a chain (D, magenta; E, azure; F, green; G, faded red) and tube (colored: C, by chain; polar H, white; N, dark blue; O, red; S, yellow). The small molecules are represented by sticks (gray for <b>12</b>, cyan for <b>13</b>, violet for <b>14</b>, orange for <b>20</b>, black for <b>21</b>, khaki for <b>22</b>) and balls (colored: C, as for the sticks; polar H, white; N, dark blue; O, red). The hydrogen bonds between the ligand and Aβ(25–35) are represented by the dashed black lines.</p>
Full article ">
17 pages, 600 KiB  
Review
Reproductive Health in Women with Major β-Thalassemia: Evaluating Ovarian Reserve and Endocrine Complications
by Vasileios Tsilionis, Efthalia Moustakli, Stefanos Dafopoulos, Athanasios Zikopoulos, Sotirios Sotiriou, Athanasios Zachariou and Konstantinos Dafopoulos
Metabolites 2024, 14(12), 717; https://doi.org/10.3390/metabo14120717 - 20 Dec 2024
Viewed by 483
Abstract
Thalassemia is an autosomal recessive hereditary chronic hemolytic anemia characterized by a partial or complete deficiency in the synthesis of alpha- or beta-globin chains, which are essential components of adult hemoglobin. Mutations in the globin genes lead to the production of unstable globin [...] Read more.
Thalassemia is an autosomal recessive hereditary chronic hemolytic anemia characterized by a partial or complete deficiency in the synthesis of alpha- or beta-globin chains, which are essential components of adult hemoglobin. Mutations in the globin genes lead to the production of unstable globin chains that precipitate within cells, causing hemolysis. This shortens the lifespan of mature red blood cells (RBCs) and results in the premature destruction of RBC precursors in the bone marrow. Regular red blood cell transfusions are the standard treatment for thalassemia. However, these transfusions can lead to increased iron overload, which can impair vital systems such as the liver, heart, ovaries, and endocrine system. Focusing on female reproductive endocrinology, recurrent blood transfusions can cause iron accumulation in the pituitary and hypothalamus, leading to hypogonadotropic hypogonadism (HH), the most common endocrinopathy in these patients, affecting 40–91% of women. Recurrent transfusions and the resulting iron overload can also lead to oxidative stress and ovarian damage in patients with beta-thalassemia major (BTM). Despite advancements in iron chelation therapy, hypothalamic–pituitary damage associated with HH contributes to subfertility and sexual dysfunction, often with little to no recovery. In women exposed to gonadotoxic drugs, particularly those with BTM, anti-Mullerian hormone (AMH)—a marker of ovarian reserve—is frequently used to assess ovarian damage. This review aims to explore the pathophysiology of β-thalassemia and its major clinical manifestations, with a focus on endocrine complications and their impact on ovarian reserve. It also investigates how metabolomics can provide insights into the disease’s metabolic alterations and inform current and emerging therapeutic strategies to mitigate complications and optimize patient outcomes, potentially leading to more effective and personalized treatments. Full article
Show Figures

Figure 1

Figure 1
<p>The primary cause of infertility in female patients with β-thalassemia major (BTM) seems to be attributable to both direct and indirect effects of iron overload. Dysfunction in the female reproductive axis arises from the synergistic impacts of increased oxidative stress and iron deposition, which result from a significant imbalance between pro-oxidants and antioxidants.</p>
Full article ">
15 pages, 882 KiB  
Review
Sex and Gender Differences in Iron Chelation
by Sarah Allegra, Stefano Comità, Antonella Roetto and Silvia De Francia
Biomedicines 2024, 12(12), 2885; https://doi.org/10.3390/biomedicines12122885 - 18 Dec 2024
Viewed by 670
Abstract
Background/Objectives: In the absence of physiological mechanisms to excrete excessive iron, the administration of iron chelation therapy is necessary. Age and hormones have an impact on the absorption, distribution, metabolism, and excretion of the medications used to treat iron excess, resulting in notable [...] Read more.
Background/Objectives: In the absence of physiological mechanisms to excrete excessive iron, the administration of iron chelation therapy is necessary. Age and hormones have an impact on the absorption, distribution, metabolism, and excretion of the medications used to treat iron excess, resulting in notable sex- and gender-related variances. Methods: Here, we aimed to review the literature on sex and gender in iron overload assessment and treatment. Results: The development of iron chelators has shown to be a successful therapy for lowering the body’s iron levels and averting the tissue damage and organ failure that follows. Numerous studies have described how individual factors can impact chelation treatment, potentially impact therapeutic response, and/or result in inadequate chelation or elevated toxicity; however, most of these data have not considered male and female patients as different groups, and particularly, the effect of hormonal variations in women have never been considered. Conclusions: An effective iron chelation treatment should take into account sex and gender differences. Full article
(This article belongs to the Special Issue Gender Medicine and Pharmacology, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Iron metabolism scheme. Holo-transferrin (Holo-Tf) is created when apo-Tf binds to Fe3+. On the cell surface, Holo-Tf and transferrin receptor 1 (TfR1) form a complex, which is then endocytosed. In the endosome, Fe3+ is liberated from Holo-Tf by a proton pump and converted into Fe2+ by six-transmembrane epithelial antigen of the prostate 3 (Steap3). Divalent metal transporter 1 (DMT1) transports Fe3+ into the cytosol by crossing the endosomal membrane. To begin a new cycle, the apo-Tf released is recycled back to the plasma membrane. The newly acquired iron enters the cytosolic labile iron pool (LIP) and is distributed into various cellular compartments. Unused cellular iron is either exported by ferroportin or retained in ferritin.</p>
Full article ">Figure 2
<p>Scheme of hepcidin synthesis. HFE is removed from TFR1 by Tf-Fe2, which then combines with TFR2 and HJV to stimulate hepcidin through bone morphogenetic protein (BMP)/SMAD signaling. Hepcidin is activated by HJV as a co-receptor for the cytokines BMP2 and BMP6. This route involves BMP receptors type I (BMPRI) and type II (BMPRII). To trigger the production of genes controlled by BMP-responsive elements, such as hepcidin, SMAD4 translocate to the nucleus. Matriptase 2, a serine protease encoded by TMPRSS6, cleaves and produces a soluble form of HJV, suppressing BMP/SMAD signaling to hepcidin. Through the interaction of the STAT3 pathway and interleukin-6 (IL6) with its receptor (IL6R), infection and inflammation significantly boost the synthesis of hepcidin.</p>
Full article ">
16 pages, 3231 KiB  
Article
Monovalent and Divalent Designs of Copper Radiotheranostics Targeting Fibroblast Activation Protein in Cancer
by Pawan Thapa, Sashi Debnath, Anjan Bedi, Madhuri Parashar, Paulina Gonzalez, Joshua Reus, Hans Hammers and Xiankai Sun
Cancers 2024, 16(24), 4180; https://doi.org/10.3390/cancers16244180 - 15 Dec 2024
Viewed by 555
Abstract
Background: Fibroblast activation protein (FAP)-targeted theranostic radiopharmaceuticals have shown desired tumor-to-background organ selectivity due to the ubiquitous presence of FAP within the tumor microenvironment. However, suboptimal tumor retention and fast clearance have hindered their use to deliver effective cancer therapies. With well-documented [...] Read more.
