Nothing Special   »   [go: up one dir, main page]

Next Issue
Volume 11, October-2
Previous Issue
Volume 11, September-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 11, Issue 19 (October-1 2022) – 253 articles

Cover Story (view full-size image): The transport of low-density lipoprotein (LDL) through the endothelium is a key step in atherosclerosis development, but it is notorious that phenotypic differences exist between endothelial cells (ECs) originating from different vascular beds. Here, we systematically compared brain versus aortic ECs regarding their interaction with LDL and observed that both EC types bind and internalize LDL. However, whereas aortic ECs degrade very small amounts of LDL and transcytose the majority, brain EC degrade but do not transport LDL. We found that the LDLR–clathrin pathway leads to LDL degradation in either EC type, whereas ALK1, which promotes LDL transport in aortic ECs, limits LDL degradation in brain ECs. Finally, neither SR-BI nor caveolin-1, which promote LDL uptake and transport into aortic ECs, limit LDL binding and association in brain ECs. View this paper
  • 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:
16 pages, 3151 KiB  
Article
MEG3 Expression Indicates Lymph Node Metastasis and Presence of Cancer-Associated Fibroblasts in Papillary Thyroid Cancer
by Sina Dadafarin, Tomás C. Rodríguez, Michelle A. Carnazza, Raj K. Tiwari, Augustine Moscatello and Jan Geliebter
Cells 2022, 11(19), 3181; https://doi.org/10.3390/cells11193181 - 10 Oct 2022
Cited by 8 | Viewed by 2947
Abstract
Papillary thyroid cancer is the most common endocrine malignancy, occurring at an incidence rate of 12.9 per 100,000 in the US adult population. While the overall 10-year survival of PTC nears 95%, the presence of lymph node metastasis (LNM) or capsular invasion indicates [...] Read more.
Papillary thyroid cancer is the most common endocrine malignancy, occurring at an incidence rate of 12.9 per 100,000 in the US adult population. While the overall 10-year survival of PTC nears 95%, the presence of lymph node metastasis (LNM) or capsular invasion indicates the need for extensive neck dissection with possible adjuvant radioactive iodine therapy. While imaging modalities such as ultrasound and CT are currently in use for the detection of suspicious cervical lymph nodes, their sensitivities for tumor-positive nodes are low. Therefore, advancements in preoperative detection of LNM may optimize the surgical and medical management of patients with thyroid cancer. To this end, we analyzed bulk RNA-sequencing datasets to identify candidate markers highly predictive of LNM. We identified MEG3, a long-noncoding RNA previously described as a tumor suppressor when expressed in malignant cells, as highly associated with LNM tissue. Furthermore, the expression of MEG3 was highly predictive of tumor infiltration with cancer-associated fibroblasts, and single-cell RNA-sequencing data revealed the expression of MEG3 was isolated to cancer-associated fibroblasts (CAFs) in the most aggressive form of thyroid cancers. Our findings suggest that MEG3 expression, specifically in CAFs, is highly associated with LNM and may be a driver of aggressive disease. Full article
(This article belongs to the Special Issue Regulatory Roles of Non-coding RNAs in Cancer)
Show Figures

Figure 1

Figure 1
<p>Differential expression analysis between tumor and normal thyroid tissue. (<b>A</b>) Principal component analysis of 44 PTC and matched normal tissue. (<b>B</b>) Proportion of known driver mutations identified in tumor samples. (<b>C</b>) Mutational status, sex, TDS, and ERK score in individual tumors. TDS and ERK scale represent Log2-scaled high and low thyroid differentiation and ERK-activation based on gene signatures [<a href="#B47-cells-11-03181" class="html-bibr">47</a>]. (<b>D</b>) Pathway analysis of differentially expressed genes. (<b>E</b>) Hierarchical clustering of the top 100 differentially expressed lncRNAs (scaled normalized reads by column). FDR, false discovery rate; TDS, thyroid differentiation score.</p>
Full article ">Figure 2
<p>LNM-related co-expression analysis of PTC samples. (<b>A</b>) Relationship between WCGNA modules and LNM status. Values represent Pearson correlation coefficient and the correlation <span class="html-italic">p</span>-value (in brackets) between LNM and module eigengene. (<b>B</b>) Hallmark gene set analysis of the module most correlated with LNM status (black). (<b>C</b>) Black module constituent gene expression ranked by gene significance value related to lymph node metastasis (GS.LN) (<b>D</b>) Black module lncRNAs upregulated in LNM+ tissue but not LNM- tissue. LNM+ and LNM−, positive and negative lymph node status. * <span class="html-italic">p</span> &lt; 0.05; ns, non-significant.</p>
Full article ">Figure 3
<p><span class="html-italic">MEG3</span> expression is increased in PTC with LNM and correlates with poor outcomes (<b>A</b>) TCGA all patients (left) and <span class="html-italic">BRAF<sup>V600E</sup></span> cohort (right) survival given top (red) and bottom (blue) quartile <span class="html-italic">MEG3</span> expression. (<b>B</b>) TCGA Solid Tissue Normal <span class="html-italic">MEG3</span> expression. (<b>C</b>) <span class="html-italic">MEG3</span> fold-change expression in NYMC LNM+ and LNM-PTC.</p>
Full article ">Figure 4
<p><span class="html-italic">MEG3</span> isoform expression and association with infiltration of CAFs. (<b>A</b>) <span class="html-italic">MEG3</span> transcript isoform usage in normal (non-cancerous) tissue and NYMC PTC (LNM+/−). (<b>B</b>) Scatter plots demonstrating the correlation of <span class="html-italic">MEG3</span> expression in TCGA THCA project with tumor purity and estimated infiltration level of CAFs using TIMER2.0. EPIC output was used to represent fibroblast infiltration estimates from TIMER2.0. * <span class="html-italic">p</span> &lt; 0.05; ns, non-significant.</p>
Full article ">Figure 5
<p>Single-cell RNA-sequencing results identify cell types that express <span class="html-italic">MEG3</span>. Expression profile of <span class="html-italic">MEG3</span> in 5 ATC patient tumors represented as UMAPs with overlaid heatmaps demonstrating <span class="html-italic">MEG3</span> expression and expression in different cell subtypes in each sample. Red circles represent clusters of cells that are annotated as fibroblasts.</p>
Full article ">
22 pages, 1469 KiB  
Review
Patient Selection Approaches in FGFR Inhibitor Trials—Many Paths to the Same End?
by Peter Ellinghaus, Daniel Neureiter, Hendrik Nogai, Sebastian Stintzing and Matthias Ocker
Cells 2022, 11(19), 3180; https://doi.org/10.3390/cells11193180 - 10 Oct 2022
Cited by 11 | Viewed by 3332
Abstract
Inhibitors of fibroblast growth factor receptor (FGFR) signaling have been investigated in various human cancer diseases. Recently, the first compounds received FDA approval in biomarker-selected patient populations. Different approaches and technologies have been applied in clinical trials, ranging from protein (immunohistochemistry) to mRNA [...] Read more.
Inhibitors of fibroblast growth factor receptor (FGFR) signaling have been investigated in various human cancer diseases. Recently, the first compounds received FDA approval in biomarker-selected patient populations. Different approaches and technologies have been applied in clinical trials, ranging from protein (immunohistochemistry) to mRNA expression (e.g., RNA in situ hybridization) and to detection of various DNA alterations (e.g., copy number variations, mutations, gene fusions). We review, here, the advantages and limitations of the different technologies and discuss the importance of tissue and disease context in identifying the best predictive biomarker for FGFR targeting therapies. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of FGFR signaling and impact of various alterations. (<b>A</b>) Physiologic signaling upon ligand binding leads to various downstream signaling cascades affecting cellular survival, growth, migration, metabolism and interaction with cellular microenvironment. (<b>B</b>) Point mutations (marked in red) lead to constitutive activation by either affecting the extracellular ligand-binding domain or the intracellular tyrosine kinase domains. Signaling becomes independent of FGF ligand binding. (<b>C</b>) Gene fusions, rearrangements or translocations on DNA level (marked in dark green) lead to ligand-independent constitutive activation of the kinase domains by adding alternative kinase elements. (<b>D</b>) Gene amplification by DNA copy number alterations leads to higher expression of the receptor, providing more opportunities for ligands to bind and to activate the signaling cascade. It is noteworthy that all shown alterations also lead to increased mRNA expression levels but not all alterations lead to receptor overexpression. AKT: synonymous Protein Kinase B; β-cat: β-catenin; FGFR: fibroblast growth factor receptor; JAK: janus kinase; MAPK: mitogen-activated protein kinase; mTOR: mammalian/mechanistic target of rapamycin; PI3K: phosphoinositide-3-kiase; RAF: rapidly accelerated fibrosarcoma; RAS: rat sarcoma; STAT: signal transducer and activator of transcription; WNT: wingless and Int-1.</p>
Full article ">Figure 2
<p>Prevalence of FGFR alterations in selected tumor types. For each tumor type, the prevalence of amplifications, mutations, fusions or translocations and overexpression is highlighted according to [<a href="#B34-cells-11-03180" class="html-bibr">34</a>,<a href="#B35-cells-11-03180" class="html-bibr">35</a>,<a href="#B36-cells-11-03180" class="html-bibr">36</a>,<a href="#B37-cells-11-03180" class="html-bibr">37</a>,<a href="#B38-cells-11-03180" class="html-bibr">38</a>,<a href="#B39-cells-11-03180" class="html-bibr">39</a>,<a href="#B40-cells-11-03180" class="html-bibr">40</a>,<a href="#B42-cells-11-03180" class="html-bibr">42</a>,<a href="#B43-cells-11-03180" class="html-bibr">43</a>,<a href="#B44-cells-11-03180" class="html-bibr">44</a>,<a href="#B45-cells-11-03180" class="html-bibr">45</a>,<a href="#B46-cells-11-03180" class="html-bibr">46</a>,<a href="#B47-cells-11-03180" class="html-bibr">47</a>,<a href="#B48-cells-11-03180" class="html-bibr">48</a>,<a href="#B49-cells-11-03180" class="html-bibr">49</a>,<a href="#B50-cells-11-03180" class="html-bibr">50</a>,<a href="#B51-cells-11-03180" class="html-bibr">51</a>,<a href="#B113-cells-11-03180" class="html-bibr">113</a>,<a href="#B114-cells-11-03180" class="html-bibr">114</a>,<a href="#B115-cells-11-03180" class="html-bibr">115</a>,<a href="#B116-cells-11-03180" class="html-bibr">116</a>,<a href="#B117-cells-11-03180" class="html-bibr">117</a>,<a href="#B118-cells-11-03180" class="html-bibr">118</a>,<a href="#B119-cells-11-03180" class="html-bibr">119</a>,<a href="#B120-cells-11-03180" class="html-bibr">120</a>,<a href="#B121-cells-11-03180" class="html-bibr">121</a>,<a href="#B122-cells-11-03180" class="html-bibr">122</a>,<a href="#B123-cells-11-03180" class="html-bibr">123</a>,<a href="#B124-cells-11-03180" class="html-bibr">124</a>,<a href="#B125-cells-11-03180" class="html-bibr">125</a>,<a href="#B126-cells-11-03180" class="html-bibr">126</a>]. Overexpression relates to protein overexpression as (usually) detected via immunohistochemistry. FGFR1 data marked with * for cholangiocarcinoma represent mRNA expression data. The most prevalent alteration is depicted in bold for each tumor type. FGFR: fibroblast growth factor receptor; HNSCC: Head and Neck Squamous Cell Carcinoma; NSCLC: Non-Small-Cell Lung Cancer.</p>
Full article ">Figure 3
<p>Factors influencing the selection of a predictive biomarker assay for FGFR-inhibitor therapies. Alterations in FGFR1-4 impact on the predictivity of a biomarker assay. In addition to the molecular biology of the alterations (CNV, fusion, mutation, etc.), also, the underlying tumor entity (histology), the clinical staging (e.g., muscle-invasive vs. non-muscle-invasive bladder cancer), tissue availability and the assay technology with different target readouts (protein, mRNA, DNA) determine which FGFR targeting therapy would bring benefit to a patient.</p>
Full article ">
11 pages, 1086 KiB  
Review
Mitochondrial Distress in Methylmalonic Acidemia: Novel Pathogenic Insights and Therapeutic Perspectives
by Svenja Aline Keller and Alessandro Luciani
Cells 2022, 11(19), 3179; https://doi.org/10.3390/cells11193179 - 10 Oct 2022
Cited by 5 | Viewed by 3090
Abstract
Mitochondria are highly dynamic, double-membrane-enclosed organelles that sustain cellular metabolism and, hence, cellular, and organismal homeostasis. Dysregulation of the mitochondrial network might, therefore, confer a potentially devastating vulnerability to high-energy-requiring cell types, contributing to a broad variety of hereditary and acquired diseases, which [...] Read more.
Mitochondria are highly dynamic, double-membrane-enclosed organelles that sustain cellular metabolism and, hence, cellular, and organismal homeostasis. Dysregulation of the mitochondrial network might, therefore, confer a potentially devastating vulnerability to high-energy-requiring cell types, contributing to a broad variety of hereditary and acquired diseases, which include inborn errors of metabolism, cancer, neurodegeneration, and aging-associated adversities. In this Review, we highlight the biological functions of mitochondria-localized enzymes, from the perspective of understanding the pathophysiology of the inherited disorders destroying mitochondrial homeostasis and cellular metabolism. Using methylmalonic acidemia (MMA) as a paradigm of mitochondrial dysfunction, we discuss how mitochondrial-directed signaling pathways sustain the physiological homeostasis of specialized cell types and how these may be disturbed in disease conditions. This Review also provides a critical analysis of molecular underpinnings, through which defects in the autophagy-mediated quality control and surveillance systems contribute to cellular dysfunction, and indicates potential therapeutic strategies for affected tissues. These insights might, ultimately, advance the discovery and development of new therapeutics, not only for methylmalonic acidemia but also for other currently intractable mitochondrial diseases, thus transforming our ability to modulate health and homeostasis. Full article
(This article belongs to the Special Issue Autophagy in Kidney Homeostasis and Disease)
Show Figures

