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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (564)

Search Parameters:
Keywords = γH2AX

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 7358 KiB  
Article
Disrupted Mitochondrial Dynamics Impair Corneal Epithelial Healing in Neurotrophic Keratopathy
by Mengyi Jin, Zeyu Liu, Ruize Shi, Ya Deng, Jingwei Lin, Yuting Zhang, Lexin Lin, Yanzi Wang, Yunyi Shi, Cheng Li and Zuguo Liu
Int. J. Mol. Sci. 2025, 26(3), 1290; https://doi.org/10.3390/ijms26031290 - 3 Feb 2025
Viewed by 187
Abstract
Neurotrophic keratopathy (NK) is a degenerative corneal disease characterized by impaired corneal sensitivity and epithelial repair that is often linked to sensory nerve dysfunction. To establish a clinically relevant model and explore the mechanisms underlying NK pathogenesis, we developed a novel mouse model [...] Read more.
Neurotrophic keratopathy (NK) is a degenerative corneal disease characterized by impaired corneal sensitivity and epithelial repair that is often linked to sensory nerve dysfunction. To establish a clinically relevant model and explore the mechanisms underlying NK pathogenesis, we developed a novel mouse model through partial transection of the ciliary nerve. This approach mimics the progressive nature of NK, reproducing key clinical features such as corneal epithelial defects, reduced sensitivity, diminished tear secretion, and delayed wound healing. Using this model, we investigated how disruptions in mitochondrial dynamics contribute to corneal epithelial dysfunction and impaired repair in NK. Our findings revealed substantial disruptions in mitochondrial dynamics, including reduced expression of fusion proteins (OPA1), downregulation of fission regulators (FIS1 and MFF), and impaired mitochondrial transport, as evidenced by decreased expression of Rhot1 and Kif5b. Additionally, the downregulation of mitophagy-related genes (Pink1 and Prkn) contributed to the accumulation of dysfunctional mitochondria, leading to DNA damage and impaired corneal epithelial repair. These mitochondrial abnormalities were accompanied by increased γH2AX staining, indicative of DNA double-strand breaks and cellular stress. This study highlights the pivotal role of mitochondrial dynamics in corneal epithelial health and repair, suggesting that therapeutic strategies aimed at restoring mitochondrial function, enhancing mitophagy, and mitigating oxidative stress may offer promising avenues for treating NK. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

Figure 1
<p>Neurotrophic keratopathy model assessment. (<b>A</b>) Representative slit-lamp photographs of control and NK group corneas at 0- and 14-days post-surgery. (<b>B</b>) Fluorescein staining scores. (<b>C</b>) Tear secretion measured via phenol red cotton threads. (<b>D</b>) Corneal sensitivity measured with Cochet–Bonnet esthesiometer. Red arrows: fluorescein-stained epithelial defects. Data are presented as mean ± SD, N = 6 mice per group, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Analysis of corneal epithelial wound healing in the neurotrophic keratopathy model. (<b>A</b>) Representative slit-lamp images showing corneal epithelial wounds and fluorescein staining in the CTRL and NK groups at 0, 12, and 24 h post-wounding. (<b>B</b>) Quantitative analysis of the epithelial wound healing area. Red arrows: the unhealed epithelial defects. Data are presented as mean ± SD, N = 6 per group, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Reduced corneal nerve density in the neurotrophic keratopathy (NK) model. (<b>A</b>) Representative whole-mount staining of sub-basal nerves in the corneal epithelium for both the CTRL and NK groups. Quantitative analysis of peripheral (<b>B</b>) and central (<b>C</b>) sub-basal nerve density. Data are presented as mean ± SD, N = 5 per group, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Mitochondrial morphological alterations in corneal epithelial cells of the neurotrophic keratopathy model. (<b>A</b>) Representative images depicting Tom20 staining in corneal epithelial cells. (<b>B</b>) Mean area of mitochondria. (<b>C</b>) Mean perimeter of mitochondria. (<b>D</b>) Mean aspect ratio of mitochondria. (<b>E</b>) Mean form factor of mitochondria. (<b>F</b>) Transmission electron microscopy (TEM) visualization of mitochondrial structures. M: Mitochondria. N: nucleus. Data are presented as mean ± SD, N = 3 per group. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Quantitative PCR analysis of mitochondrial dynamics-, transport-, and mitophagy-related gene expression in corneal epithelial cells from control (CTRL) and neurotrophic keratopathy (NK) groups. (<b>A</b>,<b>B</b>) <span class="html-italic">Tim23</span> and <span class="html-italic">Tom20</span> expression levels show no significant differences. (<b>C</b>,<b>D</b>) <span class="html-italic">Drp1</span> and <span class="html-italic">Fis1</span>, key regulators of mitochondrial fission, also show no significant differences between the two groups. (<b>E</b>,<b>F</b>) <span class="html-italic">Opa1</span> and <span class="html-italic">Mfn1</span>, essential for mitochondrial fusion, are significantly downregulated in the NK group (<span class="html-italic">p</span> &lt; 0.05). (<b>G</b>) <span class="html-italic">Mfn2</span> expression remains unchanged between the two groups. (<b>H</b>,<b>J</b>) <span class="html-italic">Rhot1</span> and <span class="html-italic">Kif5b</span>, involved in mitochondrial transport, are significantly downregulated in the NK group (<span class="html-italic">p</span> &lt; 0.05), while (<b>I</b>) <span class="html-italic">Rhot2</span> shows no significant change. (<b>K</b>,<b>L</b>) <span class="html-italic">Pink1</span> and <span class="html-italic">Prkn</span>, key regulators of mitophagy, are significantly reduced in the NK group compared to the CTRL group (<span class="html-italic">p</span> &lt; 0.05). Data are presented as mean ± SD, N = 5 per group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Analysis of mitochondrial and DNA damage in NK model. (<b>A</b>) Representative immunofluorescence images showing OPA1 staining. (<b>B</b>–<b>D</b>,<b>I</b>) Western blot analyses of OPA1, FIS1, MFF, and γH2A.X. (<b>E</b>–<b>G</b>,<b>J</b>) Quantification of protein levels for OPA1, FIS1, MFF, and γH2A.X. (<b>H</b>) Representative immunofluorescence images showing γH2A.X staining in NK corneas. White arrows indicate OPA1-positive staining, while red arrows indicate γH2A.X-positive staining. Data are presented as mean ± SD, N = 3 per group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
21 pages, 13070 KiB  
Article
MK-8776 and Olaparib Combination Acts Synergistically in Hepatocellular Carcinoma Cells, Demonstrating Lack of Adverse Effects on Liver Tissues in Ovarian Cancer PDX Model
by Wiktoria Bębenek, Arkadiusz Gajek, Agnieszka Marczak, Jan Malý, Jiří Smejkal, Małgorzata Statkiewicz, Natalia Rusetska, Magdalena Bryś and Aneta Rogalska
Int. J. Mol. Sci. 2025, 26(2), 834; https://doi.org/10.3390/ijms26020834 - 20 Jan 2025
Viewed by 530
Abstract
Hepatocellular carcinoma (HCC) cells critically depend on PARP1 and CHK1 activation for survival. Combining the PARP inhibitor (PARPi) olaparib with a CHK1 inhibitor (MK-8776, CHK1i) produced a synergistic effect, reducing cell viability and inducing marked oxidative stress and DNA damage, particularly in the [...] Read more.
Hepatocellular carcinoma (HCC) cells critically depend on PARP1 and CHK1 activation for survival. Combining the PARP inhibitor (PARPi) olaparib with a CHK1 inhibitor (MK-8776, CHK1i) produced a synergistic effect, reducing cell viability and inducing marked oxidative stress and DNA damage, particularly in the HepG2 cells. This dual treatment significantly increased apoptosis markers, including γH2AX and caspase-3/7 activity. Both HCC cell lines exhibited heightened sensitivity to the combined treatment. The effect of drugs on the expression of proliferation markers in an olaparib-resistant patient-derived xenograft (PDX) model of ovarian cancer was also investigated. Ovarian tumors displayed reduced tissue growth, as reflected by a drop in proliferation marker Ki-67 levels in response to PARPi combined with CHK1i. No changes were observed in corresponding liver tissues using Ki-67 and pCHK staining, which indicates the absence of metastases and a hepatotoxic effect. Thus, our results indicate that the dual inhibition of PARP and CHK1 may prove to be a promising therapeutic approach in the treatment of primary HCC as well as OC tumors without the risk of liver metastases, especially in patients with olaparib-resistant tumor profiles. Full article
Show Figures

Figure 1

Figure 1
<p>PARPi combined with CHK1i induces lower cytotoxicity in HepG2 cells compared to Hep3B cells. (<b>A</b>) Cell viability in response to five-day treatment with PARPi (O, 1–240 µM) and CHK1i (C, 0.05–120 µM) was determined using MTT and resazurin reduction assays in hepatocellular carcinoma and PMBC cells, respectively. (<b>B</b>) The IC<sub>50</sub> values. Data are expressed as mean ± SD (n ≥ 3). (<b>C</b>,<b>D</b>) Cytotoxic effects observed in hepatocellular carcinoma and PBMC cells (120 h) after treatment with O (1, 2.5, or 5 µM) combined with C (0.05, 0.1, 0.5, 1, 2.5, and 5 µM). (<b>E</b>) Morphological changes in HCC cells in response to O (2.5 µM) and C (5 µM), or their combinations after five days of treatment. Images were captured at 10× magnification. Orange arrows show morphological changes. Data are expressed as mean ± SD (n = 3–6). Coefficient of drug interaction (CDI) values, indicating whether interaction effects are significantly synergistic (CDI &lt; 1), additive (CDI = 1.0), or antagonistic (CDI &gt; 1.0). Statistical significance was assessed using ANOVA followed by Tukey’s test. * Statistically significant changes between cells treated with the compound and control cells (<span class="html-italic">p</span> &lt; 0.05). + Statistically significant changes between cells treated with PARPi and combination treatments (O + C) (<span class="html-italic">p</span> &lt; 0.05). # Statistically significant changes between cells treated with CHK1i and combination treatments (O + C) (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>The kinetics of ROS generation in Hep3B and HepG2 cells after treatment with O (2.5 µM), C (5 µM), and the combination of O + C (2.5 µM; 5 µM) were measured immediately after adding the drugs up to 180 min, and after 48 h up to 180 min in the presence or absence of an antioxidant (NAC). The control cells (not treated) were assumed as 100%. Positive controls cells were treated with doxorubicin (DOX, 10 µM) or camptothecin (CPT, 5µM). Data are expressed as mean ± SD (n = 3–6). Statistical significance was assessed using ANOVA followed by Tukey’s test. * Statistically significant changes between cells treated with the compound compared with control cells (<span class="html-italic">p</span> &lt; 0.05). + Statistically significant changes between cells treated with PARPi and combination treatments (PARPi/CHK1i) (<span class="html-italic">p</span> &lt; 0.05). ◊ Statistically significant changes between cells treated with compound and samples preincubated with N-acetylcysteine (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>(<b>A</b>) Quantitative Western blot analysis and representative images of γH2AX, and full-length and cleaved PARP1 in HCC cells. Protein levels following treatment were quantified, with normaization to β-actin as a loading control and calculated as fold-change relative to untreated controls. (<b>B</b>) Representative images of immunofluorescence staining. HCC cells were incubated with O and C individually or O + C combination for 48 h and labeled with fluorochrome-conjugated antibodies against γH2AX (green fluorescence), RAD51, and caspase-3 (red fluorescence). Images were acquired using a confocal laser scanning microscope (magnification 63×). (<b>C</b>) Caspase-3/7 activity changes in HCC cells after single (O, C) or combined (O + C) drug administration. (Data are expressed as mean ± SD (n = 3–6). Statistical significance was assessed using ANOVA followed by Tukey’s test. * Statistically significant changes between cells treated with the compound compared with control cells (<span class="html-italic">p</span> &lt; 0.05). + Statistically significant changes between cells treated with PARPi and combination treatments (PARPi/CHK1i) (<span class="html-italic">p</span> &lt; 0.05). # Statistically significant changes between cells treated with CHK1i and combination treatments (PARPi/CHK1i) (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>O combined with C induced alanine aminotransferase activity and glutathione S-transferase after 48 h of incubation in HCC cells. Data are expressed as mean ± SD (n = 3–6). Statistical significance was assessed using ANOVA followed by Tukey’s test. * Statistically significant changes between cells treated with the compound compared with control cells (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Characterization of responses of liver tissues (<b>A</b>) and ovarian tumors (<b>B</b>), collected from mice with olaparib-resistant PDX tumors, to the tested inhibitors based on a histopathological assessment (H&amp;E-staining) and evaluation of Ki-67 and pCHK1 expression levels (IHC staining). Images were captured at 20× magnification, MICA WideFocal Live Cell, Leica Microsystems, Wetzlar, Germany).</p>
Full article ">
18 pages, 3524 KiB  
Article
Viability and Radiosensitivity of Human Tumor Cells from Breast and Colon Are Influenced by Hypericum perforatum Extract HP01
by Linda Rebecca Haake, Ahmed El Menuawy, Hannes Rennau, Frank Marthe, Urs Hähnel, Felix Bock, Guido Hildebrandt and Katrin Manda
Int. J. Mol. Sci. 2025, 26(2), 622; https://doi.org/10.3390/ijms26020622 - 13 Jan 2025
Viewed by 427
Abstract
To enhance the treatment of tumors that are resistant to radio- and chemotherapy while minimizing the side effects of radiochemotherapy, researchers are continuously seeking new active compounds for use in combination with radiotherapy. Therefore, the aim of our study was to examine the [...] Read more.
To enhance the treatment of tumors that are resistant to radio- and chemotherapy while minimizing the side effects of radiochemotherapy, researchers are continuously seeking new active compounds for use in combination with radiotherapy. Therefore, the aim of our study was to examine the cytotoxic and radiosensitizing effects of an extract from St. John’s Wort (Hypericum perforatum), referred to as HP01, on human epithelial tumor cells in vitro. The growth of MCF-7 (breast carcinoma) and HT-29 (colon carcinoma) cells was examined under the influence of HP01. In combination with radiation, the effects of HP01 on cytotoxicity and long-term survival were assessed using a colony formation assay. The number of DNA double-strand breaks was analyzed using the γH2AX assay, while cell cycle distribution was examined via flow cytometry. A growth-inhibiting and cytotoxic effect was observed for both tumor cell lines starting at a concentration of 10 µg/mL HP01. Treatment with HP01 resulted in an inhibition of clonogenic survival of tumor cells after ionizing radiation (6 Gy). The number of DNA double-strand breaks (DSBs) in tumor cells increased with HP01 treatment, but the repair of radiation-induced DNA DSBs was not affected. Cell cycle analysis revealed that HP01, in addition to radiation, enhanced G2/M arrest in MCF-7 and HT-29 cells. Overall, HP01 not only showed a growth-inhibiting effect but also demonstrated a radiosensitizing effect on human tumor cells for the first time. We conclude that the HP01-induced G2/M accumulation of cells may be the main rationale for the drug-induced radiosensitivity. It is therefore a promising candidate for combined therapy in tumor diseases and warrants further investigation. Full article
(This article belongs to the Special Issue Biological Research on Plant Bioactive Compounds)
Show Figures