Background: Fibroblast activation protein (FAP)-targeted theranostic radiopharmaceuticals have shown desired tumor-to-background organ selectivity due to the ubiquitous presence of FAP within the tumor microenvironment. However, suboptimal tumor retention and fast clearance have hindered their use to deliver effective cancer therapies. With well-documented FAP-targeting moieties and linkers appending them to optimal chelators, the development of copper radiopharmaceuticals has attracted considerable interest, given the fact that an ideal theranostic pair of copper radionuclides (64Cu: t1/2 = 12.7 h; 17.4% β+; Eβ+max = 653 keV and 67Cu: t1/2 = 2.58 d; 100% β; Eβmax = 562 keV) are available. Herein, we report our design, synthesis, and comparative evaluation of monovalent and divalent FAP-targeted theranostic conjugates constructed from our previously reported bifunctional chelator scaffold (BFS) based on 1,4,8,11-tetraaza-bicyclo [6.6.2]hexadecane-4,11-diacetic acid (CB-TE2A), which forms the most stable complex with Cu(II). Methods: After synthesis and characterization, the monovalent and divalent conjugates were radiolabeled with 64Cu for in vitro cell assays, followed by in vivo positron emission tomography (PET) imaging evaluation in relevant mouse models. Results: Both 64Cu-labeled conjugates showed high in vitro stability and anticipated FAP-mediated cell binding and internalization. The divalent one showed significantly higher FAP-specific tumor uptake than its monovalent counterpart. Conclusions: Our results demonstrate that the BFS-based multivalent approach can be practically used to generate FAP-targeted radiotheranostic agents for effective cancer diagnosis and treatment. Full article
(This article belongs to the Section Cancer Drug Development)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) UAMC1110, lead structure for FAPI development; (<b>B</b>) multivalent conjugate design for FAP-targeted copper radiotheranostics.</p>
Full article ">Figure 2
<p>Cell-based FAP-binding affinity and internalization assays of [<sup>64</sup>Cu]Cu-CB-TE2A-FAPI-04 (upper panel) and [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub> (lower panel). (<b>A</b>,<b>D</b>): FAP-specific binding assay using UAMC1110 as blockade; (<b>B</b>,<b>E</b>): Calculated ratios of total uptake vs. nonspecific uptake; (<b>C</b>,<b>F</b>): time-dependent internalization. Cell lines: RENCA-FAP (FAP<sup>+</sup>) and RENCA (FAP<sup>−</sup>).</p>
Full article ">Figure 3
<p>(<b>A</b>): Representative PET/CT images of [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub> in mice bearing RENCA-FAP (FAP<sup>+</sup>) tumors (yellow arrows indicate the tumors) at different time points. (<b>B</b>): Biodistribution of [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub> by quantitative imaging data analysis in the mice. Data presented as average %ID/g ± standard deviation (s.d.) (n = 3). (<b>C</b>): Tumor/muscle ratio at different time points. (<b>D</b>–<b>F</b>): Immunofluorescence staining assays of excised RENCA-FAP tumor tissues post imaging. The nuclei were stained with DAPI (<b>D</b>) and tumor cells with anti-FAP antibody (<b>E</b>). (<b>F</b>): Overlay of the FAP and DAPI staining.</p>
Full article ">Figure 4
<p>(<b>A</b>): Representative PET/CT images of XP-185 tumorgrafts in SCID/NOD mice (n = 4) with divalent [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub>. (<b>B</b>): Images obtained with co-injection of UAMC1110 with divalent [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub>. (<b>C</b>): Representative PET/CT images of XP-185 tumorgrafts in SCID/NOD mice (n = 4) with monovalent [<sup>64</sup>Cu]Cu-CB-TE2A-FAPI-04. Tumors are indicated with yellow arrows.</p>
Full article ">Figure 5
<p>Quantitative analysis of PET signals in tissues of interest in SCID/NOD mice bearing XP-185 tumorgrafts (n = 4). (<b>A</b>): [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub>. (<b>B</b>): [<sup>64</sup>Cu]Cu-CB-TE2A-(FAPI-04)<sub>2</sub> co-injected with UAMC1110 (tumor blocked vs. tumor unblocked at 4 h p.i., <span class="html-italic">p</span> = 0.0093.). (<b>C</b>): [<sup>64</sup>Cu]Cu-CB-TE2A-FAPI-04. Tumors are indicated with orange dotted boxes.</p>
Full article ">Scheme 1
<p>Synthetic route to monovalent and divalent FAPI-04 conjugates. Reagents and reaction condition for monovalent conjugate: FAPI-04 (1.2 equiv.), CB-TE2A(<span class="html-italic"><sup>t</sup></span>Bu)<sub>2</sub>-COOH (1 equiv.), HBTU (1 equiv.), DIPEA (5 equiv.), 24 h; TFA, 6 h. Reagents and reaction condition for divalent conjugate: FAPI-04 (3.5 equiv.), CB-TE2A(<span class="html-italic"><sup>t</sup></span>Bu)<sub>2</sub>-(COOH)<sub>2</sub> (1 equiv.), HBTU (2 equiv.), DIPEA (10 equiv.), 24 h; TFA, 3 h.</p>
Full article ">
18 pages, 3579 KiB  
Article
Construction of Cisplatin-18-Crown-6 Complexes Through Supramolecular Chemistry to Improve Solubility, Stability, and Antitumor Activity
by Yue Gao, Yeqi Huang, Chuanyu Ren, Si Xiong, Xia Guo, Ziyu Zhao, Ling Guo and Zhengwei Huang
Int. J. Mol. Sci. 2024, 25(24), 13411; https://doi.org/10.3390/ijms252413411 - 14 Dec 2024
Viewed by 423
Abstract
Cisplatin (DDP), a platinum-chelated compound renowned for its antitumor activity, is often utilized in cancer therapy. However, its real-world clinical efficacy is compromised by poor solubility and low stability, which impedes wider clinical application. Our study aimed to address these limitations of DDP [...] Read more.
