[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,001)

Search Parameters:
Keywords = patient-derived culture

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
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 263
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 ">
37 pages, 4688 KiB  
Review
Cell-Based Glioma Models for Anticancer Drug Screening: From Conventional Adherent Cell Cultures to Tumor-Specific Three-Dimensional Constructs
by Daria Lanskikh, Olga Kuziakova, Ivan Baklanov, Alina Penkova, Veronica Doroshenko, Ivan Buriak, Valeriia Zhmenia and Vadim Kumeiko
Cells 2024, 13(24), 2085; https://doi.org/10.3390/cells13242085 - 17 Dec 2024
Viewed by 344
Abstract
Gliomas are a group of primary brain tumors characterized by their aggressive nature and resistance to treatment. Infiltration of surrounding normal tissues limits surgical approaches, wide inter- and intratumor heterogeneity hinders the development of universal therapeutics, and the presence of the blood–brain barrier [...] Read more.
Gliomas are a group of primary brain tumors characterized by their aggressive nature and resistance to treatment. Infiltration of surrounding normal tissues limits surgical approaches, wide inter- and intratumor heterogeneity hinders the development of universal therapeutics, and the presence of the blood–brain barrier reduces the efficiency of their delivery. As a result, patients diagnosed with gliomas often face a poor prognosis and low survival rates. The spectrum of anti-glioma drugs used in clinical practice is quite narrow. Alkylating agents are often used as first-line therapy, but their effectiveness varies depending on the molecular subtypes of gliomas. This highlights the need for new, more effective therapeutic approaches. Standard drug-screening methods involve the use of two-dimensional cell cultures. However, these models cannot fully replicate the conditions present in real tumors, making it difficult to extrapolate the results to humans. We describe the advantages and disadvantages of existing glioma cell-based models designed to improve the situation and build future prospects to make drug discovery comprehensive and more effective for each patient according to personalized therapy paradigms. Full article
Show Figures

Figure 1

Figure 1
<p>Key molecular pathways, onco-associated molecules, and the microenvironment involved in gliomagenesis as prospective targets for glioma therapy that should be modeled in vitro.</p>
Full article ">Figure 2
<p>Application of the patient-derived glioma cell-based models.</p>
Full article ">Figure 3
<p>Approaches employed to create genetically modified glioma cell-based models and their subsequent applications. The left section shows the cell types most frequently utilized for modification, while the right section illustrates the various genetic and epigenetic modifications.</p>
Full article ">Figure 4
<p>Approaches for creating 3D cell-based glioma models. The left side of the figure shows the main cell sources for scaffold-free and scaffold-based models, while the right side shows the scaffold options for scaffold-based models.</p>
Full article ">
17 pages, 2849 KiB  
Article
The Role of Gene Expression Dysregulation in the Pathogenesis of Mucopolysaccharidosis: A Comparative Analysis of Shared and Specific Molecular Markers in Neuronopathic and Non-Neuronopathic Types of the Disease
by Karolina Wiśniewska, Magdalena Żabińska, Aneta Szulc, Lidia Gaffke, Grzegorz Węgrzyn and Karolina Pierzynowska
Int. J. Mol. Sci. 2024, 25(24), 13447; https://doi.org/10.3390/ijms252413447 - 15 Dec 2024
Viewed by 432
Abstract
Mucopolysaccharidosis (MPS) comprises a group of inherited metabolic diseases. Each MPS type is caused by a deficiency in the activity of one kind of enzymes involved in glycosaminoglycan (GAG) degradation, resulting from the presence of pathogenic variant(s) of the corresponding gene. All types/subtypes [...] Read more.
Mucopolysaccharidosis (MPS) comprises a group of inherited metabolic diseases. Each MPS type is caused by a deficiency in the activity of one kind of enzymes involved in glycosaminoglycan (GAG) degradation, resulting from the presence of pathogenic variant(s) of the corresponding gene. All types/subtypes of MPS, which are classified on the basis of all kinds of defective enzymes and accumulated GAG(s), are severe diseases. However, neuronopathy only occurs in some MPS types/subtypes (specifically severe forms of MPS I and MPS II, all subtypes of MPS III, and MPS VII), while in others, the symptoms related to central nervous system dysfunctions are either mild or absent. The early diagnosis of neuronopathy is important for the proper treatment and/or management of the disease; however, there are no specific markers that could be easily used for this in a clinical practice. Therefore, in this work, a comparative analysis of shared and specific gene expression alterations in neuronopathic and non-neuronopathic MPS types was performed using cultures of cells derived from patients. Using transcriptomic analyses (based on the RNA-seq method, confirmed by measuring the levels of a selected gene product), we identified genes (including PFN1, ADAMTSL1, and ABHD5) with dysregulated expression that are common for all, or almost all, types of MPS, suggesting their roles in MPS pathogenesis. Moreover, a distinct set of genes (including ARL6IP6 and PDIA3) exhibited expression changes only in neuronopathic MPS types/subtypes, but not in non-neuronopathic ones, suggesting their possible applications as biomarkers for neurodegeneration in MPS. These findings provide new insights into both the molecular mechanisms of MPS pathogenesis and the development of differentiation method(s) between neuronopathic and non-neuronopathic courses of the disease. Full article
(This article belongs to the Collection Feature Papers in Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Total number of transcripts with altered levels of expression (at FDR &lt; 0.1; <span class="html-italic">p</span> &lt; 0.1) in cells of different MPS types/subtypes relative to control cells, and those concerning jointly neuronopathic and non-neuronopathic types.</p>
Full article ">Figure 2
<p>Number of transcripts with altered levels of expression (at FDR &lt; 0.1; <span class="html-italic">p</span> &lt; 0.1) in cells of different MPS types/subtypes relative to control cells in relation to the number of MPS types where such differences occur (1 transcript in 11 MPS types, 8 transcripts in 10 MPS types, and so on).</p>
Full article ">Figure 3
<p>A heatmap presentation (created with HeatMapper software v. 2.8) of genes with altered expression levels in at least 10 MPS types/subtypes, for which the log<sub>2</sub> fold change value exceeded 2.5 or −2.5 (log<sub>2</sub>FC &gt; 2.5 or &lt;−2.5).</p>
Full article ">Figure 4
<p>Levels of profilin-1 (PFN1 protein, the <span class="html-italic">PFN1</span> gene product) in control cells and in fibroblast derived from all tested MPS types/subtypes, as assessed by automatic Western blotting (the WES system, based on capillary electrophoresis and immunoblotting conducted inside each capillary). Representative blots (<b>A</b>) (the picture prepared using a piece of software which is an integrated part of the WES—Automated Western Blots with Simple Western; ProteinSimple, San Jose, CA, USA) and (<b>B</b>) (quantification of results, i.e., mean values from three independent biological experiments with error bars representing SD) are demonstrated. In panel (<b>A</b>), the Total Protein Module (#DM-TP01, Protein Simple, San Jose, CA, USA) was used to determine the loading control. Statistically significant differences (in two-way ANOVA) relative to the control (at <span class="html-italic">p</span> &lt; 0.05) are indicated in panel (<b>B</b>) by asterisks.</p>
Full article ">Figure 5
<p>Number of transcripts with altered levels of expression (at FDR &lt; 0.1; <span class="html-italic">p</span> &lt; 0.1) in cells of different neuronopathic (<b>A</b>) and non-neuronopathic (<b>B</b>) MPS types/subtypes relative to control cells, with an indication of the number of specific transcripts altered in at least two neuronopathic (<b>A</b>) or non-neuronopathic (<b>B</b>) MPS types/subtypes.</p>
Full article ">Figure 6
<p>Number of transcripts with altered levels of expression (at FDR &lt; 0.1; <span class="html-italic">p</span> &lt; 0.1) in cells of different neuronopathic MPS types/subtypes relative to control cells in relation to the number of neuronopathic MPS types/subtypes where such differences occur (no (0) transcripts in 7 neuronopathic MPS types, 1 transcript in 6 neuronopathic MPS types, 5 transcripts in 5 neuronopathic MPS types, and so on).</p>
Full article ">Figure 7
<p>A heatmap presentation (created with HeatMapper software v. 2.8) of genes whose expression was altered compared to control cells in at least 5 neuronopathic MPS types/subtypes, without changes in expression in non-neuronopathic MPS types/subtypes.</p>
Full article ">
16 pages, 4073 KiB  
Article
A Repurposed Drug Selection Pipeline to Identify CNS-Penetrant Drug Candidates for Glioblastoma
by Ioannis Ntafoulis, Stijn L. W. Koolen, Olaf van Tellingen, Chelsea W. J. den Hollander, Hendrika Sabel-Goedknegt, Stephanie Dijkhuizen, Joost Haeck, Thom G. A. Reuvers, Peter de Bruijn, Thierry P. P. van den Bosch, Vera van Dis, Zhenyu Gao, Clemens M. F. Dirven, Sieger Leenstra and Martine L. M. Lamfers
Pharmaceuticals 2024, 17(12), 1687; https://doi.org/10.3390/ph17121687 - 14 Dec 2024
Viewed by 487
Abstract
Background: Glioblastoma is an aggressive and incurable type of brain cancer. Little progress has been made in the development of effective new therapies in the past decades. The blood–brain barrier (BBB) and drug efflux pumps, which together hamper drug delivery to these tumors, [...] Read more.
Background: Glioblastoma is an aggressive and incurable type of brain cancer. Little progress has been made in the development of effective new therapies in the past decades. The blood–brain barrier (BBB) and drug efflux pumps, which together hamper drug delivery to these tumors, play a pivotal role in the gap between promising preclinical findings and failure in clinical trials. Therefore, selecting drugs that can reach the tumor region in pharmacologically effective concentrations is of major importance. Methods: In the current study, we utilized a drug selection platform to identify candidate drugs by combining in vitro oncological drug screening data and pharmacokinetic (PK) profiles for central nervous system (CNS) penetration using the multiparameter optimization (MPO) score. Furthermore, we developed intracranial patient-derived xenograft (PDX) models that recapitulated the in situ characteristics of glioblastoma and characterized them in terms of vascular integrity, BBB permeability and expression of ATP-binding cassette (ABC) transporters. Omacetaxine mepesuccinate (OMA) was selected as a proof-of-concept drug candidate to validate our drug selection pipeline. Results: We assessed OMA’s PK profile in three different orthotopic mouse PDX models and found that OMA reaches the brain tumor tissue at concentrations ranging from 2- to 11-fold higher than in vitro IC50 values on patient-derived glioblastoma cell cultures. Conclusions: This study demonstrates that OMA, a drug selected for its in vitro anti-glioma activity and CNS- MPO score, achieves brain tumor tissue concentrations exceeding its in vitro IC50 values in patient-derived glioblastoma cell cultures, as shown in three orthotopic mouse PDX models. We emphasize the importance of such approaches at the preclinical level, highlighting both their significance and limitations in identifying compounds with potential clinical implementation in glioblastoma. Full article
(This article belongs to the Special Issue Therapeutic Agents for the Treatment of Tumors in the CNS)
Show Figures