Figure 1

Figure 1
<p>Growth of tumor cells treated with <span class="html-italic">Hypericum perforatum</span> extract HP01. The cells were seeded at day 0. HP01 was added to the cells at day 1 of incubation after a 24 h cell adhesion period to (<b>a</b>) MCF-7 cells and (<b>b</b>) HT-29 cells. Error bars represent the standard deviations of six (MCF-7: 1, 10, 50, 100 µg/mL HP01), three (MCF-7: 5, 20 µg/mL HP01), and four (HT-29) separate experiments; wells were assayed in triplicate in each of the different experiments. Significance was calculated for each day’s approaches (control versus treated sample). Asterisks illustrate significances: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 1 Cont.
<p>Growth of tumor cells treated with <span class="html-italic">Hypericum perforatum</span> extract HP01. The cells were seeded at day 0. HP01 was added to the cells at day 1 of incubation after a 24 h cell adhesion period to (<b>a</b>) MCF-7 cells and (<b>b</b>) HT-29 cells. Error bars represent the standard deviations of six (MCF-7: 1, 10, 50, 100 µg/mL HP01), three (MCF-7: 5, 20 µg/mL HP01), and four (HT-29) separate experiments; wells were assayed in triplicate in each of the different experiments. Significance was calculated for each day’s approaches (control versus treated sample). Asterisks illustrate significances: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 2
<p>Measurement of cytotoxicity of <span class="html-italic">Hypericum perforatum</span> extract was performed 24 h after treatment of (<b>a</b>) MCF-7 and (<b>b</b>) HT-29 cells via release of lactate dehydrogenase (LDH). Triplicates were scored for each dose. Data from four (MCF7) and three (HT-29) independent experiments are presented as mean values ± SD. Asterisks illustrate significances calculated in relation to controls without HP01 treatment (for non-irradiated experiments: 0 µg/mL HP01; 0 Gy; for irradiated experiments: 0 µg/mL HP01; 6 Gy): * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001. Hash sign illustrate significance between data from treatment with the same HP01 concentration, among non-irradiated (0 Gy) and irradiated (6 Gy) attempts (<sup>#</sup> <span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 2 Cont.
<p>Measurement of cytotoxicity of <span class="html-italic">Hypericum perforatum</span> extract was performed 24 h after treatment of (<b>a</b>) MCF-7 and (<b>b</b>) HT-29 cells via release of lactate dehydrogenase (LDH). Triplicates were scored for each dose. Data from four (MCF7) and three (HT-29) independent experiments are presented as mean values ± SD. Asterisks illustrate significances calculated in relation to controls without HP01 treatment (for non-irradiated experiments: 0 µg/mL HP01; 0 Gy; for irradiated experiments: 0 µg/mL HP01; 6 Gy): * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001. Hash sign illustrate significance between data from treatment with the same HP01 concentration, among non-irradiated (0 Gy) and irradiated (6 Gy) attempts (<sup>#</sup> <span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 3
<p>Clonogenic survival of (<b>a</b>) MCF-7 and (<b>b</b>) HT-29 cells after treatment with <span class="html-italic">Hypericum perforatum</span> extract HP01. HP01 treatment was performed 24 h after seeding and 24 h before irradiation treatment of cells. Data from at least three independent experiments are presented as normalized mean values of numbers of colonies ± SD. Asterisks illustrate significances: * <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 4
<p>DNA double-strand breaks in cells under different concentrations of <span class="html-italic">Hypericum perforatum</span> extract HP01. γH2AX foci were scored in (<b>a</b>) MCF-7 and (<b>b</b>) HT-29 cells (I) 1 h and (II) 24 h after radiation with 6 Gy or 0 Gy (control) and treatment with different HP01 concentrations (20 µg/mL, 50 µg/mL). Data from three independent experiments are presented as mean values ± SD. Asterisks illustrate significances: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 5
<p>Cell cycle distribution of <span class="html-italic">Hypericum perforatum</span> extract HP01-treated MCF-7 (<b>a</b>) and HT-29 (<b>b</b>) cells in combination with ionizing radiation (6 Gy) or non-irradiation (0 Gy). HP01 was added to the cells 24 h before irradiation. Cells were fixed 24 h after irradiation (48 h HP01 treatment). The proportion of sub-G1-phase cells was less than 0.5% in each case. For the three independent experiments significances were calculated in relation to controls without HP01 treatment (for non-irradiated experiments: 0 µg/mL HP01; 0 Gy; for irradiated experiments: 0 µg/mL HP01; 6 Gy) and illustrated by asterisks (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01). Hash signs illustrate significance between data from treatment with the same HP01 concentration among non-irradiated (0 Gy) and irradiated (6 Gy) attempts (<sup>#</sup> <span class="html-italic">p</span> ≤ 0.05, <sup>##</sup> <span class="html-italic">p</span> ≤ 0.01, <sup>###</sup> <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">
23 pages, 4232 KiB  
Article
MYC Overexpression Enhances Sensitivity to MEK Inhibition in Head and Neck Squamous Cell Carcinoma
by Cuicui Yang, Xiaowu Pang, Shaolei Teng, Shamel Wilson, Xinbin Gu and Guiqin Xie
Int. J. Mol. Sci. 2025, 26(2), 588; https://doi.org/10.3390/ijms26020588 - 12 Jan 2025
Viewed by 498
Abstract
MEK inhibitors, such as trametinib, have shown therapeutic potential in head and neck squamous cell carcinoma (HNSCC). However, the factors influencing cancer cell sensitivity and resistance to MEK inhibition remain poorly understood. In our study, we observed that MEK inhibition significantly reduced the [...] Read more.
MEK inhibitors, such as trametinib, have shown therapeutic potential in head and neck squamous cell carcinoma (HNSCC). However, the factors influencing cancer cell sensitivity and resistance to MEK inhibition remain poorly understood. In our study, we observed that MEK inhibition significantly reduced the expression of MYC, a transcription factor critical for the therapeutic response. MYC overexpression markedly enhanced the sensitivity of HNSCC cells to trametinib, as evidenced by delayed wound healing and reduced colony formation. Cell cycle analysis revealed that trametinib induced a G1 phase arrest, whereas MYC overexpression accelerated cell cycle progression, with a reduced induction of p27 and p21 and diminished decreases in E2F1 and phospho-Ser2/5 levels. Flow cytometry and protein analyses demonstrated that MYC overexpression amplified trametinib-induced apoptosis and DNA damage, as evidenced by elevated levels of pro-apoptotic markers (p53, cleaved PARP, and BIM) and γH2AX. In vivo xenograft models confirmed these findings, showing increased sensitivity to trametinib in MYC-overexpressing tumors. Moreover, MEK inhibition increased autophagy in HNSCC cells, a factor critical for therapeutic resistance. Inhibiting trametinib-induced autophagy further enhanced apoptotic cell death. These findings suggest that MYC expression and autophagy play crucial roles in HNSCC’s response to MEK inhibition. Combining trametinib with autophagy inhibition may improve therapeutic outcomes in HNSCC. Full article
(This article belongs to the Special Issue New Wave of Cancer Therapeutics: Challenges and Opportunities)
Show Figures

Figure 1

Figure 1
<p>Multiple anti-cancer effects induced by trametinib through MEK/MAPK inhibition in JHU11 HNSCC cells. Total protein extracts were prepared from JHU11 cells treated with either vehicle or 200 nM trametinib for 48 h. Western blot analysis was performed to detect (<b>A</b>) pERK1/2 (<b>a</b>), total ERK1/2 (<b>b</b>), pEGFR (<b>c</b>), MYC (<b>d</b>), and β-Actin (<b>e</b>). (<b>B</b>) Protein band intensities of (<b>a</b>) pERK/ERK, (<b>b</b>) pEGFR, and (<b>c</b>) MYC were quantified and normalized to the corresponding β-actin to compare vehicle- and trametinib-treated cells. (<b>C</b>,<b>D</b>) γH2A.X (<b>a</b>) and β-Actin (<b>Cb</b>); (<b>E</b>,<b>F</b>) p21 (<b>a</b>), p27 (<b>b</b>), cPARP/PARP (<b>c</b>), BIM (<b>d</b>), p53 (<b>e</b>), and β-Actin (<b>Ef</b>); (<b>G</b>,<b>H</b>) pSer2/5 (<b>a</b>), pSer7 (<b>b</b>), and β-Actin (<b>Gc</b>). Protein band intensities for the ratio of cPARP/PARP, pEGFR, MYC, γH2A.X, p21, p27, BIM, p53, pSer2/5, and pSer7 were normalized to β-actin correspondingly and compared between vehicle-treated and trametinib-treated cells. Data from independent experiments are presented as mean ± SD (n = 3~4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 2
<p>MYC overexpression renders JHU22 HNSCC cells more sensitive to trametinib. (<b>A</b>) Overall survival percentage of HNSCC patients with (solid line) versus without MYC amplification (dotted line) (<span class="html-italic">p</span> = 0.013). (<b>B</b>) Increased MYC mRNA expression is observed in HNSCC patients with MYC amplification. All data are from the TCGA database (279 samples), Nature 2015, accessed via cBioPortal.org [<a href="#B27-ijms-26-00588" class="html-bibr">27</a>,<a href="#B28-ijms-26-00588" class="html-bibr">28</a>]. (<b>C</b>) Total protein extracts were obtained from JHU22-LV and JHU22-MYC cells treated with either vehicle or 200 nM trametinib for 48 h. Western blot analysis was performed to detect MYC and β-Actin. (<b>D</b>) MYC band intensities were quantified and normalized to the corresponding β-actin to compare vehicle- and trametinib-treated cells (n = 4). (<b>E</b>) A dose-escalation study was conducted to evaluate the effects of increasing concentrations of trametinib on cell viability in JHU22-LV and JHU22-MYC cells, with treatment lasting for 7 days. Images represent two replicates. (<b>F</b>) IC50 values for trametinib (<b>a</b>,<b>b</b>) and erlotinib (<b>c</b>,<b>d</b>) were determined in JHU22-LV and JHU22-MYC cells, respectively. The red dotted line indicates 50% viability, corresponding to the IC50 value. (<b>G</b>) Colony formation assays were performed to assess the proliferative ability of JHU22-LV (<b>a</b>,<b>c</b>) and JHU22-MYC (<b>b</b>,<b>d</b>) cells following treatment with either vehicle or trametinib (200 nM) for 7 days. (<b>H</b>) Bar graphs and statistical analysis of the colony coverage (Total area, % of plate). (<b>I</b>) Wound healing assays were used to examine changes in migratory capacity between JHU22-LV and JHU22-MYC cells, with treatments of either vehicle or trametinib (200 nM) conducted at 0, 6, and 24 h. (<b>J</b>) Line graphs and statistical analysis of wound healing assays. Data from independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-LV vehicle-treated controls. ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 3
<p>MYC overexpression alters the molecular response of HNSCC cells to trametinib. Total protein extracts were prepared from JHU22-LV and JHU22-MYC cells treated with either vehicle or trametinib (200 nM) for 48 h. (<b>A</b>) Western blot analysis was performed to detect pERK1/2 (<b>a</b>), total ERK1/2 (<b>b</b>), pEGFR (<b>c</b>) and β-Actin (<b>d</b>). (<b>B</b>) The pERK1/2/ERK1/2 ratio (<b>a</b>) or protein band intensity of pEGFR (<b>b</b>) was quantified and normalized to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells (n = 4). (<b>C</b>) Western blot analysis was also conducted to detect γH2A.X (<b>a</b>), pSer2/5 (<b>b</b>), pSer7 (<b>c</b>), and β-actin (<b>d</b>) in JHU22-LV and JHU22-MYC cells treated with vehicle or trametinib for 48 h. (<b>D</b>) The protein band intensities of γH2A.X (<b>a</b>), pSer2/5 (<b>b</b>), and pSer7 (<b>c</b>) were quantified and normalized to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells. Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-LV vehicle-treated controls. ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 4
<p>MYC overexpression in JHU22 HNSCC cells promotes cell cycle progression and apoptotic cell death in response to trametinib. (<b>A</b>) JHU22-LV and JHU22-MYC cells were treated with either vehicle or trametinib (200 nM) for 48 h, and flow cytometric analysis of cell cycle phases was performed following DAPI staining. Blue: G1; Yellow: S Phase; Green: G2/M; Magenta: Sum. (<b>B</b>) The percentage of cells in the G1 or G2/M phase in both JHU22-LV and JHU22-MYC cells was quantified (n = 4). (<b>C</b>) Total protein extracts were obtained from JHU22-LV and JHU22-MYC cells following treatment with either vehicle or trametinib (200 nM) for 48 h. Western blot analysis was conducted to detect p21 (<b>a</b>), p27 (<b>b</b>), E2F1 (<b>c</b>), and β-actin (<b>d</b>). (<b>D</b>) Protein band intensities of p21 (<b>a</b>), p27 (<b>b</b>), and E2F1 (<b>c</b>) were quantified and normalized to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells (n = 4). (<b>E</b>) JHU22-LV (<b>a</b>,<b>c</b>) and JHU22-MYC (<b>b</b>,<b>d</b>) cells were treated with either vehicle or trametinib (200 nM) for 48 h and then collected for annexin V and PI staining, followed by flow cytometric analysis to assess apoptotic cells. Pseudo color plots are used to indicate cell density or number. (<b>F</b>) The populations of apoptotic cells in both early and late stages were quantified (n = 4). (<b>G</b>) Western blot analysis was performed to detect cPARP/PARP (<b>a</b>), BIM (<b>b</b>), p53 (<b>c</b>), and β-actin (<b>d</b>) in JHU22-LV and JHU22-MYC cells. (<b>H</b>) The cPARP/PARP ratio (<b>a</b>) or protein band intensities of BIM (<b>b</b>) and p53 (<b>c</b>) were quantified relative to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells. Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-LV vehicle-treated controls. # <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 compared to JHU22-MYC vehicle-treated controls. ns: not significant. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 5
<p>Inhibition of trametinib-induced autophagy enhances apoptotic cell death in HNSCC cells. (<b>A</b>) Total protein extracts were prepared from JHU22-LV and JHU22-MYC cells treated with either vehicle or trametinib (200 nM) for 48 h. Western blot analysis was performed to detect LC3bI/II (<b>a</b>), p62 (<b>b</b>), and β-actin (<b>c</b>). (<b>B</b>) Protein band intensities of LC3bII (<b>a</b>) and p62 (<b>b</b>) were quantified relative to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells (n = 4). (<b>C</b>) JHU22-LV and JHU22-MYC cells were treated with vehicle (<b>a</b>,<b>e</b>), trametinib (<b>b</b>,<b>f</b>; 200 nM), HCQ (<b>c</b>,<b>g</b>; 10 µM), or the combination of trametinib and HCQ (<b>d</b>,<b>h</b>) for 48 h, then collected for annexin V and PI staining and analyzed via flow cytometry to assess early and late-stage apoptosis. (<b>D</b>) The populations of apoptotic cells in both early and late stages were quantified (n = 4). (<b>E</b>) Total protein extracts were prepared from JHU22-LV and JHU22-MYC cells treated with either vehicle, trametinib (200 nM), HCQ (10 µM), or the combination of trametinib and HCQ for 48 h. Western blot analysis was performed to detect cPARP/PARP and β-actin. (<b>F</b>) The ratio of cPARP/PARP was quantified to compare the effects between vehicle-treated and trametinib-treated cells. Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-LV vehicle-treated controls. ## <span class="html-italic">p</span> &lt; 0.01; ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. ns: not significant. Abbreviations: Veh: Vehicle, Tram: Trametinib, HCQ: hydroxychloroquine.</p>
Full article ">Figure 6
<p>Impact of trametinib on xenograft tumors overexpressing MYC in HNSCC. JHU22-LV and JHU22-MYC cells were injected subcutaneously into athymic nude mice to form xenograft tumors (n = 4). (<b>A</b>) Representative images of the gross tumors. (<b>B</b>) Average body weights of the mice were monitored during tumor development. (<b>C</b>) Tumor sizes were measured every 2–3 days following injection and calculated using the formula: length × width<sup>2</sup> × 0.5. (<b>D</b>) Comparison of the average weights of dissected tumors from mice. (<b>E</b>) IHC images showing MYC (JHU22-LV: a1, a2; JHU22-MYC: a3, a4), Ki-67 (JHU22-LV: b1, b2; JHU22-MYC: b3, b4), γH2A.X (JHU22-LV: c1, c2; JHU22-MYC: c3, c4), and cleaved-Casp3 (JHU22-LV: d1, d2; JHU22-MYC: d3, d4) in the HNSCC xenograft tumors treated with Veh or Tram, respectively. Scale bar = 30 µm (insets). (<b>F</b>) Quantification of the IHC staining for MYC (<b>a</b>), Ki-67 (<b>b</b>), γH2A.X (<b>c</b>), and cleaved-Caspase 3 (<b>d</b>). Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-LV vehicle-treated controls. ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib, cleaved-Casp3: cleaved-Caspase 3.</p>
Full article ">
14 pages, 2641 KiB  
Article
Germline Single-Nucleotide Polymorphism GFI1-36N Causes Alterations in Mitochondrial Metabolism and Leads to Increased ROS-Mediated DNA Damage in a Murine Model of Human Acute Myeloid Leukemia
by Jan Vorwerk, Longlong Liu, Theresa Helene Stadler, Daria Frank, Helal Mohammed Mohammed Ahmed, Pradeep Kumar Patnana, Maxim Kebenko, Eva Dazert, Bertram Opalka, Nikolas von Bubnoff and Cyrus Khandanpour
Biomedicines 2025, 13(1), 107; https://doi.org/10.3390/biomedicines13010107 - 5 Jan 2025
Viewed by 880
Abstract
Background/Objectives: GFI1-36N represents a single-nucleotide polymorphism (SNP) of the zinc finger protein Growth Factor Independence 1 (GFI1), in which the amino acid serine (S) is replaced by asparagine (N). The presence of the GFI1-36N gene variant is associated with a reduced DNA [...] Read more.
Background/Objectives: GFI1-36N represents a single-nucleotide polymorphism (SNP) of the zinc finger protein Growth Factor Independence 1 (GFI1), in which the amino acid serine (S) is replaced by asparagine (N). The presence of the GFI1-36N gene variant is associated with a reduced DNA repair capacity favoring myeloid leukemogenesis and leads to an inferior prognosis of acute myeloid leukemia (AML) patients. However, the underlying reasons for the reduced DNA repair capacity in GFI1-36N leukemic cells are largely unknown. Since we have demonstrated that GFI1 plays an active role in metabolism, in this study, we investigated whether increased levels of reactive oxygen species (ROS) could contribute to the accumulation of genetic damage in GFI1-36N leukemic cells. Methods: We pursued this question in a murine model of human AML by knocking in human GFI1-36S or GFI1-36N variant constructs into the murine Gfi1 gene locus and retrovirally expressing MLL-AF9 to induce AML. Results: Following the isolation of leukemic bone marrow cells, we were able to show that the GFI1-36N SNP in our model is associated with enhanced oxidative phosphorylation (OXPHOS), increased ROS levels, and results in elevated γ-H2AX levels as a marker of DNA double-strand breaks (DSBs). The use of free radical scavengers such as N-acetylcysteine (NAC) and α-tocopherol (αT) reduced ROS-induced DNA damage, particularly in GFI1-36N leukemic cells. Conclusions: We demonstrated that the GFI1-36N variant is associated with extensive metabolic changes that contribute to the accumulation of genetic damage. Full article
(This article belongs to the Special Issue Molecular Research on Acute Myeloid Leukemia (AML) Volume II)
Show Figures