Cisplatin (DDP), a platinum-chelated compound renowned for its antitumor activity, is often utilized in cancer therapy. However, its real-world clinical efficacy is compromised by poor solubility and low stability, which impedes wider clinical application. Our study aimed to address these limitations of DDP through host–guest supramolecular chemistry approaches. We explored the potential of 18-crown-6 as the host molecule to solubilize and stabilize DDP, the guest molecule. Utilizing techniques such as UV–visible spectroscopy, Fourier-transform infrared spectroscopy, Raman spectroscopy, and molecular docking, we conducted a comprehensive analysis on the physical state and inclusion mode of the DDP@18-crown-6 complex. Phase solubility studies and Job’s plot confirmed that the DDP@18-crown-6 complex significantly enhanced the aqueous solubility of DDP, with an optimal 1:1 binding ratio. Stability analyses revealed that this complex markedly improved the stability of DDP in pure water. Meanwhile, the stabilization effects of DDP@18-crown-6 were remarkably elevated when combined with 0.9% sodium chloride. In vitro antitumor assays in A549 cell lines demonstrated that the DDP@18-crown-6 complex outperformed raw DDP in cytotoxicity, showing a significantly lower IC50 value. This research offered a promising strategy for DDP solubilization and stabilization, facilitating its anticancer therapeutic efficacy. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of cisplatin (<b>A</b>) and 18-crown-6 (<b>B</b>), and “crown” shaped like 18-crown-6 (<b>C</b>).</p>
Full article ">Figure 2
<p>The optimization of preparation time (<b>A</b>) and rotation rate (<b>B</b>) of DDP@18-crown-6 complex (<span class="html-italic">n</span> = 3) and optical photograph of cisplatin dispersed or dissolved (<b>C</b>) in ultrapure water (a) or 18-crown-6 aqueous solution (b). ANOVA or <span class="html-italic">t</span> tests were utilized to determine whether there are significant differences between the data. <span class="html-italic">p</span> value style: ** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>The UV–visible spectrogram of cisplatin, 18-crown-6, and the DDP@18-crown-6 complex.</p>
Full article ">Figure 4
<p>The FT-IR spectrogram (<b>A</b>) and Raman spectrogram (<b>B</b>) of cisplatin (a), 18-crown-6 (b), and DDP@18-crown-6 complex (c).</p>
Full article ">Figure 5
<p>The optimal binding conformation of cisplatin to 18-crown-6. The C, N, O, H, Cl, and Pt atoms are colored in cyan, blue, red, white, green, and orange, respectively. Orange dashed lines are coordinate covalent bonds, and blue dashed lines are hydrogen bonds.</p>
Full article ">Figure 6
<p>Phase solubility diagram (<b>A</b>) for cisplatin with 18-crown-6 in aqueous solution (<span class="html-italic">n</span> = 3) and UV–visible spectrogram of the complex at different 18-crown-6 concentrations (<b>B</b>). The linear regression equation for the phase solubility curves was <span class="html-italic">Y</span> = 0.03151<span class="html-italic">X</span> + 6.036 (<b>A</b>) and the correlation coefficient, <span class="html-italic">R</span><sup>2</sup>, was greater than 0.99.</p>
Full article ">Figure 7
<p>Job’s plot (<b>A</b>) (<span class="html-italic">n</span> = 3) and binding structural schematic (<b>B</b>) of cisplatin and 18-crown-6 at <span class="html-italic">X</span><sub>m</sub> = 0.9. The C, N, O, H, Cl, and Pt atoms are colored in cyan, blue, red, white, green, and orange, respectively. Orange dashed lines are coordinate covalent bonds. Red dashed lines indicate electrostatic interaction, and blue dashed lines are hydrogen bonds. ANOVA or <span class="html-italic">t</span> tests were utilized to determine whether there are significant differences between the data. <span class="html-italic">p</span> value style: **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>(<b>A</b>) Time-evolution stability of cisplatin (in H<sub>2</sub>O), cisplatin (in 0.9% NaCl aqueous solution), DDP@18-crown-6 complex (in H<sub>2</sub>O), and DDP@18-crown-6 complex (0.9% in NaCl aqueous solution); and (<b>B</b>) time-evolution stability of cisplatin at different 18-crown-6 concentrations (<span class="html-italic">n</span> = 3). ANOVA or <span class="html-italic">t</span> tests were utilized to determine whether there are significant differences between the data. <span class="html-italic">p</span> value style: ns: not significant (<span class="html-italic">p</span> &gt; 0.05); ** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>(<b>A</b>) Photostability of cisplatin (in H<sub>2</sub>O), cisplatin (in 0.9% NaCl aqueous solution), DDP@18-crown-6 complex (in H<sub>2</sub>O), and DDP@18-crown-6 complex (in 0.9% NaCl aqueous solution); and (<b>B</b>) photostability of cisplatin at different 18-crown-6 concentrations (<span class="html-italic">n</span> = 3). ANOVA or <span class="html-italic">t</span> tests were utilized to determine whether there are significant differences between the data. <span class="html-italic">p</span> value style: ns: not significant (<span class="html-italic">p</span> &gt; 0.05); **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 10
<p>In vitro cytotoxicity of cisplatin and DDP@18-crown-6 complex (<b>A</b>), and 18-crown-6 (<b>B</b>). (<span class="html-italic">n</span> = 3) ANOVA or <span class="html-italic">t</span> tests were utilized to determine whether there are significant differences between the data. <span class="html-italic">p</span> value style: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
10 pages, 219 KiB  
Case Report
A Novel Pathogenic Sense Variant in Exon 7 of the HK1 Gene in a Patient with Hexokinase Deficiency and Gilbert Syndrome
by Magdalena Bartnik, Weronika Pawlik, Beata Burzyńska, Konrad Wasilewski, Elżbieta Kamieńska and Tomasz Urasiński
Genes 2024, 15(12), 1576; https://doi.org/10.3390/genes15121576 - 7 Dec 2024
Viewed by 550
Abstract
Background: Hexokinase (HK) deficiency is a rare autosomal recessively inherited disease manifested by chronic nonspherocytic hemolytic anemia. Most patients present with a mild to severe course of the disease (fetal hydrocephalus, neonatal hyperbilirubinemia, severe anemia). We reviewed 37 cases of patients with hexokinase [...] Read more.