Figure 1

Figure 1
<p>Pharmacokinetic analysis of OMA in vivo indicates accumulation in the brain tumor tissue. LC/MS/MS analysis determined the drug concentration after a single dose of 1.5 mg/kg of OMA in tumor-bearing mice. (<b>A</b>) Assessment of omacetaxine’s concentration in plasma (ng/mL), (<b>B</b>) liver (ng/g), (<b>C</b>) brain tumor tissue (ng/g) at 1, 2 and 4 h and (<b>D</b>) B/P ratio at 1, 2 and 4 h in GS832 PDX model (n = 5–10). (<b>E</b>) Assessment of drug concentration in plasma (ng/mL), (<b>F</b>) liver (ng/g) and (<b>G</b>) brain tumor tissue (ng/g) at 1 h and (<b>H</b>) B/P ratio at 1 h in GS607 PDX model (n = 4). Assessment of drug concentration in (<b>I</b>) plasma (ng/mL), (<b>J</b>) liver (ng/g) and (<b>K</b>) brain tumor tissue (ng/g) at 1 h and (<b>L</b>) B/P ratio at 1 h in GBM8 PDX model (n = 7).</p>
Full article ">Figure 2
<p>Assessment of the BBB permeability in three glioblastoma PDX models. The permeability of the BBB was evaluated by T2W, T1W pre- and post-gadolinium MRI imaging. Histological visualization of the tumor by H/E staining of mouse brain coronal slices. MRI images were obtained 70 days post-tumor implantation for GS607 and GS832-PDX, and 30 days post-implantation for GBM8. Black dashed lines indicate the tumor areas in the mouse brain slices, while red arrows denote the tumor regions in the MRI images. The number of mice used in this experiment was 3 per model.</p>
Full article ">Figure 3
<p>Characterization of the vascular integrity of the glioblastoma PDX models, GS607, GS832 and GBM8. The integrity of the vasculature was assessed by immunofluorescence staining for ZO-1 and GLUT-1, and GSC infiltration by Nestin staining. The number of mice used in this experiment varied between 3 and 6 per model. Scale bar 100 μm.</p>
Full article ">Figure 4
<p>(<b>A</b>) Assessment of P-gp and BCRP transporter’s expression in the glioblastoma PDX models by IHC staining, magnification ×20 and scale bar 100 μm. Red arrows indicate the expression of the P-gp and BCRP transporters in the mouse brain tumor areas. The number of mice used in this experiment varied between 3 to 8 per model. (<b>B</b>) Assessment of omacetaxine’s in vitro affinity for P-gp and BCRP transporters by LC/MS-MS. The graphs depict the concentration of OMA as a percentage (<span class="html-italic">y</span>-axis) in the basal and apical compartments of LLC cells expressing murine Abcb1a and human ABCB1 and MDCK cells expressing murine Abcg2 and human ABCG2, over time in hours (<span class="html-italic">x</span>-axis). Zosuquidar was added to MDCK cells to inhibit endogenous (canine) P-gp. All experiments were performed in 3 biological replicates.</p>
Full article ">Figure 4 Cont.
<p>(<b>A</b>) Assessment of P-gp and BCRP transporter’s expression in the glioblastoma PDX models by IHC staining, magnification ×20 and scale bar 100 μm. Red arrows indicate the expression of the P-gp and BCRP transporters in the mouse brain tumor areas. The number of mice used in this experiment varied between 3 to 8 per model. (<b>B</b>) Assessment of omacetaxine’s in vitro affinity for P-gp and BCRP transporters by LC/MS-MS. The graphs depict the concentration of OMA as a percentage (<span class="html-italic">y</span>-axis) in the basal and apical compartments of LLC cells expressing murine Abcb1a and human ABCB1 and MDCK cells expressing murine Abcg2 and human ABCG2, over time in hours (<span class="html-italic">x</span>-axis). Zosuquidar was added to MDCK cells to inhibit endogenous (canine) P-gp. All experiments were performed in 3 biological replicates.</p>
Full article ">
23 pages, 3853 KiB  
Article
Keratinocyte-Mediated Antigen Presentation in Psoriasis: Preliminary Insights from In Vitro Studies
by Katarzyna Zima, Dorota Purzycka-Bohdan, Aneta Szczerkowska-Dobosz and Magdalena Gabig-Cimińska
Int. J. Mol. Sci. 2024, 25(24), 13387; https://doi.org/10.3390/ijms252413387 - 13 Dec 2024
Viewed by 439
Abstract
Antigen presentation plays a critical role in the pathogenesis of immune-mediated disorders. This study aimed to investigate the effects of IFN-γ and a cytokine mix (5MIX: IL-1α, IL-17A, IL-22, OsM, and TNF-α) on the antigen-presenting capabilities of keratinocytes, with a specific focus on [...] Read more.
Antigen presentation plays a critical role in the pathogenesis of immune-mediated disorders. This study aimed to investigate the effects of IFN-γ and a cytokine mix (5MIX: IL-1α, IL-17A, IL-22, OsM, and TNF-α) on the antigen-presenting capabilities of keratinocytes, with a specific focus on immune-mediated dermatological conditions such as psoriasis (Ps). To achieve this, keratinocytes were treated with IFN-γ and 5MIX, and their impact on the expression of key antigen-presentation molecules, HLA-DRα and CD74, was assessed. Transcriptomic analysis revealed that IFN-γ alone altered the expression of 254 genes, highlighting its central role in modulating immune responses, including the recruitment of immune cells and regulation of inflammation. Temporal experiments further demonstrated that IFN-γ and 5MIX enhanced early endocytic activity and lysosomal degradation pathways, both essential for effective antigen presentation and T-cell activation. To extend these findings to a clinical context, a co-culture model using keratinocytes derived from psoriatic patients was established. This model revealed increased cytokine production following antigen stimulation, indicating robust and consistent CD4+ and naïve T-cell responses. These results elucidate the complex dynamics of cytokine signaling and antigen presentation in keratinocytes, providing insights into potential therapeutic strategies for immune-mediated skin disorders like Ps. Full article
(This article belongs to the Special Issue Immunological and Molecular Networks in the Skin and Skin Diseases)
Show Figures

Figure 1

Figure 1
<p>Elevation of HLA-DRα and CD74 expression in IFN-γ stimulated Normal Human Epidermal Keratinocytes (NHEKs) 2D in vitro model. The effect of interferon gamma (IFN-γ; 500 U/mL); 5MIX: interleukin (IL)-1α, IL-17A, IL-22, oncostatin M (OsM), and tumor necrosis factor alpha (TNF-α), at 2 ng/mL each; and the combination of 5MIX (2 ng/mL each) with IFN-γ (500 U/mL) on the levels of HLA-DRα and CD74 (<b>A</b>) proteins, in an in vitro 2D model of NHEK. The graphs represent the results of densitometric analyses normalized to β-actin, presented as a percentage of the control, non-treated cells (<b>B</b>). Representative immunofluorescence images (<b>C</b>) show the subcellular localization of HLA-DRα (green) with DAPI-stained nuclei (blue). Ten microscopic fields, each comprising 50–100 cells, were arbitrarily chosen for imaging. The scale bar equals 35.6 µm. All experiments were conducted in biological triplicates (<span class="html-italic">n</span> = 3), and comparisons with <span class="html-italic">p</span> &lt; 0.05 are displayed.</p>
Full article ">Figure 2
<p>Synergistic enhancement of ovalbumin (OVA, used as a model antigen) internalization, through colocalization with early endosomal and lysosomal markers was analyzed in Normal Human Epidermal Keratinocytes (NHEKs) treated with interferon gamma (IFN-γ; 500 U/mL) and/or 5MIX, a cytokine mix comprising interleukin (IL)-1α, IL-17A, IL-22, oncostatin M (OsM), and tumor necrosis factor alpha (TNF-α), each at 2 ng/mL, in a time-dependent study. Ten microscopic fields, each containing 50–100 cells, were arbitrarily chosen for each of the three separate experiments (<span class="html-italic">n</span> = 3), and representative images were captured. The scale bar equals 10 µm. The colocalization of OVA with the early endosome marker 1 (EEA1) and lysosomal-associated membrane protein 1 (LAMP1) was evaluated at four time points: 15 min (<b>A</b>), 30 min (<b>B</b>), 45 min (<b>C</b>), and 60 min (<b>D</b>) using an in vitro 2D NHEK model. Quantification of colocalization levels was performed using Pearson’s correlation coefficient (<b>E</b>). All experiments were conducted in biological triplicates (<span class="html-italic">n</span> = 3), and comparisons with <span class="html-italic">p</span> &lt; 0.05 are indicated.</p>
Full article ">Figure 3
<p>The transcriptomic landscape of Normal Human Epidermal Keratinocytes (NHEKs) unveils differential expression paradigms in response to 5MIX (interleukin (IL)-1α, IL-17A, IL-22, oncostatin M (OsM), and tumor necrosis factor alpha (TNF-α), at 2 ng/mL each) and/or interferon gamma (IFN-γ) at 500 U/mL stimulation. A heatmap visualization of gene expression data from NHEKs treated with 5MIX; IFN-γ at 500 U/mL; and a combination of 5MIX and IFN-γ (<b>A</b>). The intensity of color reflects the level of gene expression: deep red represents higher expression levels, while dark blue signifies lower expression levels. Panel (<b>B</b>) illustrates a Venn diagram comparing the number of differentially expressed genes (DEGs) across the three experimental conditions: 5MIX, IFN-γ, and 5MIX + IFN-γ. The numbers in each section represent the count of unique or overlapping DEGs in the different treatment groups. IFN-γ treatment resulted in 254 DEGs, 5MIX in 17 DEGs, and the combination of 5MIX + IFN-γ modulated 164 DEGs. Overlapping regions indicate genes shared among the conditions. The Venn diagram was generated using the InteractiVenn web-based tool [<a href="#B13-ijms-25-13387" class="html-bibr">13</a>]. All experiments were conducted in biological triplicates (<span class="html-italic">n</span> = 3), and comparisons with <span class="html-italic">p</span> &lt; 0.05 are displayed.</p>
Full article ">Figure 4
<p>Differential gene expression in Normal Human Epidermal Keratinocytes (NHEKs) in response to 5MIX (interleukin (IL)-1α, IL-17A, IL-22, oncostatin M (OsM), and tumor necrosis factor alpha (TNF-α), at 2 ng/mL each) and/or interferon gamma (IFN-γ) at 500 U/mL stimulation. Volcano plots illustrate the statistical significance versus the magnitude of gene expression changes in NHEKs upon treatment with 5MIX (<b>A</b>), IFN-γ (<b>B</b>), and their combination (<b>C</b>). Each point on the plots represents a gene, with the <span class="html-italic">x</span>-axis displaying the log2 fold change and the y-axis showing the negative log10 of the adjusted <span class="html-italic">p</span>-value. Genes with statistically significant differential expression are highlighted in blue and annotated with their respective gene symbols. The horizontal dashed lines represent thresholds for adjusted <span class="html-italic">p</span>-values: <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">p</span> &lt; 0.05, and <span class="html-italic">p</span> &lt; 0.10. These thresholds demarcate varying levels of statistical significance, with genes above the dashed lines meeting the respective significance criteria. All experiments were conducted in biological triplicates (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Pathway enrichment analysis of differentially expressed genes (DEGs) in Normal Human Epidermal Keratinocytes (NHEKs) treated with 5MIX (interleukin (IL)-1α, IL-17A, IL-22, oncostatin M (OsM), and tumor necrosis factor alpha (TNF-α), at 2 ng/mL each) and/or interferon gamma (IFN-γ) at 500 U/mL. Bubble plots illustrate Reactome pathway enrichment for DEGs in NHEKs upon treatment with 5MIX (<b>A</b>), IFN-γ (<b>B</b>), and their combination (<b>C</b>), compared to the control (CTRL). The x-axis shows the Reactome pathways, while the y-axis represents the significance level of enrichment (−log10 of the false discovery rate (FDR)). Bubble sizes correspond to the number of genes associated with each pathway, while the color gradient indicates FDR values, with darker blue representing higher statistical significance and lighter green reflecting less significant enrichment. All analyses were conducted using the STRING platform (<a href="https://string-db.org/" target="_blank">https://string-db.org/</a>; accessed on 23 November 2024) [<a href="#B14-ijms-25-13387" class="html-bibr">14</a>], and all experiments were performed in biological triplicates (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Treatment with 5MIX (IL-1α, IL-17A, IL-22, oncostatin M (OsM), and TNF-α at 2 ng/mL each) and/or IFN-γ at 500 U/mL, in combination with ovalbumin (OVA), significantly modulates cytokine production in naïve CD4+ and CD4+ lymphocytes. The levels of IFN-γ, IL-2, IL-10, and IL-17A cytokines in the supernatants obtained from a co-culture of Normal Human Epidermal Keratinocytes (NHEKs) and normal CD4+ lymphocytes (<b>A</b>), and IL-2, IL-10, and IL-17A in the supernatants from a co-culture of keratinocytes and normal naïve CD4+ lymphocytes (<b>B</b>). NHEKs were stimulated with a combination of 5MIX; IFN-γ; and the combination of 5MIX with IFN-γ for 24 h. Subsequently, OVA at 100 µg/mL was introduced as the antigen. After 24 h, the cultures were washed, and CD4+ lymphocytes were added for an additional 24 h. The quantification of cytokine production was performed using Luminex<sup>®</sup> assays. All experiments were conducted in biological triplicates (<span class="html-italic">n</span> = 3), and comparisons with <span class="html-italic">p</span> &lt; 0.05 are displayed.</p>
Full article ">Figure 7
<p>Biological interventions exhibit the modulation of immunological responses in psoriasis compared to the control, as evidenced by slightly reduced cytokine production following antigen stimulation. The levels of interferon gamma (IFN-γ), interleukin (IL)-2, IL-17A, IL-17F, and IL-22 in the supernatants from a co-culture of Psoriatic Human Epidermal Keratinocytes (PHEKs) and CD4+ lymphocytes (<b>A</b>), and IL-2, IL-10, and IL-17A in the supernatants from a co-culture of PHEKs and naïve CD4+ lymphocytes (<b>B</b>), measured from autologous, untreated psoriatic patients (UT), those subjected to topical treatment (TT), and those receiving biological treatment (BT). The keratinocytes were stimulated with IFN-γ (500 U/mL) for 24 h. Subsequently, ovalbumin (OVA) at 100 µg/mL was introduced as the antigen. After 24 h, the cultures were washed, and CD4+ lymphocytes were added for an additional 24 h. Cytokine production quantification was carried out using Luminex<sup>®</sup> assays. Comparisons yielding <span class="html-italic">p</span>-values less than 0.05 are presented. Statistical significance between groups (UT vs. TT and UT vs. BT) is 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). Numerical <span class="html-italic">p</span>-values are provided for comparisons within each group (before and after OVA stimulation). All experiments were conducted in biological replicates (<span class="html-italic">n</span> = 13), with <span class="html-italic">n</span> = 5 for UT, <span class="html-italic">n</span> = 4 for TT, and <span class="html-italic">n</span> = 4 for BT.</p>
Full article ">
15 pages, 2991 KiB  
Article
Elevated IL-6 Expression in Autologous Adipose-Derived Stem Cells Regulates RANKL Mediated Inflammation in Osteoarthritis
by Hyun-Joo Lee, Dae-Yong Kim, Hyeon jeong Noh, Song Yi Lee, Ji Ae Yoo, Samuel Jaeyoon Won, Yoon Sang Jeon, Ji Hoon Baek and Dong Jin Ryu
Cells 2024, 13(24), 2046; https://doi.org/10.3390/cells13242046 - 11 Dec 2024
Viewed by 362
Abstract
Interleukin-6 (IL-6) expression in mesenchymal stem cells (MSCs) has been shown to play a pivotal role in modulating cartilage regeneration and immune responses, particularly in the context of diseases that involve both degenerative processes and inflammation, such as osteoarthritis (OA). However, the precise [...] Read more.
Interleukin-6 (IL-6) expression in mesenchymal stem cells (MSCs) has been shown to play a pivotal role in modulating cartilage regeneration and immune responses, particularly in the context of diseases that involve both degenerative processes and inflammation, such as osteoarthritis (OA). However, the precise mechanism through which IL-6 and other immune-regulatory factors influence the therapeutic efficacy of autologous adipose-derived stem cells (ASCs) transplantation in OA treatment remains to be fully elucidated. This study aims to investigate the relationship between IL-6 expression in autologous ASCs isolated from OA patients and their impact on immune modulation, particularly focusing on the regulation of Receptor Activator of Nuclear factor Kappa-Β Ligand (RANKL), a key mediator of immune-driven cartilage degradation in OA. Autologous ASCs were isolated from the stromal vascular fraction (SVF) of adipose tissue obtained from 22 OA patients. The isolated ASCs were cultured and characterized using reverse transcription polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and flow cytometry to the phenotype and immune regulatory factors of MSCs. Based on IL-6 expression levels, ASCs were divided into high and low IL-6 expression groups. These groups were then co-cultured with activated peripheral blood mononuclear cells (PBMCs) to evaluate their immune-modulatory capacity, including the induction of regulatory T cells, inhibition of immune cell proliferation, and regulation of key cytokines, such as interferon-gamma (IFN-γ). Additionally, RANKL expression, a critical factor in osteoclastogenesis and cartilage degradation, was assessed in both ASC groups. High IL-6-expressing ASCs demonstrated a significantly greater capacity to inhibit immune cell proliferation and IFN-γ production compared to their low IL-6-expressing counterparts under co-culture conditions. Moreover, the group of ASCs with high IL-6 expression showed a marked reduction in RANKL expression, suggesting enhanced potential to control osteoclast activity and subsequent cartilage defect in OA. Conclusion: Autologous ASCs with elevated IL-6 expression exhibit enhanced immunomodulatory properties, particularly in regulating over-activated immune response and reducing osteoclastogenesis through RANKL suppression. These findings indicate that selecting ASCs based on IL-6 expression could enhance the therapeutic efficacy of ASC-based treatments for OA by mitigating immune-driven joint inflammation and cartilage degradation, potentially slowing disease progression. Full article
Show Figures