Figure 1

Figure 1
<p>Increased OCR/ECAR ratio in the presence of the <span class="html-italic">GFI1-36N</span> SNP in both the human chronic myeloid leukemia (CML) cell line K562 and in the murine model of human acute myeloid leukemia (AML). (<b>A</b>): Oxygen consumption rate (OCR) was highest in <span class="html-italic">GFI1-36N</span>-K562 (ki/ki) cells while (<b>B</b>): extracellular acidification rate (ECAR) remained unchanged between <span class="html-italic">GFI1-36N</span> (ki/ki), <span class="html-italic">GFI1-36N</span> (+/ki), and <span class="html-italic">GFI1-36S</span> cells. (<b>C</b>): Increased OCR/ECAR ratio in <span class="html-italic">GFI1-36N</span>-K562 cells. (<b>D</b>): OCR and (<b>E</b>): ECAR were both increased in <span class="html-italic">GFI1-36N-MLL-AF9</span> cells compared to <span class="html-italic">GFI1-36S-MLL-AF9</span> controls. (<b>F</b>): Elevated OCR/ECAR ratio in <span class="html-italic">GFI1-36N-MLL-AF9</span> cells. FCCP: carbonyl cyanide-<span class="html-italic">p</span>-trifluoromethoxyphenylhydrazone; R + A: rotenone + antimycin A; 2-DG: 2-deoxyglucose. Mean ± SEM; ns: not significant; * <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">n</span> = 3–4.</p>
Full article ">Figure 2
<p>Alterations in mitochondrial metabolism. Increased (<b>A</b>): reactive oxygen species (ROS) and (<b>B</b>): mitochondrial membrane potential (MMP) with (<b>C</b>): decreased mitochondrial mass (mito-mass) in <span class="html-italic">GFI1-36N-MLL-AF9</span> cells. Mean ± SEM; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 3
<p>Radical scavengers N-acetylcysteine (NAC) and α-tocopherol (αT) reduced DNA damage in <span class="html-italic">GFI1-36N</span> leukemic cells. Leukemic bone marrow (BM) cells were incubated for 48 h with (<b>A</b>,<b>B</b>): 10 mM NAC or H<sub>2</sub>O or with (<b>C</b>,<b>D</b>): 30 μM αT or DMSO. After irradiation of the cells with 3 Gy, γ-H2AX MFI was determined by flow cytometry in <span class="html-italic">GFI1-36S</span> and <span class="html-italic">GFI1-36N</span> leukemic cells. B and D: After normalization to data from the untreated control, treated <span class="html-italic">GFI1-36N</span> leukemic cells showed fewer DNA double-strand breaks (DSBs) than <span class="html-italic">GFI1-36S</span> leukemic controls. Mean ± SEM; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 4
<p>Mass spectrometry discovered enrichment of pro-oncogenic metabolites in <span class="html-italic">GFI1-36N</span> leukemic cells. (<b>A</b>): Top 25 upregulated metabolite sets and (<b>B</b>)<b>:</b> upregulated metabolites in <span class="html-italic">GFI1-36N-MLL-AF9</span> cells. Mean ± SEM; ns: not significant; * <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">n</span> = 2–3.</p>
Full article ">Figure 5
<p>Relative cell count of <span class="html-italic">GFI1-36S-</span> and <span class="html-italic">GFI1-36N-MLL-AF9</span> cells after drug treatment normalized to control. <span class="html-italic">GFI1-36N-MLL-AF9</span> cells did not respond to metformin treatment and showed decreased sensitivity against treatment with UK5099, etoxomir, and BPTES. Mean ± SEM, ns: not significant; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; <span class="html-italic">n</span> = 3–6.</p>
Full article ">
14 pages, 13854 KiB  
Article
Comparison of Radio- and Phototoxicity in Association with an Enhancing Effect of the Photosensitizers Psoralen, Trioxsalen and Ortho-Iodo-Hoechst33258 on FaDu, PC-3, 4T1 and B16-F10 Cells
by Katja Tietze, Florian Brandt, Kerstin Wetzig, Lisa Hübinger, Marc Pretze, Ralph Alexander Bundschuh and Jörg Kotzerke
Biomedicines 2025, 13(1), 73; https://doi.org/10.3390/biomedicines13010073 - 31 Dec 2024
Viewed by 424
Abstract
Background: Energy delivered at different wavelengths causes different types of damage to DNA. Methods: PC-3, FaDu, 4T1 and B16-F10 cells were irradiated with different wavelengths of ultraviolet light (UVA/UVC) and ionizing radiation (X-ray). Furthermore, different photosensitizers (ortho-iodo-Hoechst33258/psoralen/trioxsalen) were tested for their amplifying effect. [...] Read more.
Background: Energy delivered at different wavelengths causes different types of damage to DNA. Methods: PC-3, FaDu, 4T1 and B16-F10 cells were irradiated with different wavelengths of ultraviolet light (UVA/UVC) and ionizing radiation (X-ray). Furthermore, different photosensitizers (ortho-iodo-Hoechst33258/psoralen/trioxsalen) were tested for their amplifying effect. Survival fraction and damage analysis using the γH2A.X assay (double-strand breaks) and the ELISA assay (cyclobutane pyrimidine dimers) were compared. Results: The PC-3 cells were found to be the most sensitive cells to the treatment strategies used. FaDu and PC-3 showed a strong sensitivity to UVA. Analysis of the damage showed that the cell lines exhibited different sensitivities. Conclusions: Thus, an enhancing effect of photosensitizers (PS) in combination with UVA could be demonstrated in some cases. However, this is cell- and dose-dependent. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Figure 1
<p>Nuclear staining of 4T1 with oIH, pso and tri. Subsequently, images were captured with the AxioObserver Z.1 fluorescence microscope (Carl Zeiss AG, Oberkochen, Germany; filter set 49; <math display="inline"><semantics> <mi mathvariant="normal">λ</mi> </semantics></math><sub>Ex</sub> = 365 nm, <math display="inline"><semantics> <mi mathvariant="normal">λ</mi> </semantics></math><sub>Em</sub> = 445 nm). Scale: 50 <math display="inline"><semantics> <mi mathvariant="normal">μ</mi> </semantics></math>m. Resolution: 1388 × 1040 pixel, 300 dpi. (<b>a</b>,<b>d</b>,<b>g</b>) DIC with oIH, pso and tri; (<b>b</b>,<b>e</b>,<b>h</b>) fluorescence; (<b>c</b>,<b>f</b>,<b>i</b>) merge images.</p>
Full article ">Figure 2
<p>Colony formation assay after treatment with X-rays (<b>a</b>), UVC (<b>b</b>), UVA (<b>c</b>) and with PS + UVA on FaDu (<b>d</b>), on PC-3 (<b>e</b>), on 4T1 (<b>f</b>), on B16-F10 (<b>g</b>).</p>
Full article ">Figure 3
<p>Colony formation assay and damage analysis as a comparison of PC-3 after treatment with X-rays (<b>a</b>), UVC (<b>b</b>), UVA (<b>c</b>), oIH and UVA (<b>d</b>), pso and UVA (<b>e</b>) and tri and UVA (<b>f</b>).</p>
Full article ">Figure A1
<p>Colony formation assay and damage analysis as a comparison of FaDu after treatment with X-rays (<b>a</b>), UVC (<b>b</b>), UVA (<b>c</b>), oIH and UVA (<b>d</b>), pso and UVA (<b>e</b>) and tri and UVA (<b>f</b>).</p>
Full article ">Figure A2
<p>Colony formation assay and damage analysis as a comparison of 4T1 after treatment with X-rays (<b>a</b>), UVC (<b>b</b>), UVA (<b>c</b>), oIH and UVA (<b>d</b>), pso and UVA (<b>e</b>) and tri and UVA (<b>f</b>).</p>
Full article ">Figure A3
<p>Colony formation assay and damage analysis as a comparison of B16-F10 after treatment with X-rays (<b>a</b>), UVC (<b>b</b>), UVA (<b>c</b>), oIH and UVA (<b>d</b>), pso and UVA (<b>e</b>) and tri and UVA (<b>f</b>).</p>
Full article ">Figure A3 Cont.
<p>Colony formation assay and damage analysis as a comparison of B16-F10 after treatment with X-rays (<b>a</b>), UVC (<b>b</b>), UVA (<b>c</b>), oIH and UVA (<b>d</b>), pso and UVA (<b>e</b>) and tri and UVA (<b>f</b>).</p>
Full article ">
18 pages, 14945 KiB  
Article
Long-Term Therapeutic Effects of 225Ac-DOTA-E[c(RGDfK)]2 Induced by Radiosensitization via G2/M Arrest in Pancreatic Ductal Adenocarcinoma
by Mitsuyoshi Yoshimoto, Kohshin Washiyama, Kazunobu Ohnuki, Ayano Doi, Miki Inokuchi, Motohiro Kojima, Brian W. Miller, Yukie Yoshii, Anri Inaki and Hirofumi Fujii
Pharmaceutics 2025, 17(1), 9; https://doi.org/10.3390/pharmaceutics17010009 - 24 Dec 2024
Viewed by 577
Abstract
Background: Alpha radionuclide therapy has emerged as a promising novel strategy for cancer treatment; however, the therapeutic potential of 225Ac-labeled peptides in pancreatic cancer remains uninvestigated. Methods: In the cytotoxicity study, tumor cells were incubated with 225Ac-DOTA-RGD2. [...] Read more.
Background: Alpha radionuclide therapy has emerged as a promising novel strategy for cancer treatment; however, the therapeutic potential of 225Ac-labeled peptides in pancreatic cancer remains uninvestigated. Methods: In the cytotoxicity study, tumor cells were incubated with 225Ac-DOTA-RGD2. DNA damage responses (γH2AX and 53BP1) were detected using flowcytometry or immunohistochemistry analysis. Biodistribution and therapeutic studies were carried out in BxPC-3-bearing mice. Results: 225Ac-DOTA-RGD2 demonstrated potent cytotoxicity against cells expressing αvβ3 or αvβ6 integrins and induced G2/M arrest and γH2AX expression as a marker of double-stranded DNA breaks. 225Ac-DOTA-RGD2 (20, 40, 65, or 90 kBq) showed favorable pharmacokinetics and remarkable tumor growth inhibition without severe side effects in the BxPC-3 mouse model. In vitro studies revealed that 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 swiftly induced G2/M arrest and elevated γH2AX expression. Furthermore, to clarify the mechanism of successful tumor growth inhibition for a long duration in vivo, we investigated whether short-term high radiation exposure enhances radiation sensitivity. Initially, a 4 h induction treatment with 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 enhanced both cytotoxicity and γH2AX expression with 0.5 kBq/mL of 225Ac-DOTA-RGD2 compared to a treatment with only 0.5 kBq/mL of 225Ac-DOTA-RGD2. Meanwhile, the γH2AX expression induced by 5 or 10 kBq/mL of 225Ac-DOTA-RGD2 alone decreased over time. Conclusions: These findings highlight the potential of using 225Ac-DOTA-RGD2 in the treatment of intractable pancreatic cancers, as its ability to induce G2/M cell cycle arrest enhances radiosensitization, resulting in notable growth inhibition. Full article
(This article belongs to the Section Clinical Pharmaceutics)
Show Figures

