[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
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

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

Search Results (2,409)

Search Parameters:
Keywords = liver toxicity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 700 KiB  
Article
Comparison of Hepatic Function and Chemotherapy-Induced Side Effects Between Pegylated Liposomal Doxorubicin (PLD), Topotecan (TOPO), and Gemcitabine in Platinum-Resistant Ovarian Cancer (PROC)
by Radu-Dumitru Dragomir, Marina Adriana Mercioni, Șerban Negru, Dorel Popovici, Sorin Săftescu, Andiana Roxana Blidari and Ioan Sas
J. Pers. Med. 2025, 15(1), 39; https://doi.org/10.3390/jpm15010039 - 19 Jan 2025
Viewed by 282
Abstract
Background/Objectives: Platinum-resistant ovarian cancer (PROC) is a major therapeutic challenge, as it responds poorly to standard platinum-based treatment, has limited treatment options, and offers a generally unfavorable prognosis. Chemotherapeutic agents like pegylated liposomal doxorubicin (PLD), topotecan (TOPO), and gemcitabine (GEM) are used [...] Read more.
Background/Objectives: Platinum-resistant ovarian cancer (PROC) is a major therapeutic challenge, as it responds poorly to standard platinum-based treatment, has limited treatment options, and offers a generally unfavorable prognosis. Chemotherapeutic agents like pegylated liposomal doxorubicin (PLD), topotecan (TOPO), and gemcitabine (GEM) are used for this setting, but with varying efficacy and toxicity profiles, leading to an increasing need to understand the optimal balance between treatment effectiveness and tolerability for improving patient outcomes. This study evaluates the efficacy and side effects of PLD, TOPO, and GEM, focusing on progression-free survival (PFS), overall survival (OS), and safety profiles. Methods: We conducted a retrospective observational study that included 856 PROC patients treated with PLD (n = 383), TOPO (n = 352), or GEM (n = 121) at the OncoHelp Oncology Center from January 2018 to December 2023. Inclusion criteria encompass diagnosis, prior platinum therapy, and Eastern Cooperative Oncology Group (ECOG) status (0–2). Treatment protocols followed standard dosing, with adjustments for toxicity. Primary endpoints included PFS and OS, with safety assessed by incidence of grade 3 and 4 toxicities per CTCAE v5.0. Kaplan–Meier analysis and Cox regression were used to compare survival, and statistical significance was set at p < 0.05. Results: TOPO showed higher toxicity than PLD and GEM, including liver damage, hematological and non-hematological side effects, while PLD induced more skin toxicity. In terms of survival, minor differences were seen between the three chemotherapeutic agents, with a slight advantage for PLD for better disease control. Conclusions: Given the comparable results in OS across the regimens, treatment decisions should be based on other factors such as patient tolerance and quality of life. Full article
(This article belongs to the Special Issue Personalized Medicine in Gynecology and Obstetrics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Platinum-resistant ovarian cancer study design.</p>
Full article ">
19 pages, 1036 KiB  
Review
Additive Effects of Glutathione in Improving Antibiotic Efficacy in HIV–M.tb Co-Infection in the Central Nervous System: A Systematic Review
by Leena Nabipur, Michael Mouawad and Vishwanath Venketaraman
Viruses 2025, 17(1), 127; https://doi.org/10.3390/v17010127 - 17 Jan 2025
Viewed by 340
Abstract
Background: HIV and tuberculosis (TB) co-infection poses a significant health challenge, particularly when involving the central nervous system (CNS), where it leads to severe morbidity and mortality. Current treatments face challenges such as drug resistance, immune reconstitution inflammatory syndrome (IRIS), and persistent inflammation. [...] Read more.
Background: HIV and tuberculosis (TB) co-infection poses a significant health challenge, particularly when involving the central nervous system (CNS), where it leads to severe morbidity and mortality. Current treatments face challenges such as drug resistance, immune reconstitution inflammatory syndrome (IRIS), and persistent inflammation. Glutathione (GSH) has the therapeutic potential to enhance treatment outcomes by improving antibiotic efficacy, reducing inflammation, and mitigating immune dysfunction. Methods: Relevant studies were identified through systematic searches of PubMed, Elsevier, WHO, and related databases. Inclusion criteria focused on preclinical and clinical research examining GSH or its precursors in HIV, TB, or co-infection, with emphasis on microbial control, immune modulation, and CNS-related outcomes. Results: Preclinical studies showed that GSH improves macrophage antimicrobial function, reduces oxidative stress, and limits Mycobacterium tuberculosis (M.tb) growth. Animal models demonstrated reduced bacterial burden in the lungs, liver, and spleen with GSH supplementation, along with enhanced granuloma stability. Clinical studies highlighted increased TH1 cytokine production, reduced inflammatory markers, and improved CD4+ T cell counts in HIV–M.tb co-infected patients. N-acetylcysteine (NAC), a GSH precursor, was shown to significantly enhance the efficacy of first-line TB antibiotics and mitigate treatment-associated toxicity. Discussion: GSH shows promise as an adjunct therapy for HIV–M.tb co-infection, particularly for cases involving the CNS, where it may improve immune recovery and reduce inflammation. However, evidence is limited by small sample sizes and a lack of randomized trials. Future research should focus on developing CNS-directed GSH formulations and evaluating its integration into current treatment protocols to address the dual burden of HIV and TB, ultimately improving patient outcomes. Full article
(This article belongs to the Special Issue HIV and Tuberculosis (TB) Coinfection)
Show Figures

Figure 1

Figure 1
<p>Identification of studies via databases using PRISMA flow diagram.</p>
Full article ">Figure 2
<p>The role of glutathione (GSH) in immune modulation and defense.</p>
Full article ">
16 pages, 2681 KiB  
Article
Toxicology Effects of Cadmium in Pomacea canaliculate: Accumulation, Oxidative Stress, Microbial Community, and Transcriptome Analysis
by Mingxin Qiu, Xiaoyang Bi, Yuanyang Liu, Huashou Li, Dongqin Li and Guikui Chen
Int. J. Mol. Sci. 2025, 26(2), 751; https://doi.org/10.3390/ijms26020751 - 17 Jan 2025
Viewed by 410
Abstract
Cadmium (Cd) pollution poses an important problem, but limited information is available about the toxicology effects of Cd on freshwater invertebrates. We investigated the accumulation, oxidative stress, microbial community changes, and transcriptomic alterations in apple snails (Pomacea canaliculata) under Cd stress. The [...] Read more.
Cadmium (Cd) pollution poses an important problem, but limited information is available about the toxicology effects of Cd on freshwater invertebrates. We investigated the accumulation, oxidative stress, microbial community changes, and transcriptomic alterations in apple snails (Pomacea canaliculata) under Cd stress. The snails were exposed to the 10 μg/L Cd solution for 16 days, followed by a 16-day elimination period. Our results showed that the liver accumulated the highest Cd concentration (17.41 μg/g), followed by the kidneys (8.00 μg/g) and intestine-stomach (6.68 μg/g), highlighting these tissues as primary targets for Cd accumulation. During the elimination period, Cd concentrations decreased in all tissues, with the head-foot and shell exhibiting over 30% elimination rates. Cd stress also resulted in reduced activities of superoxide dismutase (SOD), catalase (CAT), and glutathione transferase (GST) compared to the control group. Notably, even after 16 days of depuration, the enzyme activities did not return to normal levels, indicating persistent toxicological effects. Cd exposure significantly reduced the diversity of gut microbiota in P. canaliculata. Moreover, transcriptome analysis identified differentially expressed genes (DEGs) primarily associated with lysosome function, motor proteins, protein processing in the endoplasmic reticulum, drug metabolism via cytochrome P450 (CYP450), arachidonic acid metabolism, and ECM–receptor interactions. These findings suggest that Cd stress predominantly disrupts cellular transport and metabolic processes. Overall, our study provides comprehensive insights into the toxicological impact of Cd on P. canaliculata and emphasizes the importance of understanding the mechanisms underlying Cd toxicity in aquatic organisms. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