Figure 1

Figure 1
<p>The characterization of ASC-derived OA patients. (<b>A</b>) FACS analysis showing the expression of MSC characterization markers in representative ASCs used in this study. Green histograms represent staining with isotype-matched control antibodies, while red histograms depict the specific expression of each indicated marker. (<b>B</b>) Immunofluorescence staining for FABP4, Osteocalcin, and Aggrecan in ASCs induced to undergo adipogenic, osteogenic, and chondrogenic differentiation, respectively. DAPI was used for nuclear staining. Scale bars: FABP4, 400 μm; Osteocalcin, 200 μm; Aggrecan, 400 μm).</p>
Full article ">Figure 2
<p>Gene and protein expression analysis of ASCs related to immunoregulatory factor, IL-6, TGF-b, and CCL2. The analysis was conducted on ASCs isolated from 22 OA patients. Gene and protein expression levels are presented relative to the levels observed in ASCs isolated from patients S01 to S22. Expression of (<b>A</b>) IL-6, (<b>B</b>) TGF-β, and (<b>C</b>) CCL2 in ASCs. The left panel shows mRNA expression levels, and the right panel presents protein expression both relative to gene and protein levels in ASCs isolated from patients S01 to S22.</p>
Full article ">Figure 3
<p>Comparative analysis of IL-6 expression in ASCs from two groups of osteoarthritis (OA) patients based on IL-6 levels. Based on the IL-6 protein expression, the ASCs of OA patients were divided into two groups: ASCs of 5 patients with high IL-6 expression (ASC<sup>IL-6H</sup>) and five patients with low IL-6 expression (ASC<sup>IL-6L</sup>). (<b>A</b>) Individual IL-6 protein expression values in each patient, divided into the ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup> groups. (<b>B</b>) The average levels of protein expression for IL-6, TGF-β, and CCL2 in the ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup> groups. (<b>C</b>) Table summarizing MSC characterization marker expression for the ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup> groups. The experiments were conducted in triplicates. The results are shown as the mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Comparison of Regulatory T Cell Induction between ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. (<b>A</b>) Flow cytometry analysis of CD4<sup>+</sup>CD25<sup>+</sup>FoxP3<sup>+</sup> regulatory T cells (Tregs) under allogeneic mixed lymphocyte reaction (MLR) cultures with the addition of ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. Five ASCs from each group were cultured under MLR conditions to determine Treg inductivity. (<b>B</b>) In the presence of ASC<sup>IL-6 H</sup> and ASC<sup>IL-6L</sup>, the bars represent the median and range of results obtained from the five ASCs in each group co-cultured under MLR conditions. (<b>C</b>) Cytokine levels in the supernatant of MLR cultures, as measured by CBA assay. The levels of IFN-γ, TNF-α, IL-10, and IL-17 were quantified. The experiments were conducted in triplicates. The results are shown as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Comparison of Immunomodulatory effect between ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. PBMCs stimulated with phytohaemagglutinin (PHA) were cultured alone or in the presence of the five ASCs from each group, ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. (<b>A</b>) After 72 h of co-culture, the proliferating cells and CD25 expression were analyzed by flow cytometer. (<b>B</b>) In the presence of ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>, the bars represent the median and range of results obtained from the five ASCs in each group co-cultured with PHA-stimulated PBMCs. (<b>C</b>) The production of IFN-g, TNF-a, IL-10, and IL-17 in the supernatant of PHA stimulation was analyzed by CBA assay. The experiments were conducted in triplicates. The results are shown as the mean ± SD. * <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 6
<p>Regulatory Effect on RANKL of immune cells by IL-6 Expression in ASCs. PBMCs stimulated with cultured alone PHA or in the presence of the five ASCs from each group: ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. After 72 h of co-culture, The RANKL expression of T cells was analyzed by flow cytometer. (<b>A</b>) The gating strategy for RANKL expression of CD3<sup>+</sup> T cells under PHA stimulation. (<b>B</b>) Flow cytometry analysis for RANKL expression of CD3<sup>+</sup> T cells. (<b>C</b>) In the presence of ASC_IL-6H and ASC_IL-6L, the bars represent the median and range of results obtained from the five ASCs in each group co-cultured with PHA-stimulated PBMCs. The experiments were conducted in triplicates. The results are shown as the mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
15 pages, 2776 KiB  
Article
Obesity-Related Inflammation Reduces Treatment Sensitivity and Promotes Aggressiveness in Luminal Breast Cancer Modulating Oxidative Stress and Mitochondria
by Pere Miquel Morla-Barcelo, Lucas Melguizo-Salom, Pilar Roca, Mercedes Nadal-Serrano, Jorge Sastre-Serra and Margalida Torrens-Mas
Biomedicines 2024, 12(12), 2813; https://doi.org/10.3390/biomedicines12122813 - 11 Dec 2024
Viewed by 460
Abstract
Background: Obesity, characterized by the secretion of several pro-inflammatory cytokines and hormones, significantly increases the risk of developing breast cancer and is associated with poorer outcomes. Mitochondrial and antioxidant status are crucial in both tumor progression and treatment response. Methods: This study investigates [...] Read more.
Background: Obesity, characterized by the secretion of several pro-inflammatory cytokines and hormones, significantly increases the risk of developing breast cancer and is associated with poorer outcomes. Mitochondrial and antioxidant status are crucial in both tumor progression and treatment response. Methods: This study investigates the impact of an ELIT cocktail (17β-estradiol, leptin, IL-6, and TNFα), which simulates the obesity-related inflammation condition in postmenopausal women, using a 3D culture model. We examined the effects of ELIT exposure on mammosphere formation, oxidative stress and mitochondrial markers, and treatment sensitivity in luminal (T47D, MCF7) and triple-negative (MDA-MB-231) breast cancer cell lines. After that, 3D-derived cells were re-cultured under adherent conditions focusing on the mechanisms leading to dissemination and drug sensitivity. Results: Our results indicated that ELIT condition significantly increased mammosphere formation in luminal breast cancer cell lines (from 3.26% to 6.38% in T47D cell line and 0.68% to 2.32% in MCF7 cell line) but not in the triple-negative MDA-MB-231 cell line. Further analyses revealed a significant decrease in mitochondrial and antioxidant-related markers, particularly in the T47D cell line, where higher levels of ESR2, three-fold increased by ELIT exposure, may play a critical role. Importantly, 3D-derived T47D cells exposed to ELIT showed reduced sensitivity to tamoxifen and paclitaxel, avoiding a 34.2% and 75.1% reduction in viability, respectively. Finally, through in silico studies, we identified specific biomarkers, including TOMM20, NFE2L2, CAT, and ESR2, correlated with poor prognosis in luminal breast cancer. Conclusions: Taken together, our findings suggest that antioxidant and mitochondrial markers are key factors that reduce treatment sensitivity in obesity-related luminal breast cancer. The identified biomarkers may serve as valuable tools for the prognosis and development of more effective therapies in these patients. Full article
Show Figures

