[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Previous Issue
Volume 13, December-2
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Cells, Volume 14, Issue 1 (January-1 2025) – 10 articles

  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
18 pages, 20003 KiB  
Article
ST8SIA6 Sialylates CD24 to Enhance Its Membrane Localization in BRCA
by Jinxia He, Fengchao Zhang, Baihai Wu and Wengong Yu
Cells 2025, 14(1), 9; https://doi.org/10.3390/cells14010009 (registering DOI) - 26 Dec 2024
Abstract
CD24, a highly sialylated glycosyl-phosphatidyl-inositol (GPI) cell surface protein that interacts with sialic acid-binding immunoglobulin-like lectins (Siglecs), serves as an innate immune checkpoint and plays a crucial role in inflammatory diseases and tumor progression. Recently, cytoplasmic CD24 has been observed in samples from [...] Read more.
CD24, a highly sialylated glycosyl-phosphatidyl-inositol (GPI) cell surface protein that interacts with sialic acid-binding immunoglobulin-like lectins (Siglecs), serves as an innate immune checkpoint and plays a crucial role in inflammatory diseases and tumor progression. Recently, cytoplasmic CD24 has been observed in samples from patients with cancer. However, whether sialylation governs the subcellular localization of CD24 in cancer remains unclear, and the impact of CD24 expression and localization on the clinical prognosis of cancer remains controversial. Here, we performed a systematic pan-cancer analysis of the gene expression levels and clinical correlation of CD24. Our analysis revealed that CD24 was highly expressed in breast tumor tissues and tumor cells, significantly shortening patient survival time. However, this correlation was not evident in other types of cancer. Additionally, a correlation analysis of CD24 levels with sialyltransferases (STs) revealed that ST8SIA6 is the key ST affecting CD24 sialylation. Further investigation demonstrated that ST8SIA6 directly modified CD24, promoting its localization to the cell membrane. Taken together, these findings elucidate, for the first time, the mechanisms by which ST8SIA6 regulates CD24 subcellular localization, providing new insights into the biological functions and applications of CD24. Full article
Show Figures

Figure 1

Figure 1
<p>The flowchart illustrates the pan-cancer analysis of CD24. Abbreviations: IHC, immunohistochemistry; RNA-seq, RNA sequencing; DBs, databases; TILs, tumor-infiltrating lymphocytes.</p>
Full article ">Figure 2
<p>Expression analysis of the <span class="html-italic">CD24</span> gene in various human cancers. (<b>A</b>) The RNA expression levels of <span class="html-italic">CD24</span> in human cancers were analyzed using the TIMER algorithm. The statistical significance computed by the Wilcoxon test is annotated by the number of asterisks. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) The RNA expression levels of <span class="html-italic">CD24</span> in various human cancer cell lines were analyzed using data from the Human Protein Atlas database.</p>
Full article ">Figure 3
<p>The clinical relevance of <span class="html-italic">CD24</span> gene expression levels across various cancer types. (<b>A</b>) The clinical correlation between <span class="html-italic">CD24</span> gene expression levels and various types of cancer was analyzed using the TIMER algorithm, incorporating key clinical factors, including age, tumor stage, and tumor purity. A heatmap was drawn to show the normalized coefficient of the gene in the Cox model. Z-Score &gt; 0, <span class="html-italic">p</span> &lt; 0.05, increased risk; Z-Score &lt; 0, <span class="html-italic">p</span> &lt; 0.05, decreased risk; <span class="html-italic">p</span> &gt; 0.05, not significant. * <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>B</b>) The impact of <span class="html-italic">CD24</span> levels on the survival rates of patients with BRCA, MESO, and SKCM. HR represents the hazard ratio. (<b>C</b>) The impact of <span class="html-italic">CD24</span> levels on the survival rates of patients COAD, LGG, and UCS. HR represents the hazard ratio. (<b>D</b>) Spearman’s correlations between <span class="html-italic">CD24</span> gene expression levels and TILs, including activated CD8<sup>+</sup> T cells, activated CD4<sup>+</sup> T cells, natural killer cells, activated dendritic cells, macrophages, monocytes, and neutrophils, across various types of human cancers were obtained from the TISIDB database. Rho represents the Spearman’s correlation coefficient. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <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 4
<p>The subcellular localization of CD24 protein was analyzed using data from the Human Protein Atlas database. (<b>A</b>) The 13 cancer tissues with the highest levels of CD24 in the cytoplasm and membrane, namely, THCA, HNSC, TGCT, CESC, STAD, BLCA, OV, lung cancer (LUAD and LUSC), PRAD, renal cancer (KICH, KIRC, and KIRP), UCEC, PAAD, and BRCA. (<b>B</b>) Representative immunohistochemical staining images of changes in membranous and cytoplasmic CD24 levels in high- and low-stage tumors from BLCA and PARD patients are shown (scale bar, 100 μm and 25 μm). (<b>C</b>) Representative immunohistochemical staining images of CD24 in the nucleus (scale bar, 100 μm and 20 μm). (<b>D</b>) Representative immunohistochemical staining images of membranous and cytoplasmic CD24 in THCA, TGCT, OV, and BRCA (scale bar, 100 μm and 25 μm).</p>
Full article ">Figure 5
<p>The correlations between <span class="html-italic">CD24</span> levels and the levels of ST8 family members in human cancer were analyzed using the TIMER database. (<b>A</b>) <span class="html-italic">CD24</span> levels were positively correlated with <span class="html-italic">ST8SIA2</span> and <span class="html-italic">ST8SIA6</span> levels in BRCA. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <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>B</b>) The correlations between <span class="html-italic">CD24</span> and <span class="html-italic">ST8SIA6</span> levels were analyzed in various human cancers. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <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 6
<p>The correlations between <span class="html-italic">Siglec-7/9/10</span> levels and the abundance of tumor-infiltrating macrophages in BRCA (<b>A</b>) and other human cancers (<b>B</b>) were analyzed using the TIMER database. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <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 7
<p>CD24 is directly modified by ST8SIA6. (<b>A</b>) The co-localization of CD24 and Siglec-E Fc fluorescence was observed in 4T1 cells (scale bar, 10 μm). (<b>B</b>) The co-localization of CD24 and Siglec-E fluorescence was observed in TNBC tumor tissues (scale bar, 20 μm and 10 μm). (<b>C</b>) The co-localization of CD24 and ST8SIA6 fluorescence was observed in 4T1 cells (scale bar, 10 μm). (<b>D</b>) The co-localization of CD24 and ST8SIA6 fluorescence was observed in TNBC tumor tissues (scale bar, 50 μm and 10 μm).</p>
Full article ">Figure 8
<p>The impact of ST8SIA6 on the subcellular localization and expression of CD24. (<b>A</b>) The expression of ST8SIA6 was detected by western blotting in <span class="html-italic">St8sia6</span>-overexpressing (OE-<span class="html-italic">St8sia6</span>) and <span class="html-italic">St8sia6</span>-knockout (sg<span class="html-italic">St8sia6</span>) 4T1 cells. (<b>B</b>) The expression of ST8SIA6 was detected by immunofluorescence in OE-<span class="html-italic">St8sia6</span> and sg<span class="html-italic">St8sia6</span> 4T1 cells (scale bar, 25 μm). (<b>C</b>) The effects of ST8SIA6 on CD24 expression were detected by flow cytometry. (<b>D</b>) Quantitative analysis of CD24 expression in 4T1 cells. The graph shows the median fluorescence intensity (MFI) of CD24. Data are presented as the mean ± standard error of the mean. The <span class="html-italic">p</span>-values were calculated using a one-way analysis of variance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. (<b>E</b>) The effect of ST8SIA6 on the subcellular localization of CD24 was determined by immunofluorescence (scale bar, 10 μm).</p>
Full article ">
14 pages, 1541 KiB  
Article
Chromosomal Abnormalities in Miscarriages and Maternal Age: New Insights from the Study of 7118 Cases
by Anna A. Pendina, Mikhail I. Krapivin, Olga G. Chiryaeva, Lubov’ I. Petrova, Elizaveta P. Pashkova, Arina V. Golubeva, Andrei V. Tikhonov, Alla S. Koltsova, Ekaterina D. Trusova, Dmitrii A. Staroverov, Andrey S. Glotov, Olesya N. Bespalova and Olga A. Efimova
Cells 2025, 14(1), 8; https://doi.org/10.3390/cells14010008 - 26 Dec 2024
Abstract
Chromosomal abnormalities of the embryo are the most common cause of first-trimester pregnancy loss. In this single-center study, we assessed the frequency and the spectrum of chromosomal abnormalities in miscarriages for each year of maternal age from 23 to 44. Cytogenetic data were [...] Read more.