Figure 1
<p>Cadmium accumulation and elimination in the different tissues of <span class="html-italic">Pomacea canaliculata</span> on day-16 and day-32. (<b>a</b>) Cd concentration in shell; (<b>b</b>) Cd concentration in head-foot; (<b>c</b>) Cd concentration in heart; (<b>d</b>) Cd concentration in gonads; (<b>e</b>) Cd concentration in intestine-stomach; (<b>f</b>) Cd concentration in liver; (<b>g</b>) Cd concentration in kidneys. * indicates a significant difference compared with the control (* <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; 0.01). # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 are relative to day 32.</p>
Full article ">Figure 2
<p>(<b>a</b>) Superoxide dismutase (SOD), (<b>b</b>) catalase (CAT), and (<b>c</b>) glutathione transferase (GST) changes in <span class="html-italic">Pomacea canaliculata</span> on day 16 and day 32, respectively. * indicates a significant difference compared with the control (* <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; 0.01). ### <span class="html-italic">p</span> &lt; 0.001 is relative to day 32.</p>
Full article ">Figure 3
<p>Changes in alpha diversity of <span class="html-italic">Pomacea canaliculata</span>. (<b>a</b>) Chao index of species richness; (<b>b</b>) observed features in the microbiota; (<b>c</b>) Shannon index of species diversity; and (<b>d</b>) Simpson index of species diversity. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>The taxonomic composition of <span class="html-italic">Pomacea canaliculata</span> gut microbiota at the phylum (<b>a</b>) and genus levels (<b>b</b>).</p>
Full article ">Figure 5
<p>GO annotations of single genes of the transcriptome, where each annotated sequence was assigned at least one of the following GO terms: biological process (BP), cellular component (CC), and molecular function (MF).</p>
Full article ">Figure 6
<p>RNA-Seq analysis to identify the differentially expressed genes of <span class="html-italic">P. canaliculata</span> upon Cd treatment. (<b>a</b>) Volcano plot of DEGs; (<b>b</b>) heatmap of DEGs; (<b>c</b>) KEGG pathway enrichment analysis of DEGs; (<b>d</b>) GO function analysis of DEGs. Pathway enrichment bubble plot colors represent adjusted <span class="html-italic">p</span>-values, and bubble sizes represent the number of genes enriched.</p>
Full article ">
20 pages, 677 KiB  
Review
In Vivo and In Vitro Models of Hepatic Fibrosis for Pharmacodynamic Evaluation and Pathology Exploration
by Yanting Hu, Zhongrui Zhang, Akida Adiham, Hong Li, Jian Gu and Puyang Gong
Int. J. Mol. Sci. 2025, 26(2), 696; https://doi.org/10.3390/ijms26020696 - 15 Jan 2025
Viewed by 540
Abstract
Hepatic fibrosis (HF) is an important pathological state in the progression of chronic liver disease to end-stage liver disease and is usually triggered by alcohol, nonalcoholic fatty liver, chronic hepatitis viruses, autoimmune hepatitis (AIH), or cholestatic liver disease. Research on novel therapies has [...] Read more.
Hepatic fibrosis (HF) is an important pathological state in the progression of chronic liver disease to end-stage liver disease and is usually triggered by alcohol, nonalcoholic fatty liver, chronic hepatitis viruses, autoimmune hepatitis (AIH), or cholestatic liver disease. Research on novel therapies has become a hot topic due to the reversibility of HF. Research into the molecular mechanisms of the pathology of HF and potential drug screening relies on reliable and rational biological models, mainly including animals and cells. Hence, a number of modeling approaches have been attempted based on human dietary, pathological, and physiological factors in the development of HF. In this review, classical and novel methods of modeling HF in the last 10 years were collected from electronic databases, including Web of Science, PubMed, ScienceDirect, ResearchGate, Baidu Scholar, and CNKI. Animal models of HF are usually induced by chemical toxicants, special diets, pathogenic microorganisms, surgical operations, and gene editing. The advantages and limitations of hepatic stellate cells (HSCs), organoids, and 3D coculture-based HF modeling methods established in vitro were also proposed and summarized. This information provides a scientific basis for the discovery of the pathological mechanism and treatment of HF. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Summary of animal models induced by specific diets for studying HF. Single factor, MCD, GAN; combined diet, HFHCD, CDAHFD, OYC-NASH2, STZ+HFD; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; HSCs, hepatic stellate cells; ↑, up-regulated expression.</p>
Full article ">
20 pages, 6034 KiB  
Article
Therapeutic Potential of Clove Oil in Mitigating Cadmium-Induced Hepatorenal Toxicity Through Antioxidant, Anti-Inflammatory, and Antiapoptotic Mechanisms
by Inas M. Elgharib, Fatma M. Abdelhamid, Gehad E. Elshopakey, Hatem Sembawa, Talat A. Albukhari, Waheed A. Filimban, Rehab M. Bagadood, Mohamed E. El-Boshy and Engy F. Risha
Pharmaceuticals 2025, 18(1), 94; https://doi.org/10.3390/ph18010094 - 14 Jan 2025
Viewed by 363
Abstract
Hazardous heavy metals, particularly cadmium (Cd), are widely distributed in the environment and cause oxidative stress in various animal and human organs. Clove oil (CLO), a common aromatic spice, has been used as a traditional medication as it has potent anti-inflammatory, antioxidant, and [...] Read more.
Hazardous heavy metals, particularly cadmium (Cd), are widely distributed in the environment and cause oxidative stress in various animal and human organs. Clove oil (CLO), a common aromatic spice, has been used as a traditional medication as it has potent anti-inflammatory, antioxidant, and hepatoprotective properties. Background/Objectives: This study aimed to investigate the antioxidant, antiapoptotic, and anti-inflammatory effects of clove oil (CLO) against hepatorenal toxicity induced by cadmium (Cd). Methods: Twenty rats were equally divided into four groups: a control group, a Cd group treated with 15 mg/kg b.wt CdCl2, a CLO group administered 200 mg/kg b.wt CLO, and a Cd+CLO group. All groups were orally treated for 4 weeks. Results: Cadmium (Cd) exposure caused anemia and hepatorenal damage, as evidenced by increased serum levels of urea, creatinine, uric acid, total bilirubin (including its direct and indirect fractions), and elevated activities of liver enzymes such as alanine transaminase (ALT), aspartate transaminase (AST), and alkaline phosphatase (ALP). However, total protein and albumin levels decreased. Furthermore, there was a decrease in the levels of glutathione, glutathione transferase, and catalase in the liver antioxidant profiles. Meanwhile, malondialdehyde levels increased. Cadmium toxicity caused elevated expression of liver apoptosis markers, such as tumor necrosis factor-alpha (TNF-α) and caspase-3, and inflammation. CLO ameliorated the oxidative effects of Cd through decreasing urea (27.4%), creatinine (41.6%), liver enzymes, and hepatic apoptotic markers while increasing levels of total protein, albumin, and hepatic values of SOD (60.37%), CAT (64.49%), GSH (50.41%), and GST (9.16%). Conclusions: Hematological and biochemical parameters, as well as the antioxidant system, improved following clove oil treatment, leading to a reduction in hepatorenal damage. Therefore, it is possible to conclude that CLO protects rats from inflammation, apoptosis, and hepatorenal oxidative damage caused by Cd poisoning. Comprehensive translational research is required to validate CLO’s efficacy and safety of use in humans. Future studies should focus on elucidating the precise molecular mechanisms, optimal dosing strategies, and potential synergistic effects of CLO with other therapeutic agents. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