Background: Hexokinase (HK) deficiency is a rare autosomal recessively inherited disease manifested by chronic nonspherocytic hemolytic anemia. Most patients present with a mild to severe course of the disease (fetal hydrocephalus, neonatal hyperbilirubinemia, severe anemia). We reviewed 37 cases of patients with hexokinase deficiency described so far, focusing on the severity of the disease, clinical presentation, treatment applied, and genetic test results. Methods: We present a 10-year-old girl who initially presented with symptoms of weakness, excessive fatigue, and yellowing of the skin and sclerae. Genetic testing detected the (TA)7 variant in both alleles of the UGT1A1 gene and diagnosed Gilbert’s disease. In the follow-up, red cell hemolysis was observed. The diagnosis was extended, and tests for red cell enzymopathy were performed and a reduced level of hexokinase—0.65 IU/gHb (normal 0.78–1.57) was found. Next-generation sequencing revealed a new sense-change variant in exon 7 in the hexokinase gene not previously reported in databases. Results: Up to this date, only around 37 cases of hexokinase deficiency associated with hereditary nonspherocytic hemolytic anemia have been documented around the world. Diagnosing hexokinase deficiency involves clinical evaluation, laboratory testing, and genetic analysis. Management focuses on treating symptoms and preventing complications; there is no cure for the underlying enzyme deficiency. In patients with severe anemia, the treatment is multiple blood transfusions followed by iron chelation therapy. Conclusions: Understanding and diagnosing hexokinase deficiency is critical for providing appropriate care and improving the quality of life for affected individuals. Full article
(This article belongs to the Special Issue Variations of Rare Genetic Diseases)
19 pages, 2853 KiB  
Article
Clinical Features, Microbiological Characteristics, and Drug Sensitivity Analysis of Rare Human Spinal Pythiosis Strain
by Mingliang Li, Donglin Zhu, Qiuyue Diao, Xiaoyun Liu, Xiaogang Bi, Jianwen Dong, Jian Sun, Yun Xi and Kouxing Zhang
J. Fungi 2024, 10(12), 812; https://doi.org/10.3390/jof10120812 - 22 Nov 2024
Viewed by 913
Abstract
Pythiosis, a rare and formidable infectious disease caused by Pythium insidiosum, is characterized by profound uncertainties in achieving definitive diagnoses, suboptimal outcomes, and an exceptionally high mortality rate. Here, we present a rare case of human spinal pythiosis in southern China. With [...] Read more.
Pythiosis, a rare and formidable infectious disease caused by Pythium insidiosum, is characterized by profound uncertainties in achieving definitive diagnoses, suboptimal outcomes, and an exceptionally high mortality rate. Here, we present a rare case of human spinal pythiosis in southern China. With advanced metagenomic sequencing technology, Pythium insidiosum was pinpointed as the causative pathogen. We discovered that the inoculation of either tissue fragments or homogenate yielded more successful results and enabled a moderate extension of the culture duration to 5–10 days through an exhaustive comparison of diverse inoculation and culture conditions for general clinical specimens. A pronounced genetic affinity of the isolated strain towards the Pythium insidiosum strain MCC 13 was detected after a comprehensive whole-genome sequencing analysis. Antifungal agents exhibited negligible sensitivity towards Pythium insidiosum in an antimicrobial susceptibility test. Conversely, antibacterial agents such as oxazolidinones, tetracyclines, macrolides, and amphenicols demonstrated varying degrees of sensitivity, albeit with most of their minimum inhibitory concentrations (MICs) substantially surpassing the safe concentration ranges for effective clinical treatment. Notably, tigecycline stood out as a promising candidate, exhibiting favorable therapeutic effects at moderate concentrations, making it a potential drug of choice for the control of pythiosis. A combined susceptibility test suggested that combinations of tetracyclines with macrolides, oxazolidinones, and amphenicols exhibited synergistic antibacterial effects, with the combination of doxycycline and trimethoprim–sulfamethoxazole (TMP-SMX) in particular playing a pivotal role. To our surprise, the MICs of iron chelators, specifically deferiprone and deferoxamine, against the strain were exceedingly low, which led to the speculation that exogenous iron chelators may have competitively inhibited the iron-chelating enzymes of the strain. The research derived from this single, rare case has certain limitations, but considering that there are currently no reports of invasive infections of deep organs in humans caused by Pythium insidiosum, the above findings can offer novel insights into the treatment of invasive pythiosis. Combination therapy based on tetracyclines, especially tigecycline, the use of TMP-SMX, and the adjunctive use of iron chelators, represent promising approaches to tackle the clinical challenges in the treatment of invasive pythiosis. However, further studies, including similar cases of spinal pythiosis and in vivo trials, are still needed to validate them. In addition, while paying attention to the therapeutic potentials of the above plans, we should also closely monitor the risks and side effects that may arise from excessive MICs or the expanded use of related drugs during the treatment process. Full article
(This article belongs to the Section Fungal Pathogenesis and Disease Control)
Show Figures

Figure 1

Figure 1