Figure 1

Figure 1
<p>The absence of the enzyme MMUT leads to accumulation of organic acids and mitochondrial abnormalities. Mutations in the <span class="html-italic">MMUT</span> gene encoding the mitochondrial enzyme methylmalonyl-coenzyme A mutase, which mediates the terminal step of branched chain amino acid and odd-chain lipid catabolism, trigger the accumulation of metabolites (e.g., methylmalonic acid, propionic acid, and 2-methylcitric acid) and lack of anaplerosis. This leads to morphologically abnormal mitochondria with disorganized cristae, decreased production of ATP (red arrows) and exaggerated generation of ROS/oxidative stress, ultimately causing severe organ dysfunctions that primarily affect brain, liver, and kidney.</p>
Full article ">Figure 2
<p>Working model depicting the link between MMUT, mitochondria, mitophagy, and epithelial homeostasis in healthy and MMA-affected kidney cells. In MMA-affected kidney cells and zebrafish, the deficiency of the enzyme MMUT and the resulting accumulation of toxic organic acids trigger mitochondrial abnormalities, which are characterized by a collapse of the mitochondrial membrane potential (ΔΨm, red arrows), impaired ATP production/bioenergetics (red arrows) and augmented generation of mitochondrial ROS and oxidative stress. Faulty execution of PINK1‒Parkin-mediated mitophagy induced by MMUT deficiency impedes the delivery of damaged mitochondria and their dismantling by autophagy–lysosome degradation systems. This, in turn, promotes the accumulation of damaged and/or dysfunctional, ROS-overproducing mitochondria that, ultimately, trigger cellular and kidney damage. The treatments with mitochondria-targeted ROS scavengers mito-TEMPO or MitoQ repair mitochondrial dysfunctions, neutralizes epithelial damage in MMA cells, and improves disease-relevant phenotypes in <span class="html-italic">mmut</span>-deficient zebrafish.</p>
Full article ">
22 pages, 4644 KiB  
Article
ROCK Inhibitor (Y-27632) Abolishes the Negative Impacts of miR-155 in the Endometrium-Derived Extracellular Vesicles and Supports Embryo Attachment
by Islam M. Saadeldin, Bereket Molla Tanga, Seonggyu Bang, Chaerim Seo, Okjae Koo, Sung Ho Yun, Seung Il Kim, Sanghoon Lee and Jongki Cho
Cells 2022, 11(19), 3178; https://doi.org/10.3390/cells11193178 - 10 Oct 2022
Cited by 6 | Viewed by 2648
Abstract
Extracellular vesicles (EVs) are nanosized vesicles that act as snapshots of cellular components and mediate cellular communications, but they may contain cargo contents with undesired effects. We developed a model to improve the effects of endometrium-derived EVs (Endo-EVs) on the porcine embryo attachment [...] Read more.
Extracellular vesicles (EVs) are nanosized vesicles that act as snapshots of cellular components and mediate cellular communications, but they may contain cargo contents with undesired effects. We developed a model to improve the effects of endometrium-derived EVs (Endo-EVs) on the porcine embryo attachment in feeder-free culture conditions. Endo-EVs cargo contents were analyzed using conventional and real-time PCR for micro-RNAs, messenger RNAs, and proteomics. Porcine embryos were generated by parthenogenetic electric activation in feeder-free culture conditions supplemented with or without Endo-EVs. The cellular uptake of Endo-EVs was confirmed using the lipophilic dye PKH26. Endo-EVs cargo contained miR-100, miR-132, and miR-155, together with the mRNAs of porcine endogenous retrovirus (PERV) and β-catenin. Targeting PERV with CRISPR/Cas9 resulted in reduced expression of PERV mRNA transcripts and increased miR-155 in the Endo-EVs, and supplementing these in embryos reduced embryo attachment. Supplementing the medium containing Endo-EVs with miR-155 inhibitor significantly improved the embryo attachment with a few outgrowths, while supplementing with Rho-kinase inhibitor (RI, Y-27632) dramatically improved both embryo attachment and outgrowths. Moreover, the expression of miR-100, miR-132, and the mRNA transcripts of BCL2, zinc finger E-box-binding homeobox 1, β-catenin, interferon-γ, protein tyrosine phosphatase non-receptor type 1, PERV, and cyclin-dependent kinase 2 were all increased in embryos supplemented with Endo-EVs + RI compared to those in the control group. Endo-EVs + RI reduced apoptosis and increased the expression of OCT4 and CDX2 and the cell number of embryonic outgrowths. We examined the individual and combined effects of RI compared to those of the miR-155 mimic and found that RI can alleviate the negative effects of the miR-155 mimic on embryo attachment and outgrowths. EVs can improve embryo attachment and the unwanted effects of the de trop cargo contents (miR-155) can be alleviated through anti-apoptotic molecules such as the ROCK inhibitor. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Obtaining the endometrial-derived extracellular vesicles (Endo-EVs). (<b>A</b>) Porcine endometrium (<span class="html-italic">n</span> = 6) was retrieved from the uterus (*) of diestrus sows (as indicated by corpora lutea, the black arrow); (<b>B</b>) primary endometrium cell culture was established (white arrow) from the endometrial tissue flakes (yellow arrow). On day-8 of primary outgrowths (Scale bar = 50 µm), the tissue chops were removed, and the cells were cultured in a serum-free culture medium to collect the conditioned medium for Endo-EVs isolation. (<b>C</b>) Endo-EVs were isolated by targeted nanofiltration method and were characterized by ZetaView nanoflow cytometry and nanoparticle tracking analysis and showed an average diameter of 115.6 ± 28.4 nm with a concentration of 1.1 × 10<sup>8</sup> particles/mL (dilution factor is 20X). (<b>D</b>) Endo-EVs were visualized by transmission electron microscope and showed bilipid vesicles (white arrow).</p>
Full article ">Figure 2
<p>Images of gel electrophoresis of mRNA and miRNA of the endometrium and their derived extracellular vesicles (Endo-EVs). The PCR products were electrophorized in agarose gel (2%), and the bands were visualized using a 100 bp DNA ladder as reference. The band expression of the snRNA (U6) and GAPDH were used as housekeeping genes for miRNA and mRNA, respectively. We contrasted the expression in Endo-EVs and found that some mRNAs were not expressed in the Endo-EVs such as GAPDH and catenin. For more details about the PCR product size, please refer to <a href="#cells-11-03178-t001" class="html-table">Table 1</a> in <a href="#sec2-cells-11-03178" class="html-sec">Section 2</a>.</p>
Full article ">Figure 3
<p>Cellular uptake of Endo-EVs. Endo-EVs were stained with a lipophilic live-imaging dye PKH26, and the free dye was removed by washing during isolation. Plain conditioned medium was processed in the same way as the isolated EVs and worked as negative control (Control). Attached embryonic cells were incubated with the stained control and Endo-EVs for 24 h and then were stained with DAPI and visualized by fluorescence microscope. White arrows indicate the presence of cytoplasmic stained EVs surrounding the nuclei. Scale bar = 20 µm.</p>
Full article ">Figure 4
<p>The effects of endometrial EVs (Endo-EVs) and ROCK inhibitor (RI) on porcine embryo development. (<b>A</b>) Day-7 zona-free embryos (<span class="html-italic">n</span> = 20, 3 replicates) were cultured on Matrigel-coated dishes in microdrops of culture medium in a humidified atmosphere of 5% CO<sub>2</sub> for 36 h. The control group was cultured in a plain culture medium while the RI group in a medium supplemented with Y-27632 (10 µg/mL) and EVs group in a medium supplemented with Endo-EVs of 2.6 × 106 particles/mL. In the combined group, embryos were cultured in a medium supplemented with both RI and EVs of the same working concentrations. Scale bar = 100 µm. All groups were imaged in a bright field before staining with TUNEL assay and contrasted with DAPI stain. White arrows indicate the apoptotic cells; (<b>B</b>–<b>D</b>) graphs show the embryo attachment, cell number, and apoptosis among the groups, respectively. Values (mean ± SD) were compared with ANOVA followed by Tukey’s test to determine the difference among the groups. Values denoted by ‡, #, §, and * were considered statistically different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>The effect of combined treatment of Endo-EVs and ROCK inhibitor (RI) on embryonic outgrowths. (<b>A</b>) Day-7 zona-free porcine embryos (<span class="html-italic">n</span> =18 for 3 replicates) were cultured on Matrigel-coated dishes in microdrops of culture medium in a humidified atmosphere of 5% CO<sub>2</sub> for 5 days. Scale bar = 50 µm; (<b>B</b>) the averages of percentages of embryonic cell outgrowths were calculated on day-2, day-3, and day-5 (mean ± S.E.M.) and compared with Student’s <span class="html-italic">t</span>-test. Asterisk (*) indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>The effect of combined treatment of Endo-EVs with ROCK inhibitor (RI) on OCT4 and CDX2 expression in cultured embryonic cells. (<b>A</b>) Day-7 zona-free porcine embryos (<span class="html-italic">n</span> = 18 for 3 replicates) were cultured on Matrigel-coated dishes in microdrops of culture medium in a humidified atmosphere of 5% CO<sub>2</sub> for 5 days and then incubated with primary antibodies specific to OCT4 and CDX2 followed by corresponding specific secondary antibodies. Scale bar = 50 µm. (<b>B</b>,<b>C</b>) Images of CDX2 and OCT4, respectively, were analyzed with ImageJ software to compare the pixels of the fluorescence intensity in the same exposure time, contrast, and area of analysis. The values were normalized to the control group as an arbitrary unit to show the fold of change between the groups. Values (mean ± S.E.M.) were analyzed with Student’s <span class="html-italic">t</span>-test. Asterisk (*) indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>Relative quantitative analysis (RT-qPCR) of mRNA transcripts expressed in the embryos treated with Endo-EVs and RI. Five blastocysts from each group for 4 replicates were used for qPCR analysis. The means were normalized to the control group and expressed as arbitrary units. Data were expressed as mean ± S.E.M. and the difference between the two groups was compared with Student’s <span class="html-italic">t</span>-test. Values denoted by an asterisk (*) were considered statistically significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 8
<p>Investigating the effects of the miR-155 inhibitor on embryonic attachment and development. (<b>A</b>) MiR-155 inhibitor was designed (<a href="#cells-11-03178-t001" class="html-table">Table 1</a>) and transfected to day-7 zona-free porcine embryos. Embryos were cultured on Matrigel-coated dishes in microdrops of culture medium containing Endo-EVs in a humidified atmosphere of 5% CO<sub>2</sub> for 5 days. Control Endo-EVs group was treated the same as the miR-155 group except for the absence of RNA sequence during transfection. Scale bar = 50 µm. All groups were imaged in a bright field before staining with TUNEL assay and contrasted with DAPI stain; (<b>B</b>–<b>D</b>) graphs show the embryo attachment, cell number, and apoptosis among the groups, and values (mean ± SEM) were compared with Student’s <span class="html-italic">t</span>-test to determine the difference among the groups. Values denoted by an asterisk (*) were considered statistically significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 9
<p>Investigating the effects of miR-155 mimic and ROCK inhibitor (RI) on embryonic attachment and development. (<b>A</b>) MiR-155 mimic duplex was designed (<a href="#cells-11-03178-t001" class="html-table">Table 1</a>) and transfected to day-7 zona-free porcine embryos. Embryos were cultured on Matrigel-coated dishes in microdrops of culture medium in a humidified atmosphere of 5% CO<sub>2</sub> for 5 days. The three groups were treated the same as the miR-155 mimic group except for the absence of RNA sequence during transfection of the RI group. Scale bar = 50 µm. All groups were imaged in a bright field before staining with TUNEL assay and contrasted with DAPI stain. White arrows indicate the apoptotic cells; (<b>B</b>–<b>D</b>) graphs show the embryo attachment, cell number, and apoptosis among the groups. Data were expressed as means ± S.E.M. Values (mean ± SD) were compared with ANOVA followed by Tukey’s test to determine the difference among the groups. Values denoted by symbols #, §, and * were considered statistically significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 10
<p>The impacts of targeting porcine endogenous retrovirus (PERV) expression in porcine endometrium through CRISPR/Cas9 on the derived EVs and the subsequent embryo development. (<b>A</b>) Endometrium was transfected with CRISP/Cas9 vector for 24 h;(<b>A</b>’,<b>A</b>’’) to compare the green fluorescence protein expression (white arrow, see <a href="#app1-cells-11-03178" class="html-app">Supplementary Figure S1</a> for the vector details) in control and mutated cells, respectively. The resulting cells were used to isolate Endo-EVs as mentioned previously. Scale bar = 50 µm. PERV targeted endometrium showed 5-fold and 4.5-fold reduction in the mRNA expression in both endometrium (<b>B</b>) and their derived EVs (<b>C</b>), respectively, while they showed upregulated miR-155 about 6-fold and 5-fold in endometrium (<b>B</b>) and their derived EVs (<b>C</b>), respectively. (<b>D</b>); (<b>E</b>) On day-5, embryo treated with PERV-targeted and PERV-diminished EVs showed low percentages of embryo attachment and outgrowths (<b>F</b>). Values were expressed as means ± S.E.M. and the difference between the two groups was compared with Student’s <span class="html-italic">t</span>-test. Values denoted by an asterisk (*) were considered statistically significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
16 pages, 3794 KiB  
Article
Ethylene Activates the EIN2-EIN3/EIL1 Signaling Pathway in Tapetum and Disturbs Anther Development in Arabidopsis
by Ben-Shun Zhu, Ying-Xiu Zhu, Yan-Fei Zhang, Xiang Zhong, Keng-Yu Pan, Yu Jiang, Chi-Kuang Wen, Zhong-Nan Yang and Xiaozhen Yao
Cells 2022, 11(19), 3177; https://doi.org/10.3390/cells11193177 - 10 Oct 2022
Cited by 10 | Viewed by 2792
Abstract
Ethylene was previously reported to repress stamen development in both cucumber and Arabidopsis. Here, we performed a detailed analysis of the effect of ethylene on anther development. After ethylene treatment, stamens but not pistils display obvious developmental defects which lead to sterility. [...] Read more.
Ethylene was previously reported to repress stamen development in both cucumber and Arabidopsis. Here, we performed a detailed analysis of the effect of ethylene on anther development. After ethylene treatment, stamens but not pistils display obvious developmental defects which lead to sterility. Both tapetum and microspores (or microsporocytes) degenerated after ethylene treatment. In ein2-1 and ein3-1 eil1-1 mutants, ethylene treatment did not affect their fertility, indicating the effects of ethylene on anther development are mediated by EIN2 and EIN3/EIL1 in vivo. The transcription of EIN2 and EIN3 are activated by ethylene in the tapetum layer. However, ectopic expression of EIN3 in tapetum did not induce significant anther defects, implying that the expression of EIN3 are regulated post transcriptional level. Consistently, ethylene treatment induced the accumulation of EIN3 in the tapetal cells. Thus, ethylene not only activates the transcription of EIN2 and EIN3, but also stabilizes of EIN3 in the tapetum to disturb its development. The expression of several ethylene related genes was significantly increased, and the expression of the five key transcription factors required for tapetum development was decreased after ethylene treatment. Our results thus point out that ethylene inhibits anther development through the EIN2-EIN3/EIL1 signaling pathway. The activation of this signaling pathway in anther wall, especially in the tapetum, induces the degeneration of the tapetum and leads to pollen abortion. Full article
(This article belongs to the Special Issue Pollen Development)
Show Figures

Figure 1

Figure 1
<p>The fertility was disturbed after exogenous ethylene treatment. (<b>A</b>) Inflorescence before ethylene treatment. Note that the start position of inflorescence (flower with anthers at stage 13) was marked with a red line so as to observe the elongation of siliques after 14 days. (<b>B</b>–<b>E</b>) Inflorescence of Col without ET treatment (<b>B</b>), with ethylene (100 µL L<sup>−1</sup>) treatment for 6 h (<b>C</b>), 12 h (<b>D</b>), and 24 h (<b>E</b>). After ET treatment, these plants grew for 14 days under normal conditions. Arrows indicate the closed buds or short siliques without seeds. The developmental stage of anthers in each bud before ethylene treatment is marked next to the corresponding siliques or flower buds. Bars = 2 cm in B–E. (<b>F</b>) Col flower. Bar = 1.0 mm. (<b>G</b>,<b>H</b>) Col flower after ethylene (100 µL L<sup>−1</sup>) treatment for 12 h (<b>G</b>) and 24 h (<b>H</b>). Note that filaments are shortened after 12 h or 24 h ethylene treatment, and the development of the anther of stage 8 is abnormal after 24 h ethylene treatment. Bars = 1.0 mm in (<b>G</b>,<b>H</b>).</p>
Full article ">Figure 2
<p>Alexander staining of anthers after 24 h of ethylene treatment. (<b>A</b>–<b>I</b>) Alexander staining of stage 5 to stage 12 anthers after 24 h treatment with ethylene (100 µL L<sup>−1</sup>). Bars = 50 µm. After ethylene treatment, plants were transferred to normal condition for further growth. The flower buds in the inflorescence gradually mature, and Alexander staining was carried out to stain the anthers of each mature flower.</p>
Full article ">Figure 3
<p>Ethylene treatment resulted in the degeneration of tapetum and microspores. (<b>A</b>–<b>G</b>) Semi-thin sections of Col anther before and after ethylene treatment (100 µL L<sup>−1</sup> for 24 h). Note for the degeneration of tapetal layer and microspores. Bars = 20 µm. Ms: microsporocyte; Tds: tetrad; Msp: microspores; Ep: epidermis; En: endothecium; ML: middle layer; T: tapetum; Vt: Vacuolated tapetum; Dmsp: degenerated microspores.</p>
Full article ">Figure 4
<p><span class="html-italic">etr1-7 ers1-2</span> double mutant showed abnormal tapetum and microspores. (<b>A</b>–<b>F</b>) Semi-thin sections of anthers from stage 5 to stage 12 of <span class="html-italic">etr1-7 ers1-2</span> mutant. Bars = 20 µm. Ep: epidermis; En: endothecium; ML: middle layer; T: Tapetum; Dt: Degenerated tapetum; Abp: Aborted pollen.</p>
Full article ">Figure 5
<p>The inflorescence of <span class="html-italic">ein2-1</span> and <span class="html-italic">ein3-1 eil1-1</span> is insensitive to exogenous ethylene treatment. (<b>A</b>–<b>D</b>) The inflorescence of <span class="html-italic">ein2-1</span> (<b>A</b>), <span class="html-italic">ein3-1eil1-1</span> (<b>B</b>), <span class="html-italic">ein3-1</span> (<b>C</b>) and <span class="html-italic">eil1-1</span> (<b>D</b>) after treated with ethylene (100 µL L<sup>−1</sup>) for 24 h. After ethylene treatment, these plants were transferred to normal growth condition for 14 days. Bars = 2 cm.</p>
Full article ">Figure 6
<p>The expression patterns of p<span class="html-italic">EIN2</span>:<span class="html-italic">VENUS</span> and p<span class="html-italic">EIN3</span>:<span class="html-italic">VENUS</span> reporters during anther development. (<b>A</b>,<b>B</b>) Fluorescence images of the p<span class="html-italic">EIN2</span>:<span class="html-italic">VENUS</span> (<b>A</b>) and p<span class="html-italic">EIN3</span>:<span class="html-italic">VENUS</span> (<b>B</b>) before and after ethylene treatment. The dashed box in the close-up region indicate the tapetum layer in both (<b>A</b>,<b>B</b>). Note for the VENUS signals were appeared in tapetum and were enhanced in epidermis and endothecium after ethylene treatment. The green channel showed the VENUS signals and the red fluorescence channel showed auto-fluorescence of chlorophyll in epidermis (Ep) and endothecium (En). T: tapetum. Bars = 30 μm.</p>
Full article ">Figure 7
<p>Ectopic transcription of <span class="html-italic">EIN2</span> and <span class="html-italic">EIN3</span> in tapetum did not influence pollen formation. (<b>A</b>) Alexander staining of pollens of p<span class="html-italic">DYT1</span>/Col, p<span class="html-italic">DYT1</span>:<span class="html-italic">EIN2</span>/Col, and p<span class="html-italic">DYT1</span>:<span class="html-italic">EIN3</span>/Col transgenic plants. Note, all of the three transgenic populations showed two phenotypes: plants showed normal pollens and plants showed partial pollen lethality phenotype. Bars = 50 µm. (<b>B</b>) Left panel: The percentage of plants with abnormal anthers in each group of the transgenic population. Middle panel: Real-time quantitative RT-PCR analysis of <span class="html-italic">EIN2</span> transcript levels in three independent lines of p<span class="html-italic">DYT1</span>:<span class="html-italic">EIN2</span>/Col transgenic plants. Right panel: Real-time quantitative RT-PCR analysis of <span class="html-italic">EIN3</span> transcript levels in three independent lines of p<span class="html-italic">DYT1</span>:<span class="html-italic">EIN3</span>/Col transgenic plants. β-tubulin was used as an internal control for normalization. Data are mean ± SEM of three biological replicates with technical duplicates for each.</p>
Full article ">Figure 8
<p>The expression pattern of <span class="html-italic">EIN3</span> in anthers after ethylene treatment. Fluorescence images of the p<span class="html-italic">EIN3</span>:<span class="html-italic">EIN3-GFP</span> in anthers. The green channel showed the GFP signal and the red fluorescence channel showed auto-fluorescence of chlorophyll in epidermal and endothecium. The blue, white, and red arrows indicate the GFP signal in the nuclei of tapetum, epidermis and endothecium, respectively. The green arrow indicate the GFP signal in the nuclei of filament. Ep: epidermis; En: endothecium; ML: middle layer; T: tapetum. Bars = 30 μm.</p>
Full article ">Figure 9
<p>Quantitative RT-PCR analysis of <span class="html-italic">ERFs</span>, <span class="html-italic">SAGs</span> and tapetum development-related key transcription factors after ethylene treatment. (<b>A</b>) Relative expression of <span class="html-italic">EIN2</span>, <span class="html-italic">EIN3</span>, <span class="html-italic">ERFs</span> and <span class="html-italic">SAGs</span> genes in the inflorescence of Col after ethylene treatment for 24 h. (<b>B</b>) The expression of the five key transcription factor genes in tapetum development is reduced after ethylene treatment. Relative expression of <span class="html-italic">AMS</span>, <span class="html-italic">DYT1</span>, <span class="html-italic">TDF1</span>, <span class="html-italic">MS188</span>, and <span class="html-italic">MS1</span> in the inflorescence of Col-0 after ethylene treatment for 24 h. Col treated with air for 24 h was used as a mock. Gene expression was relative to that of the mock. β-tubulin was used as an internal control for normalization. Data are mean ± SEM of 3 biological replicates with technical duplicates for each. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (<span class="html-italic">t</span>-test).</p>
Full article ">
11 pages, 448 KiB  
Communication
An Inflammatory Signature to Predict the Clinical Benefit of First-Line Cetuximab Plus Platinum-Based Chemotherapy in Recurrent/Metastatic Head and Neck Cancer
by Stefano Cavalieri, Mara Serena Serafini, Andrea Carenzo, Silvana Canevari, Deborah Lenoci, Federico Pistore, Rosalba Miceli, Stefania Vecchio, Daris Ferrari, Cecilia Moro, Andrea Sponghini, Alessia Caldara, Maria Cossu Rocca, Simona Secondino, Gabriella Moretti, Nerina Denaro, Francesco Caponigro, Emanuela Vaccher, Gaetana Rinaldi, Francesco Ferraù, Paolo Bossi, Lisa Licitra and Loris De Ceccoadd Show full author list remove Hide full author list
Cells 2022, 11(19), 3176; https://doi.org/10.3390/cells11193176 - 10 Oct 2022
Viewed by 2140
Abstract
Epidermal growth factor receptor (EGFR) pathway has been shown to play a crucial role in several inflammatory conditions and host immune-inflammation status is related to tumor prognosis. This study aims to evaluate the prognostic significance of a four-gene inflammatory signature in recurrent/metastatic (R/M) [...] Read more.
Epidermal growth factor receptor (EGFR) pathway has been shown to play a crucial role in several inflammatory conditions and host immune-inflammation status is related to tumor prognosis. This study aims to evaluate the prognostic significance of a four-gene inflammatory signature in recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) patients treated with the EGFR inhibitor cetuximab plus chemotherapy. The inflammatory signature was assessed on 123 R/M HNSCC patients, enrolled in the multicenter trial B490 receiving first-line cetuximab plus platinum-based chemotherapy. The primary endpoint of the study was progression free survival (PFS), while secondary endpoints were overall survival (OS) and objective response rate (ORR). The patient population was subdivided into 3 groups according to the signature score groups. The four-genes-signature proved a significant prognostic value, resulting in a median PFS of 9.2 months in patients with high vs. 6.2 months for intermediate vs. 3.9 months for low values (p = 0.0016). The same findings were confirmed for OS, with median time of 18.4, 13.4, and 7.5 months for high, intermediate, and low values of the score, respectively (p = 0.0001). When ORR was considered, the signature was significantly higher in responders than in non-responders (p = 0.0092), reaching an area under the curve (AUC) of 0.65 (95% CI: 0.55–0.75). Our findings highlight the role of inflammation in the response to cetuximab and chemotherapy in R/M-HNSCC and may have translational implications for improving treatment selection. Full article
Show Figures

Figure 1

Figure 1
<p>CONSORT diagram.</p>
Full article ">Figure 2
<p>Prognostic value of the inflammation signature. (<b>A</b>) Kaplan–Meier curves of B490 patients’ PFS associated to the four-gene inflammatory signature. (<b>B</b>) Kaplan–Meier curves of B490 patients’ OS associated to the four-gene inflammatory signature. Curve separation was assessed by the log-rank test. Samples were stratified based on tertiles: high (red), intermediate (yellow) and low (gray) inflammatory score groups.</p>
Full article ">Figure 3
<p>The inflammatory score as continuous variable was associated to cetuximab response. (<b>A</b>) Boxplot of the inflammatory scores in responders (complete responders + partial responders, CR + PR) and non-responder (stable disease + progressive disease, SD + PD). The inflammatory score is based on scaled expression of the four genes retrieved from normalized RNAseq data, (<b>B</b>) receiver operating characteristics (ROC) curve and area under the curve (AUC) of the four-gene inflammatory signature.</p>
Full article ">
13 pages, 640 KiB  
Article
KRAS G12D Mutation Subtype in Pancreatic Ductal Adenocarcinoma: Does It Influence Prognosis or Stage of Disease at Presentation?
by Henry Shen, Joanne Lundy, Andrew H. Strickland, Marion Harris, Michael Swan, Christopher Desmond, Brendan J. Jenkins and Daniel Croagh
Cells 2022, 11(19), 3175; https://doi.org/10.3390/cells11193175 - 10 Oct 2022
Cited by 23 | Viewed by 3228
Abstract
Background: KRAS G12D mutation subtype is present in over 40% of pancreatic ductal adenocarcinoma (PDAC), one of the leading global causes of cancer death. This retrospective cohort study aims to investigate whether detection of the KRAS G12D mutation subtype in PDAC patients [...] Read more.
Background: KRAS G12D mutation subtype is present in over 40% of pancreatic ductal adenocarcinoma (PDAC), one of the leading global causes of cancer death. This retrospective cohort study aims to investigate whether detection of the KRAS G12D mutation subtype in PDAC patients is a determinant of prognosis across all stages of disease. Methods: We reviewed the medical records of 231 patients presenting with PDAC at a large tertiary hospital, and compared survival using the Kaplan Meier, log-rank test and Cox proportional hazards regression model. Results: KRAS G12D mutation subtype was not significantly associated with poorer survival compared across the whole population of PDAC patients (p = 0.107; HR 1.293 95% CI (0.946–1.767)). However, KRAS G12D patients who were resectable had a shorter median survival time of 356 days compared to all other genotypes (median survival 810 days) (p = 0.019; HR 1.991 95% CI (1.121–3.537)). Conclusions: KRAS G12D patients who were resectable at diagnosis had shorter survival compared to all other PDAC patients. These data suggest that KRAS G12D may be a clinically useful prognostic biomarker of PDAC. Full article
(This article belongs to the Special Issue Targeting RAS-Dependent Cancers)
Show Figures

Figure 1

Figure 1
<p>Kaplan Meier curves and respective numbers at risk. G12D subtype associated with poorer survival in patients NCCN staged as resectable. (<b>a</b>) G12D subtype associated with poorer survival in patients NCCN staged as operable. (<b>b</b>) G12D subtype associated with poorer survival in patients who received surgery. G12D absent includes both other <span class="html-italic">KRAS</span> subtypes and <span class="html-italic">KRAS</span> wild-type.</p>
Full article ">
26 pages, 3314 KiB  
Review
Mitochondrial VDAC1: A Potential Therapeutic Target of Inflammation-Related Diseases and Clinical Opportunities
by Hang Hu, Linlin Guo, Jay Overholser and Xing Wang
Cells 2022, 11(19), 3174; https://doi.org/10.3390/cells11193174 - 10 Oct 2022
Cited by 29 | Viewed by 7060
Abstract
The multifunctional protein, voltage-dependent anion channel 1 (VDAC1), is located on the mitochondrial outer membrane. It is a pivotal protein that maintains mitochondrial function to power cellular bioactivities via energy generation. VDAC1 is involved in regulating energy production, mitochondrial oxidase stress, Ca2+ [...] Read more.
The multifunctional protein, voltage-dependent anion channel 1 (VDAC1), is located on the mitochondrial outer membrane. It is a pivotal protein that maintains mitochondrial function to power cellular bioactivities via energy generation. VDAC1 is involved in regulating energy production, mitochondrial oxidase stress, Ca2+ transportation, substance metabolism, apoptosis, mitochondrial autophagy (mitophagy), and many other functions. VDAC1 malfunction is associated with mitochondrial disorders that affect inflammatory responses, resulting in an up-regulation of the body’s defensive response to stress stimulation. Overresponses to inflammation may cause chronic diseases. Mitochondrial DNA (mtDNA) acts as a danger signal that can further trigger native immune system activities after its secretion. VDAC1 mediates the release of mtDNA into the cytoplasm to enhance cytokine levels by activating immune responses. VDAC1 regulates mitochondrial Ca2+ transportation, lipid metabolism and mitophagy, which are involved in inflammation-related disease pathogenesis. Many scientists have suggested approaches to deal with inflammation overresponse issues via specific targeting therapies. Due to the broad functionality of VDAC1, it may become a useful target for therapy in inflammation-related diseases. The mechanisms of VDAC1 and its role in inflammation require further exploration. We comprehensively and systematically summarized the role of VDAC1 in the inflammatory response, and hope that our research will lead to novel therapeutic strategies that target VDAC1 in order to treat inflammation-related disorders. Full article
(This article belongs to the Collection The Pathomechanism of Mitochondrial Diseases)
Show Figures

Figure 1

Figure 1
<p><b>VDAC1 regulates inflammatory pathogenesis.</b> Mitochondria are the center of energy generation, the TCA cycle, glycolysis and lipid metabolism. VDAC1 is the fundamental component that maintains mitochondrial function. VDAC1 plays an important role in the regulation of apoptosis, mtDNA release, Ca<sup>2+</sup> signaling, TCA cycle, glycolysis, lipid metabolism and mitophagy. Impaired mitochondrial homeostasis with dysfunctional signal networks results in inflammatory pathogenesis and mitochondrial diseases. <b>Abbreviations:</b> ACSL: long-chain acyl-CoA synthase; BAX: Bcl-2-associated X protein; CPT1a: carnitine palmitoyltransferase 1A; Cyto c: cytochrome c; HK: hexokinase; LC3: microtubule-associated proteins 1A/1B light chain 3; IMM: inner mitochondrial membrane; MOMP: mitochondrial outer membrane permeabilization; mtDNA: mitochondrial DNA; PINK1: PTEN-induced putative kinase 1; TCA cycle: tricarboxylic acid cycle; Ub: ubiquitin; VDAC1: voltage-dependent anion channel 1.</p>
Full article ">Figure 2
<p><b>VDAC1 mediates apoptosis and mtDNA release to promote cytokines expression and inflammatory pathogenesis.</b> (<b>A</b>) Bcl-2 family member, BAX, interacts with VDAC1 to release cytochrome c into the cytoplasm, promoting apoptosis. (<b>B</b>) MOMP induces proteasomal degradation of IAPs, which causes NIK to further induce the pro-inflammatory NF-κB signal and activate caspase-1/8; this in turn results in the maturation of pro-inflammatory factor IL-1β. (<b>C</b>) Mitochondrial overproduced ROS oxidize mtDNA to fomtDNA. The mtDNA and fomtDNA pass the VDAC1 oligomers channel or the oligomerization BAX pore into the cytoplasm. The released mtDNA/fomtDNA induce the cGAS-STING pathway to promote interferon gene expressions via TBK1-IRF3 to up-regulate IFN-α/β, or through TBK1-NF-κB to enhance TNF-α, IL-6 and IL-12. Additionally, mtDNA interacts with TLR9 and promotes TNF-α, IL-6 and IL-12 expression via NF-κB signaling. Moreover, the released mtDNA induces the NLRP3 inflammasome and AIM2 inflammasome to enhance caspase-1/8 activation to promote IL-1β/IL-18 maturation. <b>Abbreviations:</b> AIM2: absent in melanoma 2; BAX: Bcl-2-associated X protein; cGAS: cyclic GMP-AMP synthase; Cyto c: cytochrome c; fomtDNA: oxidized mtDNA fragments; IAP: inhibitors of apoptosis; IFN: interferon; IL: interleukin; IRF3: interferon regulatory factor 3; IMM: inner mitochondrial membrane; MOMP: mitochondrial outer membrane permeabilization; mtDNA: mitochondrial DNA; NF-κB: nuclear factor-κB; NIK: NF-κB induced kinase; NLRP3: nucleotide-binding domain and leucine-rich repeat (LRR) containing P3; ROS: reactive oxygen species; STING: stimulator of interferon genes; TLR9: Toll-like receptor 9; TBK1: TANK-binding kinase 1; TNF: tumor necrosis factor; VDAC1: voltage-dependent anion channel 1.</p>
Full article ">Figure 3
<p><b>VDAC1 is involved in Ca<sup>2+</sup> transportation, lipid metabolism, glycolysis</b><b>, TCA cycle</b><b>and in inflammatory responses.</b> (<b>A</b>) VDAC1 regulates Ca<sup>2+</sup> transportation, glycolysis, TCA cycle and lipid metabolism. VDAC1 transports Ca<sup>2+</sup> between the mitochondria and cytoplasm to maintain calcium homeostasis. In the energy generation system, the VDAC1 pore maintains substrates, metabolites, biomolecules, etc., in a balanced manner to sustain salutogenesis. (<b>B</b>) Ca<sup>2+</sup> signaling affects the inflammatory responses of neutrophils, macrophages, dendritic cells and CD4<sup>+</sup> T cells. (<b>C</b>) Inflammatory responses of macrophages, dendritic cells and CD4<sup>+</sup> T cells are promoted by glycolysis/TCA cycle energy generation pathways. <b>Abbreviations:</b> VDAC1: voltage-dependent anion channel 1; TRPML1: also known as MCOLN1, mucolipin TRP cation channel 1; GRP75: glucose-regulated protein 75; IP3R: inositol 1,4,5-trisphosphate receptor; DJ1: deglycase DJ-1, also known as Parkinson disease protein 7, is encoded by the PARK7 gene in human; RyR2: ryanodine receptor 2; CPT1a: carnitine palmitoyltransferase 1A; ACSL: long-chain acyl-CoA synthase; TCA cycle: tricarboxylic acid cycle; HK: hexokinase; ATP: adenosine triphosphate; ADP: adenosine diphosphate; NADH: nicotinamide adenine dinucleotide hydrogen; PEP: phosphoenolpyruvate; Th: T helper.</p>
Full article ">Figure 4
<p><b>VDAC1, PINK1/Parkin signaling and mitophagy.</b> (<b>A</b>) PINK1/Parkin targets damaged mitochondria, ubiquitinates VDAC1, and ultimately degrades damaged mitochondria by promoting mitophagy. (<b>B</b>) Mitophagy modulates NLRP3, MAVS and mtDNA release, affecting the immune response. NLRP3 activates caspase-1 to promote IL-1β/IL-18 maturation. MAVS enhance IFN-α/β expression. Meanwhile, mitophagy suppresses NLRP3, MAVS and mtDNA release, which results in reduced cytokines release. Additionally, mitophagy can also promote mtDNA release, which affects cytokines expression. <b>Abbreviations:</b> IL: interleukin; IFN: interferon; LC3: microtubule-associated proteins 1A/1B light chain 3; MAVS: mitochondrial antiviral signaling protein; NLRP3: nucleotide-binding domain and leucine-rich repeat (LRR) containing P3; PINK1: PTEN-induced putative kinase 1;; Ub: ubiquitin; VDAC1: voltage-dependent anion channel 1.</p>
Full article ">
17 pages, 24126 KiB  
Article
Captopril Alleviates Chondrocyte Senescence in DOCA-Salt Hypertensive Rats Associated with Gut Microbiome Alteration
by Lok Chun Chan, Yuqi Zhang, Xiaoqing Kuang, Mohamad Koohi-Moghadam, Haicui Wu, Theo Yu Chung Lam, Jiachi Chiou and Chunyi Wen
Cells 2022, 11(19), 3173; https://doi.org/10.3390/cells11193173 - 10 Oct 2022
Cited by 2 | Viewed by 2497
Abstract
Gut microbiota is the key controller of healthy aging. Hypertension and osteoarthritis (OA) are two frequently co-existing age-related pathologies in older adults. Both are associated with gut microbiota dysbiosis. Hereby, we explore gut microbiome alteration in the Deoxycorticosterone acetate (DOCA)-induced hypertensive rat model. [...] Read more.
Gut microbiota is the key controller of healthy aging. Hypertension and osteoarthritis (OA) are two frequently co-existing age-related pathologies in older adults. Both are associated with gut microbiota dysbiosis. Hereby, we explore gut microbiome alteration in the Deoxycorticosterone acetate (DOCA)-induced hypertensive rat model. Captopril, an anti-hypertensive medicine, was chosen to attenuate joint damage. Knee joints were harvested for radiological and histological examination; meanwhile, fecal samples were collected for 16S rRNA and shotgun sequencing. The 16S rRNA data was annotated using Qiime 2 v2019.10, while metagenomic data was functionally profiled with HUMAnN 2.0 database. Differential abundance analyses were adopted to identify the significant bacterial genera and pathways from the gut microbiota. DOCA-induced hypertension induced p16INK4a+ senescent cells (SnCs) accumulation not only in the aorta and kidney (p < 0.05) but also knee joint, which contributed to articular cartilage degradation and subchondral bone disturbance. Captopril removed the p16INK4a + SnCs from different organs, partially lowered blood pressure, and mitigated cartilage damage. Meanwhile, these alterations were found to associate with the reduction of Escherichia-Shigella levels in the gut microbiome. As such, gut microbiota dysbiosis might emerge as a metabolic link in chondrocyte senescence induced by DOCA-triggered hypertension. The underlying molecular mechanism warrants further investigation. Full article
(This article belongs to the Special Issue Cellular Senescence in Aging and Aging-Related Diseases)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Blood pressure measured every 2 weeks from week 0 to week 14 of the Control, DOCA, and DOCA + Captopril groups. The treatment of DOCA starts from w0. In week 9, DOCA + Captopril groups began to feed captopril (<span class="html-italic">n</span> = 8 for each group). The results of p16 staining in the aorta (<b>B</b>) and kidney (<b>D</b>) with red boxes highlighting the p16-positive cells. (<b>C</b>,<b>E</b>) show the percentage of p16-positive cells in the aorta and kidney, respectively. (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>The p16 staining results of cartilage (<b>A</b>), synovium (<b>C</b>), meniscus (<b>E</b>), and secondary spongiosa (<b>G</b>) are shown with the red boxes indicating the p16- positive regions. The percentage of p16-positive cells in the (<b>B</b>) cartilage, (<b>D</b>) synovium, (<b>F</b>) meniscus, and (<b>H</b>) secondary spongiosa in Control (red), DOCA (green), and Captopril-treated group (blue) are plotted. (<b>I</b>,<b>J</b>) The results of p53 staining of cartilage and the percentage of p53-positive cells in the sample. The MMP13 staining of the (<b>K</b>,<b>L</b>) cartilage and (<b>M</b>,<b>N</b>) synovium of the Control, DOCA, and DOCA + Captopril groups, respectively. (*** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>(<b>A</b>) The 3D bone structure of secondary spongiosa under micro-CT with the (<b>B</b>) bone volume ratio (BV/TV), (<b>C</b>) trabecular number (Tb.N), (<b>D</b>) trabecular separation (Tb.Sp), and (<b>E</b>) trabecular thickness (Tb.Th) calculated for the Control, DOCA, and DOCA+Captopril groups, respectively. (*** <span class="html-italic">p</span> &lt; 0.001) (<b>F</b>) The heatmap of the mean values of the top 50 radiomic features quantifying the texture of secondary spongiosa.</p>
Full article ">Figure 4
<p>(<b>A</b>) Relative abundance TAXA plot of the top 20 most abundant genus-level taxonclassified sequences from gut bacterial 16S rRNA gene amplicon sequencing (<span class="html-italic">n</span> = 24). (<b>B</b>) The gut microbial genus richness of the Control, DOCA, and DOCA + Captopril groups. A Mann–Whitney U test was conducted, showing a statistically insignificant (<span class="html-italic">p</span> = 0.414) difference between the richness of the control and DOCA groups, while a marginally significant difference could be observed among the DOCA and DOCA + Captopril groups (<span class="html-italic">p</span> = 0.065). (<b>C</b>) Cladogram to demonstrate the linear discriminant effect size of the significant genera. (<b>D</b>) Gut microbial genus correlation networks of the Control, DOCA, and DOCA + Captopril groups, respectively. (<b>E</b>) Density and (<b>F</b>) Shannon entropy of the correlation networks of the Control (red), DOCA (green), and DOCA + Captopril (blue) groups. All the network parameters were compared among the groups through one-way ANOVA with Tukey HSD posthoc analysis (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>(<b>A</b>) Heatmap of the log-fold-change of the top 13 genus having differential abundance among the Control, DOCA, and DOCA + Captopril groups. (<b>B</b>) The plot of the first 2 principal components (PCs) of the identified bacterial genera’s abundance, with the first PC explaining 18% variance and the second PC explaining 12% variance. (<b>C</b>–<b>F</b>) Box plots show the abundance of the 4 highlighted bacterial genera, and the <span class="html-italic">p</span>-values of pairwise comparison are reported. (* <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>(<b>A</b>) Heatmap of the log-fold-change of the top 32 differentially expressed pathways identified from the taxonclassified sequences from gut bacterial shotgun metagenomic sequencing among the Control, DOCA, and DOCA + Captopril groups. (<b>B</b>) The plot of the first 2 principal components (PCs) of the 32 identified bacterial pathways, with the first PC explaining 18% variance and the second PC explaining 12% variance. (<b>C</b>,<b>D</b>) Box plots demonstrate the expression of the 2 identified bacterial pathways in each group with the <span class="html-italic">p</span>-values of the group comparison reported. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 7
<p>Multi-omics correlation networks between the selected metagenomic bacterial pathways, 16S bacterial genus, phenotypes secondary spongiosa, and blood pressure of all experimental groups. The nodes are colored in orange for the pathways enriched from metagenomic data, cyan for bacterial genus identified from 16S rRNA Gene Sequencing, blue for secondary spongiosa phenotype, and pink for blood pressure. The edges of the networks are defined by Spearman’s correlation coefficients with Benjamini and Yekutieli-adjusted <span class="html-italic">p</span>-value &lt; 0.05. Greater intensity and thickness of the edges represent the higher strength of the correlation. An index is shown on the right for the mapping between the enriched bacterial pathways and their corresponding codes.</p>
Full article ">Figure A1
<p>Average body weights of the rats of the control, DOCA-induced hypertensive, and captopril-treated groups from weeks 0 to 14.</p>
Full article ">Figure A2
<p>The results of Safranine O staining in cartilage in joints of the Control, DOCA, and DOCA + Captopril group, with the red box highlighting the regions exhibiting key differences in the staining intensity.</p>
Full article ">Figure A3
<p>(<b>A</b>) The results of p16 staining in primary spongiosa of the control, DOCA, and DOCA + Captopril groups. (<b>B</b>) The bar chart shows the percentage of p16-positive cells in the samples (*** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>) The 3D bone structure of primary spongiosa under micro-CT with the (<b>D</b>) bone volume ratio (BV/TV), (<b>E</b>) trabecular number (Tb.N), (<b>F</b>) trabecular separation (Tb.Sp), and (<b>G</b>) trabecular thickness (Tb.Th) calculated for the Control, DOCA, and DOCA + Captopril groups, respectively (*** <span class="html-italic">p</span> &lt; 0.001). (<b>H</b>) The heatmap of the top 50 radiomic features quantifying the texture of primary spongiosa. (<b>I</b>) The Sirius red staining results of p16 staining in primary spongiosa of the Control, DOCA, and DOCA + Captopril groups. DOCA and DOCA + Captopril groups show more fibrosis than Control.</p>
Full article ">Figure A4
<p>The centrality (<b>A</b>), clustering coefficient (<b>B</b>), and heterogeneity (<b>C</b>) of the correlation network between the abundance of genus level in the Control, DOCA, and DOCA + Captopril groups. Inter-group comparisons were conducted using one-way ANOVA followed by the Tukey HSD post hoc test. (<b>D</b>) The log-LDA score of the gut genera, all genera shown in the plot have significant LDA-based effect size (<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; 0.01).</p>
Full article ">
27 pages, 893 KiB  
Review
The Extracellular Matrix and Neuroblastoma Cell Communication—A Complex Interplay and Its Therapeutic Implications
by Irena Horwacik
Cells 2022, 11(19), 3172; https://doi.org/10.3390/cells11193172 - 10 Oct 2022
Cited by 10 | Viewed by 3041
Abstract
Neuroblastoma (NB) is a pediatric neuroendocrine neoplasm. It arises from the sympatho-adrenal lineage of neural-crest-derived multipotent progenitor cells that fail to differentiate. NB is the most common extracranial tumor in children, and it manifests undisputed heterogeneity. Unsatisfactory outcomes of high-risk (HR) NB patients [...] Read more.
Neuroblastoma (NB) is a pediatric neuroendocrine neoplasm. It arises from the sympatho-adrenal lineage of neural-crest-derived multipotent progenitor cells that fail to differentiate. NB is the most common extracranial tumor in children, and it manifests undisputed heterogeneity. Unsatisfactory outcomes of high-risk (HR) NB patients call for more research to further inter-relate treatment and molecular features of the disease. In this regard, it is well established that in the tumor microenvironment (TME), malignant cells are engaged in complex and dynamic interactions with the extracellular matrix (ECM) and stromal cells. The ECM can be a source of both pro- and anti-tumorigenic factors to regulate tumor cell fate, such as survival, proliferation, and resistance to therapy. Moreover, the ECM composition, organization, and resulting signaling networks are vastly remodeled during tumor progression and metastasis. This review mainly focuses on the molecular mechanisms and effects of interactions of selected ECM components with their receptors on neuroblastoma cells. Additionally, it describes roles of enzymes modifying and degrading ECM in NB. Finally, the article gives examples on how the knowledge is exploited for prognosis and to yield new treatment options for NB patients. Full article
Show Figures

Figure 1

Figure 1
<p>Anti-tumor and pro-tumor effects of selected proteoglycans in NB (see <a href="#sec3-cells-11-03172" class="html-sec">Section 3</a> for more detail) [<a href="#B50-cells-11-03172" class="html-bibr">50</a>,<a href="#B51-cells-11-03172" class="html-bibr">51</a>,<a href="#B53-cells-11-03172" class="html-bibr">53</a>,<a href="#B54-cells-11-03172" class="html-bibr">54</a>,<a href="#B56-cells-11-03172" class="html-bibr">56</a>,<a href="#B62-cells-11-03172" class="html-bibr">62</a>,<a href="#B65-cells-11-03172" class="html-bibr">65</a>,<a href="#B66-cells-11-03172" class="html-bibr">66</a>].</p>
Full article ">Figure 2
<p>Examples of factors mediating interactions of NB cells with the ECM that affect malignancy (see <a href="#sec4-cells-11-03172" class="html-sec">Section 4</a>, <a href="#sec5-cells-11-03172" class="html-sec">Section 5</a> and <a href="#sec6-cells-11-03172" class="html-sec">Section 6</a> for more detail) [<a href="#B81-cells-11-03172" class="html-bibr">81</a>,<a href="#B86-cells-11-03172" class="html-bibr">86</a>,<a href="#B87-cells-11-03172" class="html-bibr">87</a>,<a href="#B98-cells-11-03172" class="html-bibr">98</a>,<a href="#B99-cells-11-03172" class="html-bibr">99</a>,<a href="#B102-cells-11-03172" class="html-bibr">102</a>,<a href="#B103-cells-11-03172" class="html-bibr">103</a>,<a href="#B105-cells-11-03172" class="html-bibr">105</a>,<a href="#B106-cells-11-03172" class="html-bibr">106</a>,<a href="#B120-cells-11-03172" class="html-bibr">120</a>,<a href="#B122-cells-11-03172" class="html-bibr">122</a>,<a href="#B148-cells-11-03172" class="html-bibr">148</a>,<a href="#B149-cells-11-03172" class="html-bibr">149</a>,<a href="#B160-cells-11-03172" class="html-bibr">160</a>,<a href="#B163-cells-11-03172" class="html-bibr">163</a>,<a href="#B164-cells-11-03172" class="html-bibr">164</a>].</p>
Full article ">
14 pages, 2582 KiB  
Article
In Vitro Transfection of Up-Regulated Genes Identified in Favorable-Outcome Neuroblastoma into Cell Lines
by Yoko Hiyama, Emi Yamaoka, Takahiro Fukazawa, Masato Kojima, Yusuke Sotomaru and Eiso Hiyama
Cells 2022, 11(19), 3171; https://doi.org/10.3390/cells11193171 - 10 Oct 2022
Viewed by 2474
Abstract
We previously used microarrays to show that high expression of DHRS3, NROB1, and CYP26A1 predicts favorable NB outcomes. Here, we investigated whether expression of these genes was associated with suppression of NB cell (SK-N-SH, NB12, and TGW) growth. We assessed morphology [...] Read more.
We previously used microarrays to show that high expression of DHRS3, NROB1, and CYP26A1 predicts favorable NB outcomes. Here, we investigated whether expression of these genes was associated with suppression of NB cell (SK-N-SH, NB12, and TGW) growth. We assessed morphology and performed growth, colony-formation, and migration assays, as well as RNA sequencing. The effects of the transient expression of these genes were also assessed with a tetracycline-controlled expression (Tet-On) system. Gene overexpression reduced cell growth and induced morphological senescence. Gene-expression analysis identified pathways involving cellular senescence and cell adhesion. In these cells, transduced gene dropout occurred during passage, making long-term stable gene transfer difficult. Tet-On-induced gene expression caused more pronounced cell-morphology changes. Specifically, DHRS3 and NROB1 led to rapid inhibition and arrest of cell growth, though CYP26A1 did not affect cell-growth rate or cell cycle. DHRS3 arrested the cell cycle by interacting with the all-trans-retinol pathway and drove differentiation and senescence in tumors. Overexpression of these genes reduced the malignant grade of these cells. A new therapeutic strategy might be the induction of these genes, as they suppress the growth of high-risk neuroblastoma and lead to differentiation and senescence. Full article
(This article belongs to the Collection Feature Papers in 'Cell Proliferation and Division')
Show Figures

Figure 1

Figure 1
<p>Transfection efficiency in SK-N-SH, NH12, and TGW neuroblastoma cell lines. Representative images of transfected, undifferentiated neuroblastoma cell lines. (<b>A</b>) Stable expression levels of transfected gene products were assessed with fluorescent immunostaining in transfected cell lines (a–c). Expression of exogenous proteins was detected with FLAG M2 antibodies and their respective endogenous antibodies. In established cell lines, DHRS3 was expressed mainly in the plasma membrane and endoplasmic reticulum (a,b). The merged photos include nuclear staining by DAPI. (<b>B</b>) DHRS3-transfected strains were lipid-stained using Lipid Tox and visualized by transmission electron microscopy, and we found an accumulation of DHRS3 in small vesicles (arrows). These vesicles were observed shortly after transfection in NH12 and SK-N-SH cells but were absent in TGWs. In addition, CYP26A1 was also expressed in both the nucleus and the endoplasmic reticulum, and NROB1 was expressed in the nucleus (b). (<b>C</b>) The presence of the exogenous proteins was confirmed by their expected molecular weights by Western blotting.</p>
Full article ">Figure 2
<p>Cell-cycle analysis of cells by gene transfection or siRNA transduction. (<b>A</b>) Growth curves of neuroblastoma cells transfected with one of these genes selected by the favorable tumors. Overexpression of these factors reduced the growth rates. (<b>B</b>) Growth rates of siRNA transduction for each gene in these cell lines. The growth rates were not affected by siRNA transduction, but some siRNA transduction reduced the growth rates. Data points represent means ± SDs of triplicate wells. The growth rates of transfected cells were reduced, except for DHRS-transfected TGW cells. Native cell lines were confluent at 4–5 days in this culture condition. **: <span class="html-italic">p</span> &lt; 0.01, *: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Soft-agar colony-formation assay. (<b>A</b>) The colony sizes in DHRS3-, CYP26A1-, and NROB1-overexpressing clones were significantly reduced (<span class="html-italic">p</span> &lt; 0.05). The CYP26A1-overexpressing TGW cells were hard to obtain, so statistical evaluation was not performed. *: <span class="html-italic">p</span> &lt; 0.05 (<b>B</b>) Cell morphology. The sizes of colonies were obviously large in TGW cells.</p>
Full article ">Figure 4
<p>Cell-growth curves of NB cells induced by the Retro-X Tet-On Advanced System. (<b>A</b>) Growth curves of NB cell lines. Gene expression was induced with or without doxycycline. (<b>B</b>) Time course of the <span class="html-italic">DHRS3</span>-gene-expression analysis in SK-N-SH and NH12 cells using qPCR. Gene expression of DHRS3 was up-regulated until 3–6 days after Dox induction. (<b>C</b>) Immunofluorescent imaging of gene expression using the mouse-specific FLAG-M2 monoclonal antibody. Expression of each gene was obvious in all cell lines 5 days after Dox induction, but expression of DHRS3 decreased at 7 and 9 days after Dox induction. Scale bar is 5 µm. (<b>D</b>) Morphological alterations of SK-N-SH and NH-12 cells at 5 days after Dox induction. SK-N-SH and NH12 cells showed cellular enlargement. Expression of DsRed corresponded to inserted gene expression. (<b>E</b>) Cell-cycle analysis using flow cytometry in SK-N-SH cells; G0/G1 arrest was detected in Dox-induced DHRS3-expressing (DHRS3+) and NROB1-expressing (NROB1+) cells, but not in CYP26A1-expressing (CYP26A1+) cells. (<b>F</b>) Percentages of cells in G0/G1, S, and G2/M for the cells with or without Dox induction of each gene. Reductions in the percentages of S and G2/M were detected in the SK-N-SH DHRS3+ and NROB1+ cells, but not in CYP26A1+ cells.</p>
Full article ">Figure 5
<p>ATRA-inducing differentiation experiments in Dox-treated NB cells. (<b>A</b>) Expression of senescence markers in Dox-treated SK-N-SH NB cell lines. SA-β-Gal-positive cells (green). SK-N-SH cells were untreated (−Dox) or Dox-treated until day 9 (+Dox). Ectopic Dox-induced SK-N-SH cells were observed to express high levels of a senescence marker. (<b>B</b>) Expression of Dox-treated DHRS3-expressing SK-N-SH NB cells with or without ATRA treatment. After ATRA treatment, SK-N-SH cells were assessed by immunofluorescence. High-dose ATRA induced cellular enlargement, cell death, and strong expression of DHRS3. (<b>C</b>) Expression of a senescence marker in SK-N-SH cell lines after 10 μM of ATRA treatment. SA-β-Gal-positive cells (green). The senescence marker was higher in the Dox-treated SK-N-SH cells.</p>
Full article ">
17 pages, 809 KiB  
Review
Modeling Obesity-Driven Pancreatic Carcinogenesis—A Review of Current In Vivo and In Vitro Models of Obesity and Pancreatic Carcinogenesis
by Sally Kfoury, Patrick Michl and Laura Roth
Cells 2022, 11(19), 3170; https://doi.org/10.3390/cells11193170 - 10 Oct 2022
Cited by 1 | Viewed by 2945
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy with a 5-year survival rate below 10%, thereby exhibiting the worst prognosis of all solid tumors. Increasing incidence together with a continued lack of targeted treatment options will cause PDAC to be the [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy with a 5-year survival rate below 10%, thereby exhibiting the worst prognosis of all solid tumors. Increasing incidence together with a continued lack of targeted treatment options will cause PDAC to be the second leading cause of cancer-related deaths in the western world by 2030. Obesity belongs to the predominant risk factors for pancreatic cancer. To improve our understanding of the impact of obesity on pancreatic cancer development and progression, novel laboratory techniques have been developed. In this review, we summarize current in vitro and in vivo models of PDAC and obesity as well as an overview of a variety of models to investigate obesity-driven pancreatic carcinogenesis. We start by giving an overview on different methods to cultivate adipocytes in vitro as well as various in vivo mouse models of obesity. Moreover, established murine and human PDAC cell lines as well as organoids are summarized and the genetically engineered models of PCAC compared to xenograft models are introduced. Finally, we review published in vitro and in vivo models studying the impact of obesity on PDAC, enabling us to decipher the molecular basis of obesity-driven pancreatic carcinogenesis. Full article
(This article belongs to the Special Issue Metabolic Regulation: Cell Growth and Proliferation)
Show Figures

Figure 1

Figure 1
<p>Overview of in vitro and in vivo models of obesity and PDAC.</p>
Full article ">
15 pages, 2948 KiB  
Article
The Flavone Cirsiliol from Salvia x jamensis Binds the F1 Moiety of ATP Synthase, Modulating Free Radical Production
by Lavinia Carlini, Gabriele Tancreda, Valeria Iobbi, Federico Caicci, Silvia Bruno, Alfonso Esposito, Daniela Calzia, Stefano Benini, Angela Bisio, Lucia Manni, Anna Schito, Carlo Enrico Traverso, Silvia Ravera and Isabella Panfoli
Cells 2022, 11(19), 3169; https://doi.org/10.3390/cells11193169 - 9 Oct 2022
Cited by 11 | Viewed by 2161
Abstract
Several studies have shown that mammalian retinal rod outer segments (OS) are peculiar structures devoid of mitochondria, characterized by ectopic expression of the molecular machinery for oxidative phosphorylation. Such ectopic aerobic metabolism would provide the chemical energy for the phototransduction taking place in [...] Read more.
Several studies have shown that mammalian retinal rod outer segments (OS) are peculiar structures devoid of mitochondria, characterized by ectopic expression of the molecular machinery for oxidative phosphorylation. Such ectopic aerobic metabolism would provide the chemical energy for the phototransduction taking place in the OS. Natural polyphenols include a large variety of molecules having pleiotropic effects, ranging from anti-inflammatory to antioxidant and others. Our goal in the present study was to investigate the potential of the flavonoid cirsiliol, a trihydroxy-6,7-dimethoxyflavone extracted from Salvia x jamensis, in modulating reactive oxygen species production by the ectopic oxidative phosphorylation taking place in the OS. Our molecular docking analysis identified cirsiliol binding sites inside the F1 moiety of the nanomotor F1Fo-ATP synthase. The experimental approach was based on luminometry, spectrophotometry and cytofluorimetry to evaluate ATP synthesis, respiratory chain complex activity and H2O2 production, respectively. The results showed significant dose-dependent inhibition of ATP production by cirsiliol. Moreover, cirsiliol was effective in reducing the free radical production by the OS exposed to ambient light. We report a considerable protective effect of cirsiliol on the structural stability of rod OS, suggesting it may be considered a promising compound against oxidative stress. Full article
(This article belongs to the Special Issue Recent Advances in Metabolism and Oxidative Stress in Human Diseases)
Show Figures

Figure 1

Figure 1
<p>Molecular docking analysis of cirsiliol on F<sub>1</sub> moiety of the F<sub>1</sub>F<sub>o</sub>-ATP synthase (ATP synthase). (<b>A</b>) Molecular docking (left: conformation 1 using 2JJ2 (see also <a href="#app1-cells-11-03169" class="html-app">Supplementary Video S1</a>); right: conformation 7 using 2JIZ (see also <a href="#app1-cells-11-03169" class="html-app">Supplementary Video S2</a>)). Both conformations displayed hydrogen bonds (highlighted as green dots) with Ser-267.G. (<b>B</b>) Energy parameters assessed for conformations bearing the highest binding energy. The data show the elevated inhibition constants.</p>
Full article ">Figure 2
<p>TEM of bovine retina. (<b>A</b>) The retina exhibits photoreceptorinner (IS) and outer (OS) segments. Squared areas B and C are enlarged in panels B and C, respectively. (<b>B</b>) Part of three OS belonging to close photoreceptors. Largest gold particles (40 nm width, arrows) reveal the Ab against anti-rhodopsin; smaller gold particles (10 nm width, arrowheads) evidence the Ab against anti-ATP synthase β-subunit. A gold particle is enlarged in inset. (<b>C</b>) Details of an IS close to a OS. Note the numerous small gold particles in the mitochondrion (mt) of the IS. In the OS, two small gold particles are recognizable.</p>
Full article ">Figure 3
<p>Respiratory complexes activity, ATP synthesis and lipid peroxidation in the rod OS. All experiments were carried out on rod OS maintained either in the dark or in ambient light in the presence of 0.6 mM NADH, 10 mM succinate and 0.1 mM ADP. (<b>A</b>–<b>D</b>) Respiratory complexes activity. (<b>E</b>) ATP synthesis. (<b>F</b>) MDA levels as a lipid peroxidation marker. Data are representative of at least three independent experiments. *, *** and **** indicate a significant difference for <span class="html-italic">p</span> &lt; 0.05, 0.001 and 0.0001, respectively, between the rod OS kept in the dark or in ambient light.</p>
Full article ">Figure 4
<p>Effect of cirsiliol on F<sub>1</sub>F<sub>o</sub> -ATP synthase activity in rod OS. ATP synthesis in the rod OS after preincubation of the sample with the indicated cirsiliol concentrations (see Materials and Methods section). Data are representative of at least three independent experiments and are indicated as mean ± SD. * and **** indicate a significant difference for <span class="html-italic">p</span> &lt; 0.005 or <span class="html-italic">p</span> &lt; 0.0001, respectively, between the subsequent samples. ns indicates a non-significant statistical difference.</p>
Full article ">Figure 5
<p>Effect of cirsiliol on the respiratory complex activity in the rod OS in coupled conditions. Figure shows the activity of the four respiratory complexes. Panels (<b>A</b>–<b>D</b>) report the activity of Complexes I to IV in coupled conditions in the absence or in the presence of cirsiliol. Data are from <span class="html-italic">n</span> = 5 independent experiments and are expressed as mean ± SD. Results show a non-statistically significant difference in activity. **** indicates a significant difference for <span class="html-italic">p</span> &lt; 0.0001 between treated and untreated samples in coupled conditions.</p>
Full article ">Figure 6
<p>Effect of cirsiliol on the respiratory complex activity in the rod OS in uncoupled conditions. Figure shows the activity of the four respiratory complexes in the absence or in the presence of cirsiliol. Panels (<b>A</b>–<b>D</b>) report the activity of Complexes I to IV in uncoupled conditions (i.e., after addition of 0.01 mM nigericin plus 0.01 mM valinomycin) in the absence or in the presence of cirsiliol. Data are expressed as mean ± SD and are from <span class="html-italic">n</span> = 5 independent experiments. A non-statistically significant difference is observed.</p>
Full article ">Figure 7
<p>Effect of cirsiliol on H<sub>2</sub>O<sub>2</sub> production and rod OS integrity. (<b>A</b>) H<sub>2</sub>O<sub>2</sub> production in rod OS treated (black squares) or untreated (black circles) with 50 μM cirsiliol during 40 min of ambient light exposure. (<b>B</b>) Percentage of structure integrity of rod OS in the same condition of Panel A. *, **, **** indicate a significant difference for <span class="html-italic">p</span> &lt; 0.05, 0.01, 0.0001, respectively, between treated and untreated samples in coupled conditions. Data are expressed as mean ± SD.</p>
Full article ">
10 pages, 935 KiB  
Hypothesis
AIRE in Male Fertility: A New Hypothesis
by Jana Petrusová, Jasper Manning and Dominik Filipp
Cells 2022, 11(19), 3168; https://doi.org/10.3390/cells11193168 - 9 Oct 2022
Cited by 1 | Viewed by 1915
Abstract
Male infertility affects approximately 14% of all European men, of which ~44% are characterized as idiopathic. There is an urgency to identify the factors that affect male fertility. One such factor, Autoimmune Regulator (AIRE), a protein found in the thymus, has been studied [...] Read more.
Male infertility affects approximately 14% of all European men, of which ~44% are characterized as idiopathic. There is an urgency to identify the factors that affect male fertility. One such factor, Autoimmune Regulator (AIRE), a protein found in the thymus, has been studied in the context of central tolerance functioning as a nuclear transcription modulator, responsible for the expression of tissue-restricted antigens in specialized thymic cells that prevent autoimmunity. While its expression in the testes remains enigmatic, we recently observed that sterility in mice correlates with the absence of Aire in the testes, regardless of the deficient expression in medullary thymic epithelial cells or cells of the hematopoietic system. By assessing the Aire transcript levels, we discovered that Sertoli cells are the exclusive source of Aire in the testes, where it most likely plays a non-immune role, suggesting an unknown mechanism by which testicular Aire regulates fertility. Here, we discuss these results in the context of previous reports which have suggested that infertility observed in Aire deficient mice is of an autoimmune aetiology. We present an alternative point of view for the role of Aire in testes in respect to fertility altering the perspective of how Aire’s function in the testes is currently perceived. Full article
(This article belongs to the Section Reproductive Cells and Development)
Show Figures

Figure 1

Figure 1
<p>Depletion of Aire in spermatogonia stem cells and spermatocytes does not affect male fertility. (<b>A</b>) Male mice with conditionally depleted Aire in spermatogonia stem cells (<span class="html-italic">Vasa<sup>Cre</sup>Aire<sup>fl/fl</sup></span>) and primary spermatocytes (<span class="html-italic">Smc1β<sup>iCre</sup>Aire<sup>fl/fl</sup></span>) exhibit fertility that is comparable to wild-type males in terms of the number of pups per litter. In contrast, whole-body Aire knock-out (<span class="html-italic">Aire<sup>−/−</sup></span>) mice produced only a few pups at the beginning of their reproduction period. We established five independent breeding pairs (A–E) and monitored them for six-consecutive months (1–6, gray continuous arrows). (<b>B</b>) The size of the testes in <span class="html-italic">Vasa<sup>Cre</sup>Aire<sup>fl/fl</sup></span> as well as <span class="html-italic">Smc1β<sup>iCre</sup>Aire<sup>fl/fl</sup></span> males was fully comparable to WT controls. In contrast, six–week–old <span class="html-italic">Aire<sup>−/−</sup></span> males had significantly smaller testes. (<b>C</b>) The decrease in testes size was confirmed by assessing their weight. <span class="html-italic">p</span> &lt; 0.0001 = **** and ns = not significant.</p>
Full article ">Figure 2
<p>Sertoli cells express Aire-coding mRNA. (<b>A</b>) The cell population from within the seminiferous tubule was stained with Hoechst 33342 and visualized in Hoechst red and blue channels. A color code was applied to the following cell types: turquoise spermatogonia stem cell (SSC), green primary spermatocytes (SCI), orange secondary spermatocytes (SCII), red round spermatids and sperm (RS+S), magenta Sertoli cells (Sertoli). (<b>B</b>) A single cell suspension from (<b>A</b>) was FACS-sorted and the indicated cell-types were prepared for mRNA isolation and qRT–PCR. Only Sertoli cells expressed physiologically relevant levels of Aire mRNA. The <span class="html-italic">y</span> axis represents the relative transcript levels of <span class="html-italic">Aire</span> and marker genes normalized to the <span class="html-italic">Casc3</span> gene. (<b>C</b>) Protein immunodetection on the testicular tissue-sections showed the localization of AIRE in the nuclei of Sertoli cells (upper right panel) identified by their positivity to SOX9 (white color, upper left panel). SCP3 is a marker of primary spermatocytes (SCI). Spermatogonia stem cells (SSC), typically positioned between two Sertoli cells at the base of seminiferous tubule and round spermatid (RS), are identified as DAPI<sup>+</sup>SOX9<sup>−</sup>SCP3<sup>−</sup>, exhibiting a typical nuclear morphology [<a href="#B49-cells-11-03168" class="html-bibr">49</a>]. The dashed line demarcates the edge of the seminiferous tubule. Cell types within the diagram (lower right panel) correspond to the color scheme used in qPCR.</p>
Full article ">
17 pages, 8269 KiB  
Article
Cellular Sources and Neuroprotective Roles of Interleukin-10 in the Facial Motor Nucleus after Axotomy
by Elizabeth M. Runge, Deborah O. Setter, Abhirami K. Iyer, Eric J. Regele, Felicia M. Kennedy, Virginia M. Sanders and Kathryn J. Jones
Cells 2022, 11(19), 3167; https://doi.org/10.3390/cells11193167 - 9 Oct 2022
Cited by 3 | Viewed by 2664
Abstract
Facial motoneuron (FMN) survival is mediated by CD4+ T cells in an interleukin-10 (IL-10)-dependent manner after facial nerve axotomy (FNA), but CD4+ T cells themselves are not the source of this neuroprotective IL-10. The aims of this study were to (1) identify the [...] Read more.
Facial motoneuron (FMN) survival is mediated by CD4+ T cells in an interleukin-10 (IL-10)-dependent manner after facial nerve axotomy (FNA), but CD4+ T cells themselves are not the source of this neuroprotective IL-10. The aims of this study were to (1) identify the temporal and cell-specific induction of IL-10 expression in the facial motor nucleus and (2) elucidate the neuroprotective capacity of this expression after axotomy. Immunohistochemistry revealed that FMN constitutively produced IL-10, whereas astrocytes were induced to make IL-10 after FNA. Il10 mRNA co-localized with microglia before and after axotomy, but microglial production of IL-10 protein was not detected. To determine whether any single source of IL-10 was critical for FMN survival, Cre/Lox mouse strains were utilized to selectively knock out IL-10 in neurons, astrocytes, and microglia. In agreement with the localization data reflecting concerted IL-10 production by multiple cell types, no single cellular source of IL-10 alone could provide neuroprotection after FNA. These findings suggest that coordinated neuronal and astrocytic IL-10 production is necessary for FMN survival and has roles in neuronal homeostasis, as well as neuroprotective trophism after axotomy. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) PCR amplicon gel showing a 133 bp fragment (arrow) of exon 1 of the IL10 coding sequence, which is present in WT, absent in IL-10<sup>−/−</sup>, and restored in IL-10<sup>−/−</sup> reconstituted with WT whole splenocytes. (<b>b</b>) Average percent survival of axotomized FMN relative to the control ± SEM at 28 dpo. No significant difference was detected between IL-10<sup>−/−</sup> and IL-10<sup>−/−</sup> receiving splenocytes; survival in both groups was significantly decreased relative to WT (* <span class="html-italic">p</span> &lt; 0.05). WT <span class="html-italic">n</span> = 5, IL-10<sup>−/−</sup> <span class="html-italic">n</span> = 5, IL-10<sup>−/−</sup> + WT splenocytes <span class="html-italic">n</span> = 6.</p>
Full article ">Figure 2
<p>FMN survival in WT and IL-10/GFP reporter mice. Average percent survival of axotomized FMN relative to the control ± SEM at 28 dpo. No significant difference was detected between WT and reporter (<span class="html-italic">n</span> = 6 for both groups).</p>
Full article ">Figure 3
<p>IHC co-localization of neurons (NeuN) with GFP in IL-10/GFP reporter mouse. Dpo = days post operation, C = control (<b>left</b>) FMNuc, Ax = axotomized (<b>right</b>) FMNuc.</p>
Full article ">Figure 4
<p>Direct IHC localization of microglia (IBA1) and IL-10 in WT mice. Arrowheads at 7 and 10 dpo demonstrate microglia lying in close proximity to IL-10-positive neuronal processes. Arrows at 14 and 28 dpo indicate microglial nodules. Dpo = days post operation, C = control (<b>left</b>) FMNuc, Ax = axotomized (<b>right</b>) FMNuc.</p>
Full article ">Figure 5
<p>Fluorescent in situ hybridization for <span class="html-italic">Il10</span> and <span class="html-italic">Cx3cr1</span> mRNA transcripts.</p>
Full article ">Figure 6
<p>IHC co-localization of astrocytes (GFAP) with IL-10 using both direct IL-10 antibody and IL-10/GFP reporter. Left panels depict direct IL-10 antibody, right panels depict reporter. Arrowheads indicate areas of co-localization. Dpo = days post operation, C = control (<b>left</b>) FMNuc, Ax = axotomized (<b>right</b>) FMNuc.</p>
Full article ">Figure 7
<p>FMN survival in IL-10 cKO mice compared to littermate controls (Cre<sup>−/−</sup> or IL-10<sup>wt/wt</sup>). Average percent survival of axotomized FMN relative to control ± SEM at 28 dpo in mice lacking IL-10 in (<b>A</b>) neurons (control <span class="html-italic">n</span> = 7, cKO <span class="html-italic">n =</span> 5), (<b>B</b>) microglia (control <span class="html-italic">n</span> = 8, cKO <span class="html-italic">n =</span> 7), and (<b>C</b>) astrocytes (<span class="html-italic">n</span> = 3 for both groups). No significant differences were observed between littermates and cKO.</p>
Full article ">Figure 8
<p>(<b>A</b>) Prior to FNA, FMN constitutively produces IL-10. Microglia constitutively transcribe <span class="html-italic">Il10</span> mRNA, but protein translation is unknown. (<b>B</b>) Astrocytic IL-10 production is axotomy-induced. (<b>C</b>) The appearance of CD4+ T cells in the FMNuc promotes upregulation of IL-10R expression on resident cells [<a href="#B1-cells-11-03167" class="html-bibr">1</a>,<a href="#B9-cells-11-03167" class="html-bibr">9</a>], which may provide trophic support to injured FMN. (<b>D</b>) CD4+ T cells must express the IL-10R to modulate microglial activation and confer neuroprotection [<a href="#B9-cells-11-03167" class="html-bibr">9</a>].</p>
Full article ">
21 pages, 3788 KiB  
Article
Dynamics of Actin Cytoskeleton and Their Signaling Pathways during Cellular Wound Repair
by Shigehiko Yumura, Md. Shahabe Uddin Talukder, Mst. Shaela Pervin, Md. Istiaq Obaidi Tanvir, Takashi Matsumura, Koushiro Fujimoto, Masahito Tanaka and Go Itoh
Cells 2022, 11(19), 3166; https://doi.org/10.3390/cells11193166 - 9 Oct 2022
Cited by 4 | Viewed by 2730
Abstract
The repair of wounded cell membranes is essential for cell survival. Upon wounding, actin transiently accumulates at the wound site. The loss of actin accumulation leads to cell death. The mechanism by which actin accumulates at the wound site, the types of actin-related [...] Read more.
The repair of wounded cell membranes is essential for cell survival. Upon wounding, actin transiently accumulates at the wound site. The loss of actin accumulation leads to cell death. The mechanism by which actin accumulates at the wound site, the types of actin-related proteins participating in the actin remodeling, and their signaling pathways are unclear. We firstly examined how actin accumulates at a wound site in Dictyostelium cells. Actin assembled de novo at the wound site, independent of cortical flow. Next, we searched for actin- and signal-related proteins targeting the wound site. Fourteen of the examined proteins transiently accumulated at different times. Thirdly, we performed functional analyses using gene knockout mutants or specific inhibitors. Rac, WASP, formin, the Arp2/3 complex, profilin, and coronin contribute to the actin dynamics. Finally, we found that multiple signaling pathways related to TORC2, the Elmo/Doc complex, PIP2-derived products, PLA2, and calmodulin are involved in the actin dynamics for wound repair. Full article
(This article belongs to the Special Issue Toward Understanding Wound Repair Mechanism)
Show Figures

Figure 1

Figure 1
<p>Actin accumulation at wound sites is essential for wound repair. (<b>A</b>) Schema for the improved laserporation using gold coating. The wound diameter was set as 0.5 µm. (<b>B</b>) When the laser beam was focused on the surface of the coated coverslip, the gold coating peeled off, leaving behind a small white spot (arrow). (<b>C</b>) A typical sequence of fluorescence images of PI influx after laserporation. The wound laser beam was applied at 0 s and the duration was set at 8 ms. (<b>D</b>) Time courses of PI influx in the presence (LatA) and absence (BSS) of latrunculin A (<span class="html-italic">n</span> = 25 each). (<b>E</b>) A typical sequence of fluorescence images of a cell expressing GFP-lifeact after laserporation. The arrow shows the wound site. (<b>F</b>) Enlarged images of the rectangle in panel E 0.5 s before wounding (−0.5 s, red) and 0.5 or 3.0 s after wounding (0.5 s in upper panel and 3.0 s in lower panel, green), and the merged images. Arrows show the wound sites. (<b>G</b>) Time courses of fluorescence intensities at the wound site in the presence (LatA) and absence (BSS) of latrunculin A. The fluorescence intensities were normalized by setting the value before wounding to 1 after subtracting the background (<span class="html-italic">n</span> = 25 each). Scale bars: 10 µm for panels B, C, and D, and 1 µm for panel F.</p>
Full article ">Figure 2
<p>Actin polymerizes de novo at wound sites. (<b>A</b>) Typical sequences of fluorescence images of cells expressing GFP-lifeact in the presence of latrunculin A and/or jasplakinolide. Arrows and arrowheads show the wound sites and large actin aggregates, respectively. (<b>B</b>) Time courses of fluorescence intensities of GFP-lifeact at the wound site in the presence (Jasp) and absence (BSS) of jasplakinolide, respectively. Note that the time courses in the presence of latrunculin A are shown in <a href="#cells-11-03166-f001" class="html-fig">Figure 1</a>G. (<b>C</b>) The initiation, peak, and termination times of wound-induced actin accumulation are compared in the presence and absence of jasplakinolide (<span class="html-italic">n</span> = 25 each). Data are presented as means ± SD. ** <span class="html-italic">p ≤</span> 0.0001; <span class="html-italic">ns</span>: not significant, <span class="html-italic">p</span> &gt; 0.05. (<b>D</b>) Time courses of PI influx in the presence and absence of jasplakinolide (<span class="html-italic">n</span> = 25 each). (<b>E</b>) Time courses of fluorescence intensities of GFP-lifeact at the wound site in the presence and absence of both inhibitors (Jasp + LatA) (<span class="html-italic">n</span> = 25 each). (<b>F</b>) Time courses of fluorescence intensities of GFP-lifeact at the wound site in annexin C1-null cells. The graphs with different colors show time courses of six different cells. (<b>G</b>) Time courses of fluorescence intensities of GFP-lifeact at the wound site in wild-type cells in the presence and absence of W7 (<span class="html-italic">n</span> = 25 each).</p>
Full article ">Figure 3
<p>Dynamics of ARPs during wound repair. (<b>A</b>) Typical sequences of fluorescence images of cells expressing various (13 kinds) GFP-ARPs at wound sites. (<b>B</b>) Time courses of fluorescence intensities of GFP-ARPs and GFP-actin at wound sites (<span class="html-italic">n</span> = 25 each). (<b>C</b>) Normalized curves of panel B when each peak value is set as 100%. (<b>D</b>) Duration of appearance of individual ARPs at wound sites (red). Duration of actin dynamics (green), Ca<sup>2+</sup> influx (blue), calmodulin dynamics (blue), ESCRT component vps4 dynamics (blue), and annexin C1 dynamics (blue) are also plotted. In addition, the duration of Efa1A disappearance (orange) is plotted. Asterisks in the duration bars indicate the peak times.</p>
Full article ">Figure 4
<p>Durations of wound-induced actin dynamics in null cells or wild-type cells in the presence of inhibitors. Durations of wound-induced actin dynamics in null cells or wild-type cells in the presence of inhibitors: actin-related (red), signal-related (purple), membrane dynamics-related (blue), and microtubule-related (gray) proteins. Actual time courses of the fluorescence intensities are shown in <a href="#app1-cells-11-03166" class="html-app">Supplementary Figures S3 and S4</a>. Red asterisks indicate the cases showing significantly retarded initiation time compared with the control, and black asterisks indicate the cases showing significantly retarded termination time. * <span class="html-italic">p</span> ≤ 0.001, ** <span class="html-italic">p</span> ≤ 0.0001 (<span class="html-italic">n</span> ≥ 25 each).</p>
Full article ">Figure 5
<p>Peak amplitudes of wound-induced actin dynamics in null cells or wild-type cells in the presence of inhibitors. The amplitudes at the peaks of the actin dynamics under the same conditions as shown in <a href="#cells-11-03166-f004" class="html-fig">Figure 4</a>. Actual time courses of the fluorescence intensities are shown in <a href="#app1-cells-11-03166" class="html-app">Supplementary Figures S3 and S4</a>. Asterisks show cases with significant differences from wild-type cells in the absence of inhibitors. Data are presented as means ± SD. * <span class="html-italic">p</span> ≤ 0.001, ** <span class="html-italic">p</span> ≤ 0.0001 (<span class="html-italic">n</span> ≥ 25 each).</p>
Full article ">Figure 6
<p>Signal pathways for actin dynamics in wound repair. With reference to the signal cascades for chemotaxis, potent candidates for wound-related signals, ARPs, or other proteins we found in the present and previous studies are marked (red). Some proteins (yellow) accumulate or disappear at the wound site, but their contribution remains to be clarified using knockout or inhibitor experiments. Most of the examined proteins (green) do not contribute to wound repair.</p>
Full article ">
19 pages, 4761 KiB  
Article
Investigating Two Modes of Cancer-Associated Antigen Heterogeneity in an Agent-Based Model of Chimeric Antigen Receptor T-Cell Therapy
by Tina Giorgadze, Henning Fischel, Ansel Tessier and Kerri-Ann Norton
Cells 2022, 11(19), 3165; https://doi.org/10.3390/cells11193165 - 9 Oct 2022
Cited by 4 | Viewed by 2329
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been successful in treating liquid tumors but has had limited success in solid tumors. This work examines unanswered questions regarding CAR T-cell therapy using computational modeling, such as, what percentage of the tumor must express cancer-associated [...] Read more.
Chimeric antigen receptor (CAR) T-cell therapy has been successful in treating liquid tumors but has had limited success in solid tumors. This work examines unanswered questions regarding CAR T-cell therapy using computational modeling, such as, what percentage of the tumor must express cancer-associated antigens for treatment to be successful? The model includes cancer cell and vascular and CAR T-cell modules that interact with each other. We compare two different models of antigen expression on tumor cells, binary (in which cancer cells are either susceptible or are immune to CAR T-cell therapy) and gradated (where each cancer cell has a probability of being killed by a CAR T-cell). We vary the antigen expression levels within the tumor and determine how effective each treatment is for the two models. The simulations show that the gradated antigen model eliminates the tumor under more parameter values than the binary model. Under both models, shielding, in which the low/non-antigen-expressing cells protect high antigen-expressing cells, reduced the efficacy of CAR T-cell therapy. One prediction is that a combination of CAR T-cell therapies that targets the general population of cells as well as one that specifically targets cancer stem cells should increase its efficacy. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart of the CAR T-cell TNBC model.</p>
Full article ">Figure 2
<p>3D plots of tumors with gradated (<b>a</b>) and binary (<b>b</b>) heterogeneity plotted over time with 12.5-day intervals starting from day 12.5 and ending on day 75, the last day of our simulations. Tumor cells are plotted in blue, CAR T-cells are plotted in green, and the vasculature is plotted as red capillaries growing across the grid. The data in the gradated heterogeneity tumor progression (<b>a</b>) is from a run with 12.5% mean antigen expression tumor cells. The data in the binary heterogeneity tumor progression (<b>b</b>) is from a run with 75% antigen presenting tumor cells. CAR T-cells are introduced halfway through the simulation, on day 37.5.</p>
Full article ">Figure 3
<p>Tumor metrics over time for gradated heterogeneity (<b>A</b>) and binary heterogeneity (<b>B</b>). In both cases, the metrics include total number of tumor cells over time (<b>a</b>), total number of CAR T-cells (<b>b</b>), total number of cancer cell deaths (<b>c</b>), and total number of stem cells over time (<b>d</b>). Each iteration represents 6 h of real time. For the gradated heterogeneity metrics, 0% mean antigen expression is plotted in yellow, 12.5% mean antigen expression is plotted in light blue, 25% mean antigen expression is plotted in red, and 50% mean antigen expression is plotted in dark blue. For the binary heterogeneity metrics, 25% antigen presentation is plotted in yellow, 50% antigen presentation is plotted in light blue, 75% antigen expression is plotted in red, and 98% antigen expression is plotted in dark blue. Note: the parameters in which the line does not reach day 75 indicate that one of the tumors died out at this point and no further data was collected.</p>
Full article ">Figure 4
<p>3D plots of tumors on the last day of our simulation, day 75, with different antigen presentations. For tumor with gradated heterogeneity (<b>a</b>), the data comes from 0%, 12.5%, 25%, and 50% mean antigen presentation (left to right). Vasculature is plotted as red capillaries growing across the plane. Tumor cells with 100% chance of presenting the antigen are plotted in yellow. Tumor cells with 0% chance of presenting the antigen are plotted in red. Every cell with a chance of antigen presentation between 0% and 100% is plotted on a gradient from red to yellow. CAR T-cells are plotted in green and are introduced on day 37.5 of the simulation. For the tumors with binary heterogeneity (<b>b</b>), the data comes from runs with 25%, 50%, 75%, and 98% antigen presentation. Tumor cells with 100% antigen presentation are plotted in yellow; cells with 0% antigen presentation are plotted in red.</p>
Full article ">Figure 5
<p>Examples of antigen shielding occurring in tumors with gradated heterogeneity (<b>a</b>) and binary heterogeneity (<b>b</b>). For gradated heterogeneity, the 3D plot shows tumor cells with 100% of antigen presentation in yellow, tumor cells with 0% antigen presentation in red, and tumor cells with intermediate probabilities of antigen presentation on a gradient. For binary heterogeneity, antigen-presenting tumor cells are plotted in yellow and antigen non-presenting tumor cells in red. In both cases, the figure shows a layer (shield) of antigen non/low-presenting cells forming a shield with high antigen-presenting cells. In both cases, such a shield makes it difficult for CAR T-cells to break through that layer, as the cells making up that layer are either fully or highly immune to CAR T-cell therapy.</p>
Full article ">
19 pages, 3650 KiB  
Article
Profound Effects of Dexamethasone on the Immunological State, Synthesis and Secretion Capacity of Human Testicular Peritubular Cells
by Youli Konstantinovitch Stepanov, Jan Dominik Speidel, Carola Herrmann, Nina Schmid, Rüdiger Behr, Frank-Michael Köhn, Jan Bernd Stöckl, Ulrich Pickl, Matthias Trottmann, Thomas Fröhlich, Artur Mayerhofer and Harald Welter
Cells 2022, 11(19), 3164; https://doi.org/10.3390/cells11193164 - 9 Oct 2022
Cited by 5 | Viewed by 2405
Abstract
The functions of human testicular peritubular cells (HTPCs), forming a small compartment located between the seminiferous epithelium and the interstitial areas of the testis, are not fully known but go beyond intratesticular sperm transport and include immunological roles. The expression of the glucocorticoid [...] Read more.
The functions of human testicular peritubular cells (HTPCs), forming a small compartment located between the seminiferous epithelium and the interstitial areas of the testis, are not fully known but go beyond intratesticular sperm transport and include immunological roles. The expression of the glucocorticoid receptor (GR) indicates that they may be regulated by glucocorticoids (GCs). Herein, we studied the consequences of the GC dexamethasone (Dex) in cultured HTPCs, which serves as a unique window into the human testis. We examined changes in cytokines, mainly by qPCR and ELISA. A holistic mass-spectrometry-based proteome analysis of cellular and secreted proteins was also performed. Dex, used in a therapeutic concentration, decreased the transcript level of proinflammatory cytokines, e.g., IL6, IL8 and MCP1. An siRNA-mediated knockdown of GR reduced the actions on IL6. Changes in IL6 were confirmed by ELISA measurements. Of note, Dex also lowered GR levels. The proteomic results revealed strong responses after 24 h (31 significantly altered cellular proteins) and more pronounced ones after 72 h of Dex exposure (30 less abundant and 42 more abundant cellular proteins). Dex also altered the composition of the secretome (33 proteins decreased, 13 increased) after 72 h. Among the regulated proteins were extracellular matrix (ECM) and basement membrane components (e.g., FBLN2, COL1A2 and COL3A1), as well as PTX3 and StAR. These results pinpoint novel, profound effects of Dex in HTPCs. If transferrable to the human testis, changes specifically in ECM and the immunological state of the testis may occur in men upon treatment with Dex for medical reasons. Full article
Show Figures

Figure 1

Figure 1
<p>Consequences of Dex on cytokine levels. (<b>A</b>) HTPCs treated with 1 µM Dex for 48 h responded with the decreased secretion of IL6, IL8, MCP1 and CXCL1 compared to untreated cells (<span class="html-italic">n</span> = 1), as demonstrated by a human cytokine profiler assay. The upper panel shows the corresponding membrane spots of untreated control, while the lower panel depicts Dex (1 µM)-treated HTPCs and reveals a reduction of the signal intensity of IL6, IL8, MCP1 and CXCL1. (<b>B</b>) Using an ELISA, highly significantly decreased IL6 levels could be detected in the culture media of HTPCs from four individual patients treated for 48 h with 1 µM Dex compared to the corresponding control condition. Results were normalized to the total protein amount and statistically analyzed using a paired <span class="html-italic">t</span>-test (two-tailed paired <span class="html-italic">t</span>-test: ** <span class="html-italic">p</span> &lt; 0.01; <span class="html-italic">n</span> = 4). (<b>C</b>) Quantitative PCR revealed significantly decreased mRNA expression levels of <span class="html-italic">IL6</span>, <span class="html-italic">IL8</span>, <span class="html-italic">MCP1</span>, <span class="html-italic">MCP3</span> and <span class="html-italic">IL1B</span> in HTPCs after 24 h stimulation with 1 µM Dex. <span class="html-italic">CXCL1</span> transcripts did not change significantly compared to control (ns: not significant, * <span class="html-italic">p</span> &lt; 0.05; <span class="html-italic">n</span> = 4).</p>
Full article ">Figure 2
<p>Regulation of GR by siRNA and Dex. (<b>A</b>) Silencing of GR by siRNA: Two representative GR Western blots from two patients (P1 and P2, <span class="html-italic">n</span> = 2) show the results of HTPCs transfected with non-targeting (NT) control siRNA and GR siRNA (siGR) at two different time points (48 h and 72 h) each and emphasize the efficiency of the transfection with siGR RNA. (<b>B</b>) Transfection of HTPCs with a GR siRNA abolished the observed effect on <span class="html-italic">IL6</span> and <span class="html-italic">FKBP5</span> mRNA. Quantitative PCR data of HTPCs from three different patients (<span class="html-italic">n</span> = 3) transfected with a non-targeting control siRNA (siNT) or GR siRNA (siGR) and treated with Dex (1 µM; siNT + Dex and siGR + Dex) or an equal volume of EtOH (Basal Co and Dex) for 24 h are depicted, ## <span class="html-italic">p</span> ≤ 0.01 vs. Co cells; * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01 vs. siNT + dex cells. (<b>C</b>) Stimulation of HTPCs with Dex led to the downregulation of GR expression. In the presence of 1 µM Dex for 24 h and 48 h, GR protein in HTPCs from two different donors (P1 and P2, <span class="html-italic">n</span> = 2) was downregulated compared to control (Co), as shown by Western blot. In addition, significantly diminished <span class="html-italic">GR</span> (<span class="html-italic">n</span> = 7; upper left picture) but not <span class="html-italic">AR</span> mRNA (<span class="html-italic">n</span> = 9; lower left picture) was detected after 24 h incubation with 1 µM Dex.</p>
Full article ">Figure 3
<p>Volcano plot analysis of cellular proteomes treated with 1 µM dexamethasone for 24 h (<b>A</b>) and 72 h (<b>B</b>). The data was analyzed with a paired Student’s <span class="html-italic">t</span>-test; false-discovery rate (FDR): 0.05. Each colored dot represents a protein fulfilling the significance criteria (q-value &lt; 0.05, log<sub>2</sub> fold change &gt;|0.6|): blue–proteins less abundant in the treated group; red–proteins more abundant in the treated group. For selected significant hits, gene names are displayed. (<b>C</b>) = log<sub>2</sub> fold changes of significantly differentially abundant proteins of the 24 h-treatment and 72 h-treatment datasets. For the overlap, the 72 proteins (fulfilling the above significance criteria) from 72 h dataset were used as “base”. This was overlayed by the proteins from the 24 h dataset fulfilling only the “overlap criterion” of q-value &lt; 0.05, which additionally includes potential trends, given the lower size of this dataset.</p>
Full article ">Figure 4
<p>Volcano plot of cellular secretomes treated with 1 µM dexamethasone for 24 h (<b>A</b>) and a 72 h (<b>B</b>). The data was analyzed with a paired Student’s <span class="html-italic">t</span>-test; false-discovery rate (FDR): 0.05. Each colored dot represents a protein fulfilling the significance criteria (q-value &lt; 0.05, log<sub>2</sub> fold change &gt; |0.6|): blue—proteins less abundant in the treated group; red—proteins more abundant in the treated group. For selected significant hits, gene names are displayed.</p>
Full article ">Figure 5
<p>Bioinformatic analysis based on the functional enrichment of all merged (proteome + secretome) significantly differentially abundant proteins (<b>A</b>,<b>B</b>). 24 h-treatment, PROTEOMAPS-based analysis. (<b>a</b>,<b>b</b>): 24 h-treatment, DAVID-based analysis. (<b>C</b>,<b>D</b>): 72 h-treatment, PROTEOMAPS-based analysis. (<b>c</b>,<b>d</b>): 72 h-treatment, DAVID-based analysis. For PROTEOMAPS, proteins were annotated using GO “biological process”. The terms are shown in the legend and comprise all tiles of similar color. Annotated individual proteins are displayed in the mosaic plots. For DAVID analysis, functional enrichment terms GO_BP_all, GO_CC_all, GO_MF_all, KEGG and Reactome were used.</p>
Full article ">Figure A1
<p>Regulation of PDGFRalpha (PDGFRa) by Dex. In the presence of 1 µM Dex for 72 h, PDGFRa protein in HTPCs from three donors (P1-P3) was increased, compared to controls (Co), as shown by Western blot.</p>
Full article ">
24 pages, 1216 KiB  
Review
Multi- and Transgenerational Effects of Environmental Toxicants on Mammalian Reproduction
by Paola Rebuzzini, Gemma Fabozzi, Danilo Cimadomo, Filippo Maria Ubaldi, Laura Rienzi, Maurizio Zuccotti and Silvia Garagna
Cells 2022, 11(19), 3163; https://doi.org/10.3390/cells11193163 - 9 Oct 2022
Cited by 14 | Viewed by 3224
Abstract
Environmental toxicants (ETs) are an exogenous chemical group diffused in the environment that contaminate food, water, air and soil, and through the food chain, they bioaccumulate into the organisms. In mammals, the exposure to ETs can affect both male and female fertility and [...] Read more.
Environmental toxicants (ETs) are an exogenous chemical group diffused in the environment that contaminate food, water, air and soil, and through the food chain, they bioaccumulate into the organisms. In mammals, the exposure to ETs can affect both male and female fertility and their reproductive health through complex alterations that impact both gametogeneses, among other processes. In humans, direct exposure to ETs concurs to the declining of fertility, and its transmission across generations has been recently proposed. However, multi- and transgenerational inheritances of ET reprotoxicity have only been demonstrated in animals. Here, we review recent studies performed on laboratory model animals investigating the effects of ETs, such as BPA, phthalates, pesticides and persistent contaminants, on the reproductive system transmitted through generations. This includes multigenerational effects, where exposure to the compounds cannot be excluded, and transgenerational effects in unexposed animals. Additionally, we report on epigenetic mechanisms, such as DNA methylation, histone tails and noncoding RNAs, which may play a mechanistic role in a nongenetic transmission of environmental information exposure through the germline across generations. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the (<b>A</b>) multigenerational and (<b>B</b>) transgenerational transmission of ETs effects. F0: parental generation; F1: first filial generation: F2: second filial generation; F3: third filial generation.</p>
Full article ">Figure 2
<p>EDC exposure may alter DNA methyltransferases, histone modifiers and noncoding RNAs with downstream effects on the gene expression.</p>
Full article ">
14 pages, 1131 KiB  
Article
Crosstalk between Extracellular Matrix Stiffness and ROS Drives Endometrial Repair via the HIF-1α/YAP Axis during Menstruation
by Tao Zhang, Yan Wang, Yingnan Wang, Cuiyan Liu and Chunyang Han
Cells 2022, 11(19), 3162; https://doi.org/10.3390/cells11193162 - 9 Oct 2022
Cited by 5 | Viewed by 2554
Abstract
Although the menstrual cycle driven by sex steroid hormones is an uncomplicated physiological process, it is important for female health, fertility and regenerative biology. However, our understanding of this unique type of tissue homeostasis remains unclear. Here, we examined the biological effects of [...] Read more.
Although the menstrual cycle driven by sex steroid hormones is an uncomplicated physiological process, it is important for female health, fertility and regenerative biology. However, our understanding of this unique type of tissue homeostasis remains unclear. Here, we examined the biological effects of mechanical force by evaluating the changing trend of extracellular matrix (ECM) stiffness, and the results suggested that ECM stiffness was reduced and that breaking of mechanotransduction delayed endometrium repair in a mouse model of simulated menses. We constructed an ECM stiffness interference model in vitro to explain the mechanical force conduction mechanism during endometrial regeneration. We discovered that ECM stiffness increased the expression and nuclear transfer of YAP, which improved the creation of a microenvironment, in a manner that induced proliferation and angiogenesis for endometrial repair by activating YAP. In addition, we observed that physiological endometrial hypoxia occurs during the menstrual cycle and that the expression of HIF-1α was increased. Mechanistically, in addition to the classical F-actin/YAP pathway, we also found that the ROS/HIF-1α/YAP axis was involved in the transmission of mechanical signals. This study provides novel insights into the essential menstrual cycle and presents an effective, nonhormonal treatment for menstrual disorders. Full article
(This article belongs to the Topic Cell Signaling Pathways)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p><b>ECM stiffening features of the endometrium in a mouse model of simulated menses</b>. (<b>a</b>) Diagram depicting the murine model of simulated menstruation. E2 17-β-estradiol, P4 progesterone. Timed to 0 h, 24 h, 48 h and 72 h based on P4 withdrawal time. (<b>b</b>) Representative whole uterus images (top) and histopathological images of paraffin-embedded mouse uterine sections (bottom, n = 2). Scale bar: 400 μm. (<b>c</b>,<b>d</b>) Representative images of uterine sections stained with TUNEL (top, n = 2) and BrdU (bottom, n = 2) and quantification of TUNEL+ and BrdU+ positive cells ((<b>d</b>), n = 5). Scale bars, 200 μm. The data are presented as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. (<b>e</b>) Western blot analysis of pFAK, FAK, pPMLC2 and pMLC2 expression. GAPDH was used as a loading control. (<b>f</b>) Representative images of vinculin and F-actin in mouse uterine sections. Scale bar, 20 μm. n = 4. (<b>g</b>) Endometrial histological breakdown/repair score at 24 h and 72 h in mice treated with DMSO and Cyto. B. Graphs represent the percentage of mice at each histological grade per experimental group. n = 9. Experiments were repeated n times with duplicate biological replicates.</p>
Full article ">Figure 2
<p><b>Pharmacological inhibition of YAP at menses delays repair</b>. (<b>a</b>,<b>b</b>) Representative immunofluorescence images ((<b>a</b>), n = 2) and quantification of nuclear and cytoplasmic subcellular localization ((<b>b</b>), n = 9) of YAP in uterine sections. Scale bar: 100 μm. (<b>c</b>) Western blotting for YAP in nuclear and cytoplasmic protein fractions from mouse endometrial tissue. n = 3. Lamin B1 and GAPDH were used as loading controls for nuclear and cytoplasmic proteins, respectively. (<b>d</b>) RT-qPCR analysis of CYR61 and CTGF mRNA levels. n = 3. (<b>e</b>) Endometrial histological breakdown/repair score at 24 h and 72 h in mice treated with DMSO and VP. The graphs represent the percentage of mice at each histological grade per experimental group. (<b>f</b>) Representative immunofluorescence images (left, n = 2) and quantification of YAP- and BrdU-the positive cells ((<b>f</b>), n = 5) in the uterus of mice uterine treated with DMSO or VP at 24 h and 48 h. Scale bar: 100 μm. Enlarge: 10 μm. The data are presented as the mean ± SEM. Experiments were repeated n times with duplicate biological replicates. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p><b>Hypoxia sustains HIF-1α protein stability to activate YAP</b>. (<b>a</b>) Analysis of the antioxidant indices T-AOC activity and SOD activity in endometrial tissue. n =3. (<b>b</b>) The protein levels of VEGF, CXCR4, and HIF-1α were evaluated in the uterus by Western blotting. n = 3. (<b>c</b>) Comparison of the mRNA expression levels of VEGF and CXCR4 in isolated mouse endometrial tissue during menstruation. n = 3. (<b>d</b>) Lysates of ESCs transfected with siYAP and siNC after withdrawing P4 were analyzed for the presence of the indicated proteins. n = 3. (<b>e</b>) Confocal immunofluorescence images (left, n = 2) and quantification of nuclear and cytoplasmic subcellular localization (right, n = 15) of YAP in decidualized cells transfected with siYAP. Scale bar: 100 μm. The data are presented as the mean ± SEM. Experiments were repeated n times with duplicate biological replicates. * <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><b>ECM stiffness regulates the activity of YAP/HIF-1α and ROS production in ESCs</b>. (<b>a</b>) Western blotting for YAP in nuclear and cytoplasmic protein fractions from ESCs plated on 1 kPa or 40 kPa fibronectin-coated hydrogels for 24 h, n = 3. (<b>b</b>) Confocal immunofluorescence images (left, n = 2) and quantification of nuclear and cytoplasmic subcellular localization (right, n = 15) of YAP in ESCs plated on hydrogels with different rigidities. Scale bar, 10 μm. (<b>c</b>) Western blotting for HIF-1α from decidualized cells plated on 1 kPa or 40 kPa hydrogels for 0 h and 24 h, n = 3. (<b>d</b>,<b>e</b>) Western blot analysis of HIF-1α and YAP expression in decidualized cells treated with siYAP (<b>d</b>) or VP (<b>e</b>) plated on 1 kPa or 40 kPa hydrogels for 24 h, n =3. (<b>f</b>) The intracellular ROS levels were measured by staining with DCFH-DA and then determined by flow cytometry. n = 2. The data are presented as the mean ± SEM. Experiments were repeated n times with duplicate biological replicates. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p><b>YAP activation provides the physiological environment needed for endometrial repair</b>. (<b>a</b>) Confocal immunofluorescence images (<b>left</b> n = 2) and quantification of Ki67 positive cells (<b>right</b>, n = 9) in ESCs transfected with siYAP and pcDNA 3.1 (+) YAP. Scale bars, 200 μm. (<b>b</b>,<b>c</b>) Schematic diagram of cell cycle detection results. n = 3. (<b>d</b>,<b>e</b>) Protein and mRNA expression levels of p21, cyclin D1 and CDK4 were detected in ESCs transfected with si YAP or pcDNA 3.1 (+) YAP, n = 3. (<b>f</b>) RT-qPCR analysis of Ang-2 expression levels in ESCs transfected with siYAP or pcDNA3.1(+) YAP, n = 3. The data are presented as the mean ± SEM. Experiments were repeated n times with duplicate biological replicates. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>g</b>) Schematic images depicting upregulation of ECM stiffness and ROS upon nuclear translocation and activation of YAP/HIF-1α and subsequent binding to TEAD, Ang2, and CTGFdriving endometrium repair after in the menstrual cycle.</p>
Full article ">
22 pages, 3065 KiB  
Review
Pathological Role of HDAC8: Cancer and Beyond
by Ji Yoon Kim, Hayoung Cho, Jung Yoo, Go Woon Kim, Yu Hyun Jeon, Sang Wu Lee and So Hee Kwon
Cells 2022, 11(19), 3161; https://doi.org/10.3390/cells11193161 - 9 Oct 2022
Cited by 38 | Viewed by 3920
Abstract
Histone deacetylase 8 (HDAC8) is a class I HDAC that catalyzes the deacetylation of histone and non-histone proteins. As one of the best-characterized isoforms, numerous studies have identified interacting partners of HDAC8 pertaining to diverse molecular mechanisms. Consequently, deregulation and overexpression of HDAC8 [...] Read more.
Histone deacetylase 8 (HDAC8) is a class I HDAC that catalyzes the deacetylation of histone and non-histone proteins. As one of the best-characterized isoforms, numerous studies have identified interacting partners