Figure 1
<p>Liver function tests at fourth week after clove oil treatment in rats intoxicated with cadmium (mean ± SE). Significant values are defined as those with distinct superscript letters (<span class="html-italic">p</span> &lt; 0.05). The enzymes that convert amino acids to bilirubin are (<b>A</b>) alanine aminotransferase, (<b>B</b>) aspartate aminotransferase, (<b>C</b>) alkaline phosphatase, (<b>D</b>) total bilirubin, (<b>E</b>) direct bilirubin, and (<b>F</b>) indirect bilirubin.</p>
Full article ">Figure 2
<p>Proteinogram of rats intoxicated with cadmium at the end of the fourth week after receiving clove oil treatment (mean ± SE). Significant values are defined as those with distinct superscript letters (<span class="html-italic">p</span> &lt; 0.05). (<b>A</b>) Globulin, (<b>B</b>) albumin, (<b>C</b>) total protein, and (<b>D</b>) A/G ratio.</p>
Full article ">Figure 3
<p>Microscopic images of H&amp;E-stained hepatic sections from the control group (<b>A</b>,<b>B</b>) and CLO group (<b>C</b>,<b>D</b>) demonstrating typical radially oriented hepatic cords around the central veins (CV) with normal portal regions and sinusoids (s). H&amp;E-stained hepatic sections from the Cd group (<b>E</b>,<b>F</b>) demonstrating focal areas of coagulative necrosis (black arrowheads), vascular dilation (red arrow), marked inflammation (black arrows) in the portal areas, and hydropic degeneration in hepatocytes (dashed black arrows). H&amp;E-stained hepatic sections from the Cd+CLO group (<b>G</b>,<b>H</b>) demonstrating hydropic degeneration in hepatocytes (dashed black arrows) (100 bar 100 is the low magnification, and 400 bar 50 is the high magnification).</p>
Full article ">Figure 4
<p>Microscopic images of H&amp;E-stained renal cortical sections revealing normal tubules (T) and glomeruli (G) with minimal interstitial tissue in the control group (<b>A</b>,<b>B</b>) and CLO group (<b>C</b>,<b>D</b>). H&amp;E-stained renal cortical sections from the Cd group (<b>E</b>,<b>F</b>) revealing severe hydropic degeneration of the tubular epithelium (dashed black arrows), necrosis (black arrowheads), congestion (red arrows), and marked perivascular edema (*) with many mononuclear cells infiltrating the interstitial tissue (thin black arrows). H&amp;E-stained renal cortical sections from the Cd+CLO group (<b>G</b>,<b>H</b>) showing minor perivascular edema (*) and milder hydropic degeneration in a few tubules (dashed black arrows) and some mononuclear cells infiltrating the interstitial tissue (thin black arrows) (100 bar 100 is the low magnification, and 400 bar 50 is the high magnification).</p>
Full article ">Figure 5
<p>Immunostained liver section displaying negative immunostaining against caspase-3 in the control group (<b>A</b>) and CLO group (<b>B</b>). Liver displayed strong immunostaining against caspase-3 in the Cd group (<b>C</b>). Liver sections displayed a marked decrease in immunostaining against caspase-3 in the Cd+CLO group (<b>D</b>). (IHC, DAB immunostaining, hematoxylin as a counterstain, 100×). Bars (<b>E</b>) represent the scores of caspase-3 expressions (mean ± SE). Values with different superscript letters are considered significant at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Immunostained liver section displaying negative immunostaining against TNF-α in the control group (<b>A</b>) and CLO group (<b>B</b>). Liver displayed strong immunostaining against TNF-α in the Cd group (<b>C</b>). Liver sections displayed a marked decrease in immunostaining against TNF-α in the Cd+CLO group (<b>D</b>) (IHC, DAB immunostaining, hematoxylin as a counterstain, 100×). Bars (<b>E</b>) represent the scores of TNF-α expression (mean ± SE). Values with different superscript letters are considered significant at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Experiment design.</p>
Full article ">
16 pages, 5717 KiB  
Article
Effects of Polystyrene Microplastic Exposure on Liver Cell Damage, Oxidative Stress, and Gene Expression in Juvenile Crucian Carp (Carassius auratus)
by Xiangtong Li, Yuequn Huang, Wenrong Li, Chaoyang Deng, Weiyuan Cao and Yi Yao
Toxics 2025, 13(1), 53; https://doi.org/10.3390/toxics13010053 - 12 Jan 2025
Viewed by 566
Abstract
A considerable quantity of microplastic debris exists in the environment and the toxicity of these materials has a notable impact on aquatic ecosystems. In this paper, 50–500 µm polystyrene microplastics (exposure concentrations were 200 µg/L, 800 µg/L, and 3200 µg/L concentrations) were selected [...] Read more.
A considerable quantity of microplastic debris exists in the environment and the toxicity of these materials has a notable impact on aquatic ecosystems. In this paper, 50–500 µm polystyrene microplastics (exposure concentrations were 200 µg/L, 800 µg/L, and 3200 µg/L concentrations) were selected to study the effects of polystyrene microplastics (PS-MPs) on cell morphology, detoxification enzyme activity, and mRNA expression in the liver tissues of crucian carp juveniles. The results demonstrated that: (1) Different concentrations of PS-MPs cause varying degrees of pathological and oxidative damage to liver tissue cells of crucian carp. The higher the concentration of microplastics, the lower the antioxidant enzyme (CAT, GST, SOD) activity and the greater the tissue cell damage. These results demonstrate a typical dose–effect relationship. (2) Principal component analysis and Spearman’s correlation analysis demonstrated that four components, namely glutathione S-transferase (GST) and its related genes (GSTpi, GSTα), along with catalase (CAT), contributed the most to the observed outcome. These four components demonstrated a relatively high level of responsiveness to PS-MP exposure and can be employed as ecotoxicological indicators of microplastics. (3) This experiment evaluated five genes in three treatments, which found that PS-MPs had different effects on gene expression in the liver and the tested genes were involved in different response pathways associated with virulence. In this study, the toxicity of PS-MPs to crucian carp was determined at the cellular, protein, and mRNA expression levels, and combined with principal component analysis and correlation analysis to identify response sensitivity indicators that provide a scientific basis for ecological risk assessment and the safe use of microplastics. Full article
Show Figures

Figure 1

Figure 1
<p>Cellular morphology of liver tissue of juvenile crucian carp after 32 d of stress by PS-MPs: (<b>a</b>) blank control group; (<b>b</b>) low-concentration group; (<b>c</b>) medium-concentration group; (<b>d</b>) high-concentration group (Black arrows indicate cell congestion, red arrows indicate vacuolated cells, green arrows indicate off-center nuclei, and blue arrows indicate enlarged necrotic cells, Picture magnification is 40×).</p>
Full article ">Figure 2
<p>Changes in antioxidant enzymes in liver tissues of juvenile crucian carp after 32 d of exposure to PS-MPs: (<b>a</b>) superoxide dismutase (SOD) activity, (<b>b</b>) glutathione S-transferase (GST) activity, (<b>c</b>) catalase (CAT) activity, (<b>d</b>) malondialdehyde (MDA) content. “*” indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05) and “**” indicates a highly significant difference (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 2 Cont.
<p>Changes in antioxidant enzymes in liver tissues of juvenile crucian carp after 32 d of exposure to PS-MPs: (<b>a</b>) superoxide dismutase (SOD) activity, (<b>b</b>) glutathione S-transferase (GST) activity, (<b>c</b>) catalase (CAT) activity, (<b>d</b>) malondialdehyde (MDA) content. “*” indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05) and “**” indicates a highly significant difference (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Changes in mRNA expression of antioxidant-related genes in liver tissues of juvenile crucian carp after 32 d of PS-MP exposure. “*” indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05) and “**” indicates a highly significant difference (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>Principal component analysis of antioxidant enzymes and related genes in liver tissue of juvenile crucian carp after 32 d of PS-MP exposure.</p>
Full article ">Figure 5
<p>Heatmap of correlation analysis of antioxidant enzymes and related genes in liver tissues of juvenile crucian carp after 32 d of PS-MP exposure. “*” indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05) and “**” indicates a highly significant difference (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Relationship between antioxidant enzymes and corresponding genes in juvenile crucian carp liver tissues after 32 d of PS-MP exposure: (<b>a</b>) Relationship between GST enzyme activity and <span class="html-italic">GSTpi</span> gene expression; (<b>b</b>) Relationship between GST enzyme activity and <span class="html-italic">GSTα</span> gene expression.</p>
Full article ">Figure 7
<p>Trends of mRNA expression of antioxidant-related genes in liver tissues of juvenile crucian carp after 32 d of PS-MP exposure.</p>
Full article ">
20 pages, 25587 KiB  
Article
Combining In Vitro, In Vivo, and In Silico Approaches to Explore the Effect of Ceratonia siliqua and Ocimum basilicum Rich Phenolic Formula on Lipid Metabolism and Plasma Lipoprotein Oxidation in Mice Fed a High-Fat Diet: A Follow-Up Study
by Mohammadine Moumou, Amani Tayebi, Abderrahmane Hadini, Omar M. Noman, Abdulsalam Alhalmi, Hamza Ahmoda, Souliman Amrani and Hicham Harnafi
Metabolites 2025, 15(1), 36; https://doi.org/10.3390/metabo15010036 - 10 Jan 2025
Viewed by 429
Abstract
Background/Objectives: Hyperlipidemia is a serious risk factor for cardiovascular diseases and liver steatosis. In this work, we explored the effect of an herbal formula (CBF) containing immature Ceratonia siliqua pods and Ocimum basilicum extracts on lipid metabolism disorders and lipoprotein-rich plasma (LRP) oxidation [...] Read more.
Background/Objectives: Hyperlipidemia is a serious risk factor for cardiovascular diseases and liver steatosis. In this work, we explored the effect of an herbal formula (CBF) containing immature Ceratonia siliqua pods and Ocimum basilicum extracts on lipid metabolism disorders and lipoprotein-rich plasma (LRP) oxidation in mice. Methods: The phenolic composition was determined using HPLC-DAD analysis. The antioxidant activity was studied using various in vitro methods. Acute toxicity was evaluated in mice. Importantly, the effect of the CBF on lipid metabolism disorders was investigated in a high-fat diet (HFD) hyperlipidemia mouse model. An in silico study was carried out to predict underlying mechanisms. Results: The HPLC analysis revealed gallic acid, cinnamic acid, and naringenin as major phenolics of the carob pod aqueous extract. Concerning the basil hydro-ethanolic extract, rosmarinic, chicoric, caftaric, and caffeic acids were the main phenolics. Accordingly, the CBF prevented LRP oxidation in a concentration-dependent manner. This formula is not toxic in mice (LD50 > 2000 mg/kg body weight). Moreover, animals administered the CBF at 200 mg/kg/day presented a significant decline in their body weight gain, adipose tissue weight, plasma total cholesterol, low-density lipoprotein cholesterol (LDL-C) level, and glycaemia after 10 weeks’ treatment. Accordingly, the CBF decreased the plasma atherogenic index and the LDL-C to HDL-C ratio and reduced the level of fats accumulated in the liver. The molecular docking study revealed that chicoric, rosmarinic, and caftaric acids, and naringenin bound particularly strongly to many proteins involved in the regulation of lipid and cholesterol metabolism. This includes the HMG-CoA reductase, PPARα/γ, PCSK9, Cyp7a1, and ATP-citrate lyase. Conclusions: The CBF could be a good source of natural supplements, functional foods, and pharmaceuticals effective in managing hyperlipidemia and oxidative stress and preventing their related cardiovascular disorders. Full article
(This article belongs to the Section Food Metabolomics)
Show Figures