<p>MRI scan of cervical, thoracic, spine, and lumbar spine. (<b>A</b>) Pre-first surgery (November 2022). (<b>B</b>) Pre-second surgery (December 2022). (<b>C</b>) Pre-third surgery (October 2023). (<b>D</b>) Pre-fourth surgery (January 2024). (<b>E</b>) Follow-up imaging (February 2024).</p>
Full article ">Figure 2
<p>Flowchart of the diagnosis and treatment.</p>
Full article ">Figure 3
<p>Microbiological morphology of <span class="html-italic">Pythium insidiosum</span>. (<b>A</b>) Growth curve of the <span class="html-italic">Pythium insidiosum</span> inoculated onto LB agar plates. (<b>B</b>) Morphology of the strain after 4 days of incubation at 37 °C on a 90 mm LB agar plate. (<b>C</b>) Morphology of the mycelium observed under an optical microscope (400×). (<b>D</b>) A lactophenol cotton blue staining image of the mycelium (400×). (<b>E</b>) A fluorescence staining image of the mycelium (400×). (<b>F</b>) Induced zoospore formation in the mycelium observed under an optical microscope (400×). Arrow indicates that the motile spores transformed into spherical shapes, with the loss of flagella.</p>
Full article ">Figure 4
<p>Cultivation of <span class="html-italic">Pythium insidiosum</span> (48–72 h). (<b>A</b>) Visible robust colony growth after mycelial plug inoculation. (<b>B</b>) Apparent colony growth after mycelial suspension inoculation. (<b>C</b>) No colony growth observed after spore suspension inoculation. (<b>D</b>) No colony growth observed after streak plate inoculation. (<b>E</b>) No colony growth observed after spread plate inoculation. (<b>F</b>) No colony growth observed after puncture inoculation.</p>
Full article ">Figure 5
<p>The coverage map of the <span class="html-italic">Pythium insidiosum</span> strain MCC 13.</p>
Full article ">Figure 6
<p>Phylogenetic trees show the phylogenetic position of the sample in this study within the evolutionary radiation of the genus <span class="html-italic">Pythium</span>. The highlighted red part represents the strain isolated from tissue biopsy, which showed stronger similarity with the <span class="html-italic">Pythium insidiosum</span> strain MCC 13 through blastn sequence alignment analysis.</p>
Full article ">
23 pages, 928 KiB  
Review
Beta Thalassemia in Children: Established Approaches, Old Issues, New Non-Curative Therapies, and Perspectives on Healing
by Raffaella Origa and Layal Issa
J. Clin. Med. 2024, 13(22), 6966; https://doi.org/10.3390/jcm13226966 - 19 Nov 2024
Viewed by 1050
Abstract
Despite a decrease in prevalence and incidence rates, beta thalassemia continues to represent a significant public health challenge worldwide. In high-resource settings, children with thalassemia have an open prognosis, with a high chance of reaching adulthood and old age with a good quality [...] Read more.
Despite a decrease in prevalence and incidence rates, beta thalassemia continues to represent a significant public health challenge worldwide. In high-resource settings, children with thalassemia have an open prognosis, with a high chance of reaching adulthood and old age with a good quality of life. This is achievable if transfusion therapy is properly managed, effectively mitigating ineffective erythropoiesis and its associated complications while also minimizing excessive iron accumulation. Adequate iron chelation is essential to maintain reactive forms of iron within the normal range throughout life, thus preventing organ damage caused by hemosiderosis, which inevitably results from a regular transfusion regimen. New therapies, both curative, such as gene therapy, and non-curative, such as modulators of erythropoiesis, are becoming available for patients with transfusion-dependent beta thalassemia. Two curative approaches based on gene therapy have been investigated in both adults and children with thalassemia. The first approach uses a lentivirus to correct the genetic defect, delivering a functional gene copy to the patient’s cells. The second approach employs CRISPR/Cas9 gene editing to directly modify the defective gene at the molecular level. No non-curative therapies have received approval for pediatric use. Among adults, the only available drug is luspatercept, which is currently undergoing clinical trials in pediatric populations. However, in many countries around the world, the new therapeutic options remain a mirage, and even transfusion therapy itself is not guaranteed for most patients, while the choice of iron chelation therapy depends on drug availability and affordability. Full article
(This article belongs to the Section Hematology)
Show Figures

Figure 1

Figure 1
<p>Obvious deformity of the radius and ulna in a woman with beta thalassemia who used desferrioxamine doses of up to 100 mg/kg during the first two years of life.</p>
Full article ">Figure 2
<p>Comparison between conventional therapy and new therapeutic approaches and related challenges. HSCT: hematopoietic stem cell transplantation; TMPRSS6: transmembrane serine protease 6.</p>
Full article ">
18 pages, 1230 KiB  
Review
Optimization Processes of Clinical Chelation-Based Radiopharmaceuticals for Pathway-Directed Targeted Radionuclide Therapy in Oncology
by Katsumi Tomiyoshi, Lydia J. Wilson, Firas Mourtada, Jennifer Sims Mourtada, Yuta Namiki, Wataru Kamata, David J. Yang and Tomio Inoue
Pharmaceutics 2024, 16(11), 1458; https://doi.org/10.3390/pharmaceutics16111458 - 15 Nov 2024
Viewed by 915
Abstract
Targeted radionuclide therapy (TRT) for internal pathway-directed treatment is a game changer for precision medicine. TRT improves tumor control while minimizing damage to healthy tissue and extends the survival for patients with cancer. The application of theranostic-paired TRT along with cellular phenotype and [...] Read more.