Figure 1

Figure 1
<p>In vitro cytotoxicity. (<b>a</b>) Cytotoxicity of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in human pancreatic tumor cell lines. (<b>b</b>) Comparison of cytotoxicity between <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> and <sup>225</sup>AcDOTA in BxPC-3. All assays were performed in triplicate. Data are presented as mean ± standard deviation.</p>
Full article ">Figure 2
<p>Induction of γH2AX and 53BP1 foci formation in response to increasing doses of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> at 24 h. (<b>a</b>) Representative images of γH2AX and 53BP1 foci obtained by immunofluorescence microscopy in BxPC-3 cells. Scale bar, 20 μm. (<b>b</b>) The number of γH2AX and 53BP1 foci per cell. Induction of γH2AX and 53BP1 foci in response to increasing doses of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> was monitored at 24 h. The number of γH2AX and 53BP1 foci per cell was counted, and 50–100 cells were analyzed. All assays were performed in triplicate. Data are presented as mean ± standard deviation and analyzed using a one-way analysis of variance with Dunn’s multiple-comparisons test (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Flow cytometric analysis of BxPC-3 after incubation with <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. (<b>a</b>) Representative fluorescence-activated cell sorting plots for γH2AX. The <span class="html-italic">y</span>-axis indicates γH2AX staining, and the <span class="html-italic">x</span>-axis is the DNA content. (<b>b</b>) Percentage of cells with γH2AX staining. All assays were performed in triplicate. (<b>c</b>) Percentage of cell cycle distribution (G1, S, and G2/M). All assays were performed in triplicate. Data are presented as the mean ± standard deviation (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>Biodistribution of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in BxPC-3-bearing mice. (<b>a</b>) Pharmacokinetics of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. Data are expressed as % ID/g for organs and blood and as % ID for carcass, urine, and feces. Data are shown as the mean ± standard deviation (<span class="html-italic">n</span> = 3–4). (<b>b</b>) Alpha camera imaging of intratumoral distribution and corresponding hematoxylin and eosin images. The scale bars indicate 100 μm. ID, injected dose.</p>
Full article ">Figure 5
<p>Therapeutic efficacy of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in BxPC-3-bearing mice. (<b>a</b>) Individual tumor responses. Each solid color line represents a tumor from a single mouse. (<b>b</b>) Relative tumor growth of the mice groups treated with a single dose of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> compared to the control group (untreated). Data are shown as the mean ± standard deviation. (<b>c</b>) Kaplan–Meier survival curves of the mice treated with <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. Log-rank (Mantel–Cox) test; <span class="html-italic">p</span> = 0.0192, hazard ratio [HR] 2.415, 95% CI 0.7639–7.636 (control vs. 20 kBq); <span class="html-italic">p</span> = 0.0014, HR 3.342, 95% CI 0.9631–11.60 (control vs. 40 kBq); <span class="html-italic">p</span> = 0.0002, HR 3.774, 95% CI 1.042–13.67 (control vs. 65 kBq); <span class="html-italic">p</span> = 0.0009, HR 3.786, 95% CI 1.062–13.49 (control vs. 90 kBq). (<b>d</b>) Change in body weight after administration of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. Data are shown as the mean ± standard deviation.</p>
Full article ">Figure 6
<p>Cytotoxicity, cell cycle, and γH2AX expression by low-dose <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> after 4 h of treatment with high-dose (5 or 10 kBq/mL) of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in BxPC-3 and PANC-1 cells. (<b>a</b>) Cell viability. The white, grey, and blue columns indicate the pretreatment with 0, 5, and 10 kBq/mL of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>, respectively. (<b>b</b>) Percentage of cell cycle distribution. (<b>c</b>) Time course of γH2AX expression. The significance of γH2AX expression at each time point was compared to 0, 5, or 10 kBq/mL as the control in each graph. Data represent the mean ± standard deviation (<span class="html-italic">n</span> = 2–4). * <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, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
17 pages, 5138 KiB  
Article
Lactoferrin Modulates Radiation Response Under Hypoxic Conditions, Possibly Through the Regulation of ROS Production in a Cell Type-Specific Manner
by Daitoku Murakami, Takahiro Fukazawa, Michihito Kyo, Mutsumi Miyauchi, Shigehiro Ono, Tomonao Aikawa, Nobuyuki Hirohashi and Keiji Tanimoto
Antioxidants 2025, 14(1), 1; https://doi.org/10.3390/antiox14010001 - 24 Dec 2024
Viewed by 710
Abstract
Lactoferrin (LF) is an iron-binding glycoprotein of the transferrin family and has been suggested to have a variety of biological functions, including anticancer activity. However, the effects of LF and its mechanisms in anticancer therapies, especially in radiotherapy against cancer cells under hypoxic [...] Read more.
Lactoferrin (LF) is an iron-binding glycoprotein of the transferrin family and has been suggested to have a variety of biological functions, including anticancer activity. However, the effects of LF and its mechanisms in anticancer therapies, especially in radiotherapy against cancer cells under hypoxic conditions, are not well-determined. In this study, we focused on the molecular mechanisms of LF functions in cells under hypoxic conditions. High-dose LF treatment showed cytotoxic activity in a variety of cells, including both non-cancer and cancer cells. Interestingly, hypoxic treatment increased the sensitivity to LF in some cancer cells but decreased it in non-cancer cells. LF treatment also altered sensitivity to radiation treatment: LF significantly increased the viability of irradiated KD non-cancer cells under hypoxic conditions but decreased that of HSC2 cancer cells. These effects were only observed when LF was treated within 3 h of irradiation, but not before irradiation. Importantly, knockdown of HIF1A counteracted these effects in both cell lines. Measurements of ROS activity showed that LF decreased ROS production in KD cells but increased it in HSC2 cells, resulting in a decrease in γH2AX foci in KD cells but an increase in HSC2 cells. RNA-seq and gene set enrichment analysis showed that LF treatment regulated gene expression related to the cell cycle, apoptosis, inflammation, and the NRF2 antioxidant signaling pathway. Quantitative RT-PCR confirmed the downregulation of the pro-apoptotic gene ASC in KD cells and the NRF2-regulated genes in HSC2 cells by LF treatment. Knockdown experiments confirmed the role of ASC in irradiated KD cells and NRF2 in irradiated HSC2 cells with LF treatment. In conclusion, lactoferrin was shown to affect radiation treatment by regulating apoptosis and NRF2 signaling in a cell type-specific manner under hypoxic conditions, suggesting its potential application as a protector or sensitizer for radiation therapy. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Cytotoxic activities of lactoferrin in different cell lines under normoxic or hypoxic conditions. Cytotoxic activities in cell lines under normoxic (21% O<sub>2</sub>) or hypoxic (1% O<sub>2</sub>) conditions were evaluated as IC<sub>50</sub> values using the MTT assay. Values are mean and SD (<span class="html-italic">n</span> = 3). Statistical significance is indicated by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (paired samples indicated).</p>
Full article ">Figure 2
<p>Effects of lactoferrin on radiation responses under normoxic or hypoxic conditions. (<b>A</b>) The experimental schedule is shown. Cells were cultured under normoxic (normoxia: 21% O<sub>2</sub>) or hypoxic (hypoxia: 1% O<sub>2</sub>) conditions for the indicated time periods. All cells were in normoxia when cells were irradiated. Lactoferrin was added to the cell culture medium at (<b>B</b>) 24 h before, (<b>C</b>) 0, (<b>D</b>) 1, (<b>E</b>) 2, or (<b>F</b>) 3 h after irradiation as indicated by arrowheads. After further incubation under normoxic or hypoxic conditions for 72 h, the relative cell viability of KD and HSC2 cells after γ-ray irradiation (0, 5, or 10 Gy) was evaluated using the MTT assay. (<b>G</b>) Cells were transfected with non-specific control (siNS) or siHIF1A 24 h after seeding and further incubated for 24 h before irradiation. Lactoferrin was added to the cell culture medium immediately after irradiation. After further incubation under hypoxic conditions for 72 h, the relative cell viability of KD and HSC2 cells after γ-ray irradiation (0, 5, or 10 Gy) was evaluated using the MTT assay. Values are mean and SD (<span class="html-italic">n</span> = 3). Statistical significance is indicated by * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Effects of lactoferrin on radiation-induced cell damage under hypoxic conditions. (<b>A</b>) KD or HSC2 cells (4 × 10<sup>3</sup> cells) were seeded on a 96-well plate. After 24 h culture, cells were exposed to 5 Gy of <sup>137</sup>Cs γ-rays and incubated with or without lactoferrin (10 μg/mL) for 24 h under hypoxic conditions. Reactive oxygen species (ROS) were evaluated using the DCFH-DA/H2DCFDA cellular ROS assay according to the manufacturer’s instructions and multiple images were captured using a BZ-8000 microscope. Positive cells per field were counted and calculated as relative ROS production in (<b>B</b>) KD and (<b>C</b>) HSC2 cells. Control indicates untreated cells. (<b>D</b>,<b>E</b>) KD or (<b>F</b>,<b>G</b>) HSC2 cells grown on coverslips were irradiated with 5 Gy of <sup>137</sup>Cs γ-rays under normoxic conditions and further incubated with or without lactoferrin (10 μg/mL) for 24 h under hypoxic conditions. Cells on coverslips were then fixed and stained with anti-γH2AX. Cell nuclei were stained with DAPI. Multiple images were captured using a BZ-8000 microscope, and γH2AX foci per nucleus were counted and averaged. Control indicates untreated cells. Values are mean and SD (<span class="html-italic">n</span> = 3). Statistical significance is indicated by <sup>+</sup> <span class="html-italic">p</span> &lt; 0.05 (Control vs. LF), * <span class="html-italic">p</span> &lt; 0.05 (paired samples indicated).</p>
Full article ">Figure 4
<p>Lactoferrin regulates multiple signaling pathways including apoptosis and NRF2 antioxidant responses. Comprehensive gene expression analysis followed by gene set enrichment analysis (GSEA) was performed on KD and HSC2 cells. (<b>A</b>) Gene sets with altered expression under hypoxic conditions were compared. (<b>Top</b>) Gene sets commonly altered; Middle: Gene sets altered in KD cells but not in HSC2 cells; (<b>Bottom</b>) Gene sets altered in HSC2 cells but not in KD cells. (<b>B</b>) Gene sets with altered expression by lactoferrin (LF) treatment were compared. (<b>Top</b>) Gene sets commonly altered; Middle: Gene sets altered in KD cells but not in HSC2 cells; (<b>Bottom</b>) Gene sets altered in HSC2 cells but not in KD cells. Gene sets upregulated (<b>C</b>) and downregulated (<b>D</b>) expression with lactoferrin treatment under hypoxic conditions in KD cells. Gene sets upregulated (<b>E</b>) and downregulated (<b>F</b>) expression with lactoferrin treatment under hypoxic conditions in HSC2 cells.</p>
Full article ">Figure 4 Cont.
<p>Lactoferrin regulates multiple signaling pathways including apoptosis and NRF2 antioxidant responses. Comprehensive gene expression analysis followed by gene set enrichment analysis (GSEA) was performed on KD and HSC2 cells. (<b>A</b>) Gene sets with altered expression under hypoxic conditions were compared. (<b>Top</b>) Gene sets commonly altered; Middle: Gene sets altered in KD cells but not in HSC2 cells; (<b>Bottom</b>) Gene sets altered in HSC2 cells but not in KD cells. (<b>B</b>) Gene sets with altered expression by lactoferrin (LF) treatment were compared. (<b>Top</b>) Gene sets commonly altered; Middle: Gene sets altered in KD cells but not in HSC2 cells; (<b>Bottom</b>) Gene sets altered in HSC2 cells but not in KD cells. Gene sets upregulated (<b>C</b>) and downregulated (<b>D</b>) expression with lactoferrin treatment under hypoxic conditions in KD cells. Gene sets upregulated (<b>E</b>) and downregulated (<b>F</b>) expression with lactoferrin treatment under hypoxic conditions in HSC2 cells.</p>
Full article ">Figure 5
<p>Lactoferrin regulates the gene expression of apoptosis, NRF2 signaling pathway, and DNA damage response genes. Effects of LF treatment on the expression of (<b>A</b>–<b>D</b>) apoptosis, (<b>E</b>–<b>I</b>) NRF2 signaling pathway, and (<b>J</b>–<b>M</b>) DNA damage response genes in KD and HSC2 cells under normoxic (N) and hypoxic (H) conditions for 24 h were evaluated by quantitative RT-PCR. Relative gene expression levels were calculated using <span class="html-italic">ACTB</span> expression as the denominator for each cell line (<span class="html-italic">n</span> = 3). The mean and SD are shown for all quantitative values. Statistical significance is indicated by + <span class="html-italic">p</span> &lt; 0.05 and ++ <span class="html-italic">p</span> &lt; 0.01 (N vs. H), * <span class="html-italic">p</span> &lt; 0.05, and ** <span class="html-italic">p</span> &lt; 0.01 (paired samples indicated).</p>
Full article ">Figure 6
<p>Dual role of lactoferrin in modulating radiation responses under hypoxic conditions through extrinsic apoptosis and NRF2. (<b>A</b>,<b>B</b>) KD and HSC2 cells were transfected with siRNA for non-specific (siNS), <span class="html-italic">PYCARD</span> (siASC), or <span class="html-italic">NFE2L2</span> (siNRF2) siRNA, and were incubated under normoxic (N) and hypoxic (H) conditions for 24 h. Expression of <span class="html-italic">PYCARD</span> (<span class="html-italic">ASC</span>) or <span class="html-italic">NFE2L2</span> (<span class="html-italic">NRF2</span>) was evaluated using quantitative RT-PCR. (<b>C</b>–<b>E</b>) Expression of NRF2 target genes in NRF2-knockdown KD and HSC2 cells was evaluated using quantitative RT-PCR. (<b>F</b>,<b>G</b>) To evaluate the response to radiation, cells were irradiated and treated with LF after siRNA transfection, then further incubated under hypoxic conditions for 72 h. Cell proliferation capacity was evaluated using the MTT assay. (<b>H</b>) Radiation-induced caspase-8 activation in KD and HSC2 cells with/without lactoferrin treatment under normoxic or hypoxic conditions was analyzed using immunoblots. β-actin was used as an internal loading control. Representative images of three independent experiments are shown. Values are mean and SD (<span class="html-italic">n</span> = 3). Statistical significance is indicated by + <span class="html-italic">p</span> &lt; 0.05 (N vs. H) and ++ <span class="html-italic">p</span> &lt; 0.01 (N vs. H), * <span class="html-italic">p</span> &lt; 0.05, and ** <span class="html-italic">p</span> &lt; 0.01 (paired samples indicated).</p>
Full article ">Figure 7
<p>Schematic representation of the dual role of lactoferrin in modulating radiation responses under hypoxic conditions through apoptosis and ferroptosis in non-cancer and cancer cells.</p>
Full article ">
16 pages, 2650 KiB  
Article
Insulin Sensitivity Controls Activity of Pathogenic CD4+ T Cells in Rheumatoid Arthritis
by Malin C. Erlandsson, Eric Malmhäll-Bah, Venkataragavan Chandrasekaran, Karin M. E. Andersson, Lisa M. Nilsson, Sofia Töyrä Silfverswärd, Rille Pullerits and Maria I. Bokarewa
Cells 2024, 13(24), 2124; https://doi.org/10.3390/cells13242124 - 22 Dec 2024
Viewed by 2363
Abstract
Hyperinsulinemia connects obesity, and a poor lipid profile, with type 2 diabetes (T2D). Here, we investigated consequences of insulin exposure for T cell function in the canonical autoimmunity of rheumatoid arthritis (RA). We observed that insulin levels correlated with the glycolytic index of [...] Read more.
Hyperinsulinemia connects obesity, and a poor lipid profile, with type 2 diabetes (T2D). Here, we investigated consequences of insulin exposure for T cell function in the canonical autoimmunity of rheumatoid arthritis (RA). We observed that insulin levels correlated with the glycolytic index of CD4+ cells but suppressed transcription of insulin receptor substrates, which was inversely related to insulin sensitivity. This connection between insulin levels and the glycolytic index was not seen in CD4+ cells of healthy controls. Exposure of CD4+ cells to insulin induced a senescent state recognized by cell cycle arrest and DNA content enrichment measured by flow cytometry. It also resulted in accumulation of DNA damage marker γH2AX. Insulin suppressed IFNγ production and induced the senescence-associated secretome in CD4+ cell cultures and in patients with hyperinsulinemia. Inhibition of JAK-STAT signaling (JAKi) improved insulin signaling, which activated the glycolytic index and facilitated senescence in CD4+ cell cultures. Treatment with JAKi was associated with an abundance of naïve and recent thymic emigrant T cells in the circulation of RA patients. Thus, we concluded that insulin exerts immunosuppressive ability by inducing senescence and inhibiting IFNγ production in CD4+ cells. JAKi promotes insulin effects and supports elimination of the pathogenic CD4+ cell in RA patients. Full article
(This article belongs to the Section Cellular Metabolism)
Show Figures