Figure 1

Figure 1
<p><b>Obesity-related inflammation induces aggressiveness in luminal breast cancer.</b> Mammospheres of T47D, MCF7, and MDA-MB-231 cell lines obtained after exposure to vehicle (CTRL) or ELIT cocktail (<b>A</b>). Mammosphere forming efficiency (MFE) of T47D, MCF7, and MDA-MB-231 cell lines under CTRL or ELIT conditions (<b>B</b>). mRNA expression levels of <span class="html-italic">CDH1</span> in mammospheres in CTRL or ELIT condition (<b>C</b>). Data are presented as mean ± SEM. Statistical significance was analyzed by Student’s <span class="html-italic">t</span>-test and set at * <span class="html-italic">p</span> ≤ 0.05. Enrichment analysis of Gene Sets related to aggressiveness in obese vs. lean luminal breast cancer patients from GSE189757 database (<b>D</b>,<b>E</b>). BMI: Body Mass Index; GSEA: Gene Set Enrichment Analysis.</p>
Full article ">Figure 2
<p><b>ELIT exposure modifies antioxidant, mitochondrial markers in luminal breast cancer mammospheres with high <span class="html-italic">ESR2</span> mRNA expression.</b> mRNA expression levels of oxidative stress-related genes (<b>A</b>), mitochondrial markers (<b>B</b>), and <span class="html-italic">ESR1</span> and <span class="html-italic">ESR2</span> (<b>C</b>) in T47D and MCF7 mammospheres under CTRL or ELIT condition. Data are presented as mean ± SEM. Statistical significance was analyzed by Student’s <span class="html-italic">t</span>-test and set at * <span class="html-italic">p</span> ≤ 0.05 and # <span class="html-italic">p</span> ≤ 0.1. UD: Undetected values.</p>
Full article ">Figure 3
<p><b>ELIT exposure decreases drug sensitivity of T47D 3D-derived cells.</b> Experimental design for T47D 3D-derived cells analysis (<b>A</b>). Cell viability (<b>B</b>,<b>C</b>) and H<sub>2</sub>O<sub>2</sub> production (<b>D</b>,<b>E</b>) in T47D 3D-derived cells under CTRL or ELIT condition after treatment with Tamoxifen or Paclitaxel, respectively. Enrichment analysis of Gene Sets related to drug response in obese compared to lean luminal breast cancer patients from GSE189757 database (<b>F</b>). ANOVA analysis was carried out, where E means ELIT effect; T means tamoxifen effect; P means Paclitaxel effect; and E*T or E*P mean interactive effect of ELIT with Tamoxifen or Paclitaxel, respectively. Data are presented as mean ± SEM. * Significant difference between cells treated with Tamoxifen or Paclitaxel and untreated cells (<span class="html-italic">p</span> ≤ 0.05). <span>$</span> Significant differences between CTRL and ELIT-exposed cells (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 4
<p><b>Obesity-related biomarkers associated with poor prognosis in luminal breast cancer.</b> Gene expression of <span class="html-italic">ESR2</span> (<b>A</b>) and mitochondrial (<b>B</b>) and oxidative stress (<b>C</b>) markers in luminal breast cancer responder and non-responder patients according to pCR after chemotherapy. Gene expression of <span class="html-italic">ESR2</span> (<b>D</b>) and mitochondrial (<b>E</b>) and oxidative stress (<b>F</b>) markers in luminal breast cancer responder and non-responder patients according to RFS after chemotherapy. Statistical significance was analyzed by Student’s <span class="html-italic">t</span>-test and set at <span class="html-italic">p</span> ≤ 0.05 (highlighted values).</p>
Full article ">
17 pages, 3918 KiB  
Article
Long-Term Aerobic Exercise Enhances Hepatoprotection in MAFLD by Modulating Exosomal miR-324 via ROCK1
by Yang Zhang, Qiangman Wei, Xue Geng and Guoliang Fang
Metabolites 2024, 14(12), 692; https://doi.org/10.3390/metabo14120692 - 9 Dec 2024
Viewed by 516
Abstract
Background: Insulin resistance (IR) is central to the progression of non-alcoholic fatty liver disease (MAFLD). While aerobic exercise reduces hepatic fat and enhances insulin sensitivity, the specific mechanisms—particularly those involving exosomal pathways—are not fully elucidated. Method: Exosomes were isolated from 15 MAFLD patients’ [...] Read more.
Background: Insulin resistance (IR) is central to the progression of non-alcoholic fatty liver disease (MAFLD). While aerobic exercise reduces hepatic fat and enhances insulin sensitivity, the specific mechanisms—particularly those involving exosomal pathways—are not fully elucidated. Method: Exosomes were isolated from 15 MAFLD patients’ plasma following the final session of a 12-week aerobic exercise intervention. Liver fat content was measured using MRI-PDFF, and metabolic parameters were assessed via OGTT, HOMA-IR, QUICKI, and VO2 max. Co-culture experiments evaluated the effects of exercise-derived exosomes on IR signaling pathways. miRNA microarray analysis identified miR-324, which was quantified in high-fat diet (HFD) mice with and without exercise and compared between athletes and sedentary controls. Functional assays assessed miR-324’s role in glucose and lipid metabolism, while luciferase reporter and Western blot assays confirmed ROCK1 as its direct target. Result: Aerobic exercise significantly reduced liver fat and improved insulin sensitivity in both MAFLD patients and HFD mice. Notably, exosomal miR-324 levels were lower in athletes than sedentary controls, indicating an inverse association with insulin sensitivity. Post-exercise, precursor and mature miR-324 increased in adipose tissue and decreased in muscle, suggesting its adipose origin and inverse regulation. Functional assays demonstrated that miR-324 modulates insulin resistance by targeting ROCK1. Conclusion: Exercise-induced exosomal miR-324 from adipose tissue targets ROCK1, revealing a novel mechanism by which aerobic exercise confers hepatoprotection against insulin resistance in MAFLD. These findings enhance our understanding of how exercise influences metabolic health and may inform future therapeutic strategies for managing MAFLD and related conditions. Full article
Show Figures