Targeted radionuclide therapy (TRT) for internal pathway-directed treatment is a game changer for precision medicine. TRT improves tumor control while minimizing damage to healthy tissue and extends the survival for patients with cancer. The application of theranostic-paired TRT along with cellular phenotype and genotype correlative analysis has the potential for malignant disease management. Chelation chemistry is essential for the development of theranostic-paired radiopharmaceuticals for TRT. Among image-guided TRT, 68Ga and 99mTc are the current standards for diagnostic radionuclides, while 177Lu and 225Ac have shown great promise for β- and α-TRT, respectively. Their long half-lives, potent radiobiology, favorable decay schemes, and ability to form stable chelation conjugates make them ideal for both manufacturing and clinical use. The current challenges include optimizing radionuclide production processes, coordinating chelation chemistry stability of theranostic-paired isotopes to reduce free daughters [this pertains to 225Ac daughters 221Fr and 213Bi]-induced tissue toxicity, and improving the modeling of micro dosimetry to refine dose–response evaluation. The empirical approach to TRT delivery is based on standard radionuclide administered activity levels, although clinical trials have revealed inconsistent outcomes and normal-tissue toxicities despite equivalent administered activities. This review presents the latest optimization methods for chelation-based theranostic radiopharmaceuticals, advancements in micro-dosimetry, and SPECT/CT technologies for quantifying whole-body uptake and monitoring therapeutic response as well as cytogenetic correlative analyses. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structures of clinical theranostics: DOTATATE, PSMA and FAP-2286.</p>
Full article ">Figure 2
<p>Emergent trends on how optimization processes of chelation-based radiopharmaceuticals for pathway-directed systems impact healthcare systems.</p>
Full article ">
20 pages, 3330 KiB  
Article
Extraction, Purification, Characterization, and Wound Healing Effects of Novel Prickly Pear (Opuntiaficus-indica (L.) Mill.) Heteropolysaccharides
by Naourez Ktari, Wafa Gargouri, Lobna Jlaiel, Imen Trabelsi, Sirine Ben Slima, Sana Bardaa, Farida Bendali and Riadh Ben Salah
Pharmaceuticals 2024, 17(10), 1410; https://doi.org/10.3390/ph17101410 - 21 Oct 2024
Cited by 1 | Viewed by 917
Abstract
Background: The present study undertakes the purification of a novel polysaccharide from Tunisian prickly pear (Opuntiaficus-indica (L.) Mill.) rackets (PPPRs) and the determination of its physicochemical properties, structure, antibacterial and antioxidant properties, as well as its in vitro and in vivo wound healing [...] Read more.
Background: The present study undertakes the purification of a novel polysaccharide from Tunisian prickly pear (Opuntiaficus-indica (L.) Mill.) rackets (PPPRs) and the determination of its physicochemical properties, structure, antibacterial and antioxidant properties, as well as its in vitro and in vivo wound healing potential. Methods: The PPPR was structurally analyzed by Fourier Transform Infrared Spectroscopy (FTIR) and UV/Visible Spectroscopy, revealing characteristic bands of polysaccharides. According to thin-layer chromatography (TLC), high-performance liquid chromatography (HPLC), and Gas Chromatography–Mass Spectrometry (GC–MS) analyses. Results: The crude PPPR is an heteropolysaccharide composed of glucose (62.4%), galactose (19.37%), mannose (10.24%), and rhamnose (7.98%), with an average molecular weight of 90.94 kDa. This novel polysaccharide exhibited notable antioxidant potential assessed by four different in vitro assays: the 2-diphenyl-1-picrylhydrazyl (DPPH) scavenging assay, ferric reducing power, ferrous chelating activity, and scavenging activity against 2,2′-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid (ABTS). In addition, the PPPR displayed high antibacterial activities with a MIC of 2.5 mg/mL against Salmonella Typhimurium and Pseudomonas aeruginosa, cytocompatibility properties, and non-cytotoxicity. Subsequently, the effect of the PPPR on skin wound healing was studied in a diabetic rat model induced by alloxan, revealing a significant acceleration in the wound healing process. This acceleration was evidenced by the expedited recovery of the dermis, increased formation of blood vessels, and enhanced tissue granulation. Conclusion: Therefore, the findings offer fresh perspectives on the creation of a potentially efficient and promising racket polysaccharide-based therapy for the treatment of persistent diabetic wounds. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

Figure 1
<p>Scan of the PPPR within the wavelength range of 200–800 nm.</p>
Full article ">Figure 2
<p>Fourier transform infrared spectrum of the PPPR.</p>
Full article ">Figure 3
<p>Monosaccharide composition of the PPPR. (<b>a</b>) TLC of the PPPR, (1) hydrolyzed PPPR. (2) Rhamnose, (3) xylose, (4) galactose, (5) glucose, (6) arabinose, (7) mannose, and (8) fructose were used as standards. (<b>b</b>) HPLC chromatogram profiles of PPPR.</p>
Full article ">Figure 4
<p>Gel filtration chromatographs of the PPPR.</p>
Full article ">Figure 5
<p>Differential scanning calorimetry results for the thermal behavior of the PPPR.</p>
Full article ">Figure 6
<p>Effects of the PPPR on HEK-293 cell viability.</p>
Full article ">Figure 7
<p>Antioxidant activities of the PPPR at different concentrations.(<b>a</b>) Scavenging effect on the DPPH free radical, (<b>b</b>) reducing power, (<b>c</b>) metal chelating activity, and (<b>d</b>) ABTS scavenging activity. BHT, EDTA, and Trolox were used as positive controls. All analyses were carried out in triplicate.</p>
Full article ">Figure 8
<p>(<b>a</b>) Photographs of wounds taken of diabetic rats treated with physiological serum, Cytolcentella<sup>®</sup>, glycerol, or PPPR (15 mg/mL) on days 1, 3, 5, 10, 12, and 14, (<b>b</b>) Wound healing rate (%) for different treatment groups on wounds.</p>
Full article ">Figure 9
<p>Histological hematoxylin–eosin staining analysis of wounded skin tissue sections in the diabetic rats treated with physiological serum (<b>a</b>), « Cytol Centella<sup>®</sup> » (<b>b</b>), glycerol (<b>c</b>), and PPPR (<b>d</b>) on the 14th day post-wounding (×400 magnification). Der: Dermis; Ep: Epidermis; (<span class="html-fig-inline" id="pharmaceuticals-17-01410-i001"><img alt="Pharmaceuticals 17 01410 i001" src="/pharmaceuticals/pharmaceuticals-17-01410/article_deploy/html/images/pharmaceuticals-17-01410-i001.png"/></span>): Collagen; (<span class="html-fig-inline" id="pharmaceuticals-17-01410-i002"><img alt="Pharmaceuticals 17 01410 i002" src="/pharmaceuticals/pharmaceuticals-17-01410/article_deploy/html/images/pharmaceuticals-17-01410-i002.png"/></span>): Vessels; (<span class="html-fig-inline" id="pharmaceuticals-17-01410-i003"><img alt="Pharmaceuticals 17 01410 i003" src="/pharmaceuticals/pharmaceuticals-17-01410/article_deploy/html/images/pharmaceuticals-17-01410-i003.png"/></span>): inflammatory cells.</p>
Full article ">
22 pages, 1049 KiB  
Review
Thalassemia: Pathophysiology, Diagnosis, and Advances in Treatment
by Idris Zubairu Sadiq, Fatima Sadiq Abubakar, Hauwa Salisu Usman, Aliyu Dantani Abdullahi, Bashiru Ibrahim, Babangida Sanusi Kastayal, Maryam Ibrahim and Hassan Aliyu Hassan
Thalass. Rep. 2024, 14(4), 81-102; https://doi.org/10.3390/thalassrep14040010 - 15 Oct 2024
Viewed by 5052
Abstract
Thalassemia represents a diverse group of inherited hematological disorders characterized by defective globin chain synthesis, leading to chronic anemia and associated complications. The complicated pathophysiology of beta-thalassemia involves genetic mutations or rarely deletions of the beta-globin gene on chromosome 11 whereas alpha-thalassemia involves [...] Read more.