Figure 1

Figure 1
<p>Glycolytic index of CD4+ T cells is in proportion to plasma insulin levels in rheumatoid arthritis. (<b>A</b>) Insulin signaling pathway. (<b>B</b>) Scatter plot of glycolytic index (GI) in CD4+ cells of untreated RA (n = 16) and age matched HC (n = 41). Solid line indicates median. Dotted line indicates average level. (<b>C</b>) Scatter plot of correlation between GI and plasma insulin levels of RA patients (red) and HC (black). Solid lines indicate lineal regression curve. (<b>D</b>). Scatter plot of gene expression in CD4+ cells. Solid line indicates median. (<b>E</b>) Heatmap of Spearman correlation rho values between GI, plasma insulin, and IGF1 levels with the insulin signaling genes and serological parameters. Color scale bar indicates rho value range. (<b>F</b>) Heatmap of the log2 fold change (FC) difference in gene expression of CD4+ cells with high (GI-hi, n = 34) and low (GI-lo, n = 35) GI, and between RA and HC with high GI, by RNA-Seq. <span class="html-italic">p</span>-values were obtained by DESeq2 test. Color scale bar indicates log2FC range. (<b>G</b>) Bubble diagram of transcription factor target enrichment (by FDR) among DEGs upregulated in CD4+ cells with high and low GI. (<b>H</b>) Bar diagram of biological processes (by GO:terms) enriched among DEGs upregulated in CD4+ cells with high and low GI. * <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. RA, rheumatoid arthritis; HC, healthy controls; CRP, C-reactive protein; WBC, white blood cell count; IGF1, insulin-like growth factor 1; INSR, insulin receptor; IGF1R, IGF1 receptor; IRS, insulin receptor substrate; IFNγ, interferon gamma; FDR, false discovery rate; GI, glycolytic index; VEGF, vascular endothelial growth factor.</p>
Full article ">Figure 2
<p>Insulin induces senescence and suppresses IFNγ production in CD4+ cells. CD4+ cells of 18 healthy subjects were stimulated with anti-CD3 and insulin 10 nM for 48 h. DNA content (7AAD) was analyzed by flow cytometry in large-size cell (LSC) and small-size cell (SSC) subsets. <span class="html-italic">p</span>-values were obtained by paired Wilcoxon test. (<b>A</b>) Scatter plot of DNA content by mean fluorescence intensity (MFI) of 7AAD+ cells. Scatter plot of DNA content change with insulin treatment. Con, control; Ins, insulin treated. Solid line indicates median. (<b>B</b>) Histogram of 7AAD+ cell distribution by phases of the cell cycle. Colored areas correspond to G1 (blue), S (yellow) and G2 (green) phases. (<b>C</b>) Scatter plot of 7AAD+ cell frequency in cell cycle phases. (<b>D</b>) Scatter plot of change in proliferation dye CellTrace violet (CTV) in insulin-stimulated CD4+ cells. Solid line indicates median. (<b>E</b>) Confocal microscopy image of nuclear γH2AX enrichment in insulin-treated THP1 cells. Blue line confines nuclear area. (<b>F</b>) Scatter plot of γH2AX density in nuclei of insulin-treated THP1 cells. Solid line indicates median. (<b>G</b>) Scatter plot of gene expression in insulin-treated CD4+ cells, in relative quantity (RQ) to control cell cultures. Expression was measured by qPCR. (<b>H</b>) Dotblot images of cytokine levels in pooled supernatants of insulin-treated and control cells measured by cytokine array. FC, fold change. (<b>I</b>) Scatter plot of cytokine protein levels in supernatants, by specific ELISA. (<b>J</b>) Heatmap of gene expression difference by log2 FC, by RNA-Seq. Samples are grouped by high (n = 34) and low (n = 35) glycolytic index (GI), high (n = 12) and low (n = 44) insulin, and JAKi-treated (n = 24) and non-JAKi-treated patients (Other, n = 32). <span class="html-italic">p</span>-values were obtained by DESeq2 test. Color scale bar indicates log2FC range. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>JAK/STAT-inhibitors (JAKi) increase insulin sensitivity and aggravate senescence in CD4+ cells. (<b>A</b>) Cell cycle phases and control check points. (<b>B</b>) Heatmap of transcription difference in CD4+ cell transcriptome of RA patients with high (n = 12) and low (n = 44) plasma insulin, JAKi-treated (n = 24) or non-JAKi-treated (Other, n = 32) and HC with high (n = 34) and low (n = 35) glycolytic index (GI), by RNA-Seq. Difference between groups was calculated by DESeq2-test. Color scale bar presents log2FC range. * <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. (<b>C</b>) Scatter plot of GI in CD4+ cells split by plasma insulin levels within JAKi-treated (Hi, n = 7, Lo, n = 17) and non-JAKi-treated (Other, Hi, n = 5, Lo, n = 27) RA patients. Solid line indicates median. (<b>D</b>) Heatmap of gene transcription difference in insulin signaling and GI of CD4+ cells as in B. (<b>E</b>) Scatter plot of DNA content by mean fluorescent intensity (MFI) in JAKi-treated and control cell cultures (n = 10). Scatter plot of frequency of 7AAD+ cells in different phases of the cell cycle. Scatter plot of proliferation tracer cell trace violet (CTV) intensity in JAKi-stimulated and control CD4+ cells. Solid line indicates median. (<b>F</b>) Histogram of 7AAD+ cell distribution in small-size CD4+ cells (SSC). Colored areas indicate cells stimulated with JAKi (yellow), JAKi + insulin (blue), and control culture (red line). (<b>G</b>) Box plot of DNA content change by 7AAD and population size change in paired CD4+ cell cultures stimulated with JAKi and insulin (n = 6). <span class="html-italic">p</span>-values by paired Wilcoxon test. Dotted line indicates basal level. (<b>H</b>) Scatter plot of gene transcription in CD4+ cells stimulated with anti-CD3 and tofacitinib (JAKi, 10 µM; Con, 0 µM) for 48 h. mRNA levels were analyzed by qPCR and are presented in relative quantity (RQ) to mock treated cells.</p>
Full article ">Figure 4
<p>Insulin mitigates transcriptional effect of JAK-inhibitors (JAKi) in CD4+ T cells. CD4+ cells of RA patients were isolated by positive selection, activated on anti-CD3 for 2 h, and analyzed by RNA-Seq. Serum was used for protein measurements by immunosorbent assay. Differences between the groups were analyzed by DESeq2 test and Mann–Whitney U test. (<b>A</b>) Scatter plot of normalized gene transcription in JAKi-treated (Hi, n = 7, Lo, n = 17) and non-JAKi-treated patients (Other, Hi, n = 5, Lo, n = 27), split by high (Hi) and low (Lo) insulin levels. Solid line indicates median. (<b>B</b>) Scatter plot of IL6, IL8, survivin, and VEGF levels in serum. Solid line indicates median. (<b>C</b>) Heatmap of transcription difference by log2 fold change (FC) in CD4+ cells of RA patients with high (n = 12) and low (n = 44) plasma insulin levels, JAKi-treated (n = 24) or non-JAKi-treated (n = 32), by DESeq2. Color scale bar presents log2FC range. * <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. RT emigrants, recent thymic emigrants.</p>
Full article ">
23 pages, 3368 KiB  
Article
Microbeam Radiation Therapy Bio-Dosimetry Enhanced by Novel Radiosensitiser Combinations in the Treatment of Brain Cancer
by Michael Valceski, Elette Engels, Sarah Vogel, Jason Paino, Dylan Potter, Carolyn Hollis, Abass Khochaiche, Micah Barnes, Alice O’Keefe, Matthew Cameron, Kiarn Roughley, Anatoly Rosenfeld, Michael Lerch, Stéphanie Corde and Moeava Tehei
Cancers 2024, 16(24), 4231; https://doi.org/10.3390/cancers16244231 - 19 Dec 2024
Viewed by 3014
Abstract
Background/Objectives: Brain cancer is notoriously resistant to traditional treatments, including radiotherapy. Microbeam radiation therapy (MRT), arrays of ultra-fast synchrotron X-ray beams tens of micrometres wide (called peaks) and spaced hundreds of micrometres apart (valleys), is an effective alternative to conventional treatments. MRT’s [...] Read more.
Background/Objectives: Brain cancer is notoriously resistant to traditional treatments, including radiotherapy. Microbeam radiation therapy (MRT), arrays of ultra-fast synchrotron X-ray beams tens of micrometres wide (called peaks) and spaced hundreds of micrometres apart (valleys), is an effective alternative to conventional treatments. MRT’s advantage is that normal tissues can be spared from harm whilst maintaining tumour control. Combining MRT with targeted radiosensitisers, such as nanoparticles, chemotherapeutic drugs, and halogenated pyrimidine drugs, can further improve radiotherapy by enhancing radiation damage. However, the underlying mechanisms of MRT are still being understood, which is essential to ensuring the reliable and successful use of MRT. Methods: An in vitro study was performed using γH2AX imaging, and quantification was performed via confocal microscopy and a clonogenic cell survival assay. Results: We show that methotrexate chemotherapeutics and iododeoxyuridine enhance MRT cell-killing and thulium oxide nanoparticles (TmNPs) broaden MRT peaks, and using γH2AX immunofluorescent confocal microscopy to quantify DNA damage, we further our knowledge of MRT mechanisms. γH2AX images verify the biological responses of cells aligning with the physical collimation of MRT, and we can accurately measure MRT microbeam characteristics bio-dosimetrically. The peak-to-valley dose ratio (PVDR), the ratio of the peak dose to the valley dose that characterises an MRT field, was accurately measured biologically using γH2AX imaging, despite studies previously finding this challenging. Conclusions: The measurement of biological PVDR has been performed for the first time with high-Z radiosensitisers, including nanoparticles, and several novel radiosensitiser-enhanced MRT mechanisms were discovered. Our results deepen our understanding of MRT with radiosensitisers, and can contribute to its accurate and future successful use in treating cancer. Full article
(This article belongs to the Special Issue Application of Fluorescence Imaging in Cancer)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Cell survival for various treatments and radiation fields. Clonogenic assay reveals the long-term cell survival of 9LGS in response to (<b>a</b>) various radiosensitisers (TmNPs and IUdR) and (<b>b</b>) combinations with MTX (Tm+MTX, IUdR+MTX) when irradiated. RT modalities include conventional broadbeam (CBB) orthovoltage X-rays (at lower (solid green) and higher (solid blue) dose fractions), compared with synchrotron broadbeam (SBB) X-rays (solid red) and microbeam radiation therapy (MRT), with lower and higher dose fields of 0.5 Gy (striped blue) and 5 Gy (striped yellow) (MRT doses listed are the prescribed valley doses, whereas the peak doses are PVDR = 8.9 times greater). Error bars represent the standard error of the mean (SEM) (using standard deviations at the 95% confidence interval). An average of 6 replicate samples (n = 6) is used across independent repeats.</p>
Full article ">Figure 2
<p>γH2AX immunofluorescent confocal microscopy comparing multiple MRT peaks for various radiosensitisers. Panel displays 20x dry resolution images of 9LGS cells as the maximum projection of 5-slice Z-stacks. All images are overlays of two channels, with double-strand DNA breaks (DSBs) represented by green γH2AX foci overlayed on a Hoechst 33342 nuclear counterstain (blue). Radiosensitiser treatments are displayed in columns across the panel and show DSB changes 20 min after irradiation began for all cases in 9LGS cells. Radiation treatments change down the two rows to 0.5 Gy (valley dose) synchrotron MRT (<b>top</b>) and 5 Gy (valley dose) MRT (<b>bottom</b>). PVDR = 8.9 for all. Images are representative of a set acquired for each sample time. Each sample that was imaged was fixed at 20 min post-irradiation.</p>
Full article ">Figure 3
<p>Quantification of γH2AX confocal images broken into peak and valley regions of interest. Images with 93× resolution are used to quantify DNA damage in treatments across peaks and valleys separately, compared with CBB and SBB fields. Results yielded DSB enhancement ratios for both 0.5 Gy and 5 Gy valleys and their respective peaks (PVDR = 8.9) relative to 0 Gy cells only. All results are expressed in terms of the ratio of FF of each treatment normalised to the FF of the 0 Gy control. Error bars represent the standard error of the mean (using standard deviations at the 95% confidence interval). Each collection of images for each treatment and radiation regimen was analysed to obtain an average FF across at least 6 replicate images for each treatment type for 0.5 Gy and 5 Gy MRT field fractions (MRT doses listed are the prescribed valley doses). All data points displayed in this figure represent the average of at least six quantified images across independent experimental trials (n = 6) and display error bars representing the standard error of the mean (SEM). Each sample that was imaged was fixed at 20 min post-irradiation.</p>
Full article ">Figure 4
<p>MRT beam profiles and peak widths expressed via γH2AX foci. (<b>a</b>) Beam profiles from 20× dry confocal microscopy images of MRT peaks for each treatment at both 0.5 Gy (blue) and 5 Gy (orange) (MRT doses listed are the prescribed valley doses) are shown to reveal peak broadening, with the left column showing MTX-negative treatments and the right showing MTX-positive treatments. These data are expressed as a histogram showing intensity as a percentage of the average peak (of the MRT peak) intensity vs. the off-axis lateral distance in microns. Radiation-only MRT is shown in the top row, followed by TmNP treatments in the middle and IUdR treatments at the bottom. (<b>b</b>) Peak widths are shown to quantify broadening results in subfigure (<b>a</b>) and are the average of the profile data across 16 peak segment ROIs taken across independent repeats.</p>