Figure 1

Figure 1
<p>MRI-PDFF effects and OGTT results of 12-week aerobic exercise on MAFLD patients. (<b>A</b>) MRI-PDFF imaging showing reduction in liver fat content after 12 weeks of aerobic exercise in MAFLD patients. (<b>B</b>) OGTT results indicating improved glucose tolerance in MAFLD patients post-training.</p>
Full article ">Figure 2
<p>(<b>a</b>) pet imaging with [<sup>18</sup>F]-ftha showing reduced fat accumulation in the HFD + exe group. (<b>b</b>) liver histology (H&amp;E, oil red o, and Masson’s staining) showing decreased lipid deposition and fibrosis in the HFD + exe group. (<b>c</b>–<b>e</b>) weights of liver (<b>c</b>), inguinal fat (iwat) (<b>d</b>), and epididymal fat (ewat) (<b>e</b>) reduced in the HFD + exe group. (<b>f</b>–<b>h</b>) improved liver triglycerides (tg) (<b>f</b>), ast (<b>g</b>), and alt (<b>h</b>) in the HFD + exe group. (<b>i</b>) increased liver insulin signaling (pAKT and pgsk3) in the HFD + exe group.</p>
Full article ">Figure 3
<p>Characterization and functional validation of plasma-derived exosomes in exercise-conditioned individuals. (<b>A</b>–<b>D</b>) Exosomal morphology, surface markers, and concentration, confirming the purity and consistency of exosome isolation. (<b>E</b>–<b>H</b>) Uptake of exosomes by HepG2 cells and improvements in glucose uptake, glycogen, and triglyceride levels, indicating potential insulin resistance mitigation. (<b>I</b>) Western blot showing enhanced insulin signaling in cells treated with exercise-derived exosomes.</p>
Full article ">Figure 4
<p>MiR-324-5p as a key exosomal miRNA regulating glucose and lipid metabolism. (<b>A</b>,<b>B</b>) Heatmap and relative expression of miRNAs, identifying miR-324-5p as significantly upregulated in exercise-derived exosomes. (<b>C</b>,<b>D</b>) Elevated miR-324-5p in high-glucose/high-fat-treated cells and MAFLD patients. (<b>E</b>–<b>J</b>) Functional effects of miR-324-5p overexpression and inhibition on glycogen and triglyceride content in HepG2 cells, demonstrating its role in metabolism regulation. <span class="html-italic">p</span> &lt; 0.01 (**), <span class="html-italic">p</span> &lt; 0.001 (***), and <span class="html-italic">p</span> &lt; 0.0001 (****). All statistical analyses were conducted using GraphPad Prism version 5.0, with <span class="html-italic">p</span> &lt; 0.05 considered statistically significant.</p>
Full article ">Figure 5
<p>MiR-324-5p targets ROCK1 to modulate insulin signaling pathways. (<b>a</b>–<b>c</b>) miR-324-5p binding to ROCK1 3′UTR confirmed by luciferase assay, identifying ROCK1 as a direct target. (<b>d</b>,<b>e</b>) Upregulation of ACC and PEPCK with miR-324-5p overexpression. (<b>f</b>–<b>h</b>) Western blot analysis showing ROCK1 downregulation, enhanced insulin signaling, and similar effects in ROCK1 siRNA-treated cells. <span class="html-italic">p</span> &lt; 0.05 (*). All statistical analyses were conducted using GraphPad Prism version 5.0, with <span class="html-italic">p</span> &lt; 0.05 considered statistically significant.</p>
Full article ">Figure 6
<p>Adipose tissue as the primary source of exercise-induced exosomal miR-324-5p. (<b>a</b>,<b>b</b>) Relative levels of pre- and mature miR-324 in liver, muscle, and adipose tissue, with significant post-exercise increases in adipose tissue, suggesting it as the primary source.</p>
Full article ">Figure 7
<p>Elevated exosomal miR-324-5p levels in elite athletes compared to sedentary controls. Plasma levels of exosomal miR-324-5p in athletes vs. sedentary controls, showing significantly higher levels in athletes, indicating that sustained physical training may upregulate miR-324-5p expression.</p>
Full article ">
20 pages, 5622 KiB  
Article
N-Glycoside of Indolo[2,3-a]pyrrolo[3,4-c]carbazole LCS1269 Exerts Anti-Glioblastoma Effects by G2 Cell Cycle Arrest and CDK1 Activity Modulation: Molecular Docking Studies, Biological Investigations, and ADMET Prediction
by Nikolay Kalitin, Natalia Koroleva, Anna Lushnikova, Maria Babaeva, Nadezhda Samoylenkova, Ekaterina Savchenko, Galina Smirnova, Yulia Borisova, Alexander Kostarev, Aida Karamysheva and Galina Pavlova
Pharmaceuticals 2024, 17(12), 1642; https://doi.org/10.3390/ph17121642 - 6 Dec 2024
Viewed by 453
Abstract
Background/Objectives: Indolo[2,3-a]pyrrolo[3,4-c]carbazole scaffold is successfully used as an efficient structural motif for the design and development of different antitumor agents. In this study, we investigated the anti-glioblastoma therapeutic potential of glycosylated indolocarbazole analog LCS1269 utilizing in vitro, in vivo, [...] Read more.
Background/Objectives: Indolo[2,3-a]pyrrolo[3,4-c]carbazole scaffold is successfully used as an efficient structural motif for the design and development of different antitumor agents. In this study, we investigated the anti-glioblastoma therapeutic potential of glycosylated indolocarbazole analog LCS1269 utilizing in vitro, in vivo, and in silico approaches. Methods: Cell viability was estimated by an MTT assay. The distribution of cell cycle phases was monitored using flow cytometry. Mitotic figures were visualized by fluorescence microscopy. Quantitative RT-PCR was used to evaluate the gene expression. The protein expression was assessed by Western blotting. Molecular docking and computational ADMET were approved for the probable protein target simulations and predicted pharmacological assessments, respectively. Results: Our findings clearly suggest that LCS1269 displayed a significant cytotoxic effect against diverse glioblastoma cell lines and patient-derived glioblastoma cultures as well as strongly suppressed xenograft growth in nude mice. LCS1269 exhibited more potent anti-proliferative activity toward glioblastoma cell lines and patient-derived glioblastoma cultures compared to conventional drug temozolomide. We further demonstrated that LCS1269 treatment caused the severe G2 phase arrest of cell cycle in a dose-dependent manner. Mechanistically, we proposed that LCS1269 could affect the CDK1 activity both by targeting active site of this enzyme and indirectly, in particular through the modulation of the Wee1/Myt1 and FOXM1/Plk1 signaling pathways, and via p21 up-regulation. LCS1269 also showed favorable pharmacological characteristics in in silico ADME prediction in comparison with staurosporine, rebeccamycin, and becatecarin as reference drugs. Conclusions: Further investigations of LCS1269 as an anti-glioblastoma medicinal agent could be very promising. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Molecular structures of (<b>A</b>) LCS1269, (<b>B</b>) Staurosporine, (<b>C</b>) Rebeccamycin, and (<b>D</b>) Becatecarin.</p>
Full article ">Figure 2
<p>Inhibitory effect of LCS1269 on cell viability of human glioblastoma cell lines and patient-derived glioblastoma cell cultures. (<b>A</b>–<b>C</b>) The viability of three glioblastoma cell lines treated with the indicated concentrations of LCS1269 for 72 h. (<b>D</b>) IC<sub>50</sub> values of LCS1269 for U87, U251, and T98G cell lines. (<b>E</b>–<b>G</b>) LCS1269 treatment for 72 h reduces the viability of three patient-derived glioblastoma cell cultures in a dose-dependent manner. (<b>H</b>) IC<sub>50</sub> values of LCS1269 for Gbl1, Gbl2, and Gbl3 cell cultures. Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 as compared with the untreated cells.</p>
Full article ">Figure 3
<p>Inhibitory effect of conventional anti-glioblastoma drug temozolomide on cell viability of human glioblastoma cell lines and patient-derived glioblastoma cell cultures and therapeutic potency comparison of temozolomide and LCS1269. (<b>A</b>–<b>C</b>) The viability of three glioblastoma cell lines treated with the indicated concentrations of temozolomide for 72 h. (<b>D</b>) IC<sub>50</sub> values comparison of temozolomide and LCS1269 for U87, U251, and T98G cell lines. (<b>E</b>–<b>G</b>) temozolomide treatment for 72 h reduces the viability of three patient-derived glioblastoma cell cultures in a dose-dependent manner. (<b>H</b>) IC<sub>50</sub> values comparison of temozolomide and LCS1269 for Gbl1, Gbl2, and Gbl3 cell cultures. Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 as compared with the untreated cells.</p>
Full article ">Figure 4
<p>Antitumor efficacy of LCS1269 against xenografts in U87-bearing nude mice (<span class="html-italic">n</span> = 5). (<b>A</b>) Representative image of xenograft-bearing animals (<b>upper panel</b>) and extirpated tumors (<b>lower panel</b>) from control group. (<b>B</b>) Representative image of xenograft-bearing animals (<b>upper panel</b>) and extirpated tumors (<b>lower panel</b>) from LCS1269-treated group. (<b>C</b>) Body weight changes in nude mice from control and LCS1269-treated groups. (<b>D</b>) Changes in U87 xenograft volumes in control and LCS1269-treated groups of nude mice. Data are presented as mean ± SD.</p>
Full article ">Figure 5
<p>LCS1269 promotes G2 cell cycle block in glioblastoma cell lines. (<b>A</b>) Influence of LCS1269 different concentrations treatment (0.5, 1, and 2.5 µM) for 24 h on cell cycle progression using flow cytometry. (<b>B</b>) Quantification of cell cycle phase distribution after LCS1269 treatment for 24 h. (<b>C</b>) U87, U251, and T98G cells were treated with LCS1269 for 24 h, stained with Hoechst 33258 and visualized using fluorescence microscopy (scale bar = 10 µm). (<b>D</b>) Quantitative real-time PCR data of <span class="html-italic">Aurora-B</span> gene expression in glioblastoma cell lines treated with LCS1269 in the indicated concentration for 24 h. (<b>E</b>) Western blot analysis of p-Histone H3 (Ser10) protein level after LCS1269 treatment for 24 h. β-Actin served as a loading control. Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 as compared with the untreated cells.</p>
Full article ">Figure 6
<p>LCS1269 partially affects CDK1 activity by regulation of Wee1/Myt1 and FOXM1/Plk1 signaling pathways, and p21 up-regulation. Western blot analysis of CDK1, p-CDK1, cyclin B1, Myt1, p-Wee1, Cdc25C, p-Cdc25C, p21, p27, FOXM1, Plk1, and p-Plk1 protein levels in U87, U251, and T98G cells treated with LCS1269 in indicated concentrations for 24 h. β-Actin served as a loading control.</p>
Full article ">Figure 7
<p>Ligand–protein interaction modeling based on the molecular docking of LCS1269 with CDK1. (<b>A</b>) 3D cartoon binding mode, (<b>B</b>) 2D binding mode (purple arrows show hydrogen bonds while green lines demonstrate π–π stacking interactions), and (<b>C</b>) 3D binding mode (yellow dashed lines denote hydrogen bonds; pink-blue dashed lines mark π—π stacking interactions) into the active site of CDK1 (PDB ID: 4YC6).</p>
Full article ">
22 pages, 3450 KiB  
Article
Characterization of Epithelial–Mesenchymal and Neuroendocrine Differentiation States in Pancreatic and Small Cell Ovarian Tumor Cells and Their Modulation by TGF-β1 and BMP-7
by Hendrik Ungefroren, Juliane von der Ohe, Rüdiger Braun, Yola Gätje, Olha Lapshyna, Jörg Schrader, Hendrik Lehnert, Jens-Uwe Marquardt, Björn Konukiewitz and Ralf Hass
Cells 2024, 13(23), 2010; https://doi.org/10.3390/cells13232010 - 5 Dec 2024
Viewed by 586
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis, due in part to early invasion and metastasis, which in turn involves epithelial–mesenchymal transition (EMT) of the cancer cells. Prompted by the discovery that two PDAC cell lines of the quasi-mesenchymal subtype (PANC-1, MIA [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis, due in part to early invasion and metastasis, which in turn involves epithelial–mesenchymal transition (EMT) of the cancer cells. Prompted by the discovery that two PDAC cell lines of the quasi-mesenchymal subtype (PANC-1, MIA PaCa-2) exhibit neuroendocrine differentiation (NED), we asked whether NED is associated with EMT. Using real-time PCR and immunoblotting, we initially verified endogenous expressions of various NED markers, i.e., chromogranin A (CHGA), synaptophysin (SYP), somatostatin receptor 2 (SSTR2), and SSTR5 in PANC-1 and MIA PaCa-2 cells. By means of immunohistochemistry, the expressions of CHGA, SYP, SSTR2, and the EMT markers cytokeratin 7 (CK7) and vimentin could be allocated to the neoplastic ductal epithelial cells of pancreatic ducts in surgically resected tissues from patients with PDAC. In HPDE6c7 normal pancreatic duct epithelial cells and in epithelial subtype BxPC-3 PDAC cells, the expression of CHGA, SYP, and neuron-specific enolase 2 (NSE) was either undetectable or much lower than in PANC-1 and MIA PaCa-2 cells. Parental cultures of PANC-1 cells exhibit EM plasticity (EMP) and harbor clonal subpopulations with both M- and E-phenotypes. Of note, M-type clones were found to display more pronounced NED than E-type clones. Inducing EMT in parental cultures of PANC-1 cells by treatment with transforming growth factor-β1 (TGF-β1) repressed epithelial genes and co-induced mesenchymal and NED genes, except for SSTR5. Surprisingly, treatment with bone morphogenetic protein (BMP)-7 differentially affected gene expressions in PANC-1, MIA PaCa-2, BxPC-3, and HPDE cells. It synergized with TGF-β1 in the induction of vimentin, SNAIL, SSTR2, and NSE but antagonized it in the regulation of CHGA and SSTR5. Phospho-immunoblotting in M- and E-type PANC-1 clones revealed that both TGF-β1 and, surprisingly, also BMP-7 activated SMAD2 and SMAD3 and that in M- but not E-type clones BMP-7 was able to dramatically enhance the activation of SMAD3. From these data, we conclude that in EMT of PDAC cells mesenchymal and NED markers are co-regulated, and that mesenchymal–epithelial transition (MET) is associated with a loss of both the mesenchymal and NED phenotypes. Analyzing NED in another tumor type, small cell carcinoma of the ovary hypercalcemic type (SCCOHT), revealed that two model cell lines of this disease (SCCOHT-1, BIN-67) do express CDH1, SNAI1, VIM, CHGA, SYP, ENO2, and SSTR2, but that in contrast to BMP-7, none of these genes was transcriptionally regulated by TGF-β1. Likewise, in BIN-67 cells, BMP-7 was able to reduce proliferation, while in SCCOHT-1 cells this occurred only upon combined treatment with TGF-β and BMP-7. We conclude that in PDAC-derived tumor cells, NED is closely linked to EMT and TGF-β signaling, which may have implications for the therapeutic use of TGF-β inhibitors in PDAC management. Full article
(This article belongs to the Special Issue New Insights of TGF-Beta Signaling in Cancer)
Show Figures