Figure 1

Figure 1
<p>HPLC phenolic profiles of <span class="html-italic">C. siliqua</span> (<b>A</b>) and <span class="html-italic">O. basilicum</span> (<b>B</b>) extracts.</p>
Full article ">Figure 2
<p>Chemical structures of the main phenolics from the immature carob pod aqueous extract and sweet basil hydro-ethanolic extract. 1: Quercetin (CID: 5280343); 2: rosmarinic acid (CID: 5281792); 3: gallic acid (CID: 370); 4: caftaric acid (CID: 64409397); 5: cinnamic acid (CID: 444539); 6: caffeic acid (CID 689043); 7: naringenin (CID: 439246); 8: chicoric acid (CID:528176) (Drawn using ChemDraw Pro 12 software).</p>
Full article ">Figure 3
<p>Concentration-dependent DPPH radical scavenging activity of the CBF and BHA (<b>A</b>) and the obtained IC<sub>50</sub> values (<b>B</b>). Ferric reducing power activity of the CBF and AA (<b>C</b>) and the obtained EC<sub>50</sub> values (<b>D</b>). Effect of CBF and AA on mice LRP oxidation (<b>E</b>) and the corresponding IC<sub>50</sub> values (<b>F</b>). Results are represented as mean ± SEM (<span class="html-italic">n</span> = 3). CBF: carob/basil formula; AA: ascorbic acid; BHA: butylated hydroxyanisole. * <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Effect of the CBF and fenofibrate on plasma TC (<b>A</b>), TG (<b>B</b>), LDL-C (<b>C</b>), and HDL-C (<b>D</b>) levels, the atherogenic index (<b>E</b>), LDL-C/HDL-C ratio (<b>F</b>), glycaemia (<b>G</b>), hepatic TG (<b>H</b>), and TC (<b>I</b>) levels in mice (<span class="html-italic">n</span> = 6) fed HFD for 10 consecutive weeks. NLC: normal control; HLC: hyperlipidemic control; CBFG; CBF-treated mice; FFG; fenofibrate-treated mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NLC; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. HLC.</p>
Full article ">Figure 5
<p>Heat map showed the binding energy (kcal/moL) between the CBF phenolics and main targets involved in the regulation of lipid metabolism. ACLY: ATP-citrate lyase; HMG−CoAR: β−hydroxy β−methylglutaryl−CoA reductase; PPAR: peroxisome proliferator activated receptors; LXRα: liver X receptor alpha; LPL: lipoprotein lipase; PCSK9: proprotein convertase subtilisin/kexin type 9; RXR: retinoid X receptors; Cyp7a1: cytochrome P450, family 7, subfamily a, polypeptide 1; C/EBPα: CCAAT/enhancer binding protein alpha; FXR: farnesoid X receptors; FAS: fatty acid synthase; NPC1L1: Niemann–Pick C1−Like 1.</p>
Full article ">Figure 6
<p>Interactions between CBF phenolics/fenofibrate and the main proteins involved in the regulation of lipid metabolism (3D and 2D structures). (<b>A</b>): chicoric acid/HMGCoA reductase; (<b>B</b>): rosmarinic/PPARα; (<b>C</b>): gallic acid/RXRα; (<b>D</b>): fenofibrate/PPARα.</p>
Full article ">Figure 7
<p>Interactions between CBF phenolics and main proteins involved in the regulation of lipid metabolism (3D and 2D structures). (<b>A</b>): naringenin/LXRα; (<b>B</b>): chicoric acid/FAS: (<b>C</b>): rosmarinic acid/ACLY; (<b>D</b>): naringenin/PCSK9.</p>
Full article ">
44 pages, 3456 KiB  
Review
Species Differences in the Biotransformation of Aflatoxin B1: Primary Determinants of Relative Carcinogenic Potency in Different Animal Species
by David L. Eaton, David E. Williams and Roger A. Coulombe
Toxins 2025, 17(1), 30; https://doi.org/10.3390/toxins17010030 - 9 Jan 2025
Viewed by 544
Abstract
It has been known since the early days of the discovery of aflatoxin B1 (AFB1) that there were large species differences in susceptibility to AFB1. It was also evident early on that AFB1 itself was not toxic but required bioactivation to a reactive [...] Read more.
It has been known since the early days of the discovery of aflatoxin B1 (AFB1) that there were large species differences in susceptibility to AFB1. It was also evident early on that AFB1 itself was not toxic but required bioactivation to a reactive form. Over the past 60 years there have been thousands of studies to delineate the role of ~10 specific biotransformation pathways of AFB1, both phase I (oxidation, reduction) and phase II (hydrolysis, conjugation, secondary oxidations, and reductions of phase I metabolites). This review provides a historical context and substantive analysis of each of these pathways as contributors to species differences in AFB1 hepatoxicity and carcinogenicity. Since the discovery of AFB1 as the toxic contaminant in groundnut meal that led to Turkey X diseases in 1960, there have been over 15,000 publications related to aflatoxins, of which nearly 8000 have addressed the significance of biotransformation (metabolism, in the older literature) of AFB1. While it is impossible to give justice to all of these studies, this review provides a historical perspective on the major discoveries related to species differences in the biotransformation of AFB1 and sets the stage for discussion of other papers in this Special Issue of the important role that AFB1 metabolites have played as biomarkers of exposure and effect in thousands of human studies on the toxic effects of aflatoxins. Dr. John Groopman has played a leading role in every step of the way—from initial laboratory studies on specific AFB1 metabolites to the application of molecular biomarkers in epidemiological studies associating dietary AFB1 exposure with liver cancer, and the design and conduct of chemoprevention clinical trials to reduce cancer risk from unavoidable aflatoxin exposures by alteration of specific AFB1 biotransformation pathways. This article is written in honor of Dr. Groopman’s many contributions in this area. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Basic steps in the oxidation of AFB1 to various metabolites. The human enzymes, where known, catalyzing these oxidations are listed. Each oxidation step shown in <a href="#toxins-17-00030-f001" class="html-fig">Figure 1</a> is discussed in detail below, with a focus on understanding important species differences in each oxidation step, as well as the specific enzyme isoforms that contribute to each reaction.</p>
Full article ">Figure 2
<p>Hepatic microsomal oxidation of AFB1 to various oxidative metabolites in different species. The initial rates of formation (Vo) were determined in hepatic microsomes from rat, mouse, monkey, and human microsomes under identical experimental conditions. AFBO was determined by trapping as the GSH conjugate using BHA-induced mouse liver cytosol, which contains a high level of mGSTA3-3. Each metabolite was separated and quantitated by HPLC. Rates of AFBO formation as a percentage of that observed with rat liver microsomes are also shown. The rates of formation of AFQ1, AFM1, and AFP1 were calculated as a percentage of the rate of epoxidation observed for the respective species; these values are shown above each column. (From Ramsdell and Eaton [<a href="#B16-toxins-17-00030" class="html-bibr">16</a>]). Reprinted under AACR copyright permissions to authors.</p>
Full article ">Figure 3
<p>AFB1 metabolite distribution at 1 μM and 10 μM in mouse, rat, and human hepatocytes. Isolated hepatocytes from each species were incubated for 4 h in cell culture medium. Metabolites were identified by HPLC-MS/MS. (From: Gerdemann et al. [<a href="#B6-toxins-17-00030" class="html-bibr">6</a>]; figure is reprinted under Creative Commons Attribution 4.0 International License).</p>
Full article ">Figure 4
<p>Immuno-inhibition experiments using anti-peptide antiserum against turkey P450s 1A5 and 3A37 demonstrating the relative contribution of P450 1A5 and 3A37 toward AFB1 epoxidation in turkey liver microsomes. Inhibitory effects of anti-P450 1A5 and 3A37 immune serum (5 μg/mL/nmol P450). Initial rates of exo-AFBO formation in the presence of antiserum were calculated as percentage control (treatment with pre-immune serum only). Mean ± SD. (N = 3). From: Rawl and Coulombe, [<a href="#B85-toxins-17-00030" class="html-bibr">85</a>]. Reprinted under Open Access Creative Commons Attribution.</p>
Full article ">Figure 5
<p>Phase II hydrolysis and conjugation reactions of phase I oxidation products of AFB1 biotransformation.</p>
Full article ">Figure 6
<p>Effects of co-expression of human mEH on AFB-DNA adducts in yeast also co-expressing hCYP1A2 to activate AFB1 to AFBO. Two concentrations of AFB1 were used to expose yeast cells containing human CYP1A2 and mEH cDNAs (adapted from Kelly et al. [<a href="#B138-toxins-17-00030" class="html-bibr">138</a>]. * Co-expression of mEH blocked DNA adduction with significant effect (<span class="html-italic">p</span> &lt; 0.05) at 1.25 mM AFB. Data are mean 6 SEM from samples analyzed in triplicate. (Figure available under Creative Commons Attribution 4.0 International license).</p>
Full article ">Figure 7
<p>Modulation of AFB-DNA adduct formation in the context of the GSTM1 genotype status. A total of 11 different hepatocyte preparations were examined for AFB-DNA binding. Six of the samples were GSTM1-null and five were GSTM1-positive. AFB-DNA adducts per 10<sup>7</sup> nucleotides were calculated and are shown. Each bar represents the mean and SEM. Statistical significance was determined by unpaired <span class="html-italic">t</span>-test with equal variances. Adapted from: Gross-Steinmeyer et al. [<a href="#B157-toxins-17-00030" class="html-bibr">157</a>]. Reprinted with permission from Oxford Press, Oxford, UK OX2 6DP; license #5923750542730, 7 December 2024.</p>
Full article ">Figure 8
<p>AFB-DNA adduct formation in mGstA3 knockout mice and wild-type. Mice (5 mGstA3 KO and 5 WT, 6 months of age, all males) were injected with a single dose of 5 mg/kg AFB1, dissolved in DMSO, in a volume of 100 μL/30 g of mouse weight, and euthanized 3 h later. Redrawn from: Ilic et al. [<a href="#B173-toxins-17-00030" class="html-bibr">173</a>], with permission from Elsevier Press, Berkeley, CA; license # 5923751463839, 7 December 2024.</p>
Full article ">Figure 9
<p>Reverse-phase HPLC radiochromatograms of cytosolic GST conjugation of AFBO in mouse and turkey. The top panel shows [<sup>3</sup>H]-AFBO-GST activity of BHA-induced mouse liver cytosol (500 mg protein) for comparison. The middle panel show the lack of GST-mediated [<sup>3</sup>H]-AFBO-conjugating ability of turkey hepatic cytosol (1200 mg protein). A control incubation with no cytosol is also presented (bottom panel). Even when a wide range of turkey cytosolic protein concentrations (400–1200 mg) was used, no GST-mediated trapping was detected [<a href="#B78-toxins-17-00030" class="html-bibr">78</a>]. Reprinted with permission from Elsevier Press, Berkeley, CA 94704, license # 5923770700438, 7 December 2024.</p>
Full article ">Figure 10
<p>Timeline of research interest in aflatoxins, as indicated by the number of scientific publications each year from 1963 to December 2024. Data from a PubMed search on the term “aflatoxin” or “aflatoxins”.</p>
Full article ">Figure 11
<p>Publication and citation history of Dr. John Groopman’s contributions to the past 45 years of aflatoxin research, including many papers related to species differences in biotransformation. (Figure developed from data obtained from a Web of Science citation search on “John D. Groopman” and “aflatoxins”).</p>
Full article ">
12 pages, 2080 KiB  
Article
Immunomodulatory Effect of Phage Depolymerase Dep_kpv74 with Therapeutic Potential Against K2-Hypervirulent Klebsiella pneumoniae
by Nikolay V. Volozhantsev, Maria A. Makarova, Alena S. Kartseva, Marina V. Silkina, Valentina M. Krasilnikova, Egor A. Denisenko, Alexander I. Borzilov and Victoria V. Firstova
Antibiotics 2025, 14(1), 44; https://doi.org/10.3390/antibiotics14010044 - 7 Jan 2025
Viewed by 396
Abstract
Background: The emergence of multidrug-resistant hypervirulent Klebsiella pneumoniae (hvKp) has made it difficult to treat and control infections caused by this bacterium. Previously, the therapeutic effectiveness of phage-encoded depolymerase Dep_kpv74 in a mouse model of K. pneumoniae-induced thigh soft tissue infection was [...] Read more.
Background: The emergence of multidrug-resistant hypervirulent Klebsiella pneumoniae (hvKp) has made it difficult to treat and control infections caused by this bacterium. Previously, the therapeutic effectiveness of phage-encoded depolymerase Dep_kpv74 in a mouse model of K. pneumoniae-induced thigh soft tissue infection was reported. In this study, the effect of Dep_kpv74 on blood parameters in mice, the proliferation and subpopulation composition of spleen lymphocytes, and the activity and stability of the enzyme at different pH and temperatures were further explored. Results: The stability tests showed that Dep_kpv74 remained active in the temperature range from 8 °C to 55 °C. The optimal pH value for maintaining the activity of Dep_kpv74 ranged from 5.0 to 9.0. The depolymerase was detected in the blood, spleen, and lungs of mice 10 min after intraperitoneal administration, reaching maximum activity values after 1–3 h and maintaining activity a day after administration. The introduction of Dep_kpv74 at the therapeutic dose (10 μg/mouse) or at a 10-fold higher dose did not lead to reliable changes in bloodstream cell content compared with the reference values of intact mice. The biochemical results of the studies indicated that Dep_kpv74 did not exert any toxic effects on liver and kidney functions. The results of the analysis of lymphocyte proliferative activity demonstrated that Dep_kpv74 depolymerase has a mild immunomodulatory effect. Conclusions: Thus, the results of this study provide one more confirmation that depolymerase Dep_kpv74 is a potential candidate for the treatment of infections caused by hvKp expressing K2 capsular polysaccharides. Full article
Show Figures