Full article ">Figure 4 Cont.
<p>MRT beam profiles and peak widths expressed via γH2AX foci. (<b>a</b>) Beam profiles from 20× dry confocal microscopy images of MRT peaks for each treatment at both 0.5 Gy (blue) and 5 Gy (orange) (MRT doses listed are the prescribed valley doses) are shown to reveal peak broadening, with the left column showing MTX-negative treatments and the right showing MTX-positive treatments. These data are expressed as a histogram showing intensity as a percentage of the average peak (of the MRT peak) intensity vs. the off-axis lateral distance in microns. Radiation-only MRT is shown in the top row, followed by TmNP treatments in the middle and IUdR treatments at the bottom. (<b>b</b>) Peak widths are shown to quantify broadening results in subfigure (<b>a</b>) and are the average of the profile data across 16 peak segment ROIs taken across independent repeats.</p>
Full article ">Figure 5
<p>Biological PVDR values (BPVDR) for all treatments. MRT fields at 0.5 Gy (blue) and 5 Gy (orange) doses (MRT doses listed are the prescribed valley doses) compare the biological effect of MRT peak vs. valley doses relative to the physical dosimetry. Quantitative analysis of 93x resolution γH2AX confocal images broken into peak and valley regions of interest (ROIs) allowed DSB FF factors to be obtained for peaks and valleys separately and then allowed the ratio of peak to valley to be found. All BPVDR (Equation (3)) values are taken as the average of ratios found individually for at least 6 replicate images. The red line in each graph represents the physical PVDR of 8.9. (<b>a</b>) BPVDR is shown for the case in which the broadening of the peak is considered. (<b>b</b>) BPVDR for the cases in which the peak width changes are not considered, and ROIs are fixed at 50 µm in width and centred to align with the image peak.</p>
Full article ">Figure A1
<p>γH2AX image panel of 9LGS with various treatments and radiation fields. Synchrotron broadbeam (SBB) and conventional broadbeam (CBB) orthovoltage X-rays at 5 Gy for the cell-only control (Control) and each radiosensitiser agent and combination are compared with MRT fields with 0.5 Gy and 5 Gy prescribed valley doses (peaks at PVDR = 8.9 times the valley dose). Using a confocal microscope at 93× resolution, the green channel representing γH2AX foci (correlating with DSBs) is overlayed on the blue channel representing 9LGS cell nuclei with a DAPI nuclear counterstain.</p>
Full article ">Figure A2
<p>Overlay of the ratios of the mass energy absorption coefficient of radiosensitiser elements relative to water. Both thulium and iodine coefficients of µ<sub>en</sub>/ρ for total mass energy absorption are shown normalised relative to water with the 150 kVp photon energy spectrum for the Nucletron Oldelft Therapax DXT 300 Series 3 Orthovoltage unit (Nucletron B.V., Veenendaal, The Netherlands) at the Prince of Wales Hospital, Randwick, Sydney, NSW, Australia. The data for these spectra were produced using XMuDat and SpekCalc and sourced from Boone and Chavez, 1996 [<a href="#B49-cancers-16-04231" class="html-bibr">49</a>,<a href="#B50-cancers-16-04231" class="html-bibr">50</a>,<a href="#B71-cancers-16-04231" class="html-bibr">71</a>].</p>
Full article ">
14 pages, 5269 KiB  
Article
HDAC Inhibitors Can Enhance Radiosensitivity of Head and Neck Cancer Cells Through Suppressing DNA Repair
by Jennifer Antrobus, Bethany Mackinnon, Emma Melia, Jonathan R. Hughes and Jason L. Parsons
Cancers 2024, 16(23), 4108; https://doi.org/10.3390/cancers16234108 - 7 Dec 2024
Viewed by 957
Abstract
Background/Objectives: The incidence of head and neck squamous cell carcinoma (HNSCC), currently ~800,000 cases per year worldwide, is rising. Radiotherapy remains a mainstay for the treatment of HNSCC, although inherent radioresistance, particularly in human papillomavirus (HPV)-negative disease subtypes, remains a significant barrier to [...] Read more.
Background/Objectives: The incidence of head and neck squamous cell carcinoma (HNSCC), currently ~800,000 cases per year worldwide, is rising. Radiotherapy remains a mainstay for the treatment of HNSCC, although inherent radioresistance, particularly in human papillomavirus (HPV)-negative disease subtypes, remains a significant barrier to effective treatment. Therefore, combinatorial strategies using drugs or inhibitors against specific cellular targets are necessary to enhance HNSCC radiosensitivity to lead to an improvement in patient outcomes. Given that radiotherapy acts through targeting and damaging DNA, a common strategy is to focus on enzymes within DNA-dependent cellular pathways, such as DNA damage repair. Methods: Here, we have employed a 3D spheroid model of HNSCC (FaDu) in combination with a targeted drug screen to identify novel radiosensitisers that suppress tumour growth. Results: We identified that histone deacetylases (HDACs) were prominent candidates, and subsequently identified that the HDAC inhibitors mocetinostat and pracinostat, as well as the combined HDAC–epidermal growth factor receptor inhibitor CUDC-101, were effective at radiosensitising cell models of HNSCC (FaDu, A253, UMSCC11b) through their impact on both spheroid growth and clonogenic survival assays. We also demonstrated that this combinatorial strategy leads to inhibition of the repair of DNA double-strand breaks through the neutral comet assay and γH2AX foci analysis using immunofluorescence microscopy, providing a mechanism of action through which HDAC inhibition functions in HNSCC radiosensitisation. Conclusions: We believe that this approach should be further investigated in preclinical models, in order to realise the full therapeutic potential of HDAC inhibition for the radiosensitisation of HNSCC, eventually leading to improved patient treatment efficacy and outcomes. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Screening of a selected number of FDA-approved drugs in the radiosensitisation of HNSCC cells. (<b>A</b>) Schematic of the drug-radiation screen utilising FaDu cells grown as 3D spheroids and then analysing growth over a period of 3–15 days post-seeding. Analysis of FaDu spheroid growth between 3 and 15 days post-seeding after a single 1 Gy dose of X-rays in the presence of 183 drugs at either (<b>B</b>) 0.03 µM or (<b>C</b>) 1 µM dose. Growth was analysed from a single replicate and normalised against the DMSO-treated control (first lane, yellow bar), which was set to 1.0. Indicated in green arrows are the HDAC inhibitors identified as potential radiosensitisers.</p>
Full article ">Figure 2
<p>Validation of mocetinostat, CUDC-101, and pracinostat in suppressing HNSCC spheroid growth in combination with X-ray irradiation. Growth of FaDu spheroids in the presence of (<b>A</b>) mocetinostat, (<b>B</b>) CUDC-101, and (<b>C</b>) pracinostat (all 0.2 µM) alone, and following a 2 Gy or 4 Gy dose of X-rays in comparison to the DMSO-treated control. Shown is the mean fold increase in spheroid volume ± SE across three independent experiments normalised against the volume observed on day 3 post-seeding, which was set to 1.0. (<b>D</b>–<b>F</b>) Images of the spheroids with the different treatments on days 3 and 15 post-seeding. (<b>G</b>–<b>I</b>) Fold inhibition in spheroid volume ± S.E as a function of radiation dose determined between days 3 and 15 post-seeding across three independent experiments normalised to the respective unirradiated control, which was set to 1.0. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.002 on two-sample <span class="html-italic">t</span>-tests.</p>
Full article ">Figure 3
<p>Effect of mocetinostat, CUDC-101, and pracinostat on HNSCC cell viability, clonogenic survival, and histone acetylation. (<b>A</b>–<b>C</b>) FaDu cells were treated with increasing doses of (<b>A</b>) mocetinostat, (<b>B</b>) CUDC-101, or (<b>C</b>) pracinostat (0.01–100 µM), and cell viability was measured via CellTiter Blue from three biologically independent experiments. Shown is the mean cell viability ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 on two-sample <span class="html-italic">t</span>-tests compared to the untreated controls. (<b>D</b>) FaDu, (<b>E</b>) A253, and (<b>F</b>) UMSCC11b cells were treated with mocetinostat, CUDC-101, or pracinostat (all 1 µM) in comparison to DMSO as control and clonogenic survival of cells, analysed from three independent experiments. * <span class="html-italic">p</span> &lt; 0.01, ** <span class="html-italic">p</span> &lt; 0.001 on one-sample <span class="html-italic">t</span>-tests compared to the DMSO-treated controls. (<b>G</b>) FaDu cells were treated with DMSO (Dm), 1 µM mocetinostat (Mo), CUDC-101 (Cu), or pracinostat (Pr), and either unirradiated (control) or irradiated with 4 Gy X-rays and cells harvested at 2 h post-irradiation. Histones were purified by acid extraction and analysed by immunoblotting using antibodies targeted against site-specific acetylation sites on histone H3 or H4.</p>
Full article ">Figure 4
<p>Analysis of the impact of HDAC inhibitors on the clonogenic survival of HNSCC cells post-irradiation. (<b>A</b>,<b>D</b>) FaDu, (<b>B</b>,<b>E</b>) A253, and (<b>C</b>,<b>F</b>) UMSCC11b treated with mocetinostat, CUDC-101, or pracinostat (all 1 µM) in comparison to DMSO as a control and clonogenic survival of cells with increasing doses of X-ray irradiation was analysed with three independent experiments. Shown is the mean surviving fraction ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.0005, *** <span class="html-italic">p</span> &lt; 0.0001, analysed with the LQ model.</p>
Full article ">Figure 5
<p>Mocetinostat, CUDC-101, and pracinostat cause delays in DSB repair in HNSCC cells. (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) FaDu and (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) A253 cells were treated with 1 µM CUDC-101, pracinostat, or DMSO as a control, and either unirradiated (control) or irradiated with 4 Gy X-rays and cells harvested at the various time-points post-irradiation. (<b>A</b>,<b>B</b>) Levels of DSBs were analysed directly using the neutral comet assay, with mean percentage tail DNA ± SE determined from three independent experiments. (<b>C</b>,<b>D</b>) Numbers of γH2AX foci were determined using immunofluorescence microscopy, with mean γH2AX foci/cell ± SE determined from three independent experiments. * <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 on one-sample <span class="html-italic">t</span>-tests. (<b>E</b>,<b>F</b>) Stained DNA in cells either unirradiated or 4 h post-irradiation following inhibitor treatment through gel electrophoresis. (<b>G</b>,<b>H</b>) γH2AX foci in cells either unirradiated or 8 h post-irradiation following inhibitor treatment. Scale bar is 20 µm.</p>
Full article ">
16 pages, 3617 KiB  
Article
Temporal Changes Toward Cellular Senescence in Rat Dental Pulp Stem Cells Induced by Long-Term In Vitro Culture
by Shanshan Zheng, Masato Nakagawa, Yanan Gong, Yasuhiko Matsushima, Satoshi Sasayama, Shunsuke Baba and Yoshitomo Honda
Appl. Sci. 2024, 14(23), 11376; https://doi.org/10.3390/app142311376 - 6 Dec 2024
Viewed by 601
Abstract
Rat dental pulp stem cells (DPSCs) can be used to elucidate mesenchymal stem cell (MSC) applications in regenerative medicine. However, information on rat DPSCs during long-term passage, which could lead to replicative senescence, is limited. In this study, we investigated the phenotypic changes [...] Read more.
Rat dental pulp stem cells (DPSCs) can be used to elucidate mesenchymal stem cell (MSC) applications in regenerative medicine. However, information on rat DPSCs during long-term passage, which could lead to replicative senescence, is limited. In this study, we investigated the phenotypic changes in DPSCs after 3–26 passages (3P–26P). The results show that cell morphology and nuclear size increase proportionally with passage number. The phosphorylated histone H2A.X (γ-H2A.X) positive cells (indicating DNA damage) increased significantly earlier than the 4-Hydroxynonenal (4-HNE) stained cells (indicating an abundance of intracellular reactive oxygen species). Compared to the cells subjected to 3P and 5P, the cells subjected to 15P showed reduced proliferation despite being positive for Ki67. Furthermore, cell growth was completely arrested after 26P. The senescence markers, senescence-associated β-galactosidase (SA-β-gal) and p16, exhibited similar expression patterns that were not correlated with those of p21 and urokinase-type plasminogen activator receptor (uPAR). Nearly all cells expressed SA-β-gal and p16 after 26P, whereas only half expressed p21 and uPAR. These results will contribute to understanding the characteristics of DPSCs toward replicative senescence, which are applicable to elucidate mechanisms related to regenerative medicine and stem cell aging. Full article
Show Figures