Figure 1

Figure 1
<p>Relative expression of NED markers in PDAC-derived cell lines, PANC-1, MIA PaCa-2, and BxPC-3, and in the normal pancreatic duct epithelial cell line, HPDE. The panNET-derived cell lines, BON and NT-3, were employed here as controls. Cells were lysed at different times during continuous culture and subjected to RNA isolation and analyzed by qPCR. Expression levels for the indicated genes are displayed relative to those in PANC-1 cells, set arbitrarily at 1.0. Data represent the means ± SD of 3–6 independent preparations. Please note the extra scales for BON and NT-3 cells in the SSTR2, 5 and CHGA graphs, which indicate the orders of magnitude higher expression. Data shown are the mean ± SD of at least three independent experiments. The asterisks (∗) denote significant differences (two-tailed unpaired Student’s <span class="html-italic">t</span>-test). The asterisks above BON and NT-3 cells indicate a significant difference relative to PANC-1 cells.</p>
Full article ">Figure 2
<p>NED markers are enriched in PANC-1 single-cell-derived clones with an M-phenotype over those with an E-phenotype as indicated in parentheses in (<b>B</b>). (<b>A</b>) Seven individual clones previously grouped according to their EMT state (three E-type and four M-type clones) [<a href="#B6-cells-13-02010" class="html-bibr">6</a>] were subjected to qPCR analysis for the indicated NED markers. Data represent the ratio of mean values between M and E-type clones for each marker. At 1.0, expression is equally high in M and E-type clones (indicated by the stippled line). (<b>B</b>) Immunoblot analysis of SYP in the same clones analyzed in (<b>A</b>). For quantitative analysis, the two closely spaced bands (indicated by arrows on the right-hand side) from two clones each (harvested at different times during continuous culture) were densitometrically scanned using the program NIH image. Hence, data represent the means ± SD of four bands per individual clone. Significant differences between the various clones are marked by asterisks (∗).</p>
Full article ">Figure 3
<p>IHC of the indicated NED and EMT markers in human PDAC specimens, primary tumors and a liver metastasis. CHGA, SSTR2, and SYP are primarily, and CK7 exclusively, expressed in ductal epithelial tumor cells lining the pancreatic ducts, while VIM is present in tumor cells lining the pancreatic ducts (arrows) as well as in stromal fibroblasts (considered as an internal control). Representative images are shown. For negative controls, see <a href="#app1-cells-13-02010" class="html-app">Supplementary Figure S3</a>. The scalebar represents 100 µm.</p>
Full article ">Figure 4
<p>Effect of treatment with TGF-β1 or BMP-7 on EMT and NED markers in parental PANC-1 cells. Cells were treated with TGF-β1 (5 ng/mL), BMP-7 (200 ng/mL), or vehicle (Ctrl), either singly or in combination, for 24 h and subsequently subjected to qPCR (<b>A</b>) or immunoblot (<b>B</b>) analysis of the indicated EMT- and NED-associated genes. (<b>C</b>) As in (<b>A</b>), only NED-associated markers were detected. (<b>D</b>) Five single-cell-derived clones were treated with TGF-β1 (5 ng/mL) for 24 h and subjected to qPCR analysis for the indicated markers. The graphs below the blots in (<b>B</b>) display the results of densitometry-based quantification of band intensities. Data shown (mean ± SD of three parallel wells) are each from a representative experiment out of at least three experiments performed in total. The asterisks (∗) denote significant differences (two-tailed unpaired Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 5
<p>Effect of treatment with TGF-β1 or BMP-7 on EMT and NED markers in MIA PaCa-2 cells. Cells were treated with 5 ng/mL of TGF-β1, 200 ng/mL of BMP-7, or vehicle (Ctrl) either singly, or with both growth factors simultaneously, for 24 h and subjected to qPCR (<b>A</b>) or immunoblot (<b>B</b>) analysis of the indicated EMT- and NED-associated markers. The graphs below the blots in (<b>B</b>) display the results of densitometric band quantification. Data shown (mean ± SD of three parallel wells) are from a representative experiment out of at least three experiments performed in total. The asterisks (∗) denote significant differences (two-tailed unpaired Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>Effect of treatment with TGF-β1 or BMP-7 on the activation of SMAD3 and SMAD2 in PANC-1 clones P1C3 (M-type) and P3D2 (E-type). Cells (3 wells each) were treated for 1 h with either vehicle (Control, C), TGF-β1 (T, 10 ng/mL), BMP-7 (B, 200 ng/mL), either singly or in combination (TGF-β1+BMP-7, T+B), lysed, and subjected to phospho-immunoblotting of SMAD3 (upper panels) and SMAD2 (lower panels). The graphs below the blots display the results of densitometric signal quantification. Data shown (mean ± SD of three parallel wells) are from a representative experiment out of three experiments performed in total. The asterisks (∗) denote significant differences (two-tailed unpaired Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 7
<p>Expression of EMT and NED markers in response to MET induction via TDC-IIT. Parental PANC-1 (upper graphs) and MIA PaCa-2 (lower graphs) cells were subjected to TDC-IIT or control culture (Ctrl) for 72 h followed by lysis and qPCR analysis of markers of EMT (<b>A</b>) or NED (<b>B</b>). The data represent the mean ± SD of quadruplicate wells of a representative assay out of at least three assays performed in total. The asterisks (∗) denote significant differences relative to Ctrl.</p>
Full article ">Figure 8
<p>Expression of EMT and NED markers in response to TGF-β1 and BMP-7 in SCCOHT. SCCOHT-1 cells were treated with TGF-β1 (10 ng/mL) or BMP-7 (200 ng/mL) followed by lysis and qPCR analysis of markers of EMT (A) or NED (B). The graphs represent the mean ± SD of quadruplicate wells of a representative assay out of at least three independent assays performed in total. The asterisks (∗) denote a significant difference.</p>
Full article ">Figure 9
<p>Impact of single and combined treatment with TGF-β1 and BMP-7 on functional activities in parental PANC-1, BIN-67, and SCCOHT-1 cells. (<b>A</b>) Migratory activity of parental PANC-1 cells as determined by RTCA using xCELLigence technology. PANC-1 cells received either vehicle (C) or were exposed to TGF-β1 (T, 10 ng/mL), BMP-7 (B, 200 ng/mL), or a combination of both growth factors (T+B) and ran on an xCELLigence device with CIM-plates-16 for a total of 7:45 h. Data were recorded by RTCA software and represent the mean ± SD of three parallel wells. A significant difference between the red and the green curves was first noted at the 3:25 h time point (indicated by the asterisk) and all later time points. (<b>B</b>) BIN-67 and SCCOHT-1 cells were treated for 24 h with either vehicle (C), TGF-β1 (T, 10 ng/mL), BMP-7 (B, 200 ng/mL), or a combination of both growth factors (T+B), after which cells were detached and counted. The graphs represent the mean ± SD of six independent assays (<span class="html-italic">n</span> = 6). The asterisks (∗) denote significant differences relative to C set arbitrarily to 1.00 (two-tailed unpaired Student’s <span class="html-italic">t</span>-test).</p>
Full article ">
13 pages, 2884 KiB  
Article
A High-Throughput Neurosphere-Based Colony Formation Assay to Test Drug and Radiation Sensitivity of Different Patient-Derived Glioblastoma Lines
by Manoj Kumar, Lauren C. Nassour-Caswell, Hasan Alrefai, Joshua C. Anderson, Taylor L. Schanel, Patricia H. Hicks, Rex Cardan and Christopher D. Willey
Cells 2024, 13(23), 1995; https://doi.org/10.3390/cells13231995 - 3 Dec 2024
Viewed by 571
Abstract
The gold standard assay for radiation response is the clonogenic assay, a normalized colony formation assay (CFA) that can capture a broad range of radiation-induced cell death mechanisms. Traditionally, this assay relies on two-dimensional (2D) cell culture conditions with colonies counted by fixing [...] Read more.
The gold standard assay for radiation response is the clonogenic assay, a normalized colony formation assay (CFA) that can capture a broad range of radiation-induced cell death mechanisms. Traditionally, this assay relies on two-dimensional (2D) cell culture conditions with colonies counted by fixing and staining protocols. While some groups have converted these to three-dimensional (3D) conditions, these models still utilize 2D-like media compositions containing serum that are incompatible with stem-like cell models such as brain tumor initiating cells (BTICs) that form self-aggregating spheroids in neural stem cell media. BTICs are the preferred patient-derived model system for studying glioblastoma (GBM) as they tend to better retain molecular and phenotypic characteristics of the original tumor tissue. As such, it is important that preclinical radiation studies should be adapted to BTIC conditions. In this study, we describe a series of experimental approaches for performing CFA experiments with BTIC cultures. Our results indicate that serum-free clonogenic assays are feasible for combination drug and radiation testing and may better facilitate translatability of preclinical findings. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) 39P-RT cells were seeded in the inner 60 wells of a 96-well plate at different indicated cell numbers and treated with DMSO (vehicle) or Drug A and after 2 h were irradiated with different doses of radiation. After 12 days of colony formation, colonies were stained with 0.2 μg/mL of Calcein AM (green) and imaged at 4× magnification on the Cytation 5, using 120 μm as the cutoff value for colony counting. Colony numbers are indicated in white in each well. (<b>B</b>) The semi-log clonogenic curve was plotted with standard error bars. Only the 2 Gy dose was significantly different between Drug A and vehicle (<span class="html-italic">p</span> &lt; 0.01) after analyzing two-way ANOVA. (<b>C</b>) 14P mCherry neurospheres suspension was used to seed a clear bottom 96-well black-wall plate, and after two days of incubation in a CO<sub>2</sub> incubator at 37 °C, these neurospheres were fixed with 0, 0.25, 0.47, or 0.84% PFA and then stored at 4 °C. Serial imaging was carried out using the Cytation 5 Imager from 2 h to 13 days using the 590 nm Filter/LED cube (PM 1225002), which has an excitation of 586/15 nm and an emission of 647/57 nm. Created in BioRender.com.</p>
Full article ">Figure 2
<p>(<b>A</b>) 14P NLS mCherry cells were seeded in the inner 60 wells of two 96-well plates. After 24 h, cells were treated with drug B, drug C, and their combination. After 2 h of drug treatment, one plate was irradiated with 4 Gy while the other was sham irradiated. After 12 days of colony formation, colonies were stained with 0.2 μg/mL of Calcein AM (green) and imaged at 4× magnification on the Cytation 5. Colony counts were made using a 100 μm size cutoff. (<b>B</b>) Surviving fraction was calculated for each drug/drug combination condition followed by normalization with the 4 Gy/0 μM condition (ordinary one-way ANOVA statistics are shown with SEM). (<b>C</b>) Relative colony formations are plotted with normalization only to the 0 Gy/0 μM condition (two-way ANOVA is used for statistical analysis). For (<b>B</b>,<b>C</b>), * indicates <span class="html-italic">p</span> value &lt; 0.05; ** indicates <span class="html-italic">p</span> &lt; 0.01; **** indicates <span class="html-italic">p</span> &lt; 0.0001; ‘ns’ stands for non-significant. (<b>D</b>) 14P BTIC cells were plated at 50 cells/well in 96-well plates and allowed to grow as neurospheres for 5 days. After 5 days, neurospheres were treated with drug X, and after 5 h, they were irradiated with sham (0 Gy) or 5 Gy of radiation. After 12 days of colony/neurosphere formation, colonies were stained with 0.2 μg/mL of Calcein AM (green) and imaged at 4× magnification on Cytation 5. (<b>E</b>) Relative change in colony sizes is plotted and two-way ANOVA is used for statistical analysis. * indicates <span class="html-italic">p</span> value &lt; 0.05; **** indicates <span class="html-italic">p</span> &lt; 0.0001; ‘ns’ stands for non-significant.</p>
Full article ">Figure 3
<p>(<b>A</b>,<b>C</b>) J14P and J14P-RT mCherry, 300 cells/well, were seeded in 96-well plate, and after 24 h, were irradiated with different doses of radiations (0, 2, 4, 6, and 8 Gy). After 13 days of colony/neurosphere formation, colonies were fixed with 0.84% PFA, stained with 4 μg/mL of WGA Alexa Fluor 488 (green), and imaged at 4× magnification using both green and red channels on the Cytation 5. Colonies of size 120 μm or above were counted as true colonies. (<b>B</b>,<b>D</b>) Relative colony formations are plotted and two-way ANOVA is used for statistical analysis. ** indicates <span class="html-italic">p</span> &lt; 0.01; *** indicates <span class="html-italic">p</span> &lt; 0.001; ‘ns’ stands for non-significant.</p>
Full article ">Figure 4
<p>(<b>A</b>) 14P mCherry and 14P-RT mCherry neurospheres were plated at 300 cells/well in 96-well plates and irradiated with different radiation doses in a fractionated approach (4 days between fractions). Following fixation with 0.84% PFA, cells were stained with 4 μg/mL of WGA Alexa Fluor 488 (green). (<b>B</b>) Relative colony formations are plotted and two-way ANOVA is used for statistical analysis. ** indicates <span class="html-italic">p</span> value &lt; 0.01 and ‘ns’ stands for non-significant.</p>
Full article ">
16 pages, 4154 KiB  
Article
Direct Interaction Between CD34+ Hematopoietic Stem Cells and Mesenchymal Stem Cells Reciprocally Preserves Stemness
by Rémi Safi, Tala Mohsen-Kanson, Farah Kouzi, Jamal El-Saghir, Vera Dermesrobian, Inés Zugasti, Kazem Zibara, Pablo Menéndez, Hiba El Hajj and Marwan El-Sabban
Cancers 2024, 16(23), 3972; https://doi.org/10.3390/cancers16233972 - 27 Nov 2024
Viewed by 458
Abstract
Background/Objectives: A specialized microenvironment in the bone marrow, composed of stromal cells including mesenchymal stem cells (MSCs), supports hematopoietic stem cell (HSC) self-renewal, and differentiation bands play an important role in leukemia development and progression. The reciprocal direct interaction between MSCs and CD34 [...] Read more.
Background/Objectives: A specialized microenvironment in the bone marrow, composed of stromal cells including mesenchymal stem cells (MSCs), supports hematopoietic stem cell (HSC) self-renewal, and differentiation bands play an important role in leukemia development and progression. The reciprocal direct interaction between MSCs and CD34+ HSCs under physiological and pathological conditions is yet to be fully characterized. Methods: Here, we established a direct co-culture model between MSCs and CD34+ HSCs or MSCs and acute myeloid leukemia cells (THP-1, Molm-13, and primary cells from patients) to study heterocellular communication. Results: Following MSCs-CD34+ HSCs co-culture, the expression of adhesion markers N-Cadherin and connexin 43 increased in both cell types, forming gap junction channels. Moreover, the clonogenic potential of CD34+ HSCs was increased. However, direct contact of acute myeloid leukemia cells with MSCs reduced the expression levels of connexin 43 and N-Cadherin in MSCs. The impairment in gap junction formation may potentially be due to a defect in the acute myeloid leukemia-derived MSCs. Interestingly, CD34+ HSCs and acute myeloid leukemia cell lines attenuated MSC osteoblastic differentiation upon prolonged direct cell–cell contact. Conclusions: In conclusion, under physiological conditions, connexin 43 and N-Cadherin interaction preserves stemness of both CD34+ HSCs and MSCs, a process that is compromised in acute myeloid leukemia, pointing to the possible role of gap junctions in modulating stemness. Full article
(This article belongs to the Section Tumor Microenvironment)
Show Figures