Figure 1

Figure 1
<p>Stability assay of depolymerase Dep_kpv74. (<b>A</b>) Effect of pH on the Dep_kpv74 activity; (<b>B</b>) effect of various temperatures (pH 7.4) on the Dep_kpv74 activity; (<b>C</b>) residual activity of Dep_kpv74 after incubation at 65 °C for 30 min.</p>
Full article ">Figure 2
<p>Biodistribution of Dep_kpv74 depolymerase in the blood, spleen, and lungs of outbred mice after intraperitoneal administration at a dose of 50 μg/mouse.</p>
Full article ">Figure 3
<p>Blood biochemical parameters in female BALB/c mice on days 3 and 6 after administration of 10 μg or 100 μg of Dep_kpv74 depolymerase. Control, corresponding parameters in mice not injected with depolymerase. Total protein (<b>A</b>), glucose (<b>B</b>), ALT (<b>C</b>), and AST (<b>D</b>) values are shown.</p>
Full article ">Figure 4
<p>Proliferative activity of lymphocytes in BALB/c mice on 3rd and 6th days after administration of 10 or 100 µg of depolymerase Dep_kpv74. (<b>A</b>) Unstimulated lymphocytes; (<b>B</b>–<b>D</b>) in vitro stimulation of lymphocytes by depolymerase Dep_kpv74, LPS, and ConA, respectively. Blue bars, control groups of mice without depolymerase injected; green bars, mice injected with 10 μg of depolymerase; brown bars, mice injected with 100 μg of depolymerase. <span class="html-italic">n</span> = 5 per group, means ± SD are plotted; ** <span class="html-italic">p</span> &lt; 0.001, *** <span class="html-italic">p</span> &lt; 0.0001 according to two-way ANOVA followed by Tukey’s multiple comparisons test.</p>
Full article ">Figure 5
<p>Percentage of T helper cells (CD3<sup>+</sup>CD4<sup>+</sup>), cytotoxic T cells (CD3<sup>+</sup>CD8<sup>+</sup>), and B cells (CD19<sup>+</sup>) in the mouse spleens on the 3rd (<b>A</b>) and 6th (<b>B</b>) days after administration of Dep_kpv74. Comparisons of the expression of CD69 in different cell subsets on the 3rd (<b>C</b>) and 6th (<b>D</b>) days after administration of Dep_kpv74. <span class="html-italic">n</span> = 5 per group, means ± SD are plotted; * <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 according to two-way ANOVA followed by Tukey’s multiple comparisons test. Representative flow cytometry plots are presented in <a href="#app1-antibiotics-14-00044" class="html-app">Figure S3</a>.</p>
Full article ">
16 pages, 7706 KiB  
Article
Intestinal Barrier Damage and Growth Retardation Caused by Exposure to Polystyrene Nanoplastics Through Lactation Milk in Developing Mice
by Chaoyu Zhou, Haiyan Wu, Lei Zhang, Xiao Xiao, Xiaodan Wang, Mingju Li, Runqiu Cai, Jia You, Qi Chen, Yifei Yang, Xinyuan Tian, Qianyu Bai, Yinzhu Chen, Huihui Bao and Tianlong Liu
Nanomaterials 2025, 15(1), 69; https://doi.org/10.3390/nano15010069 - 4 Jan 2025
Viewed by 563
Abstract
Microplastics, defined as plastic fragments smaller than 5 mm, degrade from larger pollutants, with nanoscale microplastic particles presenting significant biological interactions. This study investigates the toxic effects of polystyrene nanoplastics (PS-NPs) on juvenile mice, which were exposed through lactation milk and drinking water [...] Read more.
Microplastics, defined as plastic fragments smaller than 5 mm, degrade from larger pollutants, with nanoscale microplastic particles presenting significant biological interactions. This study investigates the toxic effects of polystyrene nanoplastics (PS-NPs) on juvenile mice, which were exposed through lactation milk and drinking water at concentrations of 0.01 mg/mL, 0.1 mg/mL, and 1 mg/mL. The results show that PS-NP exposure during lactation and juvenile periods caused delayed weight gain and impaired organ development, particularly in the liver and kidneys, without causing functional abnormalities or toxic injuries. The primary toxicity of PS-NPs was observed in the intestinal tract, including shortened villi, disrupted tight junctions, inhibited epithelial cell proliferation, and oxidative stress responses. These findings highlight the importance of evaluating the developmental toxicity of nanoplastics at environmentally relevant doses. Full article
(This article belongs to the Special Issue Environmental Fate, Transport and Effects of Nanoplastics)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic diagram of toxicity experiment in mice. (<b>B</b>,<b>C</b>) Weight changes of offspring mice of PDD18 and PDD46 after exposure to different doses of PS-NPs. Note: Compared with the control group, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Data were analyzed using one-way ANOVA and are expressed as mean ± SD (n = 10 for (<b>B</b>), and n = 5 for (<b>C</b>)).</p>
Full article ">Figure 2
<p>The effects of PS-NP exposure on the organs of the PDD18 (lactation period) offspring mice. (<b>A</b>–<b>D</b>) Changes in the main organ weights of the offspring mice at PDD18 after exposure to different doses of PS-NPs. Note: Compared with the control group, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. (<b>E</b>–<b>H</b>) Changes in the main organ visceral body ratio in the offspring mice of PDD18 after exposure to different doses of PS-NPs. Note: Compared with the control group, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Data were analyzed using one-way ANOVA and expressed as mean ± SD (n = 10).</p>
Full article ">Figure 3
<p>(<b>A</b>–<b>C</b>) Effects of different doses of PS-NP exposure on bone development of right hind limbs in offspring mice at PDD18 and PDD46. Asterisks indicate statistically significant differences. (<b>D</b>–<b>F</b>) Effects of different doses of PS-NP exposure on bone development of right hind limbs in offspring mice at PDD46. Data were analyzed using one-way ANOVA and are expressed as mean ± SD (n = 6).</p>
Full article ">Figure 4
<p>(<b>A</b>–<b>D</b>) Changes in liver and brain weights and visceral body ratio of PDD46 offspring mice after exposure to PS-NPs at different doses. Note: Compared with the control group, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Data were analyzed using one-way ANOVA and are expressed as mean ± SD (n = 5).</p>
Full article ">Figure 5
<p>The intestinal damage observed in the offspring mice at different stages due to PS-NP exposure. (<b>A</b>,<b>B</b>) Histological examination of offspring mice intestinal tissues with H&amp;E staining after exposure to different doses of PS-NPs. (<b>A</b>,<b>B</b>) PDD18 and PDD46, respectively. (<b>C</b>,<b>D</b>) The villus and crypt depths of the intestines in the PDD46 offspring mice. (<b>E</b>,<b>F</b>) Effects of different doses of PS-NPs on intestinal SOD and total GSH in offspring mice at PDD46 (n = 5). Note: Compared with the control group, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>The intestinal damage observed in the offspring mice at different stages due to PS-NP exposure. (<b>A</b>–<b>C</b>) Effects of different doses of PS-NP exposure on intestinal tight junction protein ZO-1 and nuclear proliferation antigen PCNA in offspring mice of PDD46. (<b>D</b>–<b>F</b>) Immunohistochemistry of intestinal ZO-1 and PCNA in offspring mice at PDD46 after exposure to different doses of PS-NPs. Note: Compared with the control group, ** <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 ">
16 pages, 2045 KiB  
Article
Identification of a New Pentafluorosulfanyl-Substituted Chalcone with Activity Against Hepatoma and Human Parasites
by Alessandra Viperino, Michael Höpfner, Nicole Edel, Ibrahim S. Al Nasr, Waleed S. Koko, Tariq A. Khan, Imen Ben Abdelmalek, Rainer Schobert, Bernhard Biersack and Bianca Nitzsche
Pharmaceuticals 2025, 18(1), 50; https://doi.org/10.3390/ph18010050 - 3 Jan 2025
Viewed by 471
Abstract
Background/Objectives: New drugs are required for the treatment of liver cancers and protozoal parasite infections. Analogs of the known anticancer active and antileishmanial 2′,4′,6′-trimethoxychalcone SU086 were prepared and investigated. Methods: The chalcones were prepared according to the Claisen–Schmidt condensation protocol and analyzed. They [...] Read more.