Chromosomal abnormalities of the embryo are the most common cause of first-trimester pregnancy loss. In this single-center study, we assessed the frequency and the spectrum of chromosomal abnormalities in miscarriages for each year of maternal age from 23 to 44. Cytogenetic data were obtained by conventional karyotyping of 7118 miscarriages in women with naturally conceived pregnancies. Chromosomal abnormalities were identified in 67.25% of miscarriages. The total incidence of chromosomal abnormalities increased with maternal aging; however, its average change for a one-year increase in maternal age differed between age spans, equaling 0.704% in the span from 23 to 37 years and 2.095% in the span from 38 to 44 years. At the age of 38 years, the incidence rate surged sharply by 14.79% up to 79.01% and then increased progressively up to 94% in 44-year-old women. The spectrum of chromosomal abnormalities in miscarriages was the same for each year of maternal age from 23 to 44 years. However, the proportions of particular chromosomal abnormalities differed between karyotypically abnormal miscarriages in younger and older women. The proportions of trisomy 16, polyploidy, monosomy X, mosaic abnormalities, and structural rearrangements decreased with increasing maternal age. In contrast, the proportions of multiple aneuploidies and regular trisomies 13, 15, 18, 21, and 22 showed an upward trend with maternal aging. To summarize, despite the increase in the total incidence of chromosomal abnormalities in miscarriages with maternal aging, the rate of change differs for younger and older women, being three times lower in the former than in the latter. Moreover, the proportion of some abnormalities in karyotypically abnormal miscarriages shows a steady growth, whereas the proportion of others becomes increasingly low with maternal aging, most probably due to the age-dependent prevalence of different molecular and cellular defects. Full article
Show Figures

Figure 1

Figure 1
<p>The incidence of chromosomal abnormalities in miscarriages in women aged 23 to 44 years with natural conceptions. The beta coefficients show the average growth in the incidence of abnormal karyotype in miscarriages for a one-year increase in maternal age at the spans of 23–37 years and 38–44 years.</p>
Full article ">Figure 2
<p>The proportions of different cytogenetic categories in karyotypically abnormal miscarriages in women aged 23 to 44 years with natural conceptions.</p>
Full article ">Figure 3
<p>Correlations between the maternal age and the incidence of different chromosomal abnormalities in miscarriages with an abnormal karyotype. Statistically significant correlations are framed (<span class="html-italic">p</span> &lt; 0.05, the nonparametric Spearman test). The beta coefficients show the average change in the incidence of chromosomal abnormality for a one-year increase in maternal age.</p>
Full article ">
28 pages, 27768 KiB  
Article
Nootkatone Derivative Nootkatone-(E)-2-iodobenzoyl hydrazone Promotes Megakaryocytic Differentiation in Erythroleukemia by Targeting JAK2 and Enhancing JAK2/STAT3 and PKCδ/MAPK Crosstalk
by Yang Pan, Feng Xiao, Chaolan Pan, Hui Song, Peng Zhao, Meijun Chen, Liejun Huang, Jue Yang and Xiaojiang Hao
Cells 2025, 14(1), 10; https://doi.org/10.3390/cells14010010 (registering DOI) - 26 Dec 2024
Abstract
Erythroleukemia, a complex myeloproliferative disorder presenting as acute or chronic, is characterized by aberrant proliferation and differentiation of erythroid cells. Although nootkatone, a sesquiterpene derived from grapefruit peel and Alaska yellow cedar, has shown anticancer activity predominantly in solid tumors, its effects in [...] Read more.
Erythroleukemia, a complex myeloproliferative disorder presenting as acute or chronic, is characterized by aberrant proliferation and differentiation of erythroid cells. Although nootkatone, a sesquiterpene derived from grapefruit peel and Alaska yellow cedar, has shown anticancer activity predominantly in solid tumors, its effects in erythroleukemia remain unexplored. This study aimed to investigate the impact of nootkatone and its derivatives on erythroleukemia. Our results demonstrate that the nootkatone derivative nootkatone-(E)-2-iodobenzoyl hydrazone (N2) significantly inhibited erythroleukemia cell proliferation in a concentration- and time-dependent manner. More importantly, N2 induced megakaryocytic differentiation, as evidenced by significant morphological changes, and upregulation of megakaryocytic markers CD41 and CD61. In vivo, N2 treatment led to a marked increase in platelet counts and megakaryocytic cell counts. Mechanistically, N2 activated a crosstalk between the JAK2/STAT3 and PKCδ/MAPK signaling pathways, enhancing transcriptional regulation of key factors like GATA1 and FOS. Network pharmacology and experimental validation confirmed that N2 targeted JAK2, and knockdown of JAK2 abolished N2-induced megakaryocytic differentiation, underscoring JAK2’s critical role in erythroleukemia differentiation. In conclusion, N2 shows great promise as a differentiation therapy for erythroleukemia, offering a novel approach by targeting JAK2-mediated signaling pathways to induce megakaryocytic differentiation. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

Figure 1
<p>Nootkatone derivative N2 inhibits cell proliferation in erythroleukemia HEL and K562 cells. (<b>A</b>) Chemical structure of nootkatone derivative N2. (<b>B</b>) Scatter diagram presentation of the influence of nootkatone and its derivatives (20 μM) on cell viability of HEL and K562 cells for 72 h. The inhibition rate was measured by MTT assays, with the 0.1% DMSO group serving as the negative control. Inhibition rate = (negative control group—treatment group)/negative control group × 100%. (<b>C</b>,<b>D</b>) HEL and K562 cells were treated with varying concentrations of the nootkatone derivative N2 for 72 h, and cell viability was evaluated using MTT assays. (<b>E</b>,<b>F</b>) The influence of N2 on the proliferation of HEL and K562 cells was quantified through MTT assays. (<b>G</b>,<b>H</b>) Effects of N2 on morphological changes in HEL and K562 cells. Magnification: ×200. Scale bar: 100 µm. Data represented the mean ± SD of 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 vs. the DMSO group.</p>
Full article ">Figure 2
<p>Nootkatone derivative N2 enhances multinucleation and CD41a expression in HEL and K562 cells. (<b>A</b>,<b>B</b>) Morphological analysis of HEL (<b>A</b>) and K562 (<b>B</b>) cells following treatment with varying concentrations of N2 for 72–96 h. Cells were subjected to Wright–Giemsa staining for morphological assessment. Magnification: ×400. Scale bar: 50 µm. (<b>C</b>,<b>D</b>) Immunofluorescence analysis was conducted on HEL (<b>C</b>) and K562 (<b>D</b>) cells treated with either DMSO or N2 for 72 h, revealing CD41a expression (green). Nuclear staining was performed using DAPI (blue). Magnification: ×400. Scale bar: 50 µm.</p>
Full article ">Figure 3
<p>Nootkatone derivative N2 increases the expression of megakaryocyte-specific markers in HEL cells. (<b>A,B</b>) Expression of CD41a and CD61 megakaryocyte-specific markers analyzed using flow cytometry in HEL cells. (<b>C</b>,<b>D</b>) Quantification of the percentage of CD41a<sup>+</sup> cells and CD61<sup>+</sup> cells in HEL cells. Data represent the mean ± SD of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group.</p>
Full article ">Figure 4
<p>Nootkatone derivative N2 increases the expression of megakaryocyte-specific markers in K562 cells. (<b>A</b>,<b>B</b>) Expression of CD41a and CD61 megakaryocyte-specific markers analyzed using flow cytometry in K562 cells. (<b>C</b>,<b>D</b>) Quantification of the percentage of CD41a<sup>+</sup> cells and CD61<sup>+</sup> cells in K562 cells. Data represent the mean ± SD of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group.</p>
Full article ">Figure 5
<p>Effect of nootkatone derivative N2 on cell cycle distribution in HEL and K562 cells. (<b>A</b>–<b>F</b>) HEL (<b>A</b>) and K562 (<b>D</b>) cells were exposed to indicated concentrations of N2 for either 72 h or 96 h. The cells were stained with PI and the percentage of cell cycle distribution was analyzed by flow cytometry. The proportions of cell cycle distribution at G1, S, and G2 phases in HEL (<b>B</b>,<b>C</b>) and K562 (<b>E</b>,<b>F</b>) cells at various time points. Data are expressed as the mean ± SD, with each experiment conducted in triplicate. *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group.</p>
Full article ">Figure 6
<p>N2 treatment was associated with an increase in polyploidy in HEL and K562 cells. (<b>A</b>–<b>D</b>) Polyploid cells in N2-treated HEL (<b>A</b>) and K562 (<b>C</b>) cells were analyzed by flow cytometry. The proportion of polyploid cells in HEL (<b>B</b>) and K562 (<b>D</b>) cells. Data are expressed as the mean ± SD. Each experiment was repeated in triplicate. * <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 vs. the DMSO group.</p>
Full article ">Figure 7
<p>Nootkatone derivative N2 activates the PKCδ/MAPK signaling pathway and its downstream megakaryocytic differentiation-related transcription factors. (<b>A</b>–<b>D</b>) Upon treating with N2 (2, 4, 8 μM), the expression levels of p-PKCδ, PKCδ, p-MEK, MEK, p-ERK, ERK, and GATA1 detected using Western blotting in HEL (<b>A</b>) and K562 (<b>C</b>) cells. Densitometry analysis of these proteins in HEL (<b>B</b>) and K562 (<b>D</b>) cells. (<b>E</b>,<b>F</b>) Effects of N2 on the mRNA expression levels of seven transcription factors relevant to megakaryocytic differentiation in HEL (<b>E</b>) and K562 (<b>F</b>) cells. All data are expressed as the mean ± SD. GAPDH was used as loading control. Each experiment was repeated in triplicate. * <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 vs. the DMSO group.</p>
Full article ">Figure 8
<p>Nootkatone derivative N2 activates the JAK2/STAT3 signaling pathway in HEL and K562 cells. (<b>A</b>) JAK2 was identified as a potential target based on the intersection of N2-predicted targets with established targets associated with acute myeloid leukemia. (<b>B</b>–<b>E</b>) HEL (<b>B</b>) and K562 (<b>C</b>) cells were treated with N2 at the indicated doses. The expression levels of p-JAK2, JAK2, p-STAT3, and STAT3 were analyzed by Western blotting. Densitometry analysis of p-JAK2, JAK2, p-STAT3, and STAT3 in HEL (<b>D</b>) and K562 (<b>E</b>) cells. All data are expressed as the mean ± SD, with GAPDH serving as the loading control. Each experiment was repeated in triplicate. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group.</p>
Full article ">Figure 9
<p>Nootkatone derivative N2 promotes megakaryocytic differentiation via activation of the JAK2/STAT3 signaling pathways. (<b>A</b>,<b>B</b>) JAK2/STAT3 pathway specific inhibitor WP1066 treatment inhibited N2-induced increases in cell size, multinucleation in HEL and K562 cells via Wright–Giemsa staining. Magnification: ×400. Scale bar: 50 µm. (<b>C</b>,<b>D</b>) HEL and K562 cells were treated with N2 (8 μM) either alone or in combination with JAK2/STAT3 inhibitor, WP1066 (1 μM), and the expression levels of CD41a and CD61 analyzed using flow cytometry. (<b>E</b>–<b>H</b>) Quantification of the percentage of CD41a<sup>+</sup> cells and CD61<sup>+</sup> cells in HEL (<b>E</b>,<b>F</b>) and K562 (<b>G</b>,<b>H</b>) cells. Data are expressed as the mean ± SD. Each experiment was repeated in triplicate. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. N2 group.</p>
Full article ">Figure 10
<p>Nootkatone derivative N2 promotes megakaryocytic differentiation via activation of the PKCδ/MAPK signaling pathways. (<b>A</b>,<b>B</b>) Treatment of HEL and K562 cells with N2 at a concentration of 8 μM, both independently and in conjunction with the PKCδ-specific inhibitor Rottlerin (1 μM). The cells were subjected to Wright–Giemsa staining for morphological assessment. Magnification: ×400. Scale bar: 50 µm. (<b>C</b>,<b>D</b>) HEL and K562 cells were treated with N2 (8 μM) in the absence or presence of Rottlerin (1 μM), and expression of CD41a and CD61 analyzed using flow cytometry. (<b>E</b>–<b>H</b>) Quantification of the percentage of CD41a<sup>+</sup> cells and CD61<sup>+</sup> cells in HEL (<b>E</b>,<b>F</b>) and K562 (<b>G</b>,<b>H</b>) cells. Data are presented as the mean ± SD. Each experiment was repeated in triplicate. *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. N2 group.</p>
Full article ">Figure 11
<p>WP1066 treatment significantly reduced N2-induced phosphorylation of JAK2, STAT3, PKCδ, MEK, and ERK, as well as the expression of GATA1. (<b>A</b>,<b>B</b>) HEL and K562 cells were treated with N2 (8 μM) and/or WP1066 (1 μM), and the protein expression levels of p-JAK2, JAK2, p-STAT3, STAT3, p-PKCδ, PKCδ, p-MEK, MEK, p-ERK, ERK, and GATA1 were detected using Western blotting. (<b>C</b>,<b>D</b>) Densitometry analysis of these proteins in HEL (<b>C</b>) and K562 (<b>D</b>) cells. Data are presented as the mean ± SD. GAPDH was used as loading control. Each experiment was repeated in triplicate. *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. N2 group.</p>
Full article ">Figure 12
<p>Rottlerin significantly diminished N2-induced PKCδ, STAT3, MEK and ERK phosphorylation. (<b>A</b>,<b>B</b>) HEL and K562 cells were treated with N2 (8 μM) in the absence or presence of Rottlerin (1 μM), and the proteins expression of p-JAK2, JAK2, p-STAT3, STAT3, p-PKCδ, PKCδ, p-MEK, MEK, p-ERK, ERK and GATA1 were detected using Western blotting. (<b>C</b>,<b>D</b>) Densitometry analysis of these proteins in HEL (<b>C</b>) and K562 (<b>D</b>) cells. Data are presented as the mean ± SD. GAPDH was used as loading control. Each experiment was repeated in triplicate. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 vs. the DMSO group. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. N2 group.</p>
Full article ">Figure 13
<p>Nootkatone derivative N2 binds to JAK2. (<b>A</b>) Molecular docking of N2 and JAK2 was conducted using AutoDock Vina 1.1.2. (<b>B</b>) CETSA was performed to assess the binding interactions between N2 and JAK2. (<b>C</b>) The stability of the JAK2 protein across varying temperatures was quantified using Western blotting analysis. (<b>D</b>) The DARTS experiments confirmed the binding of N2 to the JAK2 protein. (<b>E</b>) Densitometry analysis of JAK2. Data are presented as the mean ± SD. Each experiment was repeated in triplicate. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. lysates group, *** <span class="html-italic">p</span> &lt; 0.001 vs. pronase alone group.</p>
Full article ">Figure 14
<p>Nootkatone derivative N2-mediated megakaryocytic differentiation in erythroleukemia cells is JAK2-dependent. (<b>A</b>) Morphological analysis was conducted using Wright–Giemsa staining on LV-NC and LV-sh-JAK2 HEL cells exposed to 8 μM N2. Magnification: ×400. Scale bar: 50 µm. (<b>B</b>) Flow cytometry was employed to assess the expression levels of CD41a and CD61 in LV-NC and LV-sh-JAK2 HEL cells treated with 8 μM N2. (<b>C</b>,<b>D</b>) Quantification of the percentage of CD41a<sup>+</sup> and CD61<sup>+</sup> cells. (<b>E</b>) The expression of p-JAK2, JAK2, p-PKCδ, PKCδ, p-STAT3, STAT3, p-MEK, and MEK in LV-NC and LV-sh-JAK2 HEL cells treated with 8 μM N2 were measured by Western blotting. (<b>F</b>) Densitometry analysis of these proteins. All data are presented as the mean ± SD. Each experiment was repeated in triplicate. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. LV-NC/DMSO group. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. LV-NC/N2-8 µM group.</p>
Full article ">Figure 15
<p>Nootkatone derivative N2 accelerates megakaryocytic differentiation and suppresses erythroleukemia in vivo. (<b>A</b>) Spleen size of mice in different experimental groups. (<b>B</b>) Spleen weight presented as mean ± SD (<span class="html-italic">n</span> = 5). (<b>C</b>) Hematocrit values. (<b>D</b>) Platelet counts. (<b>E</b>) Flow cytometric analysis of CD41 and CD61 expression of spleen of each groups. (<b>F</b>,<b>G</b>) The histogram represents the percentage of CD41<sup>+</sup> and CD61<sup>+</sup> cells of spleen in each groups. (<b>H</b>) Representative H&amp;E stained images of spleen from each groups. The yellow arrow represents megakaryocytes. Magnification: ×400. Scale bar: 50 µm. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). * <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 vs. model group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. normal group.</p>
Full article ">Figure 16
<p>Nootkatone derivative N2 activates JAK2/STAT3 and PKCδ/MAPK signaling pathways. (<b>A</b>) The expression of p-JAK2, JAK2, p-PKCδ, PKCδ, p-STAT3, STAT3, p-MEK, MEK, p-ERK, and ERK of spleen tissue from each groups were measured by Western blotting. (<b>B</b>) Densitometry analysis of these proteins. (<b>C</b>) The impact of N2 on the mRNA expression of nine transcription factors related to megakaryocytic differentiation in spleen tissue. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 3). * <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 vs. model group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. normal group.</p>
Full article ">Figure 17
<p>Illustration of the role and mechanism of nootkatone derivative N2 in promoting megakaryocytic differentiation in erythroleukemia. The red arrow indicates up-regulation.</p>
Full article ">
20 pages, 3586 KiB  
Review
Hair Regeneration Methods Using Cells Derived from Human Hair Follicles and Challenges to Overcome
by Ons Ben Hamida, Moon Kyu Kim, Young Kwan Sung, Min Kyu Kim and Mi Hee Kwack
Cells 2025, 14(1), 7; https://doi.org/10.3390/cells14010007 - 25 Dec 2024
Abstract
The hair follicle is a complex of mesenchymal and epithelial cells acquiring different properties and characteristics responsible for fulfilling its inductive and regenerative role. The epidermal and dermal crosstalk induces morphogenesis and maintains hair follicle cycling properties. The hair follicle is enriched with [...] Read more.
The hair follicle is a complex of mesenchymal and epithelial cells acquiring different properties and characteristics responsible for fulfilling its inductive and regenerative role. The epidermal and dermal crosstalk induces morphogenesis and maintains hair follicle cycling properties. The hair follicle is enriched with pluripotent stem cells, where dermal papilla (DP) cells and dermal sheath (DS) cells constitute the dermal compartment and the epithelial stem cells existing in the bulge region exert their regenerative role by mediating the epithelial–mesenchymal interaction (EMI). Many studies have developed and focused on various methods to optimize the EMI through in vivo and in vitro approaches for hair regeneration. The culturing of human hair mesenchymal cells resulted in the loss of trichogenicity and inductive properties of DP cells, limiting their potential application in de novo hair follicle generation in vivo. Epithelial stem cells derived from human hair follicles are challenging to isolate and culture, making it difficult to obtain enough cells for hair regeneration purposes. Mesenchymal stem cells and epithelial stem cells derived from human hair follicles lose their ability to form hair follicles during culture, limiting the study of hair follicle formation in vivo. Therefore, many attempts and methods have been developed to overcome these limitations. Here, we review the possible and necessary cell methods and techniques used for human hair follicle regeneration and the restoration of hair follicle cell inductivity in culture. Full article
Show Figures

Figure 1

Figure 1
<p>Morphology and markers of cells isolated and cultured from human hair follicles. These are the results of staining with various markers after isolating and culturing cells from human hair follicles. The image shows dermal papilla (DP) cells after two weeks of culture, expressing a-SMA, versican, and ALP in cultured DP cells (passage 1). Outer root sheath (ORS) cells were isolated and cultured from the ORS derived from the epithelium, and the image shows the cells on day 10. Keratin 1–3 were strongly expressed, while keratin 17 and 19 were not expressed. Additionally, sebocytes were isolated and cultured from human sebaceous glands (SG), with the image showing the cells on day 10. These sebocytes strongly expressed keratin 17 and keratin 19, and lipid secretion was observed through Oil Red O and Nile Red staining, adapted with permission from Ref. [<a href="#B80-cells-14-00007" class="html-bibr">80</a>].</p>
Full article ">Figure 2
<p>Method to restore the trichogenicity of cultured human dermal papilla (DP) cells or epithelial stem cells (Epi stem cells). Cultured human epithelial stem cells (Epi stem cells) are induced into iPSCs, mixed with mouse dermal cells, and utilized in hair regeneration assays to promote hair follicle formation. Cultured dermal papilla cells (DP cells) recover their hair follicle regenerative ability through iPSC induction, 3D cultures, or treatment with small molecules, inhibitors, or exosomes. These treated DP cells are then combined with mouse epidermal cells and employed in hair regeneration assays to induce hair follicle formation.</p>
Full article ">Figure 3
<p>Currently reported methods for hair regeneration using cultured cells. This describes the models used to evaluate hair regeneration with isolated cells reported to date. Patch Assay: In this model, epidermal and dermal cells are mixed and injected subcutaneously into a nude mouse. Hair follicle formation can be observed two weeks later. Hair follicles do not form when using mouse dermal or epidermal cells alone, but mixing both types of cells results in hair follicle formation after two weeks. Chamber Assay: A chamber is inserted into a cut in the skin of a nude mouse, and a mixture of mouse dermal and epidermal cells is added to the chamber. Two weeks later, the chamber is removed, revealing a layer formed by the two cell types. By four weeks, hair follicle formation can be observed. Sandwich Assay: Dermal cells are placed between the epidermis and dermis of the footpad or sole skin, and the tissue is xenografted into another mouse. Hair follicle formation potential is then evaluated. Hair Germ Assay: Dermal and epidermal cells are mixed into a collagen gel drop, which is then injected into a nude mouse. Hair follicle formation is observed. Bearding Skin Organoid: Human pluripotent stem cells (hPSCs) are induced into iPSCs and treated with various chemicals to induce hair follicles in vitro.</p>
Full article ">
17 pages, 2689 KiB  
Article
Ezrin Polarization as a Diagnostic Marker for Circulating Tumor Cells in Hepatocellular Carcinoma
by Ibrahim Büdeyri, Olaf Guckelberger, Elsie Oppermann, Dhruvajyoti Roy, Svenja Sliwinski, Felix Becker, Benjamin Struecker, Thomas J. Vogl, Andreas Pascher, Wolf O. Bechstein, Anna Lorentzen, Mathias Heikenwalder and Mazen A. Juratli
Cells 2025, 14(1), 6; https://doi.org/10.3390/cells14010006 - 25 Dec 2024
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third leading cause of cancer-related death worldwide, with no precise method for early detection. Circulating tumor cells (CTCs) expressing the dynamic polarity of the cytoskeletal membrane protein, ezrin, have been proposed to [...] Read more.
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third leading cause of cancer-related death worldwide, with no precise method for early detection. Circulating tumor cells (CTCs) expressing the dynamic polarity of the cytoskeletal membrane protein, ezrin, have been proposed to play a crucial role in tumor progression and metastasis. This study investigated the diagnostic and prognostic potential of polarized circulating tumor cells (p-CTCs) in HCC patients. CTCs were isolated from the peripheral blood of 20 HCC patients and 18 patients with nonmalignant liver disease (NMLD) via an OncoQuick® kit and immunostained with Ezrin-Alexa Fluor 488®, CD146-PE, and CD45-APC. A fluorescence microscopy was then performed for analysis. The HCC group exhibited significantly higher levels of p-CTCs, with median values of 0.56 p-CTCs/mL, compared to 0.02 p-CTCs/mL (p = 0.03) in the NMLD group. CTCs were detected in 95% of the HCC patients, with a sensitivity of 95% and specificity of 89%. p-CTCs were present in 75% of the HCC patients, with a sensitivity of 75% and a specificity of 94%. Higher p-CTC counts were associated with the significantly longer overall survival in HCC patients (p = 0.05). These findings suggest that p-CTCs could serve as valuable diagnostic and prognostic markers for HCC. The incorporation of p-CTCs into diagnostic strategies could enhance therapeutic decision-making and improve patient outcomes. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the workflow. Created in BioRender. Juratli, M. (2023) BioRender.com/k09p009.</p>
Full article ">Figure 2
<p>Examples of polarized CTCs from individual patient samples (<b>A</b>–<b>J</b>). Anti-Ezrin-Alexa Fluor 488 (green), nuclear staining with DAPI (blue), CD146 (red), leukocyte/CD45 (violet) and merged images of all the fluorescence channels. Observed at 40× magnification.</p>
Full article ">Figure 3
<p>Boxplots comparing (<b>A</b>) CTCs and (<b>B</b>) p-CTCs between patients with HCC and those with NMLD. The lines within each box represent the median values, the boxes’ limits indicate the first and third quartiles, and the whiskers represent the smallest and largest values within 1.5 times of the IQRs from the first and third quartiles. <span class="html-italic">p</span>-values and ξ-effect sizes were determined using one-way ANOVA, and <span class="html-italic">p</span> &lt; 0.05 was considered significant.</p>
Full article ">Figure 4
<p>Kaplan–Meier analysis of HCC patients with and without p-CTCs. (<b>A)</b> Overall survival (OS) curve and (<b>B)</b> recurrence-free survival (RFS) curve. HCC patients with p-CTCs exhibited a longer mean overall survival (40 ± 8 months vs. 26 ± 22 months, <span class="html-italic">p</span> = 0.05). However, the difference in mean recurrence-free survival between HCC patients with and without p-CTCs was not statistically significant (36 ± 13 months vs. 25 ± 22 months, respectively; <span class="html-italic">p</span> = 0.20).</p>
Full article ">Figure 5
<p>Forest plot of hazard ratios for age &gt; 70 years (<b>A</b>), metastasis (<b>B</b>), p-CTC (<b>C</b>), recurrence (<b>D</b>) and tumor size &gt;5 cm (<b>E</b>) in HCC patients. Hazard ratios were calculated via univariate Cox regression analysis.</p>
Full article ">
32 pages, 2719 KiB  
Review
Metabolomics-Driven Biomarker Discovery for Breast Cancer Prognosis and Diagnosis
by Rasanpreet Kaur, Saurabh Gupta, Sunanda Kulshrestha, Vishal Khandelwal, Swadha Pandey, Anil Kumar, Gaurav Sharma, Umesh Kumar, Deepak Parashar and Kaushik Das
Cells 2025, 14(1), 5; https://doi.org/10.3390/cells14010005 - 25 Dec 2024
Abstract
Breast cancer is a cancer with global prevalence and a surge in the number of cases with each passing year. With the advancement in science and technology, significant progress has been achieved in the prevention and treatment of breast cancer to make ends [...] Read more.
Breast cancer is a cancer with global prevalence and a surge in the number of cases with each passing year. With the advancement in science and technology, significant progress has been achieved in the prevention and treatment of breast cancer to make ends meet. The scientific intradisciplinary subject of “metabolomics” examines every metabolite found in a cell, tissue, system, or organism from different sources of samples. In the case of breast cancer, little is known about the regulatory pathways that could be resolved through metabolic reprogramming. Evidence related to the significant changes taking place during the onset and prognosis of breast cancer can be obtained using metabolomics. Innovative metabolomics approaches identify metabolites that lead to the discovery of biomarkers for breast cancer therapy, diagnosis, and early detection. The use of diverse analytical methods and instruments for metabolomics includes Magnetic Resonance Spectroscopy, LC/MS, UPLC/MS, etc., which, along with their high-throughput analysis, give insights into the metabolites and the molecular pathways involved. For instance, metabolome research has led to the discovery of the glutamate-to-glutamate ratio and aerobic glycolysis as biomarkers in breast cancer. The present review comprehends the updates in metabolomic research and its processes that contribute to breast cancer prognosis and metastasis. The metabolome holds a future, and this review is an attempt to amalgamate the present relevant literature that might yield crucial insights for creating innovative therapeutic strategies aimed at addressing metastatic breast cancer. Full article
Show Figures

Figure 1

Figure 1
<p>A summary of potential non-invasive biomarkers for the early detection of breast cancer using fluid samples as the source. Abbreviations: MAPK: Mitogen-Activated Protein Kinase, ERK: Extracellular Signal-Regulated Kinase, JNK: c-Jun N-terminal Kinase, EMT: Epithelial–Mesenchymal Transition, OXPHOS: Oxidative Phosphorylation.</p>
Full article ">Figure 2
<p>Breast cancer metastasis involves multiple steps, starting with the invasion of surrounding tissues, followed by intravasation into the bloodstream or lymphatics, circulation through the body, and extravasation into distant tissues. Various factors, such as EMT, MET amplification, and abnormal TGFb production, can contribute to tumour metastasis. Abbreviations: CEA: Carcinoembryonic Antigen, FASL: Fas Ligand, OPN: Osteopontin, VEGFC: Vascular Endothelial Growth Factor C, VEGFD: Vascular Endothelial Growth Factor D, HGF: Hepatocyte Growth Factor, FRS3: Focal Adhesion Kinase Related-3, MYOZ2: Myozenin 2, RAC3GPR157: Rac Family Small GTPase 3 G Protein-Coupled Receptor 157, ZMYM6: Zinc Finger MYM-Type Protein 6, EIF3E: Eukaryotic Translation Initiation Factor 3 Subunit E, CSNK1E: Casein Kinase 1 Epsilon, ZNF510: Zinc Finger Protein 510.</p>
Full article ">Figure 3
<p>Schematic representation of various metabolic pathways involved in breast cancer prognosis.</p>
Full article ">
18 pages, 1835 KiB  
Review
Calcium Signalling in Neurological Disorders, with Insights from Miniature Fluorescence Microscopy
by Dechuan Sun, Mona Amiri, Qi Meng, Ranjith R. Unnithan and Chris French
Cells 2025, 14(1), 4; https://doi.org/10.3390/cells14010004 - 25 Dec 2024
Abstract
Neurological disorders (NDs), such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and schizophrenia, represent a complex and multifaceted health challenge that affects millions of people around the world. Growing evidence suggests that disrupted neuronal calcium signalling [...] Read more.
Neurological disorders (NDs), such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and schizophrenia, represent a complex and multifaceted health challenge that affects millions of people around the world. Growing evidence suggests that disrupted neuronal calcium signalling contributes to the pathophysiology of NDs. Additionally, calcium functions as a ubiquitous second messenger involved in diverse cellular processes, from synaptic activity to intercellular communication, making it a potential therapeutic target. Recently, the development of the miniature fluorescence microscope (miniscope) enabled simultaneous recording of the spatiotemporal calcium activity from large neuronal ensembles in unrestrained animals, providing a novel method for studying NDs. In this review, we discuss the abnormalities observed in calcium signalling and its potential as a therapeutic target for NDs. Additionally, we highlight recent studies that utilise miniscope technology to investigate the alterations in calcium dynamics associated with NDs. Full article
(This article belongs to the Special Issue New Discoveries in Calcium Signaling-Related Neurological Disorders)
Show Figures

Figure 1

Figure 1
<p>A typical structure of a conventional fluorescence microscope and the miniscope. (<b>a</b>) Conventional fluorescence microscope. (<b>b</b>) Miniscope: An LED emits light at a wavelength that excites the fluorophore, which is collimated by a half-ball lens. The excitation light passes through an excitation filter to reduce background noise before being reflected by a dichroic mirror. A GRIN lens focuses the light to activate fluorophores in neurons and collects the fluorescent signals. These signals are then directed back through the dichroic mirror and an emission filter, isolating the desired wavelength. Then, an achromatic lens focuses the signals onto a CMOS sensor.</p>
Full article ">Figure 2
<p>The miniscope facilitates the recording of neuronal calcium signals in freely moving mice. (<b>a</b>) An example of a C57BL/6 mouse wearing the UCLA miniscope (version 3). (<b>b</b>) A representative image showing hippocampal CA1 neurons captured using the miniscope. Neurons are labelled with GCamp6f. Typically, over 100 neurons can be observed within the field of view.</p>
Full article ">Figure 3
<p>An example of using the miniscope to study the calcium activity of hippocampal neurons in mice. (<b>a</b>) A mouse wearing a miniscope traverses a linear track. (<b>b</b>) An example showing that the calcium activity of hippocampal neurons exhibits spatial sensitivity when mice traverse a linear track. (<b>c</b>) An example of raw fluorescent intensity in the detected neurons captured by the miniscope. (<b>d</b>) An example of a neuron’s deconvolved calcium activity.</p>
Full article ">
19 pages, 4415 KiB  
Review
Ca2+/Calmodulin-Dependent Protein Kinase II (CaMKII) Regulates Basal Cardiac Pacemaker Function: Pros and Cons
by Tatiana M. Vinogradova and Edward G. Lakatta
Cells 2025, 14(1), 3; https://doi.org/10.3390/cells14010003 - 25 Dec 2024
Abstract
The spontaneous firing of the sinoatrial (SA) node, the physiological pacemaker of the heart, is generated within sinoatrial nodal cells (SANCs) and is regulated by a “coupled-clock” pacemaker system, which integrates a “membrane clock”, the ensemble of ion channel currents, and an intracellular [...] Read more.
The spontaneous firing of the sinoatrial (SA) node, the physiological pacemaker of the heart, is generated within sinoatrial nodal cells (SANCs) and is regulated by a “coupled-clock” pacemaker system, which integrates a “membrane clock”, the ensemble of ion channel currents, and an intracellular “Ca2+ clock”, sarcoplasmic reticulum-generated local submembrane Ca2+ releases via ryanodine receptors. The interactions within a “coupled-clock” system are modulated by phosphorylation of surface membrane and sarcoplasmic reticulum proteins. Though the essential role of a high basal cAMP level and PKA-dependent phosphorylation for basal spontaneous SANC firing is well recognized, the role of basal CaMKII-dependent phosphorylation remains uncertain. This is a critical issue with respect to how cardiac pacemaker cells fire spontaneous action potentials. This review aspires to explain and unite apparently contradictory results of pharmacological studies in the literature that have demonstrated a fundamental role of basal CaMKII activation for basal cardiac pacemaker function, as well as studies in mice with genetic CaMKII inhibition which have been interpreted to indicate that basal spontaneous SANC firing is independent of CaMKII activation. The assessment of supporting and opposing data regarding CaMKII effects on phosphorylation of Ca2+-cycling proteins and spontaneous firing of SANC in the basal state leads to the necessary conclusion that CaMKII activity and CaMKII-dependent phosphorylation do regulate basal cardiac pacemaker function. Full article
(This article belongs to the Section Cellular Metabolism)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic illustration of the coupled-clock pacemaker system and active P-CaMKII in SANCs. (<b>A</b>) Schematic presentation of ion channels “membrane clock” (including the most important currents, i.e., hyperpolarization-activated “funny” current (I<sub>f</sub>), L-type Ca<sup>2+</sup> current (I<sub>Ca,L</sub>), carried via Ca<sub>v</sub>1.2 or Ca<sub>v</sub>1.3 [<a href="#B3-cells-14-00003" class="html-bibr">3</a>], T-type Ca<sup>2+</sup> current (I<sub>Ca,T</sub>), delayed rectifier potassium current (I<sub>K</sub>), Na<sup>+</sup>/Ca<sup>2+</sup> exchange current (I<sub>NCX</sub>), sustained current (I<sub>st</sub>), etc.) and “Ca<sup>2+</sup> clock” in cardiac pacemaker cells. (<b>B</b>) Top: Illustration of spontaneous SANC action potentials, Ca<sup>2+</sup> transients, LCRs, and schematic illustration of several major ion currents involved in generation of the diastolic depolarization (DD) and interactions of molecules comprising the full coupled-clock pacemaker system. Bottom: the restitution process that defines the LCR period which is regulated by the rate of Ca<sup>2+</sup> pumping into the SR and SR Ca<sup>2+</sup> load required for activation of spontaneous release from RyRs. LCR-induced increase in local [Ca<sup>2+</sup>] beneath sarcolemma activates an inward I<sub>NCX</sub> current creating exponential increase in the DD rate (nonlinear DD). The LCR period represents the essence of the “coupled-clock” pacemaker system, which includes complex interactions between cell membrane electrogenic molecules and intracellular sarcoplasmic reticulum (SR) Ca<sup>2+</sup> cycling (see text for details). (<b>C</b>) Top: representative western blots of activated (autophosphorylated at Thr<sup>286/287</sup> site) CaMKII (P-CaMKII) and total CaMKII in rabbit SANCs and ventricular myocytes (VMs), (bottom) average values of P-CaMKII normalized to total CaMKII; (<b>D</b>) top: representative western blots of PLB phosphorylated at CaMKII-dependent Thr<sup>17</sup> site (P-PLB) and total PLB in rabbit SANCs and VMs, (bottom) average values of P-PLB normalized to total PLB; (<b>E</b>) intracellular distribution of total and autophosphorylated active (P-CaMKII) in rabbit SANCs; (<b>F</b>) top: representative western blots of RyRs phosphorylated at CaMKII-dependent Ser<sup>2814/2815</sup> site and total RyRs in the rabbit SA node and ventricular tissues; (bottom) average values of P-RyRs normalized to total RyRs expressed as percentage, assuming that the ratio P-RyRs/total RyRs in ventricular tissue is equal to 100%. * <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>,<b>D</b>,<b>F</b>) modified from [<a href="#B6-cells-14-00003" class="html-bibr">6</a>], (<b>E</b>) modified from [<a href="#B7-cells-14-00003" class="html-bibr">7</a>].</p>
Full article ">Figure 2
<p>Regulation of spontaneous Ca<sup>2+</sup> releases by cytosolic Ca<sup>2+</sup> differs in permeabilized SANCs and ventricular myocytes (VMs). (<b>A</b>) Representative images and Ca<sup>2+</sup> waveforms from bands (indicated by arrows) of SANCs and (<b>B</b>) of VMs exposed to different concentrations of cytosolic-free Ca<sup>2+</sup>. (<b>a</b>) FFT of Ca<sup>2+</sup> waveforms of SANCs in (<b>A</b>) and (<b>b</b>) FFT of the Ca<sup>2+</sup> waveforms of VMs from bands indicated by the color-matched arrows in (<b>B</b>). (<b>C</b>) Comparison of total Ca<sup>2+</sup> signal mass released by either SANCs or VMs at different cytosolic [Ca<sup>2+</sup>]<sub>c</sub>. (<b>D</b>) Average total SR Ca<sup>2+</sup> content in SANCs and VMs. (<b>E</b>) Elevation of [Ca<sup>2+</sup>]<sub>c</sub> increases phosphorylation of PLB at Thr<sup>17</sup> site in SANCs, but not in VMs. Top: Representative confocal images of permeabilized SANCs at 0 nmol/L and 150 nmol/L of [Ca<sup>2+</sup>]<sub>c</sub>, total PLB (red), and PLB phosphorylated at Thr<sup>17</sup> (green); (bottom) relative changes of phosphorylated PLB at Thr<sup>17</sup> normalized to total PLB in SANCs or VMs at different free cytosolic [Ca2+]<sub>c</sub>. * <span class="html-italic">p</span> &lt; 0.05. (<b>F</b>) Suppression of basal CaMKII activity decreases the SR Ca<sup>2+</sup> content in permeabilized SANCs. Effects of a rapid application of 20 mmol/L caffeine on representative permeabilized rabbit SANCs in the absence or presence of AIP, KN92, or KN-93. Bottom: average effects of AIP, KN-93, or KN-92 on the initial rapid component of the caffeine-induced SR Ca<sup>2+</sup> release. * <span class="html-italic">p</span> &lt; 0.05, one-way ANOVA, Newman–Keuls multiple comparison test. (<b>A</b>–<b>E</b>) modified from [<a href="#B12-cells-14-00003" class="html-bibr">12</a>]. (<b>F</b>) modified from [<a href="#B6-cells-14-00003" class="html-bibr">6</a>].</p>
Full article ">Figure 3
<p>Inhibition of CaMKII suppresses spontaneous, periodic LCRs in permeabilized SANCs. (<b>A</b>) Left: Confocal line-scan images of a representative SANC bathed in 200 nmol/L [Ca<sup>2+</sup>]<sub>c</sub> before (top) and after (bottom) superfusion with 10 μmol/L AIP. Right: AIP treatment resulted in decreased total Ca<sup>2+</sup> signal mass released by SANCs. (<b>B</b>) Spatial size of LCRs (control, yellow squares) was markedly decreased after treatment with AIP (blue squares), * <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>) Left: FFT of Ca<sup>2+</sup> oscillations (from bands indicated by arrows) in (<b>A</b>), before and after AIP treatment. Right: relative number of SANCs that generated periodic LCRs under control conditions and after AIP treatment. Logistic regression analysis demonstrated a significant difference between the two curves (<span class="html-italic">p</span> &lt; 0.002). Modified from [<a href="#B12-cells-14-00003" class="html-bibr">12</a>].</p>
Full article ">Figure 4
<p>Inhibition of CaMKII activity suppresses L-type Ca<sup>2+</sup> current but has no effect on hyperpolarization-activated funny current I<sub>f</sub>. (<b>A</b>) Average current–voltage relationships of I<sub>Ca,L</sub> in the presence or absence of the CaMKII inhibitor KN-93 (1 μmol/L); inset shows representative control recordings of I<sub>Ca,L</sub>. (<b>B</b>) Immunofluorescence intensity of autophosphorylated, active P-CaMKII in control conditions (control) and after treatment with KN-93 or AIP. * <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>) Activation (triangles) and steady-state inactivation (squares) curves of I<sub>Ca,L</sub> measured from a holding potential of –70 mV. Filled symbols show control data, and open symbols represent data recorded after 5 min treatment with 1 μmol/L KN-93. (<b>D</b>) Inhibition of CaMKII by KN-93 or AIP, but not KN-92, slows the recovery of I<sub>Ca,L</sub> from inactivation. (<b>A</b>–<b>D</b>) modified from [<a href="#B7-cells-14-00003" class="html-bibr">7</a>]. (<b>E</b>) (<b>a</b>) Traces of I<sub>f</sub> current in control (left) and following exposure to 1 μmol/L KN-93 (right). Cell capacitance is 34 pF. (<b>b</b>) Conductance–voltage relationship on I<sub>f</sub> current in control (filled squares) and in KN-93 (open squares). Current was normalized to max control current. Inset: Peak I<sub>Ca,L</sub> current (holding potential −40 mV, step to 0 mV) in the absence (black line) and presence (grey line) of 1 μmol/L KN-93. (<b>E</b>) modified from [<a href="#B54-cells-14-00003" class="html-bibr">54</a>] with permission.</p>
Full article ">Figure 5
<p>CaMKII inhibition suppresses slowly activating delayed rectifier potassium current I<sub>Ks</sub> and spontaneous beating of cardiac pacemaker cells. (<b>A</b>) Ca<sup>2+</sup>-dependent inhibitory effect of CaMKII inhibitor KN-93 on I<sub>Ks</sub>. (<b>a</b>) Time course of normalized I<sub>Ks</sub> tail currents in response to KN-93 (1 μmol/L) in cells dialyzed with Ca (+) (pCa 7) pipette solution. The inset shows original current traces recorded at the indicated time points. (<b>b</b>) I<sub>Ks</sub> in the presence of KN-93 under Ca (+) or Ca (−) (pCa 10) conditions. (<b>c</b>) Pulse protocol. Depolarizing pulses of 100 ms were applied to +30 mV from a holding potential of −50 mV at a rate of 240 pulses/min. After a cell was subjected to a train of depolarizing pulses (240 pulses), KN-93 was added to the bath solution. (<b>d</b>) I<sub>Ks</sub> at P1 (first pulse), P2, P3, P240, and P720. The dashed line indicates the zero-current level. (<b>e</b>) The summary of KN-93 effect on I<sub>Ks</sub> induced by simulated pacemaker potentials, ** <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) Effects of CaMKII inhibition on spontaneous APs in guinea pig SANCs. (<b>a</b>) Recordings of APs in the absence (left) or presence (right) of CaMKII inhibitor KN-93 or (<b>b</b>) inactive KN-93 analog KN-92; (<b>c</b>) recordings of spontaneous APs in the absence (left) or presence (right) of AIP (1 μmol/L) and after drug washout. ((<b>A</b>,<b>B</b>) modified from [<a href="#B13-cells-14-00003" class="html-bibr">13</a>], with permission). (<b>C</b>) Effects of CaMKII inhibition on spontaneous APs in rabbit SANCs. (<b>a</b>) Recordings of spontaneous APs in the absence (left) and presence (right) of the CaMKII inhibitor KN-93 (1 μmol/L). (<b>b</b>) Spontaneous APs (left) before and after (right) application of an inactive analog KN-92. (<b>c</b>) Recordings of APs in the absence (left) or presence (right) of the specific CaMKII inhibitor AIP (10 μmol/L) and (bottom) after drug washout. (<b>C</b>) modified from [<a href="#B7-cells-14-00003" class="html-bibr">7</a>].</p>
Full article ">Figure 6
<p>CaMKII inhibition decreases phosphorylation of Ca<sup>2+</sup>-cycling proteins PLB and RyRs in intact rabbit SANCs, suppresses LCRs, and prolongs the T-90 and LCR period. (<b>A</b>) Top: representative western blots of PLB phosphorylated at Thr<sup>17</sup> site and total PLB at baseline and after treatment with 1 μmol/L KN-93 or 1 μmol/L KN-92; bottom: average values of phosphorylated PLB normalized to total PLB in basal conditions, after treatment with KN-93 or KN-92. (<b>B</b>) Top: representative western blots of RyRs phosphorylated at Ser<sup>2815</sup> site and total RyRs in SA node tissue at baseline and in response to CaMKII inhibition by 1 μmol/L KN-93; bottom: average values of phosphorylated RyRs normalized to total RyRs in basal conditions and after treatment with KN-93; data are presented as % control. * <span class="html-italic">p</span> &lt; 0.05, by <span class="html-italic">t</span>-test. (<b>C</b>) Representative western blots of total PLB and PLB phosphorylated at PKA-dependent Ser<sup>16</sup> site (left) or CaMKII-dependent Thr<sup>17</sup> (right) in response to inhibition of PKA by selective PKA inhibitor peptide PKI; bottom: average values of PLB phosphorylated at Ser<sup>16</sup> site or Thr<sup>17</sup> site and normalized to total PLB after treatment with PKI; data are presented as % control. (<b>D</b>) Confocal line-scan images of a representative SANC depicting AP-induced Ca<sup>2+</sup> transients and LCRs (arrowheads) during spontaneous beating of SANCs in control and when CaMKII activity was inhibited by 10 μmol/L AIP. Normalized subsarcolemmal fluorescence averaged over an image width is shown in red and superimposed with the image. Insets define the LCR period and time to 90% decay of AP-induced Ca<sup>2+</sup> transient (T-90). Inhibition of CaMKII activity by AIP markedly suppresses LCR parameters and increases the LCR period and spontaneous cycle length; subsequently, spontaneous firing ceased. Following drug washout LCRs are restarted, and spontaneous beating recovered. (<b>E</b>,<b>F</b>) CaMKII inhibition with 1 μmol/L KN-93 or 10 μmol/L AIP, but not 1 μmol/L KN-92, markedly decreases the number of LCRs per each spontaneous cycle and the LCR size, respectively. (<b>G</b>) Histograms of the decay of AP-induced Ca<sup>2+</sup> transient, indexed by T-90, before and during CaMKII inhibition with AIP, KN-93, or inactive analog KN-92. (<b>H</b>) AIP or KN-93, but not KN-92, produced prolongation of T-90, which was paralleled by an increase in the LCR period (each symbol represents an individual SANC). * <span class="html-italic">p</span> &lt; 0.05. Modified from [<a href="#B6-cells-14-00003" class="html-bibr">6</a>].</p>
Full article ">Figure 7
<p>Bradycardia during heart failure in mice is linked to reduced CaMKII phosphorylation, decrease in Ca<sup>2+</sup> spark frequency, and amplitude of pre-transient Ca<sup>2+</sup> release. (<b>A</b>) Representative western blots and quantification of total and phosphorylated CaMKII (normalized by total CaMKII) in SAN tissues from sham and HF mice. (<b>B</b>) Representative western blots and quantification of phosphorylated PLB-T17 normalized by total PLB. (<b>C</b>) Representative western blots and quantification of RYR2-S2814 normalized to total RYR2 in SAN tissues from sham and HF mice. Blue circles in (<b>A</b>), (<b>B</b>), (<b>C</b>) highlight, respectively, basal level of CaMKII activity, PLB and RyR phosphorylation in sham operated mice. (<b>D</b>) Left: Representative line-scan images (0.25 ms per line) of spontaneous beating cells from intact SA node; arrows indicate Ca<sup>2+</sup> sparks. Right: Quantification of Ca<sup>2+</sup> spark frequency (number of sparks/s/100 μm) in sham and HF mice SAN tissues. (<b>E</b>) Quantification of the fluorescence of the fluorescence ramp (pre-transient Ca<sup>2+</sup>) was measured in SA nodes from sham and HF mice. (<b>F</b>) Representative examples of telemetric ECG traces (daytime) under basal conditions and after atropine and propranolol injection (2 mg/kg, respectively, i.p.) from sham-operated and TAC-induced HF mice. Right: Quantification of the heart rate under basal condition and upon autonomic nervous system (ANS) inhibition in the same mice (sham n = 7; HF n = 5). (<b>A</b>–<b>F</b>) modified from [<a href="#B79-cells-14-00003" class="html-bibr">79</a>], with permission.</p>
Full article ">Figure 8
<p>SANCs from wild-type mice, employed for transgenic CaMKII inhibition, have no CaMKII activity in the basal state. (<b>A</b>) Representative immunofluorescence images show absence of activated (pCaMKII) in SANCs from wild-type mice (blue arrows). ISO activates CaMKII in SANCs isolated from wild-type (WT) and AC3-C mice, but not from AC3-I mice with SA node CaMKII inhibition. Columns are as follows: 1, eGFP (expressed in AC3-C and AC3-I SANCs); 2, Thr<sup>287</sup> autophosphorylated, activated CaMKII (pCaMKII, red); 3, merge; 4, magnified images from column 2. (Scale bar, 10 μm). (<b>B</b>) Top: ECGs recorded from Langendorff-perfused hearts at baseline and after 1 μmol/L ISO. Bottom: Heart rates recorded from ECG-telemetered mice (in vivo) at rest and Langendorff-perfused hearts (ex vivo). Langendorff-perfused hearts from AC3-I and control mice (n = 5–6/group) beat at equivalent rates in the basal conditions (<span class="html-italic">p</span> = 0.318). (<b>C</b>) Consistent with the absence of basal CaMKII activity, SANC isolated from WT, AC3-C or AC3-I mouse SA nodes have the same beating rates in the basal conditions (blue oval). In response to a range of ISO concentrations, AC3-I SANC beating rates were significantly (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ANOVA) slower than controls at each ISO concentration (n = 6–10 per data point). (<b>D</b>) Representative line-scan confocal images of Rhod-2 fluorescence with simultaneously recorded spontaneous APs and spatially averaged Ca<sup>2+</sup> transients (lower) at baseline conditions and summary data for Ca<sup>2+</sup> spark frequency in basal conditions and after ISO (1 μmol/L) (n = 13–21 per group). * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 compared with baseline. (<b>A</b>–<b>D</b>) modified from [<a href="#B83-cells-14-00003" class="html-bibr">83</a>], with permission.</p>
Full article ">
22 pages, 1693 KiB  
Review
Caveolin-Mediated Endocytosis: Bacterial Pathogen Exploitation and Host–Pathogen Interaction
by Dibyasri Barman and Rishi Drolia
Cells 2025, 14(1), 2; https://doi.org/10.3390/cells14010002 - 24 Dec 2024
Abstract
Within mammalian cells, diverse endocytic mechanisms, including phagocytosis, pinocytosis, and receptor-mediated endocytosis, serve as gateways exploited by many bacterial pathogens and toxins. Among these, caveolae-mediated endocytosis is characterized by lipid-rich caveolae and dimeric caveolin proteins. Caveolae are specialized microdomains on cell surfaces that [...] Read more.
Within mammalian cells, diverse endocytic mechanisms, including phagocytosis, pinocytosis, and receptor-mediated endocytosis, serve as gateways exploited by many bacterial pathogens and toxins. Among these, caveolae-mediated endocytosis is characterized by lipid-rich caveolae and dimeric caveolin proteins. Caveolae are specialized microdomains on cell surfaces that impact cell signaling. Caveolin proteins facilitate the creation of caveolae and have three members in vertebrates: caveolin-1, caveolin-2, and caveolin-3. Many bacterial pathogens hijack caveolin machinery to invade host cells. For example, the Gram-positive facultative model intracellular bacterial pathogen Listeria monocytogenes exploits caveolin-mediated endocytosis for efficient cellular entry, translocation across the intestinal barrier, and cell–cell spread. Caveolin facilitates the internalization of group A streptococci by promoting the formation of invaginations in the plasma membrane and avoiding fusion with lysosomes, thereby aiding intracellular survival. Caveolin plays a crucial role in internalizing and modulation of host immune responses by Gram-negative bacterial pathogens, such as Escherichia coli K1, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Salmonella enterica serovar Typhimurium. Here, we summarize how bacterial pathogens manipulate the host’s caveolin system to facilitate bacterial entry and movement within and between host cells, to support intracellular survival, to evade immune responses, and to trigger inflammation. This knowledge enhances the intervention of new therapeutic targets against caveolin in microbial invasion and immune evasion processes. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic depicting the mechanism of <span class="html-italic">L. monocytogenes</span> LAP and caveolin-mediated translocation across the intestinal epithelial barrier and subsequent InlA-mediated internalization across non-phagocytic cells. LAP on <span class="html-italic">L. monocytogenes</span> binds to its host cell surface receptor heat shock protein 60 (Hsp60), inducing endocytosis of tight junction proteins, claudin-1, occludin, and the adherens junction protein E-cadherin via caveolin-1 and MLCK-mediated endocytosis. This disrupts cell junctions, allowing <span class="html-italic">L. monocytogenes</span> to pass through the paracellular spaces. InlA subsequently binds to its receptor E-cadherin at the adherens junctions to mediate transcytosis across the epithelial barrier. In non-phagocytic cells, the bacterial surface protein InlA and InlB interact with E-cadherin and c-met, leading to the cytoskeletal rearrangement via a zipper mechanism that triggers <span class="html-italic">L. monocytogenes</span> internalization through PI3-K activation and caveolin-mediated endocytosis. Figure created using Biorender and adapted from [<a href="#B43-cells-14-00002" class="html-bibr">43</a>].</p>
Full article ">Figure 2
<p>Schematic representation of the cell-to-cell spread mechanism of <span class="html-italic">L. monocytogenes</span> in phagocytic and non-phagocytic cells. In phagocytic cells (<b>left</b>), internalized actin protrusions containing <span class="html-italic">L. monocytogenes</span> secrete LLO, which disrupts phosphatidyl serine on the plasma membrane. Both actin protrusions and phosphatidyl serine-positive <span class="html-italic">L. monocytogenes</span> bind to the TIM4 receptor on the host cell surface, which causes internalization of <span class="html-italic">L. monocytogenes</span> via caveolin-mediated endocytosis. In non-phagocytic cells (<b>right</b>), when actin filament-rich protrusions containing the bacteria extend from one cell, they bind to ubiquitinated E-cadherin in adjacent cells. This binding triggers caveolae to form a flattened invagination that wraps around these bacterial protrusions, effectively engulfing them with the help of some core proteins of caveolae, such as Cav-1, Cav-2, a subset of the caveolin-associated proteins (cavin-2 and EHD2), and clathrin-interacting Epsin that assists in bending the membrane to create these invaginations. Figure created using Biorender.</p>
Full article ">Figure 3
<p>Schematics depicting the internalization mechanism of <span class="html-italic">P. gingivalis</span> and <span class="html-italic">Leptospira</span> via caveolin-mediated endocytosis. (<b>A</b>) The interaction of the virulent factor RgpA of <span class="html-italic">P. gingivalis</span> with Cav-1 in the host cell facilitates the internalization of <span class="html-italic">P. gingivalis</span> via caveolae. <span class="html-italic">P. gingivalis</span> inhibits the integrity of Mfsd2a, leading to enhanced transcytosis across the blood–brain barrier and increased Cav-1 expression, which induces albumin uptake to the cell (adapted from [<a href="#B89-cells-14-00002" class="html-bibr">89</a>]). (<b>B</b>) Leptospiral species interacts with integrin-β-1 on host cells; it triggers caveolin to form an invagination; and through the caveolae/integrin-b1-PI3K/FAK-microfilament endocytosis pathway, it enters the host cell. To avoid fusion with lysosomes, it forms leptospiral vesicles inside the host cell, and these vesicles recruit Rab5/Rab11 and Sec/Exo-SNARE proteins in endocytic recycling and vesicular transport systems for intracellular migration and finally release from the cells through a SNARE complex-mediated FAK/microfilament/microtubule endocytosis pathway. Figure created using Biorender.</p>
Full article ">
36 pages, 11803 KiB  
Article
Interplay of Transcriptomic Regulation, Microbiota, and Signaling Pathways in Lung and Gut Inflammation-Induced Tumorigenesis
by Beatriz Andrea Otálora-Otálora, César Payán-Gómez, Juan Javier López-Rivera, Natalia Belén Pedroza-Aconcha, Sally Lorena Arboleda-Mojica, Claudia Aristizábal-Guzmán, Mario Arturo Isaza-Ruget and Carlos Arturo Álvarez-Moreno
Cells 2025, 14(1), 1; https://doi.org/10.3390/cells14010001 - 24 Dec 2024
Abstract
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn’s disease, and twelve ulcerative colitis high throughput sequencing datasets using [...] Read more.
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn’s disease, and twelve ulcerative colitis high throughput sequencing datasets using R language specialized libraries and gene enrichment analyses identified a regulatory network in each inflammatory disease. IRF9 and LINC01089 in pulmonary arterial hypertension are related to the regulation of signaling pathways like MAPK, NOTCH, human papillomavirus, and hepatitis c infection. ZNF91 and TP53TG1 in Crohn’s disease are related to the regulation of PPAR, MAPK, and metabolic signaling pathways. ZNF91, VDR, DLEU1, SATB2-AS1, and TP53TG1 in ulcerative colitis are related to the regulation of PPAR, AMPK, and metabolic signaling pathways. The activation of the transcriptomic network and signaling pathways might be related to the interaction of the characteristic microbiota of the inflammatory disease, with the lung and gut cell receptors present in membrane rafts and complexes. The transcriptomic analysis highlights the impact of several coding and non-coding RNAs, suggesting their relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during lung and gut cell adaptation to inflammatory phenotypes. Full article
(This article belongs to the Topic Inflammatory Tumor Immune Microenvironment)
Show Figures

Figure 1

Figure 1
<p>Venn diagram with the transcriptomic metafirm in common and unique to each type of inflammatory disease. Created with BioRender.com.</p>
Full article ">Figure 2
<p>Coding (lilium and grey) and non-coding (purple) transcriptional regulatory network (cncTRN) of key upregulated transcription factors (TFs) and lncRNA in pulmonary arterial hypertension (PAH). Created with Cytoscape.</p>
Full article ">Figure 3
<p>Coding (lilium and grey) and non-coding (purple) transcriptional regulatory network (cncTRN) of key upregulated transcription factors in CD. Created with Cytoscape.</p>
Full article ">Figure 4
<p>Coding (lilium and grey) and non-coding (purple) transcriptional regulatory network (cncTRN) of key upregulated transcription factors in ulcerative colitis. Created with Cytoscape.</p>
Full article ">Figure 5
<p>Microbiome interaction with membrane receptor of PAH-related cells activating signaling pathways involved in transcriptional regulation during lung inflammation. In red are the upregulated genes and TFs; in black are the key upregulated TFs. Created with BioRender.com.</p>
Full article ">Figure 6
<p>Microbiome interaction with membrane receptor of CD-related cells, activating signaling pathways involved in transcriptional regulation during gut inflammation. In red are the upregulated genes and TFs; in black are the key upregulated TFs. Created with BioRender.com.</p>
Full article ">Figure 7
<p>Microbiome interaction with membrane receptor of UC-related cells, activating signaling pathways involved in transcriptional regulation during gut inflammation. In red are the upregulated genes and TFs; in black are the key upregulated TFs. Created with BioRender.com.</p>
Full article ">
Previous Issue
Back to TopTop