Figure 1

Figure 1
<p>Direct co-culture of MSCs and CD34<sup>+</sup> HSCs: (<b>A</b>) Schematic representation of the direct co-culture system between MSCs and CD34<sup>+</sup> HSCs. Representative images of HSCs alone, MSCs alone, and MSCs + CD34<sup>+</sup> HSCs after direct co-culture. Scale bar, 100 µm. (<b>B</b>) Histogram representing mRNA expression levels of adhesion and communication markers in MSCs following direct co-culture, assessed by qPCR. * <span class="html-italic">p</span> ≤ 0.05 and ** <span class="html-italic">p</span> ≤ 0.01 (<span class="html-italic">t</span>-test). (<b>C</b>) Western blot of Cx-43 and N-Cad expression in MSCs after 24 h of direct co-culture with CD34<sup>+</sup> HSCs. Results are represented as normalized expression to GAPDH in three independent experiments ± SEM. ** <span class="html-italic">p</span> ≤ 0.01 (<span class="html-italic">t</span>-test). (<b>D</b>) Expression of adhesion/communication markers and VEGF, CXCR4 in CD34<sup>+</sup> HSCs following direct co-culture, assessed by qPCR. Results are represented as normalized expression of GAPDH in three independent experiments. * <span class="html-italic">p</span> ≤ 0.05 and ** <span class="html-italic">p</span> ≤ 0.01 (<span class="html-italic">t</span>-test). (<b>E</b>) Clonogenic potential of CD34<sup>+</sup> HSCs, expressed as the number of colonies after direct co-culture with MSCs. (<b>F</b>) Cell profiling of CD34<sup>+</sup> HSCs after co-culture, showing the percentage of CD38<sup>+</sup> and CD45<sup>+</sup> cells in both suspension and adherent fractions. Green population refers to CD45<sup>+</sup> cells, red population refers to CD38<sup>+</sup> cells and purple population refers to CD45<sup>−</sup>CD38<sup>−</sup> cells.</p>
Full article ">Figure 2
<p>(<b>A</b>) Double immunostaining of Cx-43 (red), and N-Cad (green) in MSC alone and MSC + CD34<sup>+</sup> HSC following direct co-culture. Arrows point to the co-localization of the two proteins. Scale bar, 5 µm. (<b>B</b>) Cx-43 and N-Cad interaction in MSCs and HSCs after direct co-culture as detected by Duo-Link assay. The dots (red) are representatives of the close proximity of the two proteins of interest. Nuclei were stained with Hoechst 33,342 dye (blue). Scale bar, 10 µm (upper panel) and 2 µm (lower panel). (<b>C</b>) Representative flow cytometry graph showing the shift in MFI following co-culture of unlabeled HSCs with Calcein-labeled MSCs.</p>
Full article ">Figure 3
<p>Engagement in osteoblastic differentiation of MSCs after co-culture with HSCs for 24 h or 21 days: (<b>A</b>) Left panel: Expression of ALP protein in MSCs detected by immunofluorescence. Right panel: Alizarin red staining of MSCs visualized by light microscopy. Scale bar, 20 µm. (<b>B</b>) mRNA expression of ALP and Oct-4 in MSC following direct interaction with CD34<sup>+</sup> HSC for 24 h and 21 d. **** <span class="html-italic">p</span> ≤ 0.0001 (<span class="html-italic">t</span>-test).</p>
Full article ">Figure 4
<p>Direct co-culture of MSCs and AML cells: (<b>A</b>) Schematic representation of the direct co-culture system between MSCs and THP-1 and Molm-13. Scale bar, 100 µm. (<b>B</b>) mRNA and protein expression levels of Cx-43 and N-Cad in MSCs following direct co-culture with AML cell lines, assessed by qPCR and Western blot, respectively. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001 (<span class="html-italic">t</span>-test). (<b>C</b>) Representative flow cytometry graphs showing the shift in MFI following co-culture of unlabeled AML cell lines (THP-1 and Molm-13) with Calcein-labeled MSCs. (<b>D</b>) Fluorescent and light microscopy images of MSCs co-cultured with AML cell lines for ALP expression and Alizarin Red staining, respectively. Scale bar, 20 µm. (<b>E</b>) Expression of Cx-43 and N-Cad in MSCs following direct co-culture with AML primary cells (3 patients), assessed by Western blot analysis. * <span class="html-italic">p</span> ≤ 0.05 (<span class="html-italic">t</span>-test). (<b>F</b>) mRNA expression levels of Cx-43 and N-Cad in MSCs of healthy and AML patients following direct co-culture with AML cell lines, CD34<sup>+</sup> N from healthy individuals (<span class="html-italic">n</span> = 1) and CD34<sup>+</sup> AML from patients (<span class="html-italic">n</span> = 2).</p>
Full article ">
19 pages, 5090 KiB  
Article
Dissecting the Impact of Genetic Background on Oncogenic Response to Radiation Exposure in the Ptch1+/− Mouse Model
by Barbara Tanno, Emiliano Fratini, Simona Leonardi, Flavia Novelli, Valentina Pisano, Mariateresa Mancuso and Simonetta Pazzaglia
Cells 2024, 13(22), 1912; https://doi.org/10.3390/cells13221912 - 19 Nov 2024
Viewed by 671
Abstract
Medulloblastoma (MB) is a common primary brain cancer in children. The sonic hedgehog (SHH) pathway is indispensable for the normal development of the cerebellum, and MB is often caused by persistent SHH activation owing to mutations in pathway components. Patched1 (PTCH1) [...] Read more.
Medulloblastoma (MB) is a common primary brain cancer in children. The sonic hedgehog (SHH) pathway is indispensable for the normal development of the cerebellum, and MB is often caused by persistent SHH activation owing to mutations in pathway components. Patched1 (PTCH1) is the primary receptor for the SHH ligand and a negative regulator of the SHH signal transduction pathway. Mice heterozygous for the Ptch1 gene (Ptch1+/−) are predisposed to MB development. Irradiation of newborn Ptch1+/− mice dramatically increases MB occurrence. A genetic background carrying the Ptch1 mutation significantly influences the risk of developing MB. This study aims to investigate the genetic background-related mechanisms that regulate radiation-induced cellular response and oncogenesis in the cerebellum. We employed multiple approaches, including: (a) analysis of cellular radiosensitivity in granule cell precursors (GCPs), the MB cells of origin, derived from Ptch1 mice with a genetic background that is sensitive (CD1) or resistant (C57Bl/6) to the induction of radiogenic MB; (b) identification of genes differentially expressed in spontaneous and radiation-induced MBs from these two mouse strains; (c) bioinformatic analysis to correlate the expression of radiation-induced genes with survival in MB patients; and (d) examining the expression of these genes in ex vivo MBs induced by single or repeated radiation doses. We have identified a potential gene expression signature—Trp53bp1, Bax, Cyclin D1, p21, and Nanog—that influences tumor response. In ex vivo cultured spontaneous MBs, the expression levels of these genes increase after irradiation in CD1 mice, but not in mice with a C57Bl/6 genetic background, suggesting that this signature could predict tumor response to radiation therapy and help develop strategies for targeting DNA damage repair in tumors. A detailed understanding of the mechanisms behind genetic background-related susceptibility to radiation-induced oncogenic responses is crucial for translational research. Full article
Show Figures

Figure 1

Figure 1
<p>Characterization of residual DSBs and apoptosis in irradiated GCPs<sup>C57Bl-</sup><span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> and GCPs<sup>CD1-</sup><span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> at 24 h post-irradiation and the relatively unexposed controls. (<b>A</b>) Flow cytometry analysis of γH2AX in GCPs. (<b>B</b>,<b>C</b>) mRNA expression level of <span class="html-italic">Trp53bp1</span> and <span class="html-italic">Bax</span>. Evaluation of apoptosis by Annexin V assay: (<b>D</b>) plots and (<b>E</b>) table. Results are expressed as an average of 3 biological replicates ± SD; in qPCR, untreated GCPs<sup>C57Bl-</sup><span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> were taken as 1. Differences between irradiated and unirradiated GCPs of each genotype were analyzed with a Student’s <span class="html-italic">t</span>-test: show **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Characterization of growth kinetics, cell cycle, and stemness in GCPs from irradiated CD1<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> and C57Bl/6<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> mice and relative unexposed controls. (<b>A</b>) Growth curves of GCPs, in irradiated and unirradiated conditions for 8 days. (<b>B</b>) Evaluation of the expression of <span class="html-italic">Cyclin D1</span> mRNA at 24 h post-irradiation and (<b>C</b>) <span class="html-italic">p21</span> (<b>D</b>) <span class="html-italic">Nanog</span>, and (<b>E</b>) <span class="html-italic">Oct-4</span>. Neurosphere assay showing quantification (<b>F</b>) and size measurements (<b>G</b>) of neurospheres derived from irradiated GCPs after <span class="html-italic">Nanog</span> and <span class="html-italic">Oct-4</span> inhibition. The results are expressed as an average of 3 biological replicates ± SD; untreated GCPs<sup>C57Bl-</sup><span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> were taken as 1. The differences between the irradiated and corresponding unirradiated conditions of each genotype were analyzed with a Student’s <span class="html-italic">t</span>-test: show **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>p53 functional assay and flow cytometry cell cycle analysis in GCPs from 2 Gy-irradiated CD1<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> and C57Bl/6<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> mice and relative unexposed controls. (<b>A</b>) p53 functional assay on GCPs 2 h post irradiation. The results are expressed as an average of 3 biological replicates ± SD; untreated GCPs<sup>C57Bl-</sup><span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> and GCPs<sup>CD1-<span class="html-italic">Ptch1</span>+/−</sup> were taken as 100. (<b>B</b>) Flow cytometry analysis of the cell cycle at different post-irradiation times. The graphs present the mean of three experiments. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Gene expression analysis in spontaneous and radiation-induced MBs. Evaluation of mRNA expression levels of (<b>A</b>) <span class="html-italic">p53</span>, (<b>B</b>) Trp53bp1, (<b>C</b>) <span class="html-italic">Bax</span>, (<b>D</b>) <span class="html-italic">Cyclin D1</span>, (<b>E</b>) <span class="html-italic">p21</span>, (<b>F</b>) <span class="html-italic">p16</span>, (<b>G</b>) <span class="html-italic">Nanog</span>, and (<b>H</b>) <span class="html-italic">Oct-4</span>. The results are expressed as an average of 14 MBs ± SEM; untreated GCPs<sup>C57Bl-</sup><span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> were taken as 1. The differences were analyzed with a Student’s <span class="html-italic">t</span>-test: **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.5.</p>
Full article ">Figure 5
<p>Kaplan–Meier plots showing the impact of gene expression (high versus low levels) on survival in MB patients. Single gene analysis for <span class="html-italic">TP53BP1</span>, <span class="html-italic">BAX</span>, <span class="html-italic">CYCLIN D1</span>, <span class="html-italic">P21</span>, <span class="html-italic">NANOG</span>, <span class="html-italic">OCT-4</span>, and <span class="html-italic">P16</span>. Log-rank test <span class="html-italic">p</span>-values are indicated at the bottom right of each plot.</p>
Full article ">Figure 6
<p>Gene expression in spontaneous MBs from C57Bl/6<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> and CD1<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> mice after one or two 2 Gy X-ray fractions (yellow symbols). (<b>A</b>) Experimental scheme. (<b>B</b>) Expression levels of <span class="html-italic">Trp53bp1</span>, (<b>C</b>) <span class="html-italic">Bax</span>, (<b>D</b>) <span class="html-italic">Cyclin D1</span>, (<b>E</b>) <span class="html-italic">p21</span>, (<b>F</b>) <span class="html-italic">Nanog</span>, and (<b>G</b>) <span class="html-italic">Oct-4</span>. The results are expressed as an average of 3 biological replicates and expressed as means ± SE. Unirradiated C57Bl/6<sup><span class="html-italic">Ptch1</span>+/−</sup> and CD<sup><span class="html-italic">Ptch1</span>+/−</sup> spontaneous tumours are taken as 1. Differences were analyzed using an ANOVA test. Asterisks above the bars indicate comparisons between the specified groups, while asterisks over the histograms refer to comparisons with the corresponding unexposed control group: show * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Genetic background-related expression data in GCPs and MBs from <span class="html-italic">Ptch1</span><sup>+/−</sup> mice. Global recap of the frequencies of spontaneous and radiation−induced MBs (2 Gy) in C57Bl/6<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> (<b>left</b>) and CD1<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> mice (<b>right</b>) [<a href="#B12-cells-13-01912" class="html-bibr">12</a>], compared to the level of residual DSBs at 24 h (γ-H2AX and Trp53bp1<span class="html-italic">)</span> and expression of genes involved in DNA damage response (<span class="html-italic">Bax</span>, <span class="html-italic">p21</span>, <span class="html-italic">p16</span>, and <span class="html-italic">p53</span>) and in the cell cycle and stemness (<span class="html-italic">Cyclin D1</span>, <span class="html-italic">Nanog</span>, <span class="html-italic">Oct-4</span>, <span class="html-italic">p21</span>, and <span class="html-italic">p16</span>). <span class="html-italic">In vitro</span> data from sham (0 Gy) and irradiated (2 Gy) GCPs are in the yellow panels, while in vivo data from spontaneous and radiation-induced MBs are in grey. Schematic bars represent the relative expression levels compared to the expression in unexposed C57Bl/6<span class="html-italic"><sup>Ptch1</sup></span><sup>+/−</sup> control samples (Spontaneous MBs and GCPs): non-statistically different (green); downregulated genes (<span class="html-italic">p</span> &lt; 0.01) (blue); slightly upregulated genes (1.5 ≤ Relative mRNA levels &lt; 4, <span class="html-italic">p</span> &lt; 0.01) (pink); strongly upregulated genes (Relative mRNA levels &gt; 10, <span class="html-italic">p</span> &lt; 0.0001) (red).</p>
Full article ">
19 pages, 7655 KiB  
Article
Proinflammatory Cytokines Enhance the Mineralization, Proliferation, and Metabolic Activity of Primary Human Osteoblast-like Cells
by Juliana Franziska Bousch, Christoph Beyersdorf, Katharina Schultz, Joachim Windolf, Christoph Viktor Suschek and Uwe Maus
Int. J. Mol. Sci. 2024, 25(22), 12358; https://doi.org/10.3390/ijms252212358 - 18 Nov 2024
Viewed by 682
Abstract
Osteoporosis is a progressive metabolic bone disease characterized by decreased bone density and microarchitectural deterioration, leading to an increased risk of fracture, particularly in postmenopausal women and the elderly. Increasing evidence suggests that inflammatory processes play a key role in the pathogenesis of [...] Read more.
Osteoporosis is a progressive metabolic bone disease characterized by decreased bone density and microarchitectural deterioration, leading to an increased risk of fracture, particularly in postmenopausal women and the elderly. Increasing evidence suggests that inflammatory processes play a key role in the pathogenesis of osteoporosis and are strongly associated with the activation of osteoclasts, the cells responsible for bone resorption. In the present study, we investigated, for the first time, the influence of proinflammatory cytokines on the osteogenic differentiation, proliferation, and metabolic activity of primary human osteoblast-like cells (OBs) derived from the femoral heads of elderly patients. We found that all the proinflammatory cytokines, IL-1β, TNF-α, IL-6, and IL-8, enhanced the extracellular matrix mineralization of OBs under differentiation-induced cell culture conditions. In the cases of IL-1β and TNF-α, increased mineralization was correlated with increased osteoblast proliferation. Additionally, IL-1β- and TNF-α-increased osteogenesis was accompanied by a rise in energy metabolism due to improved glycolysis or mitochondrial respiration. In conclusion, we show here, for the first time, that, in contrast to findings obtained with cell lines, mesenchymal stem cells, or animal models, human OBs obtained from patients exhibited significantly enhanced osteogenesis upon exposure to proinflammatory cytokines, probably in part via a mechanism involving enhanced cellular energy metabolism. This study significantly contributes to the field of osteoimmunology by examining a clinically relevant cell model that can help to develop treatments for inflammation-related metabolic bone diseases. Full article
(This article belongs to the Topic Osteoimmunology and Bone Biology)
Show Figures