Figure 1

Figure 1
<p>Establishment of primary and senescent dental pulp stem cells (DPSCs) from 5-week-old male Wistar/ST rats. (<b>A</b>) Schematic showing preparation of primary and senescent DPSCs. (<b>B</b>) Macroscopic images of rat mandibular incisor and pulp (arrowhead). (<b>C</b>) Microscopic observation of DSPCs passaged 0 times.</p>
Full article ">Figure 2
<p>Expression patterns of surface antigens on rat DPSCs after passaging. Flow cytometric analysis used to detect surface antigens: CD34 for hematopoietic stem cells and endothelial cells marker, CD45 as leucocyte marker, and CD90 for mesenchymal stem cell marker. 3P, 5P, 15P, and 26P indicate 3, 5, 15, and 26 passages.</p>
Full article ">Figure 3
<p>Changes in rat DPSC shapes and nuclei morphologies. Microscopic observations of cell shapes (<b>A</b>) and nuclei (<b>B</b>) after 3, 5, 15, and 26 passages (3P, 5P, 15P, and 26P, respectively) and their semi-quantified data (<span class="html-italic">n</span> = 50, violin plot) on cell and nuclear size. (<b>C</b>) Orthogonal views (X–Z planes) from three-dimensional reconstruction images on nuclei and quantitative data of those volumes (<span class="html-italic">n</span> = 20, violin plot). * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Changes in F-actin in rat DPSCs. Staining of actin filament using fluorescent phalloidin.</p>
Full article ">Figure 5
<p>DNA damage within nuclei and endogenous cytosolic reactive oxygen species (ROS) production in rat DPSCs. (<b>A</b>) Representative immunofluorescence staining of phosphorylated histone H2A.X (γ-H2A.X) and 4-Hydroxynonenal (4-HNE) with DAPI. (<b>B</b>) Quantitative data for (<b>A</b>). Data presented as mean with standard deviation (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, and **** <span class="html-italic">p</span> &lt; 0.0001; one-way analysis of variance (ANOVA) with Tukey’s test. (<span class="html-italic">n</span> = 3; each data point represents percentage of positive cells (or area) calculated by counting positive cells (or area) per 50 cells (or nuclei) in random areas).</p>
Full article ">Figure 6
<p>Proliferation profiles of DPSCs. (<b>A</b>) Cell Counting Kit-8 (CCK-8) assays. Comparison of proliferation between 15P and 26P with 3P and 5P. Two-way ANOVA with Tukey’s test; <span class="html-italic">n</span> = 5. (<b>B</b>) Immunofluorescence staining of Ki67 in DPSCs and (<b>C</b>) their semi-quantified data. Data presented as mean with SD. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001; one-way ANOVA with Tukey’s test. (<span class="html-italic">n</span> = 3; each data point represents percentage of positive cells (or area) calculated by counting positive cells (or area) per 50 cells (or nuclei) in random areas).</p>
Full article ">Figure 7
<p>Expression of cellular senescence markers. Immunofluorescence staining of four senescence markers for DPSCs at low (<b>A</b>) and high (<b>B</b>) magnifications. Arrowheads: magnified cells for (<b>B</b>). (<b>C</b>) Semi-quantified data from panels A and B. Data presented as mean with 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, and **** <span class="html-italic">p</span> &lt; 0.0001; one-way ANOVA with Tukey’s test. (<span class="html-italic">n</span> = 3; each data point represents percentage of positive cells calculated by counting positive cells per 50 cells in random areas).</p>
Full article ">Figure 8
<p>Gene expression of senescence-associated genes. * <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, and **** <span class="html-italic">p</span> &lt; 0.0001; ns: no significant difference between any groups; one-way ANOVA with Tukey’s test; <span class="html-italic">n</span> = 3.</p>
Full article ">
21 pages, 2725 KiB  
Article
Impact of Gold Nanoparticles and Ionizing Radiation on Whole Chromatin Organization as Detected by Single-Molecule Localization Microscopy
by Myriam Schäfer, Georg Hildenbrand and Michael Hausmann
Int. J. Mol. Sci. 2024, 25(23), 12843; https://doi.org/10.3390/ijms252312843 - 29 Nov 2024
Viewed by 646
Abstract
In radiation tumor therapy, irradiation, on one hand, should cause cell death to the tumor. On the other hand, the surrounding non-tumor tissue should be maintained unaffected. Therefore, methods of local dose enhancements are highly interesting. Gold nanoparticles, which are preferentially uptaken by [...] Read more.
In radiation tumor therapy, irradiation, on one hand, should cause cell death to the tumor. On the other hand, the surrounding non-tumor tissue should be maintained unaffected. Therefore, methods of local dose enhancements are highly interesting. Gold nanoparticles, which are preferentially uptaken by very-fast-proliferating tumor cells, may enhance damaging. However, the results in the literature obtained from cell culture and animal tissue experiments are very contradictory, i.e., only some experiments reveal increased cell killing but others do not. Thus, a better understanding of cellular mechanisms is required. Using the breast cancer cell model SkBr3, the effects of gold nanoparticles in combination with ionizing radiation on chromatin network organization were investigated by Single-Molecule Localization Microscopy (SMLM) and applications of mathematical topology calculations (e.g., Persistent Homology, Principal Component Analysis, etc.). The data reveal a dose and nanoparticle dependent re-organization of chromatin, although colony forming assays do not show a significant reduction of cell survival after the application of gold nanoparticles to the cells. In addition, the spatial organization of γH2AX clusters was elucidated, and characteristic changes were obtained depending on dose and gold nanoparticle application. The results indicate a complex response of ALU-related chromatin and heterochromatin organization correlating to ionizing radiation and gold nanoparticle incorporation. Such complex whole chromatin re-organization is usually associated with changes in genome function and supports the hypothesis that, with the application of gold nanoparticles, not only is DNA damage increasing but also the efficiency of DNA repair may be increased. The understanding of complex chromatin responses might help to improve the gold nanoparticle efficiency in radiation treatment. Full article
(This article belongs to the Special Issue Metal Nanoparticles: From Fundamental Studies to New Applications)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Clonogenic survival of SkBr3 cells after irradiation with indicated doses of 6 MV X-rays in the presence and absence of 10 nm gold nanoparticles, respectively. A semi-logarithmic plot of cell survival is shown. The red curve corresponds to SkBr3 cells, treated with gold nanoparticles and irradiated. The blue curve corresponds to SkBr3 cells, irradiated, but not treated with gold nanoparticles. Error bars are the standard deviation of surviving fractions across three replicates. No differences in the survival fractions were found with and without gold nanoparticle incorporation. (Note: This figure was originally published in [<a href="#B31-ijms-25-12843" class="html-bibr">31</a>] and is reproduced with permission of The Licensor through PLSclear.) (<b>B</b>) SMLM image of a cell nucleus reconstructed from the orte matrix of the ALU labeling sites. The intensity of the points represents the number of next neighbors. (Scale bar: 1 µm.) (<b>C</b>) Histogram of the mean numbers of ALU signals detected in cell nuclei for different radiation conditions. Light-red columns represent control cells not treated with gold nanoparticles. Dark-red columns represent cells incubated with gold nanoparticles (“NG”). The error bars show the standard deviation of signal numbers. (<b>D</b>) Ripley distance frequency distribution of ALU loci for different treatment conditions. The curves represent the relative frequencies of the pairwise distances of all ALU point loci-measured. (Note: Control cells are those named 0 Gy in (<b>A</b>,<b>C</b>)). (<b>E</b>) PCA of the persistent homology data of the ALU loci in clusters (see peak in (<b>D</b>), which shows an increase of short distances below 200 nm). Mean values of component 1 vs. component 0 are shown. In this latent space, “component 0” is the vector in the n-dimensional orthogonal vector space of persistent imaging with the largest variability (variance). Component 1 is a vector orthogonal to component 0 with the second largest variance. The error bars of the components represent the standard deviation. The application of gold nanoparticle leads to a right shift in component 0 if the results for the same doses are compared for specimens with and without gold nanoparticle incorporation. With harsher treatment, the variation between the individual nuclei is increasing.</p>
Full article ">Figure 2
<p>(<b>A</b>) Example of a merged image of γH2AX clusters (green) within the ALU point pattern (blue; see also <a href="#ijms-25-12843-f001" class="html-fig">Figure 1</a>B). The cell nucleus was treated with gold nanoparticles and then irradiated with 2 Gy. Areas of overlap between γH2AX and ALU are indicated in red. Scale bar: 1 µm. (<b>B</b>) Histogram of the mean numbers of γH2AX signal points detected in cell nuclei for different treatment conditions. The blue columns represent the mean numbers of γH2AX signals in cells detected 30 min after irradiation. The light-blue columns are the mean numbers of signal points in cells not treated with gold nanoparticles. The dark-blue columns are the mean numbers of signal points from cells incubated with gold nanoparticles (“NG”). Error bars show the standard deviation of signal numbers. (Note: This figure was originally published in [<a href="#B31-ijms-25-12843" class="html-bibr">31</a>] and is reproduced with permission of The Licensor through PLSclear.) (<b>C</b>) Histogram of the mean numbers of γH2AX clusters detected in cell nuclei for different radiation conditions. The light-blue columns represent control cells not treated with gold nanoparticles. The dark blue columns represent cells incubated with gold nanoparticles (“NG”). The error bars show the standard deviation of signal numbers. (Note: This figure was originally published in [<a href="#B31-ijms-25-12843" class="html-bibr">31</a>] and is reproduced with permission of The Licensor through PLSclear.) (<b>D</b>) Ripley distance frequency distributions of γH2AX points for different treatment conditions. The curves show the relative frequencies of the pairwise distances of the detected points. (Note: Control cells are those named 0 Gy in (<b>A</b>,<b>C</b>).) In addition, treatment conditions of 0.5 Gy are shown, which were not included in the survival experiments of <a href="#ijms-25-12843-f001" class="html-fig">Figure 1</a>A. (<b>E</b>) PCA of the persistent homology data of the γH2AX loci in cluster (see peak in (<b>D</b>)). Mean values of component 1 vs. component 0 are shown. In this latent space, “component 0” is the vector in the n-dimensional orthogonal vector space of persistent imaging with the largest variability (variance). Component 1 is a vector orthogonal to component 0, with the second-largest variance. The error bars of the components represent the standard deviation. The application of gold nanoparticles leads to a right shift in component 0 if the data of the same irradiation dose with and without nanogold application are compared. With harsher treatment, the variation between the individual nuclei increases, as indicated by the increasing standard deviations.</p>
Full article ">Figure 3
<p>(<b>A</b>) Absolute number of fluorescent points detected per image (z-section of 500 nm) for the different experiments (contr = non-irradiated control; 1 + 1 Gy = radiation scheme 1 Gy—30 min—1 Gy; ng = gold nanoparticles; co = control without gold nanoparticles; for further details see text). Each detected point represents a fluorescent antibody against the H3K9 methylation site of heterochromatin. Blinking events within the localization precision were registered as one point. The boxplots show the mean point number of the detected nuclei (green triangle), the median (orange line), the lower and upper quantile (box), and the value range within +/−2 standard deviations (black line). The black circles refer to values that differ more than 1 box length from the median (Note: This figure and a slightly modified figure legend were originally published under CC BY license in [<a href="#B15-ijms-25-12843" class="html-bibr">15</a>].) (<b>B</b>) Ripley distance frequency distributions of H3K9me3 points (H3K9 methylation sites) for different treatment conditions. (<b>C</b>) Histogram of the relative frequencies of the next neighbors (NN) of each H3K9me3 locus. (Note for (<b>B</b>,<b>C</b>): contr = irradiated control in comparison to the same experiment with gold nanoparticles (NG); 2× 1 Gy = radiation scheme 1 Gy—30 min—1 Gy; NG control = cells with gold nanoparticles but without radiation treatment; contr-contr = control cells without gold nanoparticles and without radiation exposure.) (<b>D</b>) PCA of the persistent homology data of the H3K9me3 loci. Mean values of component 1 vs. component 0 are shown. In this latent space, “component 0” is the vector in the n-dimensional orthogonal vector space of persistent imaging with the largest variability (variance). Component 1 is a vector orthogonal to component 0 with the second-largest variance. The error bars of the components represent the standard deviation. The radiation scheme 1 Gy—30 min—1 Gy has no significant difference to the respective control, while the application of gold nanoparticles in general leads to shifts in component 0.</p>
Full article ">Figure 4
<p>PCA of the persistent homology data of the H3K9me3 loci 30 min after the application of radiation of different doses (no gold nanoparticle application). Mean values of component 1 vs. component 0 are shown. In this latent space “component 0” is the vector in the n-dimensional orthogonal vector space of persistent imaging with the largest variability (variance). Component 1 is a vector orthogonal to component 0 with the second-largest variance. The error bars represent the standard deviation.</p>
Full article ">Figure 5
<p>(<b>A</b>) Typical example of an artificial image obtained from data of the orte matrix for γH2AX signals 30 min after treatment of SkBr3 cells with gold nanoparticles and 2 Gy radiation exposure. This image is merged with the real widefield DAPI image (shadowed). (Note: This figure and a slightly modified figure legend were originally published under CC BY license in [<a href="#B15-ijms-25-12843" class="html-bibr">15</a>]). (<b>B</b>) Absolute number of fluorescent points detected per image (z-section of 500 nm) for the different experiments; (contr = non-irradiated control; 1 + 1 Gy = radiation scheme 1 Gy—30 min—1 Gy; ng = gold nanoparticles; co = control without gold nanoparticles; for further details see text). Each detected point represents a fluorescent antibody against γH2AX. Blinking events within the localization precision were registered as one point. The boxplots show the mean point number of the detected nuclei (green triangle), the median (orange line), the lower and upper quantile (box), and the value range within +/−2 standard deviations (black line). The black circles refer to values that differ more than 1 box length from the median (Note: This figure and a slightly modified figure legend were originally published under CC BY license in [<a href="#B15-ijms-25-12843" class="html-bibr">15</a>]). (<b>C</b>) Histogram of the relative frequencies of the next neighbors (NN) of each γH2AX locus. (<b>D</b>) Ripley distance frequency distributions of γH2AX points for different treatment conditions. The curves show the relative frequencies of the pairwise distances of the points. (<b>E</b>) PCA of the persistent homology data of the γH2AX loci in cluster (see peak in (<b>D</b>)). Mean values of component 1 vs. component 0 are shown. In this latent space, “component 0” is the vector in the n-dimensional orthogonal vector space of persistent imaging with the largest variability (variance). Component 1 is a vector orthogonal to component 0 with the second-largest variance. The error bars of the components represent the standard deviation. Depending on the treatment, not only the size of the γH2AX clusters is changing but also the internal topological arrangement, which results in the different component values.</p>
Full article ">Figure 6
<p>Schematic representation of the different evaluation steps of SMLM datasets. After acquisition of a time series of image frames, the coordinates and other values of all the blinking events of a cell nucleus were integrated into a matrix, the so-called “orte-matrix” (top-left). In the one-channel analysis, the coordinate values of the points were subjected to Ripley statistics. For the representative graph (top right), the point-to-point distances were calculated and represented in a normalized distance frequency histogram. In the lower part, the processes of persistent homology, persistent imaging, and principal component analysis are shown. The point pattern is transferred into a bar code description of components and holes. The lengths of the bars (difference of the α values of the end and the beginning of a particular bar) provide the lifetime; the beginning provides the birth in the one-point cloud. The one-point clouds for all cell nuclei that are considered for evaluation are transferred in pixel images (persistent imaging). Each pixel is compared for all images. The results of these comparisons span an n-dimensional orthogonal vector space. The variations of the pixel values determine the components of the principal component analysis. Finally, the outcome for the components (orthogonal vector values) with the largest variation and the second largest variation determines the latent space (graph bottom right) (for further details, see text in Material and Methods).</p>
Full article ">
16 pages, 9375 KiB  
Article
The Loss of Tafazzin Transacetylase Activity Is Sufficient to Drive Testicular Infertility
by Paige L. Snider, Elizabeth A. Sierra Potchanant, Catalina Matias, Donna M. Edwards, Jeffrey J. Brault and Simon J. Conway
J. Dev. Biol. 2024, 12(4), 32; https://doi.org/10.3390/jdb12040032 - 26 Nov 2024
Viewed by 856
Abstract
Barth syndrome (BTHS) is a rare, infantile-onset, X-linked mitochondriopathy exhibiting a variable presentation of failure to thrive, growth insufficiency, skeletal myopathy, neutropenia, and heart anomalies due to mitochondrial dysfunction secondary to inherited TAFAZZIN transacetylase mutations. Although not reported in BTHS patients, male infertility [...] Read more.
Barth syndrome (BTHS) is a rare, infantile-onset, X-linked mitochondriopathy exhibiting a variable presentation of failure to thrive, growth insufficiency, skeletal myopathy, neutropenia, and heart anomalies due to mitochondrial dysfunction secondary to inherited TAFAZZIN transacetylase mutations. Although not reported in BTHS patients, male infertility is observed in several Tafazzin (Taz) mouse alleles and in a Drosophila mutant. Herein, we examined the male infertility phenotype in a BTHS-patient-derived D75H point-mutant knockin mouse (TazPM) allele that expresses a mutant protein lacking transacetylase activity. Neonatal and adult TazPM testes were hypoplastic, and their epididymis lacked sperm. Histology and biomarker analysis revealed TazPM spermatogenesis is arrested prior to sexual maturation due to an inability to undergo meiosis and the generation of haploid spermatids. Moreover, TazPM testicular mitochondria were found to be structurally abnormal, and there was an elevation of p53-dependent apoptosis within TazPM seminiferous tubules. Immunoblot analysis revealed that TazPM gamete genome integrity was compromised, and both histone γ-H2Ax and Nucleoside diphosphate kinase-5 protein expression were absent in juvenile TazPM testes when compared to controls. We demonstrate that Taz-mediated transacetylase activity is required within mitochondria for normal spermatogenesis, and its absence results in meiotic arrest. We hypothesize that elevated TazPM spermatogonial apoptosis causes azoospermia and complete infertility. Full article