Thalassemia represents a diverse group of inherited hematological disorders characterized by defective globin chain synthesis, leading to chronic anemia and associated complications. The complicated pathophysiology of beta-thalassemia involves genetic mutations or rarely deletions of the beta-globin gene on chromosome 11 whereas alpha-thalassemia involves deletions in the HBA1 and HBA2 genes or occasionally alterations to the DNA sequence in or around these genes. These mutation and deletion effects disrupt the balance of α/β-globin chain production, resulting in ineffective erythropoiesis, hemolysis, and a cascade of clinical manifestations including anemia, bone deformities, and iron overload. Advances in diagnostic techniques have enhanced our ability to detect and characterize these mutations, facilitating early and accurate diagnoses. Current management strategies encompass regular blood transfusions, the use of hydroxyurea to improve hemoglobin levels, and iron chelation therapy to prevent iron-related organ damage. Moreover, other therapeutics such as thalidomide for those not responding to hydroxyurea, Sirolimus for patients with immunodeficiencies, and use of vitamin E as an antioxidant have proven to be effective. Innovative therapies such as gene therapy and bone marrow transplantation offer promising curative potential, opening a new era in the treatment of thalassemia. This review focuses on pathophysiological mechanisms underlying thalassemia, explores the diagnostic methodologies, and highlights recent advancements in therapeutic approaches. Full article
Show Figures

Figure 1

Figure 1
<p>Structure of hemoglobin: (<b>a</b>) 3D structure of hemoglobin, (<b>b</b>) 2D structure of hemoglobin showing α2β2 subunits of hemoglobin, (<b>c</b>) structure of hemoglobin showing binding of Fe<sup>2+</sup>, (<b>d</b>) Alpha Subunit showing number of amino acids, (<b>e</b>) Beta Subunit showing number of amino acids.</p>
Full article ">Figure 2
<p>Inherited pattern of thalassemia.</p>
Full article ">
20 pages, 1363 KiB  
Review
Review to Elucidate the Correlation between Cuproptosis-Related Genes and Immune Infiltration for Enhancing the Detection and Treatment of Cervical Cancer
by Pratibha Pandey, Seema Ramniwas, Shivam Pandey, Sorabh Lakhanpal, G. Padmapriya, Shivang Mishra, Mandeep Kaur, Ayash Ashraf, M Ravi Kumar and Fahad Khan
Int. J. Mol. Sci. 2024, 25(19), 10604; https://doi.org/10.3390/ijms251910604 - 1 Oct 2024
Viewed by 1280
Abstract
Copper is a vital trace element in oxidized and reduced forms. It plays crucial roles in numerous biological events such as redox chemistry, enzymatic reactions, mitochondrial respiration, iron metabolism, autophagy, and immune modulation. Maintaining the balance of copper in the body is essential [...] Read more.
Copper is a vital trace element in oxidized and reduced forms. It plays crucial roles in numerous biological events such as redox chemistry, enzymatic reactions, mitochondrial respiration, iron metabolism, autophagy, and immune modulation. Maintaining the balance of copper in the body is essential because its deficiency and excess can be harmful. Abnormal copper metabolism has a two-fold impact on the development of tumors and cancer treatment. Cuproptosis is a form of cell death that occurs when there is excessive copper in the body, leading to proteotoxic stress and the activation of a specific pathway in the mitochondria. Research has been conducted on the advantageous role of copper ionophores and chelators in cancer management. This review presents recent progress in understanding copper metabolism, cuproptosis, and the molecular mechanisms involved in using copper for targeted therapy in cervical cancer. Integrating trace metals and minerals into nanoparticulate systems is a promising approach for controlling invasive tumors. Therefore, we have also included a concise overview of copper nanoformulations targeting cervical cancer cells. This review offers comprehensive insights into the correlation between cuproptosis-related genes and immune infiltration, as well as the prognosis of cervical cancer. These findings can be valuable for developing advanced clinical tools to enhance the detection and treatment of cervical cancer. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Copper metabolism is regulated at both organ and cellular levels. Copper ion uptake is mediated by SLC31A2 and SLC31A1, and copper export is driven by ATP7B and ATP7A. In cells, copper is transported to different organelles (for bioavailability) via numerous copper-binding proteins (COX17, CCS, and ATOX1). The binding of MT2, GSH, and MT2 to copper can prevent the cytotoxicity of excess copper.</p>
Full article ">Figure 2
<p>Copper induces cell death in cancer therapeutics. Increased copper concentration in cancer cells increases ETC, DNA replication, glutamine transporters, harmful cuproptosis mediators, and reduced levels of positive cuproptosis mediators and antioxidant enzymes. These alterations result in cell death.</p>
Full article ">Figure 3
<p>Cuproptosis-related lncRNAs’ role in cervical cancer.</p>
Full article ">
20 pages, 3354 KiB  
Article
Iron Metabolism in Aminolevulinic Acid-Photodynamic Therapy with Iron Chelators from the Thiosemicarbazone Group
by Robert Gawecki, Patrycja Rawicka, Marta Rogalska, Maciej Serda and Anna Mrozek-Wilczkiewicz
Int. J. Mol. Sci. 2024, 25(19), 10468; https://doi.org/10.3390/ijms251910468 - 28 Sep 2024
Viewed by 623
Abstract
Iron plays a crucial role in various metabolic processes. However, the impact of 5-aminolevulinic acid (ALA) in combination with iron chelators on iron metabolism and the efficacy of ALA-photodynamic therapy (PDT) remain inadequately understood. This study aimed to examine the effect of thiosemicarbazone [...] Read more.