Figure 1

Figure 1
<p>Morphologies of the osteoblast-like cell cultures. Light microscope pictures (100× magnification, scale bars = 100 µm) of OBs from three different donors seeded at confluency (<b>A</b>) on day 1 (undifferentiated cells) and (<b>B</b>) on day 7 of osteogenesis. The arrows mark roundish-shaped cells as an example.</p>
Full article ">Figure 2
<p>Interleukin (IL)-6 and IL-8 secretion during osteogenesis. The primary human osteoblast-like cells (OBs) were incubated in osteogenesis induction medium (OIM) for 35 days and (<b>A</b>) mineralization of the extracellular matrix (ECM) was quantified by Alizarin Red S staining (n = 13, four replicates each), (<b>C</b>) accompanied by example images of corresponding microscope images (100× magnification; scale bars = 100 µm) and well plates, and (<b>B</b>) alkaline phosphatase (ALP) activity was measured every week (n = 6, 4 replicates each). The protein concentration (pg/mL) of (<b>D</b>) IL-6 and (<b>E</b>) IL-8 in the supernatant was examined by enzyme-linked immunosorbent assay (ELISA) (n = 11, two replicates). Means with standard deviation (SD) are shown, and all significances were calculated by Wilcoxon signed-rank test (<span class="html-italic">p</span> ≤ 0.05 (*), <span class="html-italic">p</span> ≤ 0.01 (**), <span class="html-italic">p</span> ≤ 0.001 (***)).</p>
Full article ">Figure 3
<p>Effects of IL-1β, IL-6, IL-8, and tumor necrosis factor (TNF)-α on the mineralization of OBs. The boxplot diagrams illustrate the relative mineralization quantified by Alizarin Red S staining of OBs (n = 9, four replicates each) incubated for 21 days with the cytokines (<b>A</b>) IL-1β, (<b>B</b>) IL-6, (<b>C</b>) IL-8, and (<b>D</b>) TNF-α at concentrations ranging from 3.125 to 250 units/mL in OIM, with the corresponding microscopic images of the calcified nodules (scale bars ≙ 100 µm). Additionally, the ALP activity on day 8 and day 10 with selected concentrations of IL-1β (250 units/mL), IL-6 (12.5 units/mL), IL-8 (12.5 units/mL), and TNF-α (250 units/mL) is shown (n = 5, four replicates each). The whiskers representing the minimum and maximum, the median lines, and the means (+) are shown. The significances refer to the control (0 units/mL) and were calculated by the Wilcoxon signed-rank test (<span class="html-italic">p</span> ≤ 0.05 (*), <span class="html-italic">p</span> ≤ 0.01 (**)).</p>
Full article ">Figure 4
<p>Reciprocal impact of the cytokines on mRNA expression in differentiated OBs. The mRNA expression of (<b>A</b>) IL-6 and (<b>B</b>) IL-8 was determined upon 21-day incubation of the cytokines IL-1β (250 units/mL), IL-6 (12.5 units/mL), IL-8 (12.5 units/mL), and TNF-α (250 units/mL) in OIM (n = 10, three replicates). The relative mRNA expression was calculated by the 2-ΔΔ Ct method and normalized on the control cells with 21-day OIM incubation without cytokines (ctrl) of each patient. The Tukey boxplot diagrams show the median line, and any values that exceed the threshold of the 75th percentile plus 1.5 times the interquartile range are plotted as statistical outliers (<b>■</b>, <b>▲</b>). The significances were calculated by the Wilcoxon signed-rank test (<span class="html-italic">p</span> ≤ 0.05 (*), <span class="html-italic">p</span> ≤ 0.01 (**)).</p>
Full article ">Figure 5
<p>Impact of proinflammatory cytokines on the cell count and proliferation rate. The cell count of OBs incubated with the cytokines IL-1β (250 units/mL), IL-6 (12.5 units/mL), IL-8 (12.5 units/mL), and TNF-α (250 units/mL) in OIM on (<b>A</b>) day 7, (<b>B</b>) day 14, and (<b>C</b>) day 21 of osteogenesis was determined by Dimethylthiazolyl Blue Tetrazolium Bromide (MTT) assay. The mean with SD is shown. The diagram in (<b>D</b>) summarizes the observed period from day 7 to day 21 (n = 8, four replicates each); the mean with SEM is shown. All data were normalized for the control (OIM without cytokines) on day 7 from each patient. (<b>E</b>) The cell proliferation rate over 48 h was measured by bromodeoxyuridine (BrdU) assay (n = 6, four replicates each). The data were normalized for the control (OIM without cytokines) from each patient, and the mean with SD is shown. The significances were calculated by the Wilcoxon signed-rank test (<span class="html-italic">p</span> ≤ 0.05 (*)).</p>
Full article ">Figure 6
<p>Impact of proinflammatory cytokines on the mitochondrial oxidative phosphorylation during osteogenesis. The (<b>A</b>) basal respiration, (<b>B</b>) adenosine triphosphate (ATP) production, and (<b>C</b>) maximal respiration were determined by Seahorse Cell Mito Stress Tests (Agilent). (<b>D</b>) All measurement time points of the assay for day 1, day 14 + OIM, and day 14 with OIM and TNF-α with the injection of assay reagents are shown. The OBs’ oxygen consumption rate (OCR) was measured on day 1 of osteogenesis (n = 16), and on day 14 without (n = 9) and with the cytokines IL-1β (250 units/mL; n = 5), IL-6 (12.5 units/mL; n = 4), IL-8 (12.5 units/mL; n = 4), and TNF-α (250 units/mL; n = 5). All experiments were performed with at least 10 technical replicates. The significances were calculated by the Mann–Whitney U test (<span class="html-italic">p</span> ≤ 0.05 (*), <span class="html-italic">p</span> ≤ 0.01 (**)).</p>
Full article ">Figure 7
<p>Impact of proinflammatory cytokines on glycolysis during osteogenesis. The (<b>A</b>) glycolysis, (<b>B</b>) glycolytic reserve, and (<b>C</b>) non-glycolytic acidification were determined by the Seahorse Glycolysis Stress Test (Agilent). (<b>D</b>) All measurement time points of the assay for day 1, day 14 + OIM, and day 14 with OIM and IL-1β with the injection of assay reagents are shown. The OBs’ extracellular acidification rate (ECAR) was measured on day 1 of osteogenesis (n = 8), and on day 14 without (n = 10, and with the cytokines IL-1β (250 units/mL; n = 6), IL-6 (12.5 units/mL; n = 6), IL-8 (12.5 units/mL; n = 6), and TNF-α (250 units/mL; n = 6). All experiments were performed with at least 10 technical replicates. The significances were calculated by the Mann–Whitney U test (<span class="html-italic">p</span> ≤ 0.05 (*), <span class="html-italic">p</span> ≤ 0.01 (**)).</p>
Full article ">
26 pages, 444 KiB  
Review
Arbovirus in Solid Organ Transplants: A Narrative Review of the Literature
by Kiran Gajurel, Reshika Dhakal and Stan Deresinski
Viruses 2024, 16(11), 1778; https://doi.org/10.3390/v16111778 - 15 Nov 2024
Viewed by 1037
Abstract
The incidence of arbovirus infections has increased in recent decades. Other than dengue, chikungunya, and West Nile viruses, the data on arbovirus in solid organ transplant (SOT) are limited to case reports, and infections in renal transplant recipients account for most of the [...] Read more.
The incidence of arbovirus infections has increased in recent decades. Other than dengue, chikungunya, and West Nile viruses, the data on arbovirus in solid organ transplant (SOT) are limited to case reports, and infections in renal transplant recipients account for most of the reported cases. Dengue and West Nile infections seem to be more severe with higher mortality in SOT patients than in the general population. Acute kidney injury is more frequent in patients with dengue and chikungunya although persistent arthralgia with the latter is less frequent. There is no clear relationship between arboviral infection and acute cellular rejection. Pre-transplant screening of donors should be implemented during increased arboviral activity but, despite donor screening and negative donor nucleic acid amplification test (NAT), donor derived infection can occur. NAT may be transiently positive. IgM tests lack specificity, and neutralizing antibody assays are more specific but not readily available. Other tests, such as immunohistochemistry, antigen tests, PCR, metagenomic assays, and viral culture, can also be performed. There are a few vaccines available against some arboviruses, but live vaccines should be avoided. Treatment is largely supportive. More data on arboviral infection in SOT are needed to understand its epidemiology and clinical course. Full article
(This article belongs to the Special Issue Viral Infections in Immunocompromised Hosts)
Back to TopTop