(This article belongs to the Special Issue Feature Papers in Journal of Developmental Biology 2025)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Tafazzin</span> mRNA expression during postnatal <span class="html-italic">wt</span> mouse testis growth. (<b>A</b>) Quantitative triplicate PCR revealed <span class="html-italic">Taz</span> levels are robust in newborn, P5, P10 and P16 testes (averaging ~3.67-fold), but is expressed at basal levels in P21, P28 and adult (4-month) testes. <span class="html-italic">Taz</span> expression was compared to housekeeping gene <span class="html-italic">Ppia</span>. qPCR data are presented as a logarithmic plot of relative expression, where a value of 1 indicates no difference in 4-month-old adult testes and values &lt; 1 indicate reduced and &gt;1 indicate increased expression. The Y-axis is the relative fold difference, and error bars represent SD. (<b>B</b>–<b>E</b>) Non-radioactive in situ hybridization detection of <span class="html-italic">Taz</span> (purple precipitate) in staged postnatal mouse testis sections revealed punctate robust expression within nascent seminiferous tubules at P5 (<b>B</b>), P16 (<b>C</b>) and P21 (<b>D</b>), but only low-level expression in P28 seminiferous tubules (<b>E</b>). <span class="html-italic">Taz</span> signal was only observed when sections were hybridized with the <span class="html-italic">Taz</span> anti-sense probe, confirming signal specificity. Serial sections were examined for comparable spatiotemporal patterns in at least three consecutive serial sections/stage. Scale bars (<b>B</b>–<b>E</b>) = 50 μm.</p>
Full article ">Figure 2
<p>Characterization of <span class="html-italic">Taz<sup>PM</sup></span> testis phenotype. (<b>A</b>) Adult 4-month testis/body weight ratio in <span class="html-italic">wt</span> (black, n = 4) verses <span class="html-italic">Taz<sup>PM</sup></span> knockin (red, n = 6) mice. (<b>B</b>–<b>G</b>) Representative images of whole <span class="html-italic">wt</span> (<b>B</b>,<b>D</b>,<b>F</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>C</b>,<b>E</b>,<b>G</b>) 4-month (<b>B</b>,<b>C</b>), P17 (<b>D</b>,<b>E</b>) and P5 (<b>F</b>,<b>G</b>) littermate testes. (<b>H</b>–<b>O</b>) Hematoxylin and eosin staining of <span class="html-italic">wt</span> (<b>H</b>,<b>J</b>,<b>L</b>,<b>N</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>I</b>,<b>K</b>,<b>M</b>,<b>O</b>) testes (<b>H</b>,<b>I</b>) and epididymis (<b>J</b>,<b>K</b>) from 4-month-old mice, as well as P28 (<b>L</b>,<b>M</b>) and P17 (<b>N</b>,<b>O</b>) testis sections. (<b>P</b>,<b>Q</b>) Analysis of 4-month epidydimal <span class="html-italic">wt</span> and <span class="html-italic">Taz<sup>PM</sup></span> content numbers ((<b>P</b>), n = 7/genotype) and resultant staining of sperm head nuclei DNA chromatin (blue DAPI-positive), mitochondria-rich midpiece (green MitoTracker-positive) and tail (red actin-positive) within contents. (<b>R</b>,<b>S</b>) Oil red-O staining of lipid deposition in P28 <span class="html-italic">wt</span> (<b>R</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>S</b>) testes. Scale bars (<b>B</b>,<b>C</b>,<b>E</b>) = 1 mm; (<b>D</b>,<b>F</b>,<b>G</b>) = 500 μm; (<b>H</b>,<b>I</b>,<b>K</b>,<b>R</b>,<b>S</b>) = 20 μm; (<b>J</b>,<b>K</b>) = 50 μm; (<b>L</b>,<b>M</b>,<b>N</b>,<b>O</b>,<b>Q</b>) = 10 μm. Statistical significance set at **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Hook1 and Dazl biomarker analysis. (<b>A</b>–<b>F</b>) Immunostaining using Hook1 antibody (signal is brown DAB precipitate) expression in 4-month (<b>A</b>,<b>B</b>), P17 (<b>C</b>,<b>D</b>) and P5 (<b>E</b>,<b>F</b>) <span class="html-italic">wt</span> (<b>A</b>,<b>C</b>,<b>E</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>B</b>,<b>D</b>,<b>F</b>) testis sections. (<b>G</b>–<b>L</b>) Immunostaining using Dazl antibody (signal is brown DAB precipitate) expression in 4-month (<b>G</b>,<b>H</b>), P17 (<b>I</b>,<b>J</b>) and P5 (<b>K</b>,<b>L</b>) <span class="html-italic">wt</span> (<b>G</b>,<b>I</b>,<b>K</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>H</b>,<b>J</b>,<b>L</b>) testis sections. Scale bars (<b>A</b>,<b>B</b>,<b>E</b>,<b>F</b>,<b>K</b>,<b>L</b>) = 20 μm; (<b>C</b>,<b>D</b>,<b>G</b>,<b>H</b>,<b>I</b>,<b>J</b>) = 50 μm.</p>
Full article ">Figure 4
<p>Molecular analysis of <span class="html-italic">Taz<sup>PM</sup></span> testis phenotype. (<b>A</b>,<b>B</b>) Western evaluation of Taz and Vdac protein levels in triplicate <span class="html-italic">wt</span> and <span class="html-italic">Taz<sup>PM</sup></span> P28 testes ((<b>A</b>), n = 3 per genotype). Both mitochondria Hsp70 and housekeeping Gapdh were used as independent loading controls, with Taz; Gapdh quantification and statistical analysis (via two-tailed t-test) shown (<b>B</b>). The Y-axis is the relative fold difference. (<b>C</b>,<b>D</b>) Representative electron microscopy images showing P28 <span class="html-italic">wt</span> normal (<b>C</b>) and <span class="html-italic">Taz<sup>PM</sup></span> swollen/abnormal (<b>D</b>) mitochondrial morphology (n = 4/genotype). (<b>E</b>) Ultra-performance liquid chromatography analysis (n = 5/genotype) revealed that ATP, ADP, AMP, and NAD<sup>+</sup> levels as well as ATP/ADP and ATP/AMP ratios remain unchanged, but total adenine nucleotides (Ad Sum) are reduced (<span class="html-italic">p</span> = 0.05) in <span class="html-italic">Taz<sup>PM</sup></span> (red) verses <span class="html-italic">wt</span> (black) testes (n = 5/genotype). Scale bars (<b>C</b>,<b>D</b>) = 500 nm.</p>
Full article ">Figure 5
<p>Spermatogenic failure in <span class="html-italic">Taz<sup>PM</sup></span> testes. (<b>A</b>–<b>D</b>) Non-radioactive in situ hybridization detection of <span class="html-italic">Catsper1</span> (<b>A</b>,<b>B</b>) and <span class="html-italic">Catsper4</span> (<b>C</b>,<b>D</b>) mRNA in immature P28 <span class="html-italic">wt</span> (<b>A</b>,<b>C</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>B</b>,<b>D</b>) testis sections. (<b>E</b>–<b>H</b>) <span class="html-italic">Cracd</span> mRNA in P28 (<b>E</b>,<b>F</b>) and P17 (<b>G</b>,<b>H</b>) <span class="html-italic">wt</span> (<b>E</b>,<b>G</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>F</b>,<b>H</b>) testis sections. (<b>I</b>–<b>L</b>) <span class="html-italic">Nme5</span> mRNA in P28 (<b>I</b>,<b>J</b>) and P17 (<b>K</b>,<b>L</b>) <span class="html-italic">wt</span> (<b>I</b>,<b>K</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>J</b>,<b>L</b>) testis sections. Scale bars (<b>A</b>–<b>J</b>) = 20 μm; (<b>K</b>,<b>L</b>) = 10 μm.</p>
Full article ">Figure 6
<p><span class="html-italic">Taz<sup>PM</sup></span> testes exhibit persistent p53-associated apoptosis. (<b>A</b>–<b>F</b>) Immunohistological detection of phosphohistone H3 cell proliferation marker (punctate brown DAD-positivity) in 4-month (<b>A</b>,<b>B</b>), P28 (<b>C</b>,<b>D</b>) and P17 (<b>E</b>,<b>F</b>) <span class="html-italic">wt</span> (<b>A</b>,<b>C</b>,<b>E</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>B</b>,<b>D</b>,<b>F</b>) testis sections. (<b>G</b>–<b>L</b>) TUNEL apoptosis marker expression (punctate brown DAD-positivity) in methyl green counterstained P28 (<b>G</b>,<b>H</b>), P17 (<b>I</b>,<b>J</b>) and P5 (<b>K</b>,<b>L</b>) <span class="html-italic">wt</span> (<b>G</b>,<b>I</b>,<b>K</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>H</b>,<b>J</b>,<b>L</b>) testis sections. (<b>M</b>,<b>N</b>). Western analysis of total p53, phospho p53<sup>S392</sup>, Sirt1, γ-H2Ax (both upper ubiquitinated and lower phospho γ-H2Ax isoforms) and Nme5 protein levels was performed in triplicate for <span class="html-italic">wt</span> and <span class="html-italic">Taz<sup>PM</sup></span> P28 testes ((<b>M</b>), n = 3 per genotype). Housekeeping Gapdh was used as the loading control, with quantification and statistical analysis (via two-tailed <span class="html-italic">t</span>-tests for all except Sirt1/Nme5 which used one-tailed t-tests due to <span class="html-italic">wt</span> variability) shown (<b>N</b>). Scale bars (<b>A</b>–<b>J</b>) = 50 μm; (<b>K</b>,<b>L</b>) = 20 μm.</p>
Full article ">
20 pages, 8899 KiB  
Article
B Chromosome Transcriptional Inactivation in the Spermatogenesis of the Grasshopper Eyprepocnemis plorans
by Juan Luis Santos, María Teresa Parra, Sara Arévalo, Andrea Guajardo-Grence, Jesús Page, José Ángel Suja, Carlos García de la Vega and Alberto Viera
Genes 2024, 15(12), 1512; https://doi.org/10.3390/genes15121512 - 25 Nov 2024
Viewed by 679
Abstract
Background/Objectives: We analyzed the relationship between synapsis, recombination, and transcription during the spermatogenesis of the grasshopper Eyprepocnemis plorans carrying B chromosomes (type B1). Methods: The progression of synapsis was interpreted according to the dynamics of the cohesin subunit SMC3 axes. DNA double-strand breaks [...] Read more.
Background/Objectives: We analyzed the relationship between synapsis, recombination, and transcription during the spermatogenesis of the grasshopper Eyprepocnemis plorans carrying B chromosomes (type B1). Methods: The progression of synapsis was interpreted according to the dynamics of the cohesin subunit SMC3 axes. DNA double-strand breaks were revealed by RAD51 immunolabeling, while transcriptional activity was determined by the presence of RNA polymerase II phosphorylated at serine 2 (pRNApol II) immunolabeling. The two repressive epigenetic modifications, histone H3 methylated at lysine 9 (H3K9me3) and histone H2AX phosphorylated at serine 139 (γ-H2AX), were employed to reveal transcriptional inactivity. Results: During prophase I, spermatocytes with one B1 chromosome showed overall transcription except in the regions occupied by both the X and the B1 chromosomes. This transcriptional inactivity was accompanied by the accumulation of repressive epigenetic modifications. When two B1 chromosomes were present, they could appear as a fully synapsed monochiasmatic bivalent, showing intense H3K9me3 labeling and absence of pRNApol II, while γ-H2AX labeling was similar to that shown by the autosomes. Conclusions: According to our results, B1 transcriptional inactivation was triggered in spermatogonia, long before the beginning of meiosis, and was accompanied by H3K9me3 heterochromatinization that was maintained throughout spermatogenesis. Moreover, when two B1 were present, the transcriptional inactivation did not preclude synapsis and recombination achievement by these chromosomes. Full article
(This article belongs to the Section Cytogenomics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Dynamics of the B1 univalent and bivalent revealed by SMC3 immunolabeling. Prophase I spread spermatocytes from individuals with a B1 univalent (<b>A</b>–<b>E</b>) or a B1 bivalent (<b>F</b>–<b>J</b>). The positions of the sex chromosome (X), the B1 univalent (B), the B1 bivalent (B<sub>II</sub>) and of autosomal bivalents (Au) are indicated. Nuclei are counterstained with DAPI (blue). (<b>A</b>) At leptotene, SMC3 axes (white) are present all along the chromosomes. Thus, the identification of the X and B1 chromosomes was impeded by the imbrication of the SMC3 axes. (<b>B</b>) During zygotene, SMC3 autosomal axes were gradually paired into thicker filaments except at the X and B1 chromosomes. (<b>C</b>) At pachytene, autosomal bivalents displayed fully paired SMC3 axes, whereas a single thin SMC3 axis was observed in the X and B1 chromosomes. (<b>D</b>) 300% magnification of the nuclear region occupied by the X and B1 chromosomes. (<b>E</b>) Electron microscopy microphotograph of a pachytene spermatocyte showing the unsynapsed X and B1 chromosomes. (<b>F</b>) Leptotene spermatocytes with thin SMC3 axes all over the nucleus. (<b>G</b>) During zygotene, the SMC3 axes of the B1 chromosomes were gradually paired, as in the case of autosomes. (<b>H</b>) Pachytene spermatocyte showing fully paired SMC3 axes all along the autosomal bivalents, whereas pairing delay was observable in one of the ends of the B1 bivalent. (<b>I</b>) 300% magnifications of selected B1 bivalents with unpaired ends (red arrowheads), or with complete paired SMC3 axes as the autosomes (Au). In contrast, the X univalent exhibited a single unpaired SMC3 axis. (<b>J</b>) Electron microscopy microphotograph of a pachytene spermatocyte with fully synapsed B1 bivalent. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 2
<p>Meiotic reciprocal recombination in B1 chromosomes. Double immunolabeling of SMC3 (white) and RAD51 (red) in squashed spermatocytes counterstained with DAPI (blue) in individuals without B1 chromosomes (0B) and bearing one B1 (1B) or two B1 chromosomes (2B). The positions of the sex chromosome (X), the B1 univalent (B), the B1 bivalent (B<sub>II</sub>) and autosomal bivalents (Au) are indicated. Note that in prophase I stages, the X and B1 chromosomes are heteropycnotic positive chromatin bodies after DAPI staining. (<b>A</b>) Pachytene spermatocyte of a 0B male. RAD51 foci are visible along paired SMC3 axes of autosomes, but not over the X chromosome. (<b>B</b>) Pachytene spermatocyte of a 1B male. RAD51 foci are located over the fully paired SMC3 axes of autosomes but not over the unpaired SMC3 axes of the X and B1 chromosomes. (<b>C</b>) 300% magnification of the nuclear region occupied by the X and B1 chromosomes (yellow arrowheads signal RAD51 focus on an autosomal bivalent). (<b>D</b>) Zygotene spermatocyte of a 2B male. RAD51 foci were found over paired and unpaired SMC3 axes of autosomal bivalents and the B1 bivalent. (<b>E</b>) 300% magnification of the nuclear region occupied by the X chromosome and the B1 bivalent. Yellow and green arrowheads signal RAD51 foci located on autosomal and B1 bivalents, respectively. (<b>F</b>) Early/mid pachytene spermatocyte of a 2B male. Autosomal and B1 bivalents displayed RAD51 foci over their paired SMC3 axes. (<b>G</b>) 300% magnification of the nuclear region occupied by the X chromosome and the B1 bivalent. Yellow and green arrowheads signal RAD51 foci located on autosome and B bivalents, respectively. (<b>H</b>) Metaphase I spermatocyte of a 2B1 male. SMC3 appeared at the interchromatid domain of autosomal and B1 bivalents and was interrupted at chiasma sites. (<b>I</b>) 300% magnification of the X chromosome and the B1 bivalent. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 3
<p>Transcriptional activity of B1 chromosomes during prophase I stages. Double immunolabelings of SMC3 (white) and pRNApol II (red) in prophase I spread spermatocytes counterstained with DAPI (blue) from individuals with one B1 (<b>A</b>–<b>D</b>) or two B1 chromosomes (<b>E</b>–<b>H</b>). The positions of the sex chromosome (X), the B1 univalent (B) and the B1 bivalent (B<sub>II</sub>) are indicated, and in (<b>B</b>–<b>D</b>,<b>F</b>–<b>H</b>), their positions are outlined. (<b>A</b>) In leptotene, pRNApol II labeling appeared spread throughout most of the nucleus although reduced at some regions (yellow arrowheads). (<b>B</b>–<b>D</b>) During zygotene (<b>B</b>,<b>C</b>) and pachytene (<b>D</b>), pRNApol II labeling covered most of the autosomal chromatin irrespectively of their pairing status. The labeling was clearly reduced only in particular regions possibly corresponding to chromosome ends (yellow arrowheads). pRNApol II labeling is absent in the regions occupied by both the X and the B1 univalents. (<b>E</b>,<b>F</b>) The pattern of the pRNApol II labeling in individuals with two B1 chromosomes is identical to that described above. Significantly, the region occupied by the B1s does not present any signal from leptotene up to pachytene. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 4
<p>H3K9me3 labeling during prophase I stages. Double immunolabelings of SMC3 (white) and H3K9me3 (green) in prophase I spread spermatocytes from individuals with one (<b>A</b>–<b>D</b>) and two B1 chromosomes (<b>E</b>–<b>H</b>). The positions of the sex chromosome (X), the B1 univalent (B) and the B1 bivalent (B<sub>II</sub>) are indicated. (<b>A</b>) At leptotene, an intense H3K9me3 signal was visible in a particular nuclear region and discrete accumulations appeared in the rest of the nucleus. (<b>B</b>–<b>D</b>) From zygotene (<b>B</b>,<b>C</b>) and up to pachytene (<b>D</b>), the large H3K9me3 labeling was maintained and was located in the regions occupied by the X and B1 chromosomes. Additional H3K9me3 signals were positioned at some ends of the SMC3 axes of autosomal bivalents (yellow arrowheads). (<b>E</b>–<b>H</b>) In individuals carrying two B1 chromosomes, the H3K9me3 labeling was identical to that described above. The large signal corresponded to the positions of the X chromosome and the B1 bivalent. Moreover, some ends of autosome SMC3 axes presented bright foci (yellow arrowheads). Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 5
<p>γ-H2AX labeling during prophase I stages. Double immunolabelings of SMC3 (white) and γ-H2AX (purple) in prophase I spread spermatocytes from individuals with one B1 (<b>A</b>–<b>D</b>) and two B1 chromosomes (<b>E</b>–<b>K</b>). The positions of the sex chromosome (X), the B1 univalent (B), the B1 bivalent (B<sub>II</sub>) and autosomes (Au) are indicated. (<b>A</b>) γ-H2AX labeling became visible at leptotene as discrete flares over the unpaired SMC3 axes. (<b>B</b>) At early zygotene, γ-H2AX labeling was only observable over the yet unpaired SMC3 axes of the autosomal bivalents. Both the X and B1 chromosomes did not exhibit γ-H2AX signal. (<b>C</b>) By late zygotene, as DSBs repair proceeded, γ-H2AX was restricted to the last unpaired regions of the SMC3 autosomal axes and covered the unpaired SMC3 axes of the X and B1. (<b>D</b>) In early/mid pachytene, discrete γ-H2AX foci were positioned over the trajectories of the fully paired autosomal SMC3 axes. In contrast, the X and B1 univalents presented γ-H2AX ribbons covering their single SMC3 axes. (<b>E</b>) In individuals with two B1 chromosomes, γ-H2AX labeling become visible at leptotene. (<b>F</b>) By early zygotene, γ-H2AX labeling spread over the nucleus, covering unpaired autosomal SMC3 axes. (<b>G</b>) At late zygotene, γ-H2AX distribution was limited to the last unpaired regions of the SMC3 axes of autosomes and the B1 bivalent and over the single unpaired axis of the X chromosome. (<b>H</b>,<b>I</b>) Early and mid-pachytene spermatocytes presented γ-H2AX labeling as foci over paired SMC3 axes at autosomal bivalents and as a more continuous ribbon over the unpaired SMC3 axis of the X chromosome. (<b>J</b>,<b>K</b>) 300% magnifications of the nuclear region occupied by the X and B1 bivalent during early (<b>J</b>) and mid-pachytene (<b>K</b>). The B1 bivalent appeared labeled by γ-H2AX over the unpaired ends of the SMC3 axes (yellow arrowheads in (<b>J</b>)). By late pachytene (<b>K</b>), the B1 bivalent was fully paired and there was no γ-H2AX labeling. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 6
<p>Transcriptional activity in <span class="html-italic">E. plorans</span> spermatogonial cells<b>.</b> Double immunolabelings of H3K9me3 (green) with either γ-H2AX (purple) or pRNApol II (red) in squashed spermatogonia counterstained with DAPI (blue) from males without (0B), with one (1B) or two (2B) B1 chromosomes<b>.</b> The position of the sex chromosome (X) is indicated. (<b>A</b>,<b>B</b>) Spermatogonia from 0B individuals presented H3K9me3 accumulated at the X chromosome and at certain nuclear regions (white arrowheads). γ-H2AX was not detected and pRNApol II rendered faint labeling over the nucleus, except at those regions labeled with H3K9me3 (white arrowheads). (<b>C</b>–<b>F</b>) In nuclei of 1B and 2B individuals, H3K9me3 labeled several autosomal regions (white arrowheads), and the X chromosome, but γ-H2AX was not detected. Moreover, H3K9me3 labeling corresponded with regions of reduced pRNApol II labeling. (<b>G</b>) Scattered dot plot of the corrected total nuclear fluorescence of H3K9me3 immunolabeling in spermatogonia from individuals presenting 0, 1 or 2 B1 chromosomes. For each class, the individual data (n = 18) and median with 95% CI are depicted. Statistical significance was assessed using an ANOVA test (<span class="html-italic">p</span> &lt; 0.0001; ***) and Tukey’s multiple comparisons test. Bar in (<b>A</b>), 10 µm.</p>
Full article ">
Back to TopTop