Iron plays a crucial role in various metabolic processes. However, the impact of 5-aminolevulinic acid (ALA) in combination with iron chelators on iron metabolism and the efficacy of ALA-photodynamic therapy (PDT) remain inadequately understood. This study aimed to examine the effect of thiosemicarbazone derivatives during ALA treatment on specific genes related to iron metabolism, with a particular emphasis on mitochondrial iron metabolism genes. In our study, we observed differences depending on the cell line studied. For the HCT116 and MCF-7 cell lines, in most cases, the decrease in the expression of selected targets correlated with the increase in protoporphyrin IX (PPIX) concentration and the observed photodynamic effect, aligning with existing literature data. The Hs683 cell line showed a different gene expression pattern, previously not described in the literature. In this study, we collected an extensive analysis of the gene variation occurring after the application of novel thiosemicarbazone derivatives and presented versatile and effective compounds with great potential for use in ALA-PDT. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Iron metabolism in a cell considering the molecular targets studied in this paper (created in BioRender). PPIX—protoporphyrin IX, FECH—ferrochelatase, HO-1—heme oxygenase, FTMT—ferritin, FXN—frataxin, ICSU—iron-sulfur cluster assembly enzyme, MFRN1/2—mitoferrin 1/2, ABCB8—ATP-binding cassette subfamily B member 8.</p>
Full article ">Figure 2
<p>The structures of the TSCs and Cp94 examined in this study.</p>
Full article ">Figure 3
<p>Accumulation of PPIX after treatment with 5-ALA, TSCs, Cp94, and their combinations on (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The red dotted line on the graph depicts the fluorescence intensity of PPIX following treatment with 5-ALA alone. The data are presented as the means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Absorption titration spectra of selected compounds (50 μM) with iron (III) ions in water: (<b>A</b>)—Cp94; (<b>B</b>)—TSC-34; (<b>C</b>)—TSC-109; (<b>D</b>)—TSC-113; (<b>E</b>)—TSC-116. Blue arrows indicate a decrease in band intensity for the test compound, while red arrows highlight an increase in band intensity for the complex. Measurements were performed at room temperature 4 h after the samples had been prepared.</p>
Full article ">Figure 5
<p>Phototoxic effect after treatment with 5-ALA, TSCs, Cp94, and their combinations on (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Expression of FECH in the (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Expression of HO-1 in the (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>Expression of FTMT in the (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>Expression of FXN in the (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>Expression of MFRN 1 and 2 genes on the (<b>A</b>,<b>D</b>): HCT116, (<b>B</b>,<b>E</b>): MCF-7 and (<b>C</b>,<b>F</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <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; 0.0001.</p>
Full article ">Figure 11
<p>Expression of DMT1 in the (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 12
<p>Expression of ABCB8 in the (<b>A</b>): HCT116, (<b>B</b>): MCF-7, and (<b>C</b>): Hs683 cell lines. The data are presented as means ± standard deviation from three independent experiments and analyzed using one-way ANOVA with Tukey’s post hoc test, indicating significance levels as follows: * <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; 0.0001.</p>
Full article ">
34 pages, 1412 KiB  
Review
A Descriptive Review of the Antioxidant Effects and Mechanisms of Action of Berberine and Silymarin
by Ana María García-Muñoz, Desirée Victoria-Montesinos, Pura Ballester, Begoña Cerdá and Pilar Zafrilla
Molecules 2024, 29(19), 4576; https://doi.org/10.3390/molecules29194576 - 26 Sep 2024
Cited by 1 | Viewed by 2019
Abstract
Oxidative stress is a key factor in the development of chronic diseases such as type 2 diabetes, cardiovascular diseases, and liver disorders. Antioxidant therapies that target oxidative damage show significant promise in preventing and treating these conditions. Berberine, an alkaloid derived from various [...] Read more.
Oxidative stress is a key factor in the development of chronic diseases such as type 2 diabetes, cardiovascular diseases, and liver disorders. Antioxidant therapies that target oxidative damage show significant promise in preventing and treating these conditions. Berberine, an alkaloid derived from various plants in the Berberidaceae family, enhances cellular defenses against oxidative stress through several mechanisms. It activates the AMP-activated protein kinase (AMPK) pathway, which reduces mitochondrial reactive oxygen species (ROS) production and improves energy metabolism. Furthermore, it boosts the activity of key antioxidant enzymes like superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), thus protecting cells from oxidative damage. These actions make berberine effective in managing diseases like type 2 diabetes, cardiovascular conditions, and neurodegenerative disorders. Silymarin, a flavonolignan complex derived from Silybum marianum, is particularly effective for liver protection. It activates the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, enhancing antioxidant enzyme expression and stabilizing mitochondrial membranes. Additionally, silymarin reduces the formation of ROS by chelating metal ions, and it also diminishes inflammation. This makes it beneficial for conditions like non-alcoholic fatty liver disease (NAFLD) and alcohol-related liver disorders. This review aims to highlight the distinct mechanisms by which berberine and silymarin exert their antioxidant effects. Full article
(This article belongs to the Special Issue Antioxidant Activity of Natural Products: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Mechanisms of Antioxidant Action of Berberine and Silymarin.</p>
Full article ">Figure 2
<p>Systemic health effects of berberine and silymarin.</p>
Full article ">
Back to TopTop