Background/Objectives: New drugs are required for the treatment of liver cancers and protozoal parasite infections. Analogs of the known anticancer active and antileishmanial 2′,4′,6′-trimethoxychalcone SU086 were prepared and investigated. Methods: The chalcones were prepared according to the Claisen–Schmidt condensation protocol and analyzed. They were tested for activity against two liver cancer cell lines (HepG2 and HuH-7) and protozoal parasites (Toxoplasma gondii and Leishmania major). Unspecific toxicity and expression of Hsp90 and Hsp70 upon treatment were analyzed in liver cancer cells. Results: A new chalcone, 2′,4′,6′-trimethoxy-3-pentafluorosulfanylchalcone (246TMP-3SF5), with a pentafluorosulfanyl (SF5) substituent showed pronounced activities against liver cancer cells and T. gondii parasites which were superior to the activities of the parent chalcone SU086 in these models. In contrast, SU086 and its anthracene analog 2′,4′,6′-trimethoxy-9-anthracenylchalcone (246TMP-Anth) were most active against L. major promastigotes. The new SF5-substituted chalcone behaved like the known Hsp90 inhibitor 17-AAG and upregulated Hsp70 expression in liver cancer cells. Conclusions: The SF5-substituted SU086 analog has potential to become a new drug for the therapy of hepatoma and toxoplasmosis. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Structures of the core compound <span class="html-italic">trans</span>-chalcone and of notable natural or synthetic chalcone derivatives with known anticancer activities.</p>
Full article ">Figure 2
<p>Structures of chalcones used in this study for anti-hepatoma and antiparasitic testing.</p>
Full article ">Figure 3
<p>Colony formation after 14 days of growth under chalcone 246TMP-3SF5 treatment or given inhibitor at the indicated concentrations in HepG2 (<b>A</b>) and HuH-7 cells (<b>B</b>). Colony count was assessed using ImageJ and quantified for HepG2 (<b>C</b>) and HuH-7 cells (<b>D</b>). Results are shown as means ± SEM of <span class="html-italic">n</span> = 4 independent experiments. **** <span class="html-italic">p</span> ≤ 0.0001; one-way ANOVA Dunnett’s post-hoc test.</p>
Full article ">Figure 4
<p>LDH assay with chalcone 246TMP-3SF5 and comparison with the Hsp90 inhibitor 17-AAG after 6 and 24 h of treatment at the indicated concentrations in HepG2 and HuH-7 cells. Data are given as changes in percentage compared to LDH release of untreated controls. Results are shown as means ± SEM of <span class="html-italic">n</span> = 3 independent experiments.</p>
Full article ">Figure 5
<p>Western Blot images showing Hsp90, Hsp70 and β-actin expression levels in HCC cell lines upon treatment with 246TMP-3SF5 and comparison compound 17-AAG for 24 h at indicated concentrations. Representative Western blot images of <span class="html-italic">n</span> = 3 independent experiments are shown.</p>
Full article ">
32 pages, 6318 KiB  
Review
Molecular Mechanisms of Phthalate-Induced Hepatic Injury and Amelioration by Plant-Based Principles
by Lalit Kumar Singh, Rashmi Pandey, Nikhat Jamal Siddiqi and Bechan Sharma
Toxics 2025, 13(1), 32; https://doi.org/10.3390/toxics13010032 - 2 Jan 2025
Viewed by 1036
Abstract
Phthalates are the emerging environmental toxicants derived from phthalic acid and its constituents, which are moderately present in plastics and many personal care products. Phthalate exposure occurs through various environmental factors, including air, water, and soil, with absorption facilitated via ingestion, inhalation, and [...] Read more.
Phthalates are the emerging environmental toxicants derived from phthalic acid and its constituents, which are moderately present in plastics and many personal care products. Phthalate exposure occurs through various environmental factors, including air, water, and soil, with absorption facilitated via ingestion, inhalation, and dermal contact. Upon exposure, phthalates become bioavailable within the biological systems and undergo biotransformation and detoxification processes in the liver. The physicochemical properties of phthalates indicate their lipophilicity, environmental persistence, and bioaccumulation potential, influencing their absorption, distribution, and hepatic biotransformation. The prolonged exposure to phthalates adversely influences the biological redox system by altering the levels of the enzymatic and non-enzymatic antioxidants, molecular signaling pathways, and causing hepatic pathogenesis. The strategies to combat phthalate-induced toxicity include avoiding exposure to these compounds and using plant-based bioactive molecules such as polyphenols, which possess therapeutic potential as antioxidants, suppress inflammatory cascades, prevent oxidative damage, and stabilize cellular integrity. This review presents a comprehensive and updated account of the chemical, biochemical, immunological, and toxicological properties of phthalates, along with novel plant-based therapeutic strategies to mitigate the phthalate-induced adverse effects on living systems. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The structure and synthesis of phthalate. Phthalic acid undergoes esterification with alcohol in the presence of an acid catalyst, producing phthalate esters and water as a byproduct.</p>
Full article ">Figure 2
<p>Pathways and routes of human exposure to phthalates released from different sources in the environment.</p>
Full article ">Figure 3
<p>Systematic presentation of metabolic pathways of phthalates in the liver, focusing on the conversion of diester phthalates (HMwP esters) into the corresponding monoester phthalates (LMwP esters). The hydroxylation and oxidation reactions of diesters are catalyzed by the phase I enzymes, while in this context the conjugation reactions of phase II are catalyzed by UDP-glucuronyl transferase.</p>
Full article ">Figure 4
<p>A schematic illustration delineating the toxicological impact of phthalates in the liver and its molecular mechanisms on the onset of inflammation and oxidative stress. This figure suggested that phthalates alleviated oxidative stress and inflammatory pathways and modulated the signaling cascades associated with DNA damage. AMPK: adenosine monophosphate-activated protein kinase, ATP: adenosine tri-phosphate, pAMPK: phosphorylated adenosine monophosphate-activated protein kinase, SREBP: sterol regulatory element-binding protein, FAS: fatty acid synthase, HSL: hormone-sensitive lipase, GPAT: glycerol-3-phosphate acyltransferase, TG: triglyceride, NAFLD: non-alcohol fatty liver disease, TGF-β: transforming growth factor-beta, JAK/STAT: janus kinase-signal transducer and activator of transcription. PPAR-γ: peroxisome proliferator-activated receptor gamma, NF-kB: nuclear factor kappa B, α-SMA: alpha-smooth muscle actin, COL III and I: collagen type III and I, Acot1: acyl-CoA thioesterase 1.</p>
Full article ">
28 pages, 5217 KiB  
Article
Rapid Multi-Well Evaluation of Assorted Materials for Hydrogel-Assisted Giant Unilamellar Vesicle Production: Empowering Bottom-Up Synthetic Biology
by Cherng-Wen Darren Tan, Magdalena Schöller and Eva-Kathrin Ehmoser
Gels 2025, 11(1), 29; https://doi.org/10.3390/gels11010029 - 2 Jan 2025
Viewed by 365
Abstract
Giant unilamellar vesicles (GUVs) are versatile cell models in biomedical and environmental research. Of the various GUV production methods, hydrogel-assisted GUV production is most easily implemented in a typical biological laboratory. To date, agarose, polyvinyl alcohol, cross-linked dextran-PEG, polyacrylamide, and starch hydrogels have [...] Read more.
Giant unilamellar vesicles (GUVs) are versatile cell models in biomedical and environmental research. Of the various GUV production methods, hydrogel-assisted GUV production is most easily implemented in a typical biological laboratory. To date, agarose, polyvinyl alcohol, cross-linked dextran-PEG, polyacrylamide, and starch hydrogels have been used to produce GUVs. Some leach and contaminate the GUVs, while others require handling toxic material or specialised chemistry, thus limiting their use by novices. Alternative hydrogel materials could address these issues or even offer novel advantages. To facilitate discovery, we replaced the manual spreading of reagents with controlled drop-casting in glass Petri dishes and polystyrene multi-well plates, allowing us to rapidly screen up to 96 GUV-production formulations simultaneously. Exploiting this, we rapidly evaluated assorted biomedical hydrogels, including PEG-DA, cross-linked hyaluronic acid, Matrigel, and cross-linked DNA. All of these alternatives successfully produced GUVs. In the process, we also developed a treatment for recycling agarose and polyvinyl alcohol hydrogels for GUV production, and successfully encapsulated porcine liver esterase (PLE-GUVs). PLE-GUVs offer a novel method of GUV labelling and tracing, which emulates the calcein-AM staining behaviour of cells. Our results highlight the utility of our protocol for potentiating substrate material discovery, as well as protocol and product development. Full article
(This article belongs to the Special Issue Hydrogel for Tissue Engineering and Biomedical Therapeutics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>Top</b>) Diagram showing a generic drop-casting approach to hydrogel-assisted GUV production. Step 3 may be modified according to the type of hydrogel or substrate used. O/N: overnight. Note step 2, which creates the protective coat for polystyrene containers, as well as steps 4–5, which modulate the evaporation of chloroform from the amphiphile solution. (<b>Bottom</b>) Phase contrast micrographs of DC2:1 (<b>a</b>,<b>b</b>) and PC2:1 (<b>c</b>,<b>d</b>) GUVs produced using spreading (<b>a</b>,<b>c</b>) and drop-casting (<b>b</b>,<b>d</b>) of AGA hydrogel precursors and amphiphile solutions. Factors (×) indicate how many times the GUV samples have been concentrated. Scale bar = 50 µm.</p>
Full article ">Figure 2
<p>SSC-FSC scatter plots of either DC2:1 (<b>a</b>,<b>b</b>) or PC2:1 (<b>c</b>,<b>d</b>) GUVs produced on AGA hydrogels using either spreading (<b>a</b>,<b>c</b>) or drop-casting (<b>b</b>,<b>d</b>). Colours indicate regional scatter densities with red being the highest and blue being the lowest. See Methods for how P1 gate was set. (<b>e</b>) Bar graphs show yields of GUV (expressed as events/µL as determined by flow cytometry) produced on AGA hydrogels using either DC2:1 or PC2:1, as well as either drop-casting or spreading. The volume of each sample injected was determined by multiplying the flow rate with the time taken to record 10,000 events. The yield or number density of GUVs (events/µL) was then determined by dividing the number of events in the P1 gate by the sample volume injected.</p>
Full article ">Figure 3
<p>Phase contrast micrographs of GUVs made from (<b>a</b>) DC1:0, (<b>b</b>) PC1:0, (<b>c</b>) DC2:1, (<b>d</b>) PC2:1, (<b>e</b>) DP1:0, and (<b>f</b>) PPC1:0 on AGA. Factors (×) indicate how many times the GUV samples have been concentrated. Scale bar = 50 µm.</p>
Full article ">Figure 4
<p>Overlay of (<b>a</b>) FSC and (<b>b</b>) SSC histograms, as well as (<b>c</b>) SSC-FSC scatter plots of GUVs produced from DC1:0, PC1:0, DC2:1, PC2:1, DP1:0, and PPC1:0 on AGA hydrogels.</p>
Full article ">Figure 5
<p>Phase contrast micrographs of GUVs produced on AGA-PVA 1:1 hydrogels using total lipids extracted from (<b>a</b>) TF228.1.16 and (<b>b</b>) A3R5.7 cells. Micrographs are representative of duplicate experiments. Factors (×) indicate how many times the GUV samples have been concentrated. Scale bar = 50 µm.</p>
Full article ">Figure 6
<p>Phase contrast micrographs showing DC2:1 GUVs produced in polystyrene multi-well plates using (<b>a</b>) PVA, (<b>b</b>) PD100-22, (<b>c</b>) 1% HA + 10% Irgacure, (<b>d</b>) 3 mg/mL Matrigel, and (<b>e</b>) 2.5% HMW DNA + 23% PEGDGE hydrogels. Each figure shown is only a representative of the dataset being reported. Please see the relevant <a href="#app1-gels-11-00029" class="html-app">Supplementary Figures</a> for the rest of the micrographs. (<b>f</b>) Epi-fluorescence micrograph of DC2:1 + Rh-PE GUVs (red) stained with Hoechst stain (blue). Arrowheads indicate GUVs encapsulating DNA material. Factors (×) indicate how many times the GUV samples have been concentrated. Scale bar = 50 µm.</p>
Full article ">Figure 7
<p>(<b>Left</b> column) Z-stacked fluorescence intensity profiles of HMW-DNA and PEGDA hydrogels without (C8, C9) and with (D8, D9) phospholipids. (<b>Right</b> column) Corresponding bright field micrograph of the sample surface. Bar = 200 µm. Data shown are representative of triplicates.</p>
Full article ">Figure 8
<p>(<b>a</b>) Epi-fluorescence micrograph showing DC2:1-PLE GUVs produced using rehydration buffer containing porcine liver esterase (PLE) and labelled with calcein-AM (green) as well as Nile Red (red). Nile Red was used to stain the DC2:1-PLE GUV membrane. Inset: TF228.1.16 cells labelled with calcein-AM. (<b>b</b>) Epi-fluorescence image overlay of DC2:1-PLE vesicles labelled with calcein (green), calcein Red (red), and calcein Blue (blue). Inset: A3R5.7 cells labelled with calcein-AM. Factors (×) indicate how many times the GUV samples have been concentrated. Scale bar = 50 µm.</p>
Full article ">Figure 9
<p>Phase contrast micrographs showing GUVs produced from DC2:1 (<b>a</b>,<b>c</b>,<b>e</b>) and PC2:1 (<b>b</b>,<b>d</b>,<b>f</b>) on AGA-PVA 1:1, used (<b>a</b>,<b>b</b>) for the first time, (<b>c</b>,<b>d</b>) reused once, and (<b>e</b>,<b>f</b>) reused twice. Factors (×) indicate how many times the GUV samples have been concentrated. Scale bar = 50 µm.</p>
Full article ">
15 pages, 5921 KiB  
Article
Bioaccumulation Study of Cadmium and Lead in Cyprinus carpio from the Colorado River, Using Automated Electrochemical Detection
by Federico Danilo Vallese, Sofia Stupniki, Mariano Trillini, Federico Belén, María Susana Di Nezio, Alfredo Juan and Marcelo Fabian Pistonesi
Water 2025, 17(1), 77; https://doi.org/10.3390/w17010077 - 31 Dec 2024
Viewed by 401
Abstract
The monitoring of heavy metals in aquatic ecosystems is of critical importance due to the toxic effects that these elements can have on wildlife and the potential risks that they pose to human health. Rivers situated in close proximity to agricultural regions are [...] Read more.
The monitoring of heavy metals in aquatic ecosystems is of critical importance due to the toxic effects that these elements can have on wildlife and the potential risks that they pose to human health. Rivers situated in close proximity to agricultural regions are particularly susceptible to contamination from a combination of natural and anthropogenic sources. The study of bioaccumulation is of great importance for the early detection of environmental stressors. The combination of electrochemical techniques, such as square-wave anodic stripping voltammetry (SWASV), with automated flow-batch systems represents an efficient and cost-effective approach for the detection of trace metals in environmental samples. This study examines the bioaccumulation of cadmium and lead in Cyprinus carpio, a bioindicator of contamination in the Colorado River, Argentina. The fish were exposed to sublethal metal concentrations for 24, 48, and 96 h. Metal quantification was conducted using a novel automatic flow-batch system with SWASV and a bismuth film electrode. To the best of our knowledge, this constitutes the first application of this methodology on aquatic bioindicators for the assessment of metal accumulation in a natural environment. The technique demonstrated enhanced sensitivity and selectivity for the detection of trace metals. The bioaccumulation results demonstrated an increase in cadmium and lead concentrations in fish liver tissue after 96 h, reaching 10.5 µg g−1 and 11.9 µg g−1, respectively. Validation with inductively coupled plasma–atomic emission spectrometry (ICP-AES) demonstrated a satisfactory correlation, confirming the reliability of the method. This novel electrochemical approach offers enhanced accuracy and efficiency, making it a promising tool for environmental monitoring. The results indicate that Colorado River water is within safe levels for aquatic life regarding these metals. However, continuous monitoring is recommended to detect changes in contamination levels and protect ecosystem health, especially during water crises and under climate change. Full article
(This article belongs to the Special Issue Impact of Environmental Factors on Aquatic Ecosystem)
Show Figures

Figure 1

Figure 1
<p>Study area, with the Paso Alsina station as the monitoring site.</p>
Full article ">Figure 2
<p>Flow diagram of the experimental procedure: fish acclimatization, metal exposure, liver collection, and sample preparation for analysis.</p>
Full article ">Figure 3
<p>Embedded flow-batch system with electrochemical detection. MP: metering pump; EC: electrochemical cell; MS: magnetic stirrer; Buffer: buffer solution of 0.1 M of sodium acetate; BS: bismuth solution acidified with 5 mL of nitric acid; Sample: cadmium and lead standard solution or sample; Water: ultrapure water (18 MΩ).</p>
Full article ">Figure 4
<p>(<b>A</b>) Voltagram for a resulting homogenate of <span class="html-italic">Cyprinus carpio</span> taken from the Colorado River belonging to the control group. (<b>B</b>) Voltagram for a resulting homogenate of <span class="html-italic">Cyprinus carpio</span> after 48 h of exposure to metals.</p>
Full article ">
22 pages, 1692 KiB  
Article
Factors Influencing the Duration of Maintenance Therapy in Metastatic Colorectal Cancer
by Théo Fourrier, Caroline Truntzer, Morgane Peroz, Valentin Derangère, Julie Vincent, Leila Bengrine-Lefèvre, Audrey Hennequin, Rémi Palmier, David Orry, Thomas Rabel and François Ghiringhelli
Cancers 2025, 17(1), 88; https://doi.org/10.3390/cancers17010088 - 30 Dec 2024
Viewed by 503
Abstract
Background/Objectives: Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by [...] Read more.
Background/Objectives: Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by maintenance therapy with less cytotoxic molecules or a chemotherapy-free interval to reduce chemotherapy-related toxicity. In this study, the aim was to determine the patient, cancer and treatment factors that influence the duration of maintenance therapy (DMT). Methods: We collected retrospective data on a cohort of 133 patients treated at the Centre Georges François Leclerc (CGFL) cancer centre in Dijon between March 2014 and June 2022. Patients had unresectable or potentially resectable diseases. They received first-line induction treatment with chemotherapy and/or targeted therapy and maintenance treatment, defined as the interruption of at least one chemotherapy agent. Results: In the multivariate analysis, age (HR: 1.02, 95% CI 1.00–1.04, p = 0.031), N2 nodal status (HR: 1.78, 95% CI 1.09–2.89, p = 0.021) and the presence of peritoneal metastases (HR: 2.05, 95% CI 1.25–3.36, p = 0.004), as well as baseline carcino-embryonic antigen (CEA) level (HR: 1.10, 95% CI 1.00–1.20, p = 0.052), were significantly associated to poor DMT. Local treatment of liver metastases also significantly reduced the DMT (HR: 0.49, 95% CI 0.28–0.86, p = 0.013). In our cohort, induction triplet chemotherapy significantly increased the CEA delta (70% vs. 44%, p = 0.047) compared to doublet chemotherapy and led to a higher rate of liver surgery (40% vs. 21%, p = 0.014) and a trend for a higher rate of local treatment of metastases (62% vs. 45%, p = 0.059). Conclusions: Duration of maintenance therapy is determined by the initial patient and colorectal cancer characteristics. However, it is significantly increased by local treatment of liver metastases. By reducing the tumour burden, a triplet induction chemotherapy regimen increases the rate of liver metastase resection. Full article
(This article belongs to the Special Issue Contemporary Treatment of Colorectal Cancer)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Kaplan-Meier curve of the DMT depending on the induction chemotherapy regimen; (<b>b</b>) Kaplan-Meier curve of PFS depending on the induction chemotherapy regimen; (<b>c</b>) Kaplan-Meier curve of OS depending on the induction chemotherapy regimen.</p>
Full article ">Figure 2
<p>(<b>a</b>) Kaplan-Meier curve of the DMT depending on the maintenance chemotherapy regimen; (<b>b</b>) Kaplan-Meier curve of PFS depending on the maintenance chemotherapy regimen; (<b>c</b>) Kaplan-Meier curve of PFS depending on the maintenance chemotherapy regimen.</p>
Full article ">Figure 3
<p>(<b>a</b>) Kaplan-Meier curve of the DMT depending on DPYD status; (<b>b</b>) Kaplan-Meier curve of the PFS depending on DPYD status; (<b>c</b>) Kaplan-Meier curve of the OS depending on DPYD status.</p>
Full article ">Figure 4
<p>(<b>a</b>) Kaplan-Meier curve of the DMT depending on UGT1A1 status; (<b>b</b>) Kaplan-Meier curve of PFS depending on UGT1A1 status; (<b>c</b>) Kaplan-Meier curve of OS depending on UGT1A1 status.</p>
Full article ">
Back to TopTop