[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

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 (4,005)

Search Parameters:
Keywords = ERK

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 2557 KiB  
Article
Effects of Hispidulin on the Osteo/Odontogenic and Endothelial Differentiation of Dental Pulp Stem Cells
by Yeon Kim, Hyun-Joo Park, Mi-Kyoung Kim, Hyung Joon Kim, Yong-Il Kim, Soo-Kyung Bae and Moon-Kyoung Bae
Pharmaceuticals 2024, 17(12), 1740; https://doi.org/10.3390/ph17121740 - 23 Dec 2024
Abstract
Background: Human dental pulp stem cells (HDPSCs) with multi-lineage differentiation potential and migration ability are required for HDPSC-based bone and dental regeneration. Hispidulin is a naturally occurring flavonoid with diverse pharmacological activities, but its effects on biological properties of HDPSCs remain unknown. Therefore, [...] Read more.
Background: Human dental pulp stem cells (HDPSCs) with multi-lineage differentiation potential and migration ability are required for HDPSC-based bone and dental regeneration. Hispidulin is a naturally occurring flavonoid with diverse pharmacological activities, but its effects on biological properties of HDPSCs remain unknown. Therefore, we investigated the effects of hispidulin on the differentiation potential and migration ability of HDPSCs and elucidated their underlying mechanisms. Methods: The osteo/odontogenic capacity of HDPSCs was assessed using the alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining. The migration ability of HDPSCs was evaluated using a scratch wound assay. Furthermore, the endothelial differentiation of HDPSCs was examined by using a capillary sprouting assay and by assessing CD31 expression. Results: Hispidulin significantly enhanced the osteo/odontogenic differentiation of HDPSCs with increased expression of osteo/odontogenic differentiation markers. Hispidulin increased the migration of HDPSCs, which was mediated by the upregulation of C-X-C chemokine receptor type 4 (CXCR4). The treatment of HDPSCs with hispidulin enhanced the differentiation of HDPSCs into endothelial cells, as evidenced by increased capillary sprouting and endothelial marker expression. In addition, we demonstrated that hispidulin activated the ERK1/2 signaling, and its inhibition by U0126 significantly suppressed the hispidulin-induced endothelial differentiation of HDPSCs. Conclusions: These findings demonstrate that hispidulin effectively promotes the osteo/odontogenic and endothelial differentiation, and migration of HDPSCs. These results suggest that hispidulin may have potential therapeutic applications in dental pulp regeneration and tissue engineering. Full article
(This article belongs to the Special Issue Pharmacological Activities of Flavonoids and Their Analogues 2024)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of hispidulin on osteo/odontogenic differentiation and expression of osteo/odontogenic-related markers in HDPSCs. (<b>A</b>) HDPSCs were either cultured in the basic growth medium or osteogenic differentiation medium (ODM) with or without hispidulin (5 μM). ALP staining was performed on days 7 and 14. Stained cells were photographed using a phase contrast microscope at 100× magnification. ALP-positive areas were quantified by densitometry in triplicates. * <span class="html-italic">p</span> &lt; 0.01 compared to control. # <span class="html-italic">p</span> &lt; 0.05 compared to ODM. (<b>B</b>) The formation of mineralized nodules was evaluated by ARS staining and quantified through densitometric analysis in triplicate at days 14 and 21. The stained cells were imaged under 100× magnification. * <span class="html-italic">p</span> &lt; 0.01 compared to control. # <span class="html-italic">p</span> &lt; 0.01 compared to ODM. (<b>C</b>) HDPSCs were cultured with or without ODM in the presence of hispidulin for 14 days. The mRNA expression of <span class="html-italic">ALP</span>, <span class="html-italic">osteocalcin</span>, <span class="html-italic">DMP-1</span>, <span class="html-italic">and Runx-2</span> were assessed through real-time PCR analysis. All values were normalized to β-actin mRNA levels, and the expression level of the control group was designated as 1.0. * <span class="html-italic">p</span> &lt; 0.01 compared to control. ** <span class="html-italic">p</span> &lt; 0.05 compared to control. # <span class="html-italic">p</span> &lt; 0.01 compared to ODM. ## <span class="html-italic">p</span> &lt; 0.05 compared to ODM.</p>
Full article ">Figure 2
<p>Effect of hispidulin on the migration of HDPSCs. (<b>A</b>) Scratch wound migration assays were performed on HDPSCs cultured without or with hispidulin (1 or 5 μM) for 24 h. Cell migration into the scratch wound area was photographed at 100× magnification and quantified. Results are expressed as the mean values from three independent experiments per group. * <span class="html-italic">p</span> &lt; 0.05 compared with control. # <span class="html-italic">p</span> &lt; 0.01 compared with control. (<b>B</b>) HDPSCs were treated with hispidulin (1 or 5 μM) for 24 h, and the expression of CXCR4 was analyzed with real-time qPCR. All values were normalized to β-actin mRNA levels, with the control group expression set as 1.0. * <span class="html-italic">p</span> &lt; 0.05 compared with control. (<b>C</b>) Protein expression of CXCR4 was observed by western blotting using an anti-CXCR4 antibody (upper) and densitometric analysis (lower). β-actin was used as the loading control. (<b>D</b>) HDPSCs were incubated with 1 μM hispidulin alone or in combination with AMD3100 (50 μg/mL) for 24 h. Migrated cells beyond the reference line were photographed at 100× magnification and quantified. * <span class="html-italic">p &lt;</span> 0.01 compared with control. # <span class="html-italic">p</span> &lt; 0.01 compared to hispidulin.</p>
Full article ">Figure 3
<p>Effect of hispidulin on endothelial differentiation of HDPSCs. (<b>A</b>) HDPSCs implanted on a Matrigel-coated plate were treated with 1 μM hispidulin for 3, 5, and 7 days in the EGM-2MV. The numbers of sprouts were counted and imaged under 100× magnification. Each result represents the mean value of triplicate experiments in each group. * <span class="html-italic">p</span> &lt; 0.01 compared with the 3-days control. # <span class="html-italic">p</span> &lt; 0.05 compared with 5-dayscontrol. ** <span class="html-italic">p</span> &lt; 0.01 compared with the 7-days control. (<b>B</b>) The protein expression of CD31 was analyzed by western blotting. β-actin was used as a loading control. (<b>C</b>) HDPSCs were treated with 1 μM hispidulin for 3, 5, and 7 days. CD31 was measured by flow cytometry.</p>
Full article ">Figure 4
<p>Effect of hispidulin on the ERK signaling pathway in endothelial differentiation of HDPSCs. (<b>A</b>) HDPSCs were treated with hispidulin (1 μM) for the indicated times in EGM-2MV. Cell lysates were immunoblotted with antibodies against phospho-ERK and total ERK. β-actin was used as a loading control. (<b>B</b>) HDPSCs were treated with hispidulin (1 μM) alone or in combination with U0126 (10 μM). After 7 days, CD31 protein expression was analyzed by western blotting. β-actin was used as a loading control. (<b>C</b>) HDPSCs were seeded on Matrigel-coated plates and treated with hispidulin (1 μM) alone or in combination with U0126 (10 μM) in EGM-2MV. Capillary sprouting was observed after 7 days (200× magnification). Images are representative of three independent experiments. * <span class="html-italic">p</span> &lt; 0.01 compared to control. # <span class="html-italic">p</span> &lt; 0.01 compared to hispidulin.</p>
Full article ">Figure 5
<p>Schematic diagram illustrating the regulatory mechanisms of hispidulin in promoting osteo/odontogenic and endothelial differentiation, and migration of HDPSCs.</p>
Full article ">
21 pages, 4110 KiB  
Article
Succinate Regulates Endothelial Mitochondrial Function and Barrier Integrity
by Reham Atallah, Juergen Gindlhuber, Wolfgang Platzer, Rishi Rajesh and Akos Heinemann
Antioxidants 2024, 13(12), 1579; https://doi.org/10.3390/antiox13121579 (registering DOI) - 21 Dec 2024
Viewed by 317
Abstract
Endothelial dysfunction is a hallmark of several pathological conditions, including cancer, cardiovascular disease and inflammatory disorders. In these conditions, perturbed TCA cycle and subsequent succinate accumulation have been reported. The role of succinate as a regulator of immunological responses and inflammation is increasingly [...] Read more.
Endothelial dysfunction is a hallmark of several pathological conditions, including cancer, cardiovascular disease and inflammatory disorders. In these conditions, perturbed TCA cycle and subsequent succinate accumulation have been reported. The role of succinate as a regulator of immunological responses and inflammation is increasingly being recognized. Nevertheless, how endothelial cell function and phenotype are altered by elevated intracellular succinate has not been addressed yet. Thus, we employed numerous in vitro functional assays using primary HUVECs and diethyl succinate (DES), a cell membrane-permeable succinate analogue. An MTS assay 1 h post stimulation with DES suggested reduced metabolic activity in HUVECs. Concurrently, elevated production of ROS, including mitochondrial superoxide, and a reduction in mitochondrial membrane potential were observed. These findings were corroborated by Seahorse mito-stress testing, which revealed that DES acutely lowered the OCR, maximal respiration and ATP production. Given the link between mitochondrial stress and apoptosis, we examined important survival signalling pathways. DES transiently reduced ERK1/2 phosphorylation, a response that was followed by a skewed pro-apoptotic shift in the BAX to BCL2L1 gene expression ratio, which coincided with upregulating VEGF gene expression. This indicated an induction of mixed pro-apoptotic and pro-survival signals in the cell. However, the BAX/BCL-XL protein ratio was unchanged, suggesting that the cells did not commit themselves to apoptosis. An MTS assay, caspase 3/7 activity assay and annexin V/propidium iodide staining confirmed this finding. By contrast, stimulation with DES induced acute endothelial barrier permeability, forming intercellular gaps, altering cell size and associated actin filaments without affecting cell count. Notably, during overnight DES exposure gradual recovery of the endothelial barrier and cell sprouting was observed, alongside mitochondrial membrane potential restoration, albeit with sustained ROS production. COX-2 inhibition and EP4 receptor blockade hindered barrier restoration, implicating a role of COX-2/PGE2/EP4 signalling in this process. Interestingly, ascorbic acid pre-treatment prevented DES-induced acute barrier disruption independently from ROS modulation. In conclusion, succinate acts as a significant regulator of endothelial mitochondrial function and barrier integrity, a response that is counterbalanced by upregulated VEGF and prostaglandin production by the endothelial cells. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of DES on metabolic and mitochondrial function in HUVECs. (<b>A</b>) MTS assay of HUVECs stimulated with DES at indicated concentrations for 1 h (n = 3). (<b>B</b>) DHR 123 geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for selected time points (n = 5). (<b>C</b>) MitoSOX geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for selected time points (n = 3). (<b>D</b>) TMRE geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for selected time points (n = 4). For (<b>B</b>–<b>D</b>), the dotted line refers to vehicle-treated cells. (<b>E</b>) Representative microscopic image for 3 independent experiments with TMRE in HUVECs stimulated with 10 mM DES for indicated times. (<b>F</b>) Mito-stress assay of HUVECs demonstrating OCR with sequential addition of treatments. Acute response to DES, maximal respiration and ATP production were calculated (n = 5). For (<b>A</b>–<b>D</b>,<b>F</b>), data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>B</b>–<b>D</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>,<b>F</b>). For (<b>C</b>,<b>D</b>), * refers to comparison between vehicle and 10 mM DES, while # refers to comparison between vehicle and 5 mM DES. *, # <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 2
<p>Effect of DES on HUVECs viability. (<b>A</b>) Western blot of phosphorylated ERK1/2 and ratio of pERK/tERK/β-actin in HUVECs stimulated with 10 mM DES for indicated time points (n = 4). (<b>B</b>) RT-PCR of BAX, BCL2L1 and VEGF mRNA expression in HUVECs stimulated with 10 mM DES for 4 h (n = 5). (<b>C</b>) Western blot of BAX and BCL-XL in HUVECs stimulated with 10 mM DES for 6 h (n = 5). (<b>D</b>) MTS assay of HUVECs stimulated with indicated DES concentrations for 6 h (n = 3). (<b>E</b>) Caspase 3/7 activity of HUVECs post treatment with indicated DES concentrations for 16 h (n = 3). (<b>F</b>) Representative dot plot of annexin V/propidium iodide staining of HUVECs treated with indicated DES concentrations for 16 h. Live and apoptotic cells were quantified (n = 9). Data are presented as mean and SEM, with statistical significance determined using one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>,<b>D</b>–<b>F</b>) or paired <span class="html-italic">t</span>-test (<b>B</b>,<b>C</b>). * <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 3
<p>Acute effect of DES on HUVECs barrier integrity. (<b>A</b>) Resistance of HUVECs monolayer stimulated with DES at indicated concentrations (n = 4). (<b>B</b>) Immunofluorescence staining of VE-cadherin and F-actin in HUVECs stimulated with 10 mM DES at selected time points. The image is representative of 3 independent experiments. Zoomed-in images are shown on the upper right side for each condition, scale bar = 50 µm. (<b>C</b>) Image analysis demonstrating number of nuclei, gap size, average cell size and median fluorescence intensity (MFI) of phalloidin-stained stress fibres. Data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>C</b>). For (<b>A</b>), * refers to comparison between vehicle and 10 mM DES, while # refers to comparison between vehicle and 5 mM DES. * <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>Restoration of HUVECs barrier and sprouting during overnight stimulation with DES. (<b>A</b>) Resistance of HUVECs monolayer with overnight stimulation with DES at indicated concentrations (n = 6). (<b>B</b>) Spheroid sprouting assay of HUVECs stimulated with 10 mM DES for 16 h. Number of sprouts and total sprout length were calculated. The image is representative of three independent experiments. (<b>C</b>) Western blot of HIF-1α in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 8). (<b>D</b>) Western blot of phosphorylated ERK1/2 and ratio of pERK/tERK/β-actin in HUVECs stimulated with indicated DES concentrations for 16 h (n = 4). (<b>E</b>) MTS assay of HUVECs stimulated with DES at indicated concentrations for 16 h (n = 3). (<b>F</b>) DHR 123 geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 6). (<b>G</b>) MitoSOX geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 5). (<b>H</b>) TMRE geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 4). Data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>), paired <span class="html-italic">t</span>-test (<b>B</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>C</b>–<b>H</b>). ns refers to no statistical difference between vehicle and either 5 mM DES or 10 mM DES. * <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 5
<p>COX-2/PGE<sub>2</sub>/EP4 contribution to barrier recovery in HUVECs. (<b>A</b>) Western blot of COX-2 in HUVECs after 16 h of stimulation with DES at indicated concentrations (n = 6). (<b>B</b>) Radioimmunoassay for quantification of PGE<sub>2</sub> in supernatants of HUVECs stimulated with DES for 16 h (n = 3). (<b>C</b>) Resistance of HUVECs monolayer treated with diclofenac ~1 h prior to stimulation with 10 mM DES (n = 3). (<b>D</b>) Resistance of HUVECs monolayer stimulated with PGE<sub>2</sub> (n = 3). (<b>E</b>) Resistance of HUVECs monolayer treated with EP2 antagonist ~2 h post treatment with 10 mM DES (n = 3). (<b>F</b>) Resistance of HUVECs monolayer treated with EP4 antagonist ~2 h post treatment with 10 mM DES (n = 3). Data are presented as mean and SEM, with statistical significance determined using one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>,<b>B</b>) or two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>C</b>–<b>F</b>). * <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 6
<p>Ascorbic acid prevention of initial barrier drop induced by DES in HUVECs. (<b>A</b>) Resistance of HUVECs monolayer treated with ascorbic acid at demonstrated concentrations 15 min prior to stimulation with DES at indicated concentrations (n = 4). (<b>B</b>) DHR 123 geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 1 h with/without 15 min of ascorbic acid pre-treatment (n = 3). (<b>C</b>) MitoSOX geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 1 h with/without 15 min of ascorbic acid pre-treatment (n = 3). Data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>B</b>,<b>C</b>). For (<b>A</b>), * refers to comparison between 10 mM DES and AA + 10 mM DES, while # refers to comparison between 5 mM DES and AA + 5 mM DES. *, # <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. AA denotes ascorbic acid.</p>
Full article ">
21 pages, 7602 KiB  
Article
Pentagalloyl Glucose from Bouea macrophylla Suppresses the Epithelial–Mesenchymal Transition and Synergizes the Doxorubicin-Induced Anticancer and Anti-Migration Effects in Triple-Negative Breast Cancer
by Jiraporn Kantapan, Phattarawadee Innuan, Sarawut Kongkarnka, Padchanee Sangthong and Nathupakorn Dechsupa
Pharmaceuticals 2024, 17(12), 1729; https://doi.org/10.3390/ph17121729 - 20 Dec 2024
Viewed by 406
Abstract
Background: Triple-negative breast cancer (TNBC) represents an aggressive form of breast cancer with few available therapeutic options. Chemotherapy, particularly with drugs like doxorubicin (DOX), remains the cornerstone of treatment for this challenging subtype. However, the clinical utility of DOX is hampered by adverse [...] Read more.
Background: Triple-negative breast cancer (TNBC) represents an aggressive form of breast cancer with few available therapeutic options. Chemotherapy, particularly with drugs like doxorubicin (DOX), remains the cornerstone of treatment for this challenging subtype. However, the clinical utility of DOX is hampered by adverse effects that escalate with higher doses and drug resistance, underscoring the need for alternative therapies. This study explored the efficacy of pentagalloyl glucose (PGG), a natural polyphenol derived from Bouea macrophylla, in enhancing DOX’s anticancer effects and suppressing the epithelial–mesenchymal transition (EMT) in TNBC cells. Methods: This study employed diverse methodologies to assess the effects of PGG and DOX on TNBC cells. MDA-MB231 triple-negative breast cancer cells were used to evaluate cell viability, migration, invasion, apoptosis, mitochondrial membrane potential, and protein expression through techniques including MTT assays, wound healing assays, flow cytometry, Western blotting, and immunofluorescence. Results: Our findings demonstrate that PGG combined with DOX significantly inhibits TNBC cell proliferation, migration, and invasion. PGG enhances DOX-induced apoptosis by disrupting the mitochondrial membrane potential and activating caspase pathways; consequently, the activation of caspase-3 and the cleavage of PARP are increased. Additionally, the study shows that the combination treatment upregulates ERK signaling, further promoting apoptosis. Moreover, PGG reverses DOX-induced EMT by downregulating mesenchymal markers (vimentin and β-catenin) and upregulating epithelial markers (E-cadherin). Furthermore, it effectively inhibits STAT3 phosphorylation, associated with cell survival and migration. Conclusions: These results highlight the potential of PGG as an adjuvant therapy in TNBC treatment. PGG synergizes with DOX, which potentiates its anticancer effects while mitigating adverse reactions. Full article
(This article belongs to the Special Issue Adjuvant Therapies for Cancer Treatment)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The inhibitory effect of PGG and DOX on cell proliferation in TNBC cells, as assessed using the MTT assay. (<b>A</b>–<b>C</b>) Dose–response curves illustrating the cytotoxic effects of PGG on MCF-10A cells (<b>A</b>), DOX on MDA-MB231 cells (<b>B</b>), and PGG on MDA-MB231 cells (<b>C</b>) after 48 h of treatment. (<b>D</b>,<b>E</b>) Clonogenic survival analysis indicating the number of colonies formed by MDA-MB231 cells following treatment with varying concentrations of PGG (0, 2.5, 5, 10, 20, and 40 µM). Representative colony images are shown in (<b>D</b>), with quantification graphs in (<b>E</b>). The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control, while *** <span class="html-italic">p</span> &lt; 0.001 denotes a highly significant difference from the control. PGG, pentagalloyl glucose; DOX, doxorubicin.</p>
Full article ">Figure 2
<p>PGG inhibits the migration and invasion capabilities of TNBC cells. (<b>A</b>) Representative microscopic images from wound healing assays performed on MDA-MB231 cells treated with varying concentrations of PGG, captured at 0 and 48 h. (<b>B</b>) Quantification of wound closure percentages, demonstrating the impact of PGG treatment. (<b>C</b>) Transwell chamber images showing cell migration following treatment with various concentrations of PGG. (<b>D</b>) Quantification of migrated cells: migrating cells were counted in five high-power fields and averaged. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control. Scale bar = 100 µm (<b>A</b>,<b>C</b>). PGG, pentagalloyl glucose.</p>
Full article ">Figure 3
<p>Impact of combined treatment with 10, 20, and 40 µM PGG and varying concentrations of DOX on the viability of MDA-MB231 cells after 48 h, as assessed using the MTT assay (<b>A</b>). Fa-CI plot analysis depicting the interaction between DOX and PGG in MDA-MB231 cells. The dashed line at CI = 1 signifies an additive effect, while CI values less than, equal to, or greater than 1 indicate synergy, additivity, or antagonism, respectively (<b>B</b>). The effect (Fa) represents the degree of fractional inhibition associated with each combination index. PGG, pentagalloyl glucose; DOX, doxorubicin.</p>
Full article ">Figure 4
<p>The impact on apoptosis of PGG and DOX as monotherapies or in combination in TNBC cells. (<b>A</b>) Representative dot plots show the apoptotic response of MDA-MB231 cells to the indicated treatments. (<b>B</b>) Quantitative data represent the percentage of total cell death, as determined using flow cytometry. (<b>C</b>,<b>D</b>) The effects of PGG on mitochondrial membrane potential in MDA-MB231 cells were assessed using JC-1 staining: (<b>C</b>) representative images display JC-1 fluorescence across different treatment groups after 24 h, with FCCP as the positive control. Monomeric JC-1 exhibits green fluorescence, while aggregated JC-1 emits red fluorescence; (<b>D</b>) quantification of the red/green fluorescence ratio shown in a histogram. Data are presented as the mean ± SD of three independent experiments. (<b>E</b>) Western blot analysis of apoptotic markers, including Bax, Bcl-2, caspase-3, PARP, p-ERK, and t-ERK. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S1</a>. (<b>F</b>–<b>J</b>) The relative protein density values were quantified, with expression levels normalized to GAPDH as the loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control. Scale bar = 100 µm (<b>C</b>). PGG, pentagalloyl glucose; DOX, doxorubicin; PARP, poly ADP ribose polymerase; ERK, extracellular signal-regulated kinase.</p>
Full article ">Figure 4 Cont.
<p>The impact on apoptosis of PGG and DOX as monotherapies or in combination in TNBC cells. (<b>A</b>) Representative dot plots show the apoptotic response of MDA-MB231 cells to the indicated treatments. (<b>B</b>) Quantitative data represent the percentage of total cell death, as determined using flow cytometry. (<b>C</b>,<b>D</b>) The effects of PGG on mitochondrial membrane potential in MDA-MB231 cells were assessed using JC-1 staining: (<b>C</b>) representative images display JC-1 fluorescence across different treatment groups after 24 h, with FCCP as the positive control. Monomeric JC-1 exhibits green fluorescence, while aggregated JC-1 emits red fluorescence; (<b>D</b>) quantification of the red/green fluorescence ratio shown in a histogram. Data are presented as the mean ± SD of three independent experiments. (<b>E</b>) Western blot analysis of apoptotic markers, including Bax, Bcl-2, caspase-3, PARP, p-ERK, and t-ERK. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S1</a>. (<b>F</b>–<b>J</b>) The relative protein density values were quantified, with expression levels normalized to GAPDH as the loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control. Scale bar = 100 µm (<b>C</b>). PGG, pentagalloyl glucose; DOX, doxorubicin; PARP, poly ADP ribose polymerase; ERK, extracellular signal-regulated kinase.</p>
Full article ">Figure 5
<p>The impact of PGG combined with DOX on the migratory behavior of TNBC cells. (<b>A</b>) Representative microscopic images from wound healing assays and (<b>C</b>) quantification of wound closure percentages illustrating the effects of treatments with PGG (40 µM), DOX (0.75 µM), and their combination. (<b>B</b>) Transwell chamber images showing cell migration following treatment with PGG (40 µM), DOX (0.75 µM), or a combination of both. (<b>D</b>) Quantitative analysis of the number of migrating cells. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control and DOX treatment alone. Scale bar = 100 µm (<b>A</b>,<b>B</b>). PGG, pentagalloyl glucose; DOX, doxorubicin.</p>
Full article ">Figure 6
<p>Reversal of EMT and suppression of EMT marker expression by PGG in TNBC cell lines. MDA-MB231 cells were treated for 48 h, as indicated, and the expression of EMT markers was analyzed using Western blot. (<b>A</b>) Representative Western blot images showing the levels of β-catenin, vimentin, E-cadherin, and GAPDH. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S2</a>. (<b>B</b>–<b>D</b>) Quantification of band intensities from the Western blot analysis. Relative protein levels were quantified and normalized to GAPDH as the loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control and DOX-only treatment. (<b>E</b>) Immunofluorescence staining of β-catenin (green), E-cadherin (green), and vimentin (red) in MDA-MB231 cells, with nuclei counterstained using DAPI (blue). Scale bar = 100 µm.</p>
Full article ">Figure 7
<p>The effects of PGG and DOX, either alone or in combination, on STAT3 signaling proteins in TNBC cells. (<b>A</b>) Representative Western blot images showing the expression levels of phosphorylated STAT3 (p-STAT3), total STAT3 (t-STAT3), and GAPDH after treatment with PGG (40 µM), DOX (0.75 µM), or their combination for 48 h in MDA-MB231 cells. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S3</a>. (<b>B</b>) Bar graph depicting the fold change in protein expression. The relative protein densities were quantified and normalized to the GAPDH loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference compared to the control and DOX treatment alone. PGG, pentagalloyl glucose; DOX, doxorubicin; STAT3, signal transducer and activator of transcription 3; p, phosphorylated; t, total; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.</p>
Full article ">
12 pages, 6598 KiB  
Article
Different Cytotoxic Effects of Cisplatin on Pancreatic Ductal Adenocarcinoma Cell Lines
by Antonella Muscella, Luca G. Cossa, Erika Stefàno, Gianluca Rovito, Michele Benedetti, Francesco P. Fanizzi and Santo Marsigliante
Int. J. Mol. Sci. 2024, 25(24), 13662; https://doi.org/10.3390/ijms252413662 - 20 Dec 2024
Viewed by 296
Abstract
This study examined the response to cisplatin in BxPC-3, Mia-Paca-2, PANC-1, and YAPC pancreatic cancer lines with different genotypic and phenotypic characteristics, and the mechanisms associated with their resistance. BxPC-3 and MIA-PaCa-2 cell lines were the most sensitive to cisplatin, while YAPC and [...] Read more.
This study examined the response to cisplatin in BxPC-3, Mia-Paca-2, PANC-1, and YAPC pancreatic cancer lines with different genotypic and phenotypic characteristics, and the mechanisms associated with their resistance. BxPC-3 and MIA-PaCa-2 cell lines were the most sensitive to cisplatin, while YAPC and PANC-1 were more resistant. Consistently, in cisplatin-treated BxPC-3 cells, the cleavage patterns of pro-caspase-9, -7, -3, and PARP-1 demonstrated that they were more sensitive than YAPC cells. The autophagic pathway, promoting cisplatin resistance, was active in BxPC-3 cells, as demonstrated by the time-dependent conversion of LC3-I to LC3-II, whereas it was not activated in YAPC cells. In cisplatin-treated BxPC-3 cells, Bcl-2 decreased, while Beclin-1, Atg-3, and Atg-5 increased along with JNK1/2 phosphorylation. Basal levels of phosphorylated ERK1/2 in each cell line were positively correlated with cisplatin IC50 values, and cisplatin caused the activation of ERK1/2 in BxPC-3 and YAPC cells. Furthermore, ERK1/2 pharmacological inactivation increased cisplatin lethality in both BxPC-3 and YAPC cells, suggesting that p-ERK1/2 may be related to cisplatin resistance of PDAC cells. Different mechanisms and strategies are generally required to acquire drug resistance. Here, we partially explain the other response to cisplatin of BxPC-3 and YAPC cell lines by relating it to the role of ERK pathway. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Cytotoxic effects of cisplatin on pancreatic tumor lines. BxPC-3 (<b>A</b>), Mia Paca-2 (<b>B</b>), PANC-1 (<b>C</b>) and YAPC (<b>D</b>) cells were treated with different concentration of cisplatin (0.1–200 µM). Cell viability was measured with sulforhodamine B (SRB) colorimetric assay, after 12, 24, 48, or 72 h. Data are the means ± standard deviation (SD) of five independent experiments with eight replicates in each and are presented as percent of control at the corresponding time point, with the control set as 100% ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA followed by Bonferroni/Dunn post hoc tests. The dashed lines indicate the IC50 values.</p>
Full article ">Figure 2
<p>Analysis of mitochondrial membrane potential using the cationic dye JC-1 in BxPC-3 (<b>A</b>) and (<b>B</b>) YAPC cells. (<b>A</b>) Cells were incubated, or not, with 50 μM cisplatin for 24 hours and stained with 4′,6-diamidino-2-phenylindole (DAPI). The representative fields by confocal microscopy (magnification 40×) of one of four independent experiments are shown. (<b>B</b>) Quantification of the percentage of apoptotic nuclei was obtained using DAPI (means ± SD; n = 5). For both BxPC-3 and YAPC cells: <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA followed by Bonferroni/Dunn post hoc tests; values with shared letters are not significantly different according to Bonferroni/Dunn. (<b>C</b>,<b>D</b>) Fluorescent spectra of JC-1 in BxPC-3 and YAPC cells treated or not with 50 μM cisplatin for the indicated time. The data are means ± S.D. of five different experiments and are presented as red J-aggregates/green monomer JC-1 fluorescence ratio. Asterisks indicate values that are significantly different (<span class="html-italic">p</span> &lt; 0.05) from control at the same time point.</p>
Full article ">Figure 3
<p>The cleavage of caspase-9, caspase-7, caspase-3 and Parp-1 induced by cisplatin in BxPC-3 (<b>A</b>) and YAPC (<b>B</b>) cells. Cells were treated with 50 µM cisplatin for the indicated time, and then subjected to Western blotting. Incubation with anti-β-actin confirmed the equal protein loading. The results shown are representative of five different experiments. The histograms on the right are representative of five independent experiments and the densitometry results are expressed as the mean ± SD (n = 5) of the sum of the gray level values of the westerns. (<b>A</b>) <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA for all proteins. (<b>B</b>) <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA for caspase-9 and -7; <span class="html-italic">p</span> &gt; 0.05 by one-way ANOVA for caspase-3 and PARP-1. The values of histograms for full-length caspases and PARP with shared lower-case letters are not significantly different according to Bonferroni/Dunn post hoc tests. The values of histograms for cleaved caspases and PARP with shared capital case letters are not significantly different according to Bonferroni/Dunn post hoc tests.</p>
Full article ">Figure 4
<p>Cisplatin Induces Autophagy in BxPC-3 Cells. BxPC-3 (<b>A</b>) and YAPC (<b>B</b>) cells were treated with 50 µM cisplatin for different times. Cell lysates were analyzed using Western blotting, using specific antibodies. Sequential incubation with anti-β-actin confirmed the equal protein loading. Representative immunoblots of five experiments are depicted. The histograms on the right are representative of five independent experiments and the densitometry results are expressed as the mean ± SD (n = 5) of the sum of the gray level values of the westerns. (<b>A</b>) <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA for all proteins. (<b>B</b>) <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA for Atg3, Atg7, p-JNK1/2 and BCL2; <span class="html-italic">p</span> &lt; 0.01 by one-way ANOVA for Beclin-1 and <span class="html-italic">p</span> &gt; 0.05 by one-way ANOVA for LC3 I. The values of histograms with shared lower-case letters are not significantly different according to Bonferroni/Dunn post hoc tests. The values of histograms for LC3 II with shared capital case letters are not significantly different according to Bonferroni/Dunn post hoc tests.</p>
Full article ">Figure 5
<p>The effects of cisplatin on ERK1/2 activation. (<b>A</b>) BxPC-3 and YAPC cells were treated with 50 µM cisplatin for different times and cell lysates were analyzed by Western blotting, using activated ERK1/2 specific antibody. (<b>B</b>) Cells were pre-treated for 45 min with 25 µM PD98059 and then incubated or not with 50 µM cisplatin for 24 h. Then, cell viability was measured with sulforhodamine B colorimetric assay. (<b>C</b>) Basal expression levels of ERK1/2 in cell lines. Cell lysates were analyzed using Western blotting, using anti-phospho-ERK1/2 (p-ERK1/2) or anti-total ERK1/2 antibodies. (<b>D</b>) The relationship between the levels of phosphorylated ERK1/2 and the IC<sub>50</sub> of cisplatin in the pancreatic cell lines used. Sequential incubation with anti-β-actin confirmed the equal protein loading. Representative immunoblots of five experiments are depicted. The histograms on the right are representative of five independent experiments and the densitometry results are expressed as the mean ± SD (n = 5) of the sum of the gray level values of the westerns. A and B, <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA followed by Bonferroni/Dunn post hoc tests. Values with shared letters are not significantly different according to Bonferroni/Dunn. The lower-case letters refer to BxPC-3 cells and capital case letters to YAPC cells.</p>
Full article ">Figure 6
<p>The schematic diagram illustrates the intracellular signaling mechanisms in pancreatic cancer cells. (<b>A</b>) BxPC-3 cells, in response to cisplatin, activate key pathways such as apoptosis (pro-caspase-9, -7, -3 cleavage), as well as autophagy (LC3-I to LC3-II conversion) and ERK1/2 signaling, promoting survival. (<b>B</b>) YAPC cells with a KRAS mutation exhibit constitutive ERK1/2 pathway activation; consequently, there is a decreased activation pattern of caspase-9, caspase-7, and caspase-3, along with PARP-1 cleavage, representing apoptosis resistance. Additionally, YAPC cells do not show autophagy activation.</p>
Full article ">
19 pages, 8887 KiB  
Article
LPA3: Pharmacodynamic Differences Between Lysophosphatidic Acid and Oleoyl-Methoxy Glycerophosphothionate: Biased Agonism, Two Sites
by K. Helivier Solís, M. Teresa Romero-Ávila, Ruth Rincón-Heredia, Juan Carlos Martínez-Morales and J. Adolfo García-Sáinz
Receptors 2024, 3(4), 555-573; https://doi.org/10.3390/receptors3040029 - 20 Dec 2024
Viewed by 376
Abstract
Background: Lysophosphatidic acid (LPA) receptor 3 (LPA3) is involved in many physiological and pathophysiological actions of this bioactive lipid, particularly in cancer. The actions of LPA and oleoyl-methoxy glycerophosphothionate (OMPT) were compared in LPA3-transfected HEK 293 cells. Methods: [...] Read more.
Background: Lysophosphatidic acid (LPA) receptor 3 (LPA3) is involved in many physiological and pathophysiological actions of this bioactive lipid, particularly in cancer. The actions of LPA and oleoyl-methoxy glycerophosphothionate (OMPT) were compared in LPA3-transfected HEK 293 cells. Methods: Receptor phosphorylation, ERK 1/2 activation, LPA3-β-arrestin 2 interaction, and changes in intracellular calcium were analyzed. Results: Our data indicate that LPA and OMPT increased LPA3 phosphorylation, OMPT being considerably more potent than LPA. OMPT was also more potent than LPA to activate ERK 1/2. In contrast, OMPT was less effective in increasing intracellular calcium than LPA. The LPA-induced LPA3-β-arrestin 2 interaction was fast and robust, whereas that induced by OMPT was only detected at 60 min of incubation. LPA- and OMPT-induced receptor internalization was fast, but that induced by OMPT was more marked. LPA-induced internalization was blocked by Pitstop 2, whereas OMPT-induced receptor internalization was partially inhibited by Pitstop 2 and Filipin and entirely by the combination of both. When LPA-stimulated cells were rechallenged with 1 µM LPA, hardly any response was detected, i.e., a “refractory” state was induced. However, a conspicuous and robust response was observed if OMPT was used as the second stimulus. Conclusions: The differences in these agents’ actions suggest that OMPT is a biased agonist. These findings suggest that two binding sites for these agonists might exist in the LPA3 receptor, one showing a very high affinity for OMPT and another likely shared by LPA and OMPT (structural analogs) with lower affinity. Full article
Show Figures

Figure 1

Figure 1
<p>Concentration–response curves for LPA- and OMPT-induced LPA<sub>3</sub> receptor phosphorylation. Cells were incubated with the indicated concentrations of the agonists for 15 min. Receptor phosphorylation is expressed as the percentage of the baseline value. The means are plotted, and vertical lines indicate the SEM of 10 experiments performed on different days. Representative autoradiographs (<sup>32</sup>P) and Western blots (WBs) are presented above the graph.</p>
Full article ">Figure 2
<p>Concentration–response curves for LPA- and OMPT-induced ERK 1/2 phosphorylation. Cells were incubated with the indicated concentrations of the agonists for 2 min. ERK 1/2 phosphorylation is expressed as the percentage of the baseline value. The means are plotted, and vertical lines indicate the SEM of 6 experiments performed on different days. Representative Western blots for phosphorylated (pERK) and total (ERK) kinase are presented above the graph.</p>
Full article ">Figure 3
<p>Time course of LPA- and OMPT-induced ERK 1/2 phosphorylation. Cells were incubated for the times indicated with 1 µM of each agonist. ERK 1/2 phosphorylation is expressed as the percentage of the baseline value. The means are plotted, and vertical lines indicate the SEM of 6 experiments performed on different days. Representative Western blots for phosphorylated (pERK) and total (ERK) kinase are presented above the graph. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 LPA vs. OMPT.</p>
Full article ">Figure 4
<p>Time-course of LPA- and OMPT-induced LPA<sub>3</sub>-β-arrestin interaction (FRET). Cells were incubated for the times indicated with 1 µM LPA (black symbols and line) or 1 µM OMPT (red symbols and line). The baseline WT FRET index was considered as 100%. The means are plotted, and vertical lines indicate the SEM of 9–10 experiments performed on different days; 10–14 cells were analyzed for each experimental condition in all the experiments. Representative FRET index images are presented above the graph. Bars, 10 µm. *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline, ** <span class="html-italic">p</span> &lt; 0.005 vs. baseline, * <span class="html-italic">p</span> &lt; 0.05 vs. baseline (color coded).</p>
Full article ">Figure 5
<p>Time course of 1µM LPA- and 1µM OMPT-induced changes in intracellular (panel (<b>A</b>)) and plasma membrane (panel (<b>B</b>)) fluorescence. In both cases, data are presented as the percentage of the baseline values. The means are plotted, and vertical lines indicate the SEM of 4–5 experiments in which 10–14 images were taken for each condition. Representative images (fluorescence, confocal microscopy) are presented above the graph. Bars, 10 µm. *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline, ** <span class="html-italic">p</span> &lt; 0.005 vs. baseline, * <span class="html-italic">p</span> &lt; 0.05 vs. baseline, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 LPA vs. OMPT.</p>
Full article ">Figure 6
<p>Effect of Pitstop 2 on LPA and OMPT-induced internalization. Cells were preincubated for 15 min without (gray or pale red symbols and lines) or with Pitstop 2 (PIT) (black or bright red symbols and lines) before being stimulated with 1 µM LPA (panel (<b>A</b>)) or 1 µM OMPT (panel (<b>B</b>)). The means are plotted, and vertical lines indicate the SEM of 4–5 experiments in which 10–14 images were taken for each condition. Representative images (fluorescence, confocal microscopy) are presented above the graphs. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 LPA vs. OMPT.</p>
Full article ">Figure 7
<p>Effects of Pitstop 2 and Filipin on LPA-, OMPT-, and PMA-induced internalization. Cells were preincubated without any internalization inhibitor or with Pitstop 2 (PIT, 15 min, blue columns), Filipin (FIL, 60 min, yellow columns), or both agents (PIT + FIL, purple columns). After the preincubation, the cells were challenged with the agent and for the time indicated: vehicle (B, baseline, 5 min), 1 µM LPA (5 min), 1 µM OMPT (30 min), and 1 µM PMA (30 min). The baseline intracellular fluorescence was considered as 100%. The means are plotted, and vertical lines indicate the SEM of 5 experiments in which 10–14 images were taken for each condition. Representative images (fluorescence, confocal microscopy) are presented above the graphs. *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline, ** <span class="html-italic">p</span> &lt; 0.01 vs. baseline; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, indicated conditions.</p>
Full article ">Figure 8
<p>Cell proliferation as reflected by the MTT and crystal violet staining assays. Proliferation was studied without any agent (none) or with the following stimuli: 10% serum, 1 µM LPA, 1 µM PMA, 1 µM OMPT, or 100 ng/mL EGF. ** <span class="html-italic">p</span> &lt; 0.01 vs. none, *** <span class="html-italic">p</span> &lt; 0.001 vs. none; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, comparing indicated conditions.</p>
Full article ">Figure 9
<p>Increases in intracellular calcium in response to LPA and OMPT. Representative calcium tracings of cells incubated with distinct concentrations (color coded) of LPA (panel (<b>A</b>)) or OMPT (panel (<b>B</b>)). The concentration–response curves for LPA- and OMPT-induced intracellular calcium increases are presented in panel (<b>C</b>). The means are plotted, and vertical lines indicate the SEM of 5–8 distinct curves.</p>
Full article ">Figure 10
<p>Response to a second stimulation without or with an intermediate washing step. In the first two columns, cells were incubated with the vehicle, followed by a challenge with LPA or OMPT (control responses). In the second group of columns, cells were stimulated with the agonist indicated (first), and when the response vanished, the second stimulus was applied. In the third group of columns, after the cells were stimulated with the first agonist, they were extensively washed to eliminate the agent and rechallenged with the second stimulus. The concentration of LPA and OMPT was 1 µM in all cases. The means are plotted, and vertical lines indicate the SEM of 8–10 determination with cells from distinct cultures. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. vehicle+LPA, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. vehicle+OMPT, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.001 vs. vehicle+OMPT. Agonist stimulation was for 100 s (sec = seconds). Cell washing procedure took approximately 10 min and cells were challenged after washing.</p>
Full article ">Figure 11
<p>Representative calcium tracings of data that are presented in <a href="#receptors-03-00029-f010" class="html-fig">Figure 10</a>. Agonist stimulation was for 100 s (sec = seconds). Panels (<b>A</b>–<b>F</b>), continuous tracings without washing. Panels (<b>G</b>–<b>J</b>), cells were washed and the response to the second stimulus is shown. Cell washing procedure took approximately 10 min and cells were challenged after washing.</p>
Full article ">
23 pages, 7484 KiB  
Article
Unraveling the Mechanism of Impaired Osteogenic Differentiation in Osteoporosis: Insights from ADRB2 Gene Polymorphism
by Olga Krasnova, Julia Sopova, Anastasiia Kovaleva, Polina Semenova, Anna Zhuk, Daria Smirnova, Daria Perepletchikova, Olga Bystrova, Marina Martynova, Vitaly Karelkin, Olga Lesnyak and Irina Neganova
Cells 2024, 13(24), 2110; https://doi.org/10.3390/cells13242110 - 20 Dec 2024
Viewed by 276
Abstract
Osteoporosis is characterized by increased resorption and decreased bone formation; it is predominantly influenced by genetic factors. G-protein coupled receptors (GPCRs) play a vital role in bone homeostasis, and mutations in these genes are associated with osteoporosis. This study aimed to investigate the [...] Read more.
Osteoporosis is characterized by increased resorption and decreased bone formation; it is predominantly influenced by genetic factors. G-protein coupled receptors (GPCRs) play a vital role in bone homeostasis, and mutations in these genes are associated with osteoporosis. This study aimed to investigate the impact of single nucleotide polymorphism (SNP) rs1042713 in the ADRB2 gene, encoding the beta-2-adrenergic receptor, on osteoblastogenesis. Herein, using quantitative polymerase chain reaction, western immunoblotting, immunofluorescence assays, and flow cytometry, we examined the expression of ADRB2 and markers of bone matrix synthesis in mesenchymal stem cells (MSCs) derived from osteoporosis patient (OP-MSCs) carrying ADRB2 SNP in comparison with MSCs from healthy donor (HD-MSCs). The results showed significantly reduced ADRB2 expression in OP-MSCs at both the mRNA and protein levels, alongside decreased type 1 collagen expression, a key bone matrix component. Notably, OP-MSCs exhibited increased ERK kinase expression during differentiation, indicating sustained cell cycle progression, unlike that going to HD-MSC. These results provide novel insights into the association of ADRB2 gene polymorphisms with osteogenic differentiation. The preserved proliferative activity of OP-MSCs with rs1042713 in ADRB2 contributes to their inability to undergo effective osteogenic differentiation. This research suggests that targeting genetic factors may offer new therapeutic strategies to mitigate osteoporosis progression. Full article
Show Figures

Figure 1

Figure 1
<p>The morphology and phenotype of cells derived from the healthy donor’s and the osteoporotic patient’s bone samples. (<b>A</b>) Phase contrast representative image of cells derived from bone samples of a healthy donor and (<b>A’</b>) image of cells derived from bone samples of an osteoporotic patient; scale bar 400 μm. (<b>B</b>) Immunophenotype of cells derived from bone samples of a healthy donor and (<b>B’</b>) cells derived from bone samples of an osteoporotic patient. Cells are positive for mesenchymal stem cell markers CD90, CD105, and CD73, while they are negative for CD34 and CD45 blood cell markers.</p>
Full article ">Figure 2
<p>Expression of the beta-2-adrenergic receptor (ADRB2) in MSCs from the healthy donor and the osteoporotic patient bone samples. (<b>A</b>) Immunofluorescence analysis of ADRB2 expression in MSCs derived from the healthy donor’s bone samples (HD-MSCs) and (<b>A’</b>) MSCs from the osteoporotic patient’s bone samples (OP-MSCs), cultured in basal medium or osteogenic medium within 14 days; scale bar 50 μm. Abbreviations: ADRB2—beta-2-adrenergic receptor and DAPI—4′,6-diamidino-2-phenylindole. (<b>B</b>) Electron microscopy observation of ADRB2 in HD-MSCs and (<b>B’</b>) OP-MSCs cultured in basal or osteogenic media for 14 days; scale bar 1 μm. Red frames and arrows point at ADRB2 on cell membrane. Red asterisks point at ADRB2 within cells, distinct from the membrane. (<b>C</b>) Relative ADRB2 intensity in HD-MSCs and OP-MSCs cultured in the basal medium and osteogenic medium. Data are shown as mean ± SD, <span class="html-italic">n</span> &gt; 8, with the significant differences indicated with asterisks (ns—not significant, *—<span class="html-italic">p</span> &lt; 0.05). (<b>D</b>) The mRNA level of <span class="html-italic">ADRB2</span> in HD-MSCs and OP-MSCs under the basal medium or osteogenic medium conditions. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant differences indicated with asterisks (ns—not significant, **—<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). Abbreviations: HD—healthy donor, OP—osteoporotic patient, BM—basal medium, OM—osteogenic medium, and GAPDH—glyceraldehyde 3-phosphate dehydrogenase. (<b>E</b>) Western blot analysis of ADRB2 expression (<b>E’</b>) and the relative ADRB2 protein level in HD-MSCs and in OP-MSCs cultured in the basal medium and osteogenic medium. Full-length blots are presented in <a href="#app1-cells-13-02110" class="html-app">Supplementary Figure S1A,B</a>. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant differences indicated with asterisks (**—<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). (<b>F</b>) <span class="html-italic">RANKL</span> gene expression in HD-MSCs and in OP-MSCs under the basal medium and osteogenic medium conditions. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with the significant difference indicated with asterisks (****—<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3
<p>Osteogenic differentiation of HD-MSCs and OP-MSCs. (<b>A</b>,<b>A’</b>) <span class="html-italic">RUNX2</span>, <span class="html-italic">COL1A1</span>, <span class="html-italic">SP7</span>, <span class="html-italic">POSTN</span>, <span class="html-italic">ATF4</span>, and <span class="html-italic">BGLAP</span> gene expression in HD-MSCs and OP-MSCs after 7 days of osteogenic differentiation. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant differences indicated with asterisks (ns—not significant, *—<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). (<b>B</b>,<b>B’</b>) ALP activity after 14 days of osteogenic differentiation of HD-MSCs and OP-MSCs. (<b>C</b>,<b>C’</b>) Alizarin Red staining after 21 days of osteogenic differentiation of HD-MSCs and OP-MSCs. (<b>D</b>) mRNA level of <span class="html-italic">FN1</span>, <span class="html-italic">SPARC</span>, <span class="html-italic">CAD11</span>, and <span class="html-italic">FNDC3B</span> after the osteogenic differentiation of HD-MSCs and OP-MSCs. Abbreviations: HD—HD-MSCs; OP—OP-MSCs. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant differences indicated with asterisks (ns—not significant, *—<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 4
<p>Analysis of collagen type 1 expression. (<b>A</b>) Immunofluorescence analysis of RUNX2 (488 nm—green) and COL1A1 (561 nm—red) expression in HD-MSCs and (<b>A’</b>) OP-MSCs cultured in the basal medium and osteogenic medium for 14 days; scale bar 50 μm. Yellow frames point at RUNX2-positive and COL1A1-positive cells, which are shown at higher magnification in yellow insets. Yellow arrows point at RUNX2-positive cells; Abbreviations: DAPI—4′,6-diamidino-2-phenylindole. (<b>B</b>) relative intensity of RUNX2 and (<b>B’</b>) of COL1A1 in HD-MSCs and OP-MSCs cultured in the basal medium and osteogenic medium for 14 days. Data are shown as mean ± SD, <span class="html-italic">n</span> &gt; 8, with significant differences indicated with asterisks (ns—not significant, **—<span class="html-italic">p</span> &lt; 0.01, ****—<span class="html-italic">p</span> &lt; 0.0001). Abbreviations: HD—HD-MSCs; OP—OP-MSCs. (<b>C</b>) Western blot analysis of COL1A1 expression and (<b>C’</b>) relative COL1A1 protein level in HD-MSCs and OP-MSCs cultured in the basal medium and osteogenic medium for 14 days. Full-length blots are presented in <a href="#app1-cells-13-02110" class="html-app">Supplementary Figure S1C,D</a>. Statistical analysis of Western blot is represented in <a href="#app1-cells-13-02110" class="html-app">Supplementary Figure S2</a>. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant difference indicated with asterisks (*—<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). Abbreviations: HD—HD-MSCs; OP—OP-MSCs. BM—basal medium; OM—osteogenic medium.</p>
Full article ">Figure 5
<p>Expression of the beta-2-adrenergic receptor downstream targets and evaluating proliferation-related markers. (<b>A</b>) Expression of <span class="html-italic">GNAS</span>, <span class="html-italic">GNAI1</span>, <span class="html-italic">GNAI2</span>, and <span class="html-italic">GNAI3</span> genes in HD-MSCs and (<b>A’</b>) in OP-MSCs after osteogenic differentiation induction. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant differences indicated with asterisks (ns—not significant, *—<span class="html-italic">p</span> &lt; 0.05, **—<span class="html-italic">p</span> &lt; 0.01 and ****—<span class="html-italic">p</span> &lt; 0.0001). (<b>B</b>) Simplified scheme of ADRB2 signaling. (<b>C</b>) Western blot analysis of phospho-CREB1 (Ser133), total CREB1, phospho-ERK1/2 (Thr202/Tyr204), total ERK1/2 in HD-MSCs, and OP-MSCs cultured in the basal medium and the osteogenic medium for 14 days. Abbreviations: BM—basal medium; OM—osteogenic medium; p—phosphorylated; t—total. (<b>D</b>) Cell cycle phase distribution of HD-MSCs and (<b>D’</b>) cultured in the basal medium and the osteogenic medium for 7 days. (<b>E</b>) Western blot analysis of Cyclin A and Cyclin B1 expression in HD-MSCs and OP-MSCs cultured in the basal medium and the osteogenic medium for 14 days. Full-length blots are presented in the Figure(s) 1E–L. Abbreviations: BM—basal medium; OM—osteogenic medium. (<b>F</b>) The mRNA level of <span class="html-italic">CCNA1</span> in HD-MSCs and in OP-MSCs after osteogenic differentiation. Data are shown as mean ± SD, <span class="html-italic">n</span> = 3, with significant differences indicated with asterisks (***—<span class="html-italic">p</span> &lt; 0.001). Abbreviations: BM—basal medium; OM—osteogenic medium.</p>
Full article ">Figure 6
<p>The impact of propranolol treatment during osteogenic differentiation. (<b>A</b>) Cell cycle phase distribution of HD-MSCs and (<b>A’</b>) OP-MSCs cultured in the basal medium and the osteogenic medium with propranolol (10 μM) for 7 days. (<b>B</b>) Immunofluorescence analysis of COL1A1 (561 nm – red) expression in HD-MSCs and (<b>B’</b>) OP-MSCs cultured in the basal medium and the osteogenic medium, vehicle or propranolol (10 μM), for 14 days; scale bar 50 μm. Abbreviations: DAPI—4′,6-diamidino-2-phenylindole. (<b>C</b>) Relative COL1A1 intensity in HD-MSCs and OP-MSCs cultured in the basal medium and the osteogenic medium, vehicle or supplemented with propranolol (10 μM), for 14 days. Data are shown as mean ± SD, <span class="html-italic">n</span> &gt; 8, with the significant difference indicated with asterisks (ns—not significant, **—<span class="html-italic">p</span> &lt; 0.01). (<b>D</b>) Alizarin Red staining after 21 days of osteogenic differentiation, vehicle or with propranolol (10 μM) treatment of HD-MSCs and OP-MSCs. Abbreviations: BM—basal medium; OM—osteogenic medium; Veh—vehicle; PRO—propranolol.</p>
Full article ">
17 pages, 1305 KiB  
Article
CT-Scan-Assessed Body Composition and Its Association with Tumor Protein Expression in Endometrial Cancer: The Role of Muscle and Adiposity Quantities
by Cuthbert Mario Mahenge, Rand Talal Akasheh, Ben Kinder, Xuan Viet Nguyen, Faiza Kalam and Ting-Yuan David Cheng
Cancers 2024, 16(24), 4222; https://doi.org/10.3390/cancers16244222 - 18 Dec 2024
Viewed by 322
Abstract
Background: Endometrial cancer is strongly associated with obesity, and tumors often harbor mutations in major cancer signaling pathways. To inform the integration of body composition into targeted therapy paradigms, this hypothesis-generating study explores the association between muscle mass, body fat, and tumor [...] Read more.
Background: Endometrial cancer is strongly associated with obesity, and tumors often harbor mutations in major cancer signaling pathways. To inform the integration of body composition into targeted therapy paradigms, this hypothesis-generating study explores the association between muscle mass, body fat, and tumor proteomics. Methods: We analyzed data from 113 patients in The Cancer Genome Atlas (TCGA) and Cancer Proteomic Tumor Analysis Consortium (CPTAC) cohorts and their corresponding abdominal CT scans. Among these patients, tumor proteomics data were available for 45 patients, and 133 proteins were analyzed. Adiposity and muscle components were assessed at the L3 vertebral level on the CT scans. Patients were stratified into tertiles of muscle and fat mass and categorized into three groups: high muscle/low adiposity, high muscle/high adiposity, and low muscle/all adiposities. Linear and Cox regression models were adjusted for study cohort, stage, histology type, age, race, and ethnicity. Results: Compared with the high-muscle/low-adiposity group, both the high-muscle/high-adiposity (HR = 4.3, 95% CI = 1.0–29.0) and low-muscle (HR = 4.4, 95% CI = 1.3–14.9) groups experienced higher mortality. Low muscle was associated with higher expression of phospho-4EBP1(T37 and S65), phospho-GYS(S641) and phospho-MAPK(T202/Y204) but lower expression of ARID1A, CHK2, SYK, LCK, EEF2, CYCLIN B1, and FOXO3A. High muscle/high adiposity was associated with higher expression of phospho-4EBP1 (T37), phospho-GYS (S641), CHK1, PEA15, SMAD3, BAX, DJ1, GYS, PKM2, COMPLEX II Subunit 30, and phospho-P70S6K (T389) but with lower expression of CHK2, CRAF, MSH6, TUBERIN, PR, ERK2, beta-CATENIN, AKT, and S6. Conclusions: These findings demonstrate an association between body composition and proteins involved in key cancer signaling pathways, notably the PI3K/AKT/MTOR, MAPK/ERK, cell cycle regulation, DNA damage response, and mismatch repair pathways. These findings warrant further validation and assessment in relation to prognosis and outcomes in these patients. Full article
Show Figures

Figure 1

Figure 1
<p>Correlation between TSM (<b>A</b>) and TAT (<b>B</b>) with the BMI of the study participants.</p>
Full article ">Figure 2
<p>BMI distribution across the body composition groups of the study participants.</p>
Full article ">Figure 3
<p>Kaplan–Meier graph exploring the survival trend based on body composition group.</p>
Full article ">Figure 4
<p>Volcano plot of differential expression of protein tumors based on body composition groups with high muscle/low adiposity as a referent group.</p>
Full article ">
13 pages, 4490 KiB  
Article
The Potential Therapeutic Value of Aspirin in Anaplastic Thyroid Cancer
by Enke Baldini, Silvia Cardarelli, Eleonora Lori, Elena Bonati, Federica Gagliardi, Daniele Pironi, Poupak Fallahi, Alessandro Antonelli, Vito D’Andrea, Salvatore Ulisse and Salvatore Sorrenti
Cancers 2024, 16(24), 4203; https://doi.org/10.3390/cancers16244203 - 17 Dec 2024
Viewed by 335
Abstract
Background: several experimental findings and epidemiological observations indicated that aspirin/acetylsalicylic acid (ASA) may be endowed with anticancer effects against a variety of human malignancies, including thyroid carcinomas. Among these, undifferentiated/anaplastic thyroid carcinoma (ATC) is one of the most aggressive and lethal human cancers, [...] Read more.
Background: several experimental findings and epidemiological observations indicated that aspirin/acetylsalicylic acid (ASA) may be endowed with anticancer effects against a variety of human malignancies, including thyroid carcinomas. Among these, undifferentiated/anaplastic thyroid carcinoma (ATC) is one of the most aggressive and lethal human cancers, refractory to all currently available therapies. Methods: we here evaluated in a preclinical setting the effects of ASA on a panel of three ATC-derived cell lines: the CAL-62, the 8305C, and the 8505C. Results: the data obtained demonstrated the ability of ASA to inhibit, in a dose- and time-dependent manner, the proliferation of all ATC cell lines investigated, with IC50 values comprised between 2.0 and 4.3 mM. Cell growth was restrained with the same efficacy when the ASA treatment was applied to three-dimensional soft-agar cultures. In addition, ASA significantly reduced migration and invasion in two of the three ATC cell lines. We finally investigated the effects of ASA on the MAPK and PI3K/Akt signaling pathways, which are often altered in ATC. The results showed that the phosphorylation status of the Akt1/2/3 kinases was significantly reduced following ASA treatment, while ERK1/2 phosphorylation was either unaffected or slightly upregulated. Conclusions: our findings support epidemiological evidence on the anticancer potential of ASA. On this basis, further investigations should be carried out to assess the usefulness of ASA as adjuvant therapy in patients affected by ATC. Full article
(This article belongs to the Special Issue New and Future Focused Therapies for Thyroid Cancer)
Show Figures

Figure 1

Figure 1
<p>Dose-dependent inhibition of anaplastic thyroid cancer (ATC)-derived cell lines proliferation by ASA. Cells were treated with increasing doses of acetylsalicylic acid (ASA) (from 0.05 to 10 mM) for 72 h. Data are reported as the mean ± standard deviation (SD).</p>
Full article ">Figure 2
<p>Time-dependent inhibition of ATC-derived cell lines proliferation by ASA. Cells were seeded in 96-well plates, treated with ASA (10 mM for CAL-62, 5 mM for 8305C and 8505C), and measured at 24-h time intervals. Data are reported as the mean ± SD.</p>
Full article ">Figure 3
<p>Apoptotic effects of ASA on ATC-derived cell lines. Cells were seeded in 96-well plates and treated with ASA for 48 h. At the end of the incubation time, apoptosis was assessed using the Cell Death Detection ElisaPLUS kit to determine cytoplasmic histone-associated DNA fragments. The enrichment was calculated as the absorbance ratio between treated and non-treated cells. Bars represent the mean ± standard error (SE) of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Effects of ASA on the anchorage-independent growth of ATC cells. Cells were grown in a soft agar gel mixed with cell culture medium ± ASA (10 mM for CAL-62, 5 mM for 8305C and 8505C) for two weeks. Photos were finally acquired, and colonies having diameter ≥50 μm were counted. Bars represent the mean ± SE of three independent experiments. N.D., not detectable. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Effects of ASA on ATC cell migration in adherent cultures. Scratch areas were measured with the ImageJ software at different time intervals, and the speed of cell migration was calculated. Bars represent the mean ± SE of three independent experiments. **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Effects of ASA on ATC cell invasion. Cells were seeded onto PET membranes precoated with ECM in a serum-free medium and incubated for 12 h ± ASA at IC<sub>50</sub> concentrations. The complete medium was used as a chemoattractant. After removal of non-migrated cells, invading cells were fixed and stained with Crystal Violet. Bars represent the mean ± SE of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Phosphorylation status of Akt and MAPK in ATC-derived cell lines treated with ASA. Cells were incubated for 24 h ± ASA (10 mM for CAL-62, 5 mM for 8305C and 8505C), then protein extracts were prepared and analyzed by Western blot. Panel (<b>A</b>) Images from western blot. Panel (<b>B</b>) densitometric analyses. Bars represent the mean ± SE of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05; ***, <span class="html-italic">p</span> &lt; 0.001. Original western blots are presented in <a href="#app1-cancers-16-04203" class="html-app">File S1</a>.</p>
Full article ">
19 pages, 5110 KiB  
Article
Curcumin and Its Potential to Target the Glycolytic Behavior of Lactate-Acclimated Prostate Carcinoma Cells with Docetaxel
by Dongsic Choi, Jun Gi Lee, Su-Hak Heo, Moon-Kyen Cho, Hae-Seon Nam, Sang-Han Lee and Yoon-Jin Lee
Nutrients 2024, 16(24), 4338; https://doi.org/10.3390/nu16244338 - 16 Dec 2024
Viewed by 359
Abstract
Background: Dysregulated cellular metabolism is known to be associated with drug resistance in cancer treatment. Methods: In this study, we investigated the impact of cellular adaptation to lactic acidosis on intracellular energy metabolism and sensitivity to docetaxel in prostate carcinoma (PC) cells. The [...] Read more.
Background: Dysregulated cellular metabolism is known to be associated with drug resistance in cancer treatment. Methods: In this study, we investigated the impact of cellular adaptation to lactic acidosis on intracellular energy metabolism and sensitivity to docetaxel in prostate carcinoma (PC) cells. The effects of curcumin and the role of hexokinase 2 (HK2) in this process were also examined. Results: PC-3AcT and DU145AcT cells that preadapted to lactic acid displayed increased growth behavior, increased dependence on glycolysis, and reduced sensitivity to docetaxel compared to parental PC-3 and DU145 cells. Molecular analyses revealed activation of the c-Raf/MEK/ERK pathway, upregulation of cyclin D1, cyclin B1, and p-cdc2Thr161, and increased levels and activities of key regulatory enzymes in glycolysis, including HK2, in lactate-acclimated cells. HK2 knockdown resulted in decreased cell growth and glycolytic activity, decreased levels of complexes I–V in the mitochondrial electron transport chain, loss of mitochondrial membrane potential, and depletion of intracellular ATP, ultimately leading to cell death. In a xenograft animal model, curcumin combined with docetaxel reduced tumor size and weight, induced downregulation of glycolytic enzymes, and stimulated the upregulation of apoptotic and necroptotic proteins. This was consistent with the in vitro results from 2D monolayer and 3D spheroid cultures, suggesting that the efficacy of curcumin is not affected by docetaxel. Conclusions: Overall, our findings suggest that metabolic plasticity through enhanced glycolysis observed in lactate-acclimated PC cells may be one of the underlying causes of docetaxel resistance, and targeting glycolysis by curcumin may provide potential for drug development that could improve treatment outcomes in PC patients. Full article
(This article belongs to the Special Issue Effects of Plant Extracts on Human Health)
Show Figures

Figure 1

Figure 1
<p>Increased glycolytic flux in PC-3AcT and DU145AcT cells pre-adapted to lactic acid. Cellular responses were examined after culturing cells in DMEM containing 3.8 μM lactic acid for the indicated time (or 48 h, otherwise). (<b>A</b>) Percent cell viability. (<b>B</b>–<b>D</b>) Western blot analysis of cell cycle-regulatory (<b>B</b>), MEK/ERK signaling (<b>C</b>), and key regulatory enzymes in glycolysis (<b>D</b>). (<b>E</b>) Activities of hexokinase and pyruvate dehydrogenase. (<b>F</b>) Changes in glucose concentration in culture medium. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing respective PC-3 or DU145 cells was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. HK, hexokinase; PFKP, phosphofructokinase platelet; PDH, pyruvate dehydrogenase.</p>
Full article ">Figure 1 Cont.
<p>Increased glycolytic flux in PC-3AcT and DU145AcT cells pre-adapted to lactic acid. Cellular responses were examined after culturing cells in DMEM containing 3.8 μM lactic acid for the indicated time (or 48 h, otherwise). (<b>A</b>) Percent cell viability. (<b>B</b>–<b>D</b>) Western blot analysis of cell cycle-regulatory (<b>B</b>), MEK/ERK signaling (<b>C</b>), and key regulatory enzymes in glycolysis (<b>D</b>). (<b>E</b>) Activities of hexokinase and pyruvate dehydrogenase. (<b>F</b>) Changes in glucose concentration in culture medium. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing respective PC-3 or DU145 cells was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. HK, hexokinase; PFKP, phosphofructokinase platelet; PDH, pyruvate dehydrogenase.</p>
Full article ">Figure 2
<p>Mitochondrial localization of HK2 and effect of docetaxel treatment on PC-3AcT and DU145AcT cells. Cells were cultured in DMEM containing 3.8 μM lactic acid with or without docetaxel (40 nM) for 48 h. (<b>A</b>) Western blot analysis of HK2 in mitochondrial and cytosolic fractions. (<b>B</b>) Western blot analysis of complexes I–V in the mitochondrial electron transport chain. (<b>C</b>) Measurement of mitochondrial membrane potential after staining cells with rhodamine123. (<b>D</b>) Changes in intracellular ATP concentration. (<b>E</b>) Percent cell viability for cells treated with or without 40 nM docetaxel. (<b>F</b>) Annexin V-PE binding assay for cells treated with or without 40 nM docetaxel. (<b>G</b>) Measurements of mitochondrial membrane potential for cells treated with or without 40 nM docetaxel. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing respective PC-3 or DU145 cells was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. HK2, hexokinase 2; VDAC, voltage-dependent anion channel; NDUFB8, NADH-ubiquinone oxidoreductase subunit B8 (complex I); SDHB, succinate dehydrogenase complex iron sulfur subunit B (complex II); UQCRC2, ubiquinone-cytochrome C reductase core protein 2 (complex III); COX II, mitochondrial cytochrome C oxidase subunit II (complex IV); ATP5A, ATP synthase F1 subunit alpha (complex V); DTX, docetaxel.</p>
Full article ">Figure 2 Cont.
<p>Mitochondrial localization of HK2 and effect of docetaxel treatment on PC-3AcT and DU145AcT cells. Cells were cultured in DMEM containing 3.8 μM lactic acid with or without docetaxel (40 nM) for 48 h. (<b>A</b>) Western blot analysis of HK2 in mitochondrial and cytosolic fractions. (<b>B</b>) Western blot analysis of complexes I–V in the mitochondrial electron transport chain. (<b>C</b>) Measurement of mitochondrial membrane potential after staining cells with rhodamine123. (<b>D</b>) Changes in intracellular ATP concentration. (<b>E</b>) Percent cell viability for cells treated with or without 40 nM docetaxel. (<b>F</b>) Annexin V-PE binding assay for cells treated with or without 40 nM docetaxel. (<b>G</b>) Measurements of mitochondrial membrane potential for cells treated with or without 40 nM docetaxel. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing respective PC-3 or DU145 cells was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. HK2, hexokinase 2; VDAC, voltage-dependent anion channel; NDUFB8, NADH-ubiquinone oxidoreductase subunit B8 (complex I); SDHB, succinate dehydrogenase complex iron sulfur subunit B (complex II); UQCRC2, ubiquinone-cytochrome C reductase core protein 2 (complex III); COX II, mitochondrial cytochrome C oxidase subunit II (complex IV); ATP5A, ATP synthase F1 subunit alpha (complex V); DTX, docetaxel.</p>
Full article ">Figure 3
<p>Effect of HK2 knockdown alone or in combination with curcumin on glucose metabolism in PC-3AcT and Du145AcT cells. Cells were transfected with 10 nM HK2-targeting siRNA (siHK2) or stealth RNAi control (siC) for 24 h. They were then treated with or without curcumin (40 μM) in DMEM containing 3.8 μM lactic acid for 48 h. (<b>A</b>) Percent cell viability. (<b>B</b>) Western blot analysis of key regulatory enzymes in glycolysis. (<b>C</b>) Activities of hexokinase and pyruvate dehydrogenase. (<b>D</b>) Changes in glucose concentration in culture medium. (<b>E</b>) Western blot analysis of HK2 in mitochondrial and cytosolic fractions. The bar graph represents densitometric analysis of Western blot images normalized to β-actin. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing the respective siC group was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. CCM, curcumin; HK, hexokinase; PFKP, phosphofructokinase platelet; PDH, pyruvate dehydrogenase.</p>
Full article ">Figure 4
<p>Effects of HK2 knockdown alone or in combination with curcumin on mitochondrial function and programmed cell death in PC-3AcT and Du145AcT cells. Cells were transfected with 10 nM HK2-targeting siRNA (siHK2) or stealth RNAi control (siC) for 24 h. They were then treated with or without curcumin (40 μM) in DMEM containing 3.8 μM lactic acid for 48 h. (<b>A</b>) Western blot analysis of complexes I–V in mitochondrial electron transport chain. (<b>B</b>) Measurements of mitochondrial membrane potential after staining cells with rhodamine123. (<b>C</b>) Changes in intracellular ATP concentration. (<b>D</b>) Cell cycle analysis. (<b>E</b>) Annexin V-PE binding assay. (<b>F</b>) Western blot analysis of apoptosis- and necroptosis-related proteins. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing the respective siC group was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. CCM, curcumin; NDUFB8, NADH-ubiquinone oxidoreductase subunit B8 (complex I); SDHB, succinate dehydrogenase complex iron sulfur subunit B (complex II); UQCRC2, ubiquinone-cytochrome C reductase core protein 2 (complex III); COX II, mitochondrial cytochrome C oxidase subunit II (complex IV); ATP5A, ATP synthase F1 subunit alpha (complex V).</p>
Full article ">Figure 5
<p>Growth-inhibiting effect of co-treatment with curcumin and docetaxel. (<b>A</b>) Cell viability in 2D monolayer culture. Cells were cultured in DMEM containing 3.8 μM lactic acid with or without curcumin (40 μM) and docetaxel (40 nM) for 48 h. (<b>B</b>) Vitality staining of spheroids: from left to right: (i) phase-contrast image, (ii) fluorescent images of fluorescein diacetate(+) living cells in green, (iii) propidium iodide(+) dead cells in red, and (iv) merged; and spheroid cell viability. Spheroids were then treated with or without curcumin (40 µM) and docetaxel (40 nM) for 48 h in DMEM containing 3.8 μM lactic acid. (<b>C</b>) Representative mice, body weight, tumor volume, and tumor weight in PC-3-xenografted nude mice model. Mice (0.3–0.4 cm wide and 0.3–0.4 cm long) were injected intratumorally with vehicle or curcumin (15 mg/kg) and docetaxel (0.5 mg/kg) three times per week for 24 days. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing the respective control group was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. CCM/DTX, co-treatment with curcumin and docetaxel.</p>
Full article ">Figure 6
<p>Effects of co-treatment with curcumin and docetaxel on expression of glycolysis-, apoptosis-, and necroptosis-related key proteins in 2D monolayer, 3D spheroid cultures, and nude mice xenograft models. Proteins were extracted from cells, spheroids, and tumors described in <a href="#nutrients-16-04338-f005" class="html-fig">Figure 5</a>, separated on 4–12% NuPAGE gels, and subjected to Western blot analysis. (<b>A</b>) Expression levels of key regulatory enzymes of glycolysis. (<b>B</b>) Expression levels of proteins related to apoptosis and necroptosis. The bar graph represents densitometric analysis of Western blot images normalized to β-actin. Data are expressed as the mean ± standard deviation of three independent experiments. Statistical significance comparing the respective control group was considered at * <span class="html-italic">p</span> &lt; 0.05 using one-way ANOVA and Tukey’s post hoc correction. CCM/DTX, co-treatment with curcumin and docetaxel.</p>
Full article ">
18 pages, 13037 KiB  
Article
Bufadienolides from Chansu Injection Synergistically Enhances the Antitumor Effect of Erlotinib by Inhibiting the KRAS Pathway in Pancreatic Cancer
by Yanli Guo, Yu Jin, Jie Gao, Ding Wang, Yanming Wang, Liya Shan, Mengyu Yang, Xinzhi Li and Ketao Ma
Pharmaceuticals 2024, 17(12), 1696; https://doi.org/10.3390/ph17121696 - 16 Dec 2024
Viewed by 409
Abstract
Background and Objectives: The Chansu injection (CSI), a sterile aqueous solution derived from Chansu, is applied in clinical settings to support antitumor and anti-radiation treatments. CSI’s principal active components, bufadienolides (≥90%), demonstrate potential effects on pancreatic cancer (PDAC), but their underlying mechanisms remain [...] Read more.
Background and Objectives: The Chansu injection (CSI), a sterile aqueous solution derived from Chansu, is applied in clinical settings to support antitumor and anti-radiation treatments. CSI’s principal active components, bufadienolides (≥90%), demonstrate potential effects on pancreatic cancer (PDAC), but their underlying mechanisms remain unclear. This study aimed to elucidate the antitumor effects and pathways associated with CSI in PDAC. Methods: Network pharmacology and bioinformatics analyses explored CSI’s mechanisms against PDAC. MTT, colony-formation, and migration assays evaluated CSI’s impact on proliferation and migration in PANC-1 and MIA PACA-2 cells, both as a single agent and in combination with erlotinib (EGFR inhibitor). Cell cycle analysis employed flow cytometry. Animal experiments were performed on tumor-bearing mice, with targets and pathways assessed via molecular docking and western blotting. Results: CSI treatment suppressed PDAC cell proliferation and migration by inducing G2/M phase arrest. Network pharmacology, bioinformatics, and molecular docking indicated that CSI’s anti-PDAC effects may involve EGFR pathway modulation, with CSI lowering p-EGFR/KRAS/p-ERK1/2 pathway expressions in PDAC cells. Additionally, sustained KRAS activation in mediating erlotinib resistance in PDAC and CSI potentiated erlotinib’s antitumor effects through enhanced KRAS and p-ERK1/2 inhibition. CSI also enhanced erlotinib’s efficacy in tumor-bearing mice without causing detectable toxicity in renal, cardiac, or hepatic tissues at therapeutic doses. Conclusions: CSI as an adjuvant used in antitumor and anti-radiation therapies enhanced erlotinib’s antitumor effects through modulation of the KRAS pathway. CSI and erlotinib’s synergistic interaction represents a promising approach for addressing erlotinib resistance in PDAC treatment. Full article
Show Figures

Figure 1

Figure 1
<p>CSI inhibited the proliferation and migration of PDAC cells. (<b>A</b>) PANC-1 and MIA PACA-2 cells were treated with 0.1325–80 μg/mL of CSI for 24 h or 48 h, and cell viability was assessed using the MTT assay (n = 3). (<b>B</b>,<b>C</b>) Colony-formation assays were performed on PANC-1 and MIA PACA-2 cells treated with 2.5, 5 or 10 μg/mL CSI for 48 h (n = 3). (<b>D</b>,<b>E</b>) Migrating assays were conducted with PANC-1 and MIA PACA-2 cells exposed to 2.5, 5, or 10 μg/mL CSI for 48 h (n = 5). The same volume of sterile water or DMSO was added to the control group. Data were expressed as mean ± standard error (SEM). Statistical significance was defined as * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus the control group.</p>
Full article ">Figure 2
<p>CSI inhibited the growth of PDAC cells by inducing G2/M phase arrest. (<b>A</b>) PANC-1 and MIA PACA-2 cells were treated with 2.5, 5, or 10 μg/mL CSI for 24 h, and cell cycle distribution was analyzed using flow cytometry (n = 5). (<b>B</b>) The expression of cyclin B1 and CDK1 were determined using Western blot (n = 5). Data were expressed as mean ± standard error (SEM). Statistical significance was defined as * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus the control group.</p>
Full article ">Figure 3
<p>Network pharmacological and bioinformatics analysis of CSI against PDAC. (<b>A</b>) Overview of the network pharmacological and bioinformatics analysis for CSI against PDAC. (<b>B</b>) Volcano map and Venn diagram depicting differentially expressed genes from GSE62165, GSE91035, GSE15471, and GSE16515. (<b>C</b>) PPI network of CSI active compounds and anti-pancreatic cancer targets. (<b>D</b>,<b>E</b>) GO and KEGG enrichment analyses of 155 common targets for CSI in pancreatic cancer.</p>
Full article ">Figure 4
<p>Network pharmacological and bioinformatics analysis of CSI against PDAC. (<b>A</b>) Top 10 hub targets of CSI in pancreatic cancer. (<b>B</b>) Survival curves of patients with PDAC based on EGFR expression. (<b>C</b>) Targets of CSI interactions within the EGFR inhibitor resistance pathway. (<b>D</b>) Molecular docking analysis of CSI active compounds with EGFR, with erlotinib used as a positive control.</p>
Full article ">Figure 5
<p>CSI treatment reduced the expression of p-EGFR, KRAS, and p-ERK1/2. (<b>A</b>) PANC-1 and MIA PACA-2 cells were treated with 2.5, 5, or 10 μg/mL CSI for 24 h, and the protein expression levels of p-EGFR, EGFR, KRAS, p-ERK1/2, and ERK1/2 were analyzed using Western blot (n = 5). (<b>B</b>) Quantification of Western blot results for p-EGFR, EGFR, KRAS, p-ERK1/2, and ERK1/2 (n = 5). Data were expressed as mean ± standard error (SEM). Statistical significance was defined as * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus the control group.</p>
Full article ">Figure 6
<p>CSI synergistically enhanced the antitumor effect of erlotinib in vitro. Er (erlotinib), CSI (Chansu injection), and Er+CSI (erlotinib+Chansu injection). (<b>A</b>) The inhibitory effects of various concentrations of CSI (2.5, 5, 10 μg/mL) in combination with erlotinib (2, 4, 8 μM) on PANC-1 and MIA PACA-2 cells for 48 h were assessed (n = 5). (<b>B</b>) Colony-formation assays were conducted on PANC-1 cells and MIA PACA-2 treated with 10 μg/mL CSI, 2 μM erlotinib, or their combination for 48 h (n = 3). Data were expressed as mean ± standard error (SEM). Statistical significance was defined as ** <span class="html-italic">p</span> &lt; 0.01 versus the control group, and as ## <span class="html-italic">p</span> &lt; 0.01 relative to the erlotinib (Er) group, <span class="html-italic">ns</span> represented no significant difference.</p>
Full article ">Figure 7
<p>Cotreatment of CSI and erlotinib inhibited KRAS and p-ERK1/2 in PDAC cells. Er (erlotinib), CSI (Chansu injection), and Er+CSI (erlotinib+Chansu injection). (<b>A</b>,<b>B</b>) PANC-1 and MIA PACA-2 cells were treated with 10 μg/mL CSI, 2 μM erlotinib or their combination for 48 h, and cell cycle distribution was analyzed using flow cytometry assays (n = 5). (<b>C</b>) Western blot analysis of p-EGFR, EGFR, KRAS, p-ERK1/2, ERK1/2, and CDK1 (n = 5). Data were expressed as mean ± standard error (SEM). Statistical significance was defined as * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus the control group, as # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 relative to the erlotinib (Er) group, and <span class="html-italic">ns</span> represented no significant difference.</p>
Full article ">Figure 8
<p>CSI enhanced the antitumor effect of Er (erlotinib) in tumor-bearing mice. (<b>A</b>) Images of tumor samples from PDAC-bearing mice (n = 6). (<b>B</b>) Effects of CSI and erlotinib combination on tumor weight in tumor-bearing mice (n = 6). (<b>C</b>) Effects of CSI and erlotinib combination on tumor volume (n = 6). (<b>D</b>) Representative H&amp;E staining images of tumor, heart, liver, and kidney tissues. (<b>E</b>) Effect of CSI and erlotinib combination on body weight (n = 6). (<b>F</b>) Effect of CSI and erlotinib combination on the weight of the heart, liver, and kidney in mice (n = 6). (<b>G</b>) Representative IHC staining images of p-EGFR, KRAS, and p-ERK of tumor. Data were expressed as mean ± standard error (SEM). Statistical significance was defined as * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus the control group, and as ## <span class="html-italic">p</span> &lt; 0.01 relative to the erlotinib (Er) group.</p>
Full article ">
35 pages, 2075 KiB  
Review
Inflammatory Effects and Regulatory Mechanisms of Chitinase-3-like-1 in Multiple Human Body Systems: A Comprehensive Review
by Dong Liu, Xin Hu, Xiao Ding, Ming Li and Lei Ding
Int. J. Mol. Sci. 2024, 25(24), 13437; https://doi.org/10.3390/ijms252413437 - 15 Dec 2024
Viewed by 349
Abstract
Chitinase-3-like-1 (Chi3l1), also known as YKL-40 or BRP-39, is a highly conserved mammalian chitinase with a chitin-binding ability but no chitinase enzymatic activity. Chi3l1 is secreted by various cell types and induced by several inflammatory cytokines. It can mediate a series of cell [...] Read more.
Chitinase-3-like-1 (Chi3l1), also known as YKL-40 or BRP-39, is a highly conserved mammalian chitinase with a chitin-binding ability but no chitinase enzymatic activity. Chi3l1 is secreted by various cell types and induced by several inflammatory cytokines. It can mediate a series of cell biological processes, such as proliferation, apoptosis, migration, differentiation, and polarization. Accumulating evidence has verified that Chi3l1 is involved in diverse inflammatory conditions; however, a systematic and comprehensive understanding of the roles and mechanisms of Chi3l1 in almost all human body system-related inflammatory diseases is still lacking. The human body consists of ten organ systems, which are combinations of multiple organs that perform one or more physiological functions. Abnormalities in these human systems can trigger a series of inflammatory environments, posing serious threats to the quality of life and lifespan of humans. Therefore, exploring novel and reliable biomarkers for these diseases is highly important, with Chi3l1 being one such parameter because of its physiological and pathophysiological roles in the development of multiple inflammatory diseases. Reportedly, Chi3l1 plays an important role in diagnosing and determining disease activity/severity/prognosis related to multiple human body system inflammation disorders. Additionally, many studies have revealed the influencing factors and regulatory mechanisms (e.g., the ERK and MAPK pathways) of Chi3l1 in these inflammatory conditions, identifying potential novel therapeutic targets for these diseases. In this review, we comprehensively summarize the potential roles and underlying mechanisms of Chi3l1 in inflammatory disorders of the respiratory, digestive, circulatory, nervous, urinary, endocrine, skeletal, muscular, and reproductive systems, which provides a more systematic understanding of Chi3l1 in multiple human body system-related inflammatory diseases. Moreover, this article summarizes potential therapeutic strategies for inflammatory diseases in these systems on the basis of the revealed roles and mechanisms mediated by Chi3l1. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>Summary of inflammatory diseases associated with elevated levels of Chi3l1 in various human body systems. Chi3l1 is highly expressed and acts as a valuable biomarker in inflammatory conditions affecting the nervous, digestive, endocrine, reproductive, muscular, circulatory, respiratory, urinary, and skeletal systems. The arrow indicates the increased Chi3l1 levels.</p>
Full article ">Figure 2
<p>Summary of the regulatory mechanisms by which Chi3l1 is involved in multiple human body system diseases. Various signaling pathways are involved in the regulation of Chi3l1 upregulation-related inflammatory diseases. The cascades associated with specific diseases of human organ systems (e.g., respiratory, digestive, circulatory, skeletal, nervous, endocrine, reproductive, and muscular systems) are shown in (<b>A</b>), and the detailed diagrams are shown in (<b>B</b>).</p>
Full article ">Figure 3
<p>Summary of therapeutic strategies involving Chi3l1 for multiple human body system diseases. Various therapeutic methods are used to alleviate inflammatory diseases of the respiratory, digestive, circulatory, skeletal, nervous, and endocrine systems by downregulating Chi3l1 levels.</p>
Full article ">
19 pages, 4213 KiB  
Article
The Phenotype Changes of Astrocyte During Different Ischemia Conditions
by Fei Meng, Jing Cui, Peng Wang, Junhui Wang, Jing Sun and Liang Li
Brain Sci. 2024, 14(12), 1256; https://doi.org/10.3390/brainsci14121256 - 14 Dec 2024
Viewed by 557
Abstract
Objectives: Dementia is becoming a major health problem in the world, and chronic brain ischemia is an established important risk factor in predisposing this disease. Astrocytes, as one major part of the blood–brain barrier (BBB), are activated during chronic cerebral blood flow hypoperfusion. [...] Read more.
Objectives: Dementia is becoming a major health problem in the world, and chronic brain ischemia is an established important risk factor in predisposing this disease. Astrocytes, as one major part of the blood–brain barrier (BBB), are activated during chronic cerebral blood flow hypoperfusion. Reactive astrocytes have been classified into phenotype pro-inflammatory type A1 or neuroprotective type A2. However, the specific subtype change of astrocyte and the mechanisms of chronic brain ischemia are still unknown. Methods: In order to depict the phenotype changes and their possible roles during this process, a rat bilateral common carotid artery occlusion model (BCAO) was employed in the present study. Meanwhile, the signaling pathways that possibly regulate these changes were investigated as well. Results: After four-week occlusion, astrocytes in the cortex of BCAO rats were shown to be the A2 phenotype, identified by the significant up-regulation of S100a10 accompanied by the down-regulation of Connexin 43 (CX43) protein. Next, we established in vitro hypoxia models, which were set up by stimulating primary astrocyte cultures from rat cortex with cobalt chloride, low glucose, or/and fibrinogen. Consistent with in vivo data, the cultured astrocytes also transformed into the A2 phenotype with the up-regulation of S100a10 and the down-regulation of CX43. In order to explore the mechanism of CX43 protein changes, C6 astrocyte cells were handled in both hypoxia and low-glucose stimulus, in which decreased pERK and pJNK expression were found. Conclusions: In conclusion, our data suggest that in chronic cerebral ischemia conditions, the gradual ischemic insults could promote the transformation of astrocytes into A2 type instead of A1 type, and the phosphorylation of CX43 was negatively regulated by the phosphorylation of ERK and JNK. Also, our data could provide some new evidence of how to leverage the endogenous astrocytes phenotype changes during CNS injury by promoting them to be “protector” and not “culprit”. Full article
(This article belongs to the Section Molecular and Cellular Neuroscience)
Show Figures

Figure 1

Figure 1
<p>Cognitive performance tested using a Morris Water Maze in bilateral common carotid artery occlusion (BCAO) rats after 4 weeks of ligation. The rats were tested at a designed time for five consecutive days. (<b>A</b>) Mean escape latency, (<b>B</b>) mean swimming speed, (<b>C</b>) time in target quadrant, (<b>D</b>) number of platform crossover measured. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05 versus the control rats; ns, no significance (<span class="html-italic">n</span> = 7).</p>
Full article ">Figure 2
<p>Reactive astrocyte phenotype in the cortex of BCAO rats after 4 weeks of ligation. (<b>A</b>,<b>B</b>) The protein levels of GFAP were tested using Western blot, and the data were normalized to Gapdh blot. (<b>C</b>) A1 reactive astrocyte phenotypes were tested using Real-time Quantitative PCR. (<b>D</b>) C3 protein concentration was tested using Elisa. (<b>E</b>) A2 reactive astrocyte phenotypes were tested using QPCR. (<b>F</b>) S100a10 protein concentration was tested using Elisa. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05 versus the control rats; ns, no significance (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 3
<p>Reactive astrocyte function in the cortex of BCAO rats after 4 weeks of ligation. (<b>A</b>) Representative blots of CX43, Aqua4, and ApoE. (<b>B</b>–<b>D</b>) Ratios of CX43, Aqua4, and ApoE to Gapdh or Actin were calculated and compared. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05 versus the control rats; ns, no significance (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 4
<p>Cobalt chloride and fibrinogen induced toxicity in cultured cortical astrocytes of rats for 24 h. (<b>A</b>,<b>B</b>) Cell viability was determined using a CCK assay. (<b>A</b>) CoCl2 caused a dose-dependent effect on astrocyte viability (<span class="html-italic">n</span> = 3). (<b>B</b>) The effect of fibrinogen was not concentration-dependent or prominent (<span class="html-italic">n</span> = 9). Data were expressed as mean ± SEM, ** <span class="html-italic">p</span> &lt; 0.01 versus the control group; ns, no significance.</p>
Full article ">Figure 5
<p>Reactive phenotype in cultured cortical astrocytes of rats by different concentrations of CoCl2 and time duration. (<b>A</b>–<b>C</b>) After 24 h, the protein levels of C3 and S100a10 were tested using Western blot, and the data were normalized to Gapdh or Actin blot. (<b>D</b>–<b>F</b>) After 96 h, the protein levels of C3 and S100a10 were tested using Western blot, and the data were normalized to Gapdh or Actin blot. Data are expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05 versus the control group; ns, no significance (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Reactive phenotype in cultured cortical astrocytes of rats by cobalt chloride and low glucose for 24 h. (<b>A</b>–<b>D</b>) The protein levels of C3, S100a10, and GFAP were tested using Western blot and the data were normalized to Gapdh or Actin blot. (<b>E</b>,<b>F</b>) A1 and A2 reactive astrocyte phenotypes were tested using QPCR. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05,** <span class="html-italic">p</span> &lt; 0.01 versus the control group; ns, no significance (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 7
<p>Reactive phenotype in cultured cortical astrocytes of rats by fibrinogen for 24 h. (<b>A</b>,<b>B</b>) The protein levels of C3, S100a10, and GFAP were tested using Western blot, and the data were normalized to Gapdh or Actin blot. (<b>C</b>,<b>D</b>) A1 and A2 reactive astrocyte phenotypes were tested using QPCR. (<b>E</b>–<b>G</b>) Immunofluorescence staining of GFAP using control (<b>E</b>). Low concentration of fibrinogen and (<b>F</b>) high concentration of fibrinogen (<b>G</b>). (<b>H</b>) Analysis of cell area in immunofluorescence staining of GFAP. Data are expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus the control group; # <span class="html-italic">p</span> &lt; 0.05, high concentration versus low concentration of fibrinogen group; ns, no significance (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 8
<p>Function of reactive cultured cortical astrocytes of rats by cobalt chloride and low glucose for 72 h. (<b>A</b>) Representative blots of CX43, Aqua4, ApoE, and GFAP. (<b>B</b>–<b>E</b>) Ratios of CX43, Aqua4, ApoE, and GFAP to Actin or Gapdh were calculated and compared. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05 versus the control group; ns, no significance (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 9
<p>Effects of C6 cells by cobalt chloride or low glucose for 24 h. (<b>A</b>) Cell viability was determined using a CCK assay, and CoCl2 caused a dose-dependent effect on C6 cell viability. (<b>B</b>) Representative blots of CX43, Aqua4, and ApoE. (<b>C</b>–<b>E</b>) Ratios of CX43, Aqua4, and ApoE to Gapdh or Actin were calculated and compared. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus the control group; ns, no significance (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 10
<p>Signal pathway proteins of C6 cells by cobalt chloride or low glucose for 24 h. (<b>A</b>) Representative blots of pERK, pJNK, pAKT. (<b>B</b>–<b>D</b>) Ratios of pERK, pJNK, pAKT to ERK, JNK, and AKT were calculated and compared. Data were expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus the control group; ns, no significance (<span class="html-italic">n</span> = 3).</p>
Full article ">
15 pages, 6049 KiB  
Article
Slit1 Promotes Hypertrophic Scar Formation Through the TGF-β Signaling Pathway
by Hui Song Cui, Ya Xin Zheng, Yoon Soo Cho, Yu Mi Ro, Kibum Jeon, So Young Joo and Cheong Hoon Seo
Medicina 2024, 60(12), 2051; https://doi.org/10.3390/medicina60122051 - 12 Dec 2024
Viewed by 539
Abstract
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are [...] Read more.
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are delineated by an overproduction of the extracellular matrix (ECM) by activated fibroblasts, leading to anomalous fibrosis, which is a severe sequela of burns. However, the functionality of Slit1 in HTS formation remains unknown. We aimed to investigate whether Slit1 regulates fibroblasts through a fibrosis-related mechanism derived from post-burn HTS tissues and normal patient tissues. Methods: Human normal fibroblasts (HNFs) and hypertrophic scar fibroblasts (HTSFs) were extracted from normal skin and post-burn HTS tissues, with settings grouped according to the patient of origin. Cell proliferation was evaluated using a CellTiter-Glo Luminescent Cell Viability Assay Kit. Cell migration experiments were carried out using a μ-Dish insert system. Protein and mRNA expression levels were quantified by Western blot and quantitative real-time polymerase chain reaction. Results: We found increased expressions of Slit1 in HTS tissues and HTSFs compared to normal tissues and HNFs. The treatment of human recombinant Slit1 protein (rSlit1) within HNFs promoted cell proliferation and differentiation, leading to an upregulation in ECM components such as α-SMA, type I and III collagen, and fibronectin. The treatment of rSlit1 in HNFs facilitated cell migration, concurrent with enhanced levels of N-cadherin and vimentin, and a diminished expression of E-cadherin. Treatment with rSlit1 resulted in the phosphorylation of SMAD pathway proteins, including SMAD2, SMAD3, and SMAD1/5/8, and non-SMAD pathway proteins, including TAK1, JNK1, ERK1/2, and p38, in HNFs. Conclusions: Exogenous Slit1 potentiates the epithelial–mesenchymal transition and upregulates SMAD and non-SMAD signaling pathways in HNFs, leading to the development of HTS, suggesting that Slit1 is a promising new target for the treatment of post-burn HTS. Full article
(This article belongs to the Special Issue Burn Injuries and Burn Rehabilitation)
Show Figures

Figure 1

Figure 1
<p>Tissue morphology and expression of Slit1 in tissues and fibroblasts. (<b>A</b>) H&amp;E staining in normal skin and hypertrophic scar (HTS). The thickness of the epidermis in HTS appears to be greater than that of normal skin. The arrow marked out the epithelial layer of tissue. Images were acquired at ×10 magnification, scale bar = 50 μm. (<b>B</b>,<b>C</b>) Significantly increased levels of both mRNA and protein of Slit1 were observed in HTS tissue compared to those in normal tissues. ** <span class="html-italic">p</span> &lt; 0.01, vs. Normal. (<b>D</b>,<b>E</b>) Significantly increased levels of both mRNA and protein of Slit1 were observed in HTSFs compared to those in HNFs. HNFs and HTSFs were extracted from normal skin tissues and post-burn HTS tissues obtained from the same patients. ** <span class="html-italic">p</span> &lt; 0.01, vs. HNFs. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 2
<p>Effects of rSlit1 treatment on the proliferation and differentiation of HNFs. (<b>A</b>) Significantly increased proliferation of HNFs was observed following treatment with 10 and 100 ng/mL compared to DPBS-treated cells. (<b>B</b>,<b>C</b>) Significantly increased levels of both mRNA and protein of α-SMA (<span class="html-italic">ACTA2</span>) were observed in HNFs treated with 10 and 100 ng/mL of rSlit1 compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 3
<p>Effects of rSlit1 treatment on the expression of ECM components in HNFs. Significant increases of both mRNA and protein levels of (<b>A</b>,<b>B</b>) type Ⅰ collagen (<span class="html-italic">COL1AⅠ</span>), (<b>C</b>,<b>D</b>) type Ⅲ collagen (<span class="html-italic">COL3AⅠ</span>), and (<b>E</b>,<b>F</b>) fibronectin (<span class="html-italic">FN1</span>) were observed in HNFs treated with 10 and 100 ng/mL of rSlit1 compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 4
<p>Effects of rSlit1 treatment on the EMT phenotype of HNFs. The mRNA and protein levels exhibited significant increases in the expression of (<b>A</b>,<b>B</b>) vimentin (<span class="html-italic">VIM</span>) and (<b>C</b>,<b>D</b>) N-cadherin (<span class="html-italic">CDH2</span>), whereas a notable decrease in (<b>E</b>,<b>F</b>) E-cadherin (<span class="html-italic">CDH1</span>) expression was observed in HNFs treated with 10 and 100 ng/mL rSlit1, compared to those treated with DPBS. (<b>G</b>,<b>H</b>) Cell imaging demonstrated enhanced migration of HNFs treated with rSlit1 at concentrations of 10 and 100 ng/mL compared to the DPBS-treated controls. Enlarged images belong to the green box. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 5
<p>Effects of rSlit1 treatment on expression of SMAD signaling in HNFs. Significantly increased phosphorylated protein expression of (<b>A</b>,<b>B</b>) SMAD2, (<b>A</b>,<b>C</b>) SMAD, and (<b>A</b>,<b>D</b>) SMAD1/5/8 was observed in HNFs treated with 10 and 100 ng/mL rSlit1, compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 6
<p>Effects of rSlit1 treatment on expression of non-SMAD signaling in HNFs. Significantly increased phosphorylated protein expression of (<b>A</b>,<b>B</b>) TAK1, (<b>A</b>,<b>C</b>) JNK1, (<b>A</b>,<b>D</b>) ERK1/2, and (<b>A</b>,<b>E</b>) p38 was observed in HNFs treated with 10 and 100 ng/mL rSlit1 compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">
20 pages, 3801 KiB  
Review
Studies of Applications of Cold Plasma Systems in Cancer Treatment: Mechanisms of Oxidant Stress and Pathway Signaling
by David Durán Martínez, Adriana Valladares Méndez, Jesús Rivera Islas and Jessica Nayelli Sánchez-Carranza
Stresses 2024, 4(4), 896-915; https://doi.org/10.3390/stresses4040060 - 12 Dec 2024
Viewed by 490
Abstract
Cold atmospheric plasma (CAP) has gained attention as a non-invasive therapeutic option in oncology due to its selective cytotoxicity against cancer cells. CAP produces a complex mixture of reactive oxygen and nitrogen species (RONS), which induce oxidative stress, leading to various forms of [...] Read more.
Cold atmospheric plasma (CAP) has gained attention as a non-invasive therapeutic option in oncology due to its selective cytotoxicity against cancer cells. CAP produces a complex mixture of reactive oxygen and nitrogen species (RONS), which induce oxidative stress, leading to various forms of cell death, including apoptosis, necrosis, autophagy, and ferroptosis. These mechanisms allow CAP to target cancer cells effectively while sparing healthy tissue, making it a versatile tool in cancer treatment. This review explores the molecular pathways modulated by CAP, including PI3K/AKT, MAPK/ERK, and p53, which are crucial in the regulation of cell survival and proliferation. Additionally, in vivo, in vitro, and clinical studies supporting the efficacy of CAP are collected, providing additional evidence on its potential in oncological therapy. Full article
(This article belongs to the Collection Feature Papers in Human and Animal Stresses)
Show Figures

Figure 1

Figure 1
<p>CAP generation methods.</p>
Full article ">Figure 2
<p>Representative scheme of the effects on cells due to exposure to CAP.</p>
Full article ">Figure 3
<p>Differences in the effects of CAP applications on cancer cells and normal cells.</p>
Full article ">Figure 4
<p>Differentiated effects of CAP depending on the dose: low doses, healing; high doses, apoptosis in cancer cells.</p>
Full article ">Figure 5
<p>Representation of the types of cell death induced by CAP and the associated signaling pathways, highlighting the role of ROS and other mechanisms in different types of cancer.</p>
Full article ">
13 pages, 644 KiB  
Article
Biological Effect of Food for Special Medical Purposes (NutramilTM Complex) on Melanoma Cells in In Vitro Study
by Aneta Koronowicz, Katarzyna Krawczyk, Aleksandra Such, Ewelina Piasna-Słupecka, Mariola Drozdowska and Teresa Leszczyńska
Nutrients 2024, 16(24), 4287; https://doi.org/10.3390/nu16244287 - 12 Dec 2024
Viewed by 383
Abstract
Background/Objectives: Melanoma malignum is considered the most dangerous form of skin cancer, characterized by the exceptional resistance to many conventional chemotherapies. The aim of this study was to evaluate the effect of NutramilTM Complex (NC)—Food for Special Medical Purpose (FSMP), on two [...] Read more.
Background/Objectives: Melanoma malignum is considered the most dangerous form of skin cancer, characterized by the exceptional resistance to many conventional chemotherapies. The aim of this study was to evaluate the effect of NutramilTM Complex (NC)—Food for Special Medical Purpose (FSMP), on two types of melanoma cell lines, primary WM115 and malignant WM266-4. Methods: At 24 h after seeding, growth medium was replaced with a medium containing encoded treatments of NC or NC-CC (NutramilTM Complex without calcium caseinate) at various concentrations. Cells were treated for 24, 48, and 72 h. Results: Our results showed that NutramilTM Complex reduces proliferation of malignant melanoma WM266-4 cells but did not affect the proliferation of WM115 primary melanoma. This was followed by measured down-regulation of selected pro-survival proteins expression in WM266-4 cells, specifically ERK1/2, AKT-1, HSP27, Survivin, and TAK1. Interestingly, our results showed elevated levels of some pro-apoptotic proteins in both cell lines, including Bad, Smad2, p38MAPK, cleaved forms of Caspase-3/7, as well as cleaved PARP. Conclusions: Taken together, our results indicate that various melanoma cancer cell lines may respond in a different way to the same compound. They also suggest induction of apoptotic pathway by NutramilTM Complex as the most likely mechanism of its anticarcinogenic activity. Full article
(This article belongs to the Special Issue The Effect of Bioactive Compounds in Anti-inflammation)
Show Figures

Figure 1

Figure 1
<p>Cytotoxicity of NutramilTM Complex and NutramilTM Complex without calcium caseinate in human melanoma cell lines: (<b>A</b>) WM-115, (<b>B</b>) WM-266-4, and (<b>C</b>) BJ normal fibroblast cell line. Cells were exposed to 1–10% concentrations of Nutramil<sup>TM</sup> Complex (NC) or Nutramil<sup>TM</sup> Complex without calcium caseinate (NC-CC) for 24, 48, and 72 h. Data are presented as mean ± SD for n = 15. Statistical significance was determined by a <span class="html-italic">t</span>-test; * denotes <span class="html-italic">p</span> &lt; 0.05 compared to the untreated control (UC).</p>
Full article ">Figure 2
<p>The effect of Nutramil<sup>TM</sup> Complex and Nutramil<sup>TM</sup> Complex without calcium caseinate on proliferation in human melanoma cell lines (WM-115, WM-266-4) and normal fibroblast cell line (BJ). Cells WM-115 (<b>A</b>), WM266-4 (<b>B</b>), and BJ (<b>C</b>) were treated with Nutramil<sup>TM</sup> Complex (NC) or Nutramil<sup>TM</sup> Complex without calcium caseinate (NC-CC) at concentration 0, 3, 4% for 24, 48, and 72 h. Data are presented as mean ± SD for n = 12, normalized to the untreated control (UC) set as 100%. Statistical significance was determined by a <span class="html-italic">t</span>-test; * denotes <span class="html-italic">p</span> &lt; 0.05 compared to UC.</p>
Full article ">Figure 3
<p>The effect of Nutramil<sup>TM</sup> Complex and Nutramil<sup>TM</sup> Complex without calcium caseinate on expression of stress and apoptosis proteins in melanoma cells (WM-115, WM-266-4). Cells WM-115 (<b>A</b>) and WM266-4 (<b>B</b>) were treated for 48 h with 4% of Nutramil <sup>TM</sup> Complex (NC) or Nutramil <sup>TM</sup> Complex without calcium caseinate (NC-CC). Staurosporine (ST; 1.5 μM concentration) was used as positive control. The results are presented as mean ± SD, normalized to the internal reference protein (α-Tubulin), with the untreated control (UC) set as 100% expression. Statistical significance was determined using a <span class="html-italic">t</span>-test; * indicates <span class="html-italic">p</span> &lt; 0.05 compared to UC. Gene symbols and names: P44/42 MAPK (ERK1/2) phosphorylation (Thr202/Tyr204), Akt-1 phosphorylation (Ser473), Bad phosphorylation (Ser136), HSP27 phosphorylation (Ser82), Smad2 phosphorylation (Ser465/467), p53 phosphorylation (Ser15), p38 MAPK phosphorylation (Thr180/Tyr182), SAPK/JNK phosphorylation (Thr183/Tyr185), PARP cleavage (Asp214), Caspase-3 cleavage (Asp175), Caspase-7 cleavage (Asp198), IkB total, Chk-1 phosphorylation (Ser345), Chk-2 phosphorylation (Thr68), IkBα phosphorylation (Ser32/36), eIF2a phosphorylation (Ser51), TAK1 phosphorylation (Ser412), Survivin total.</p>
Full article ">
Back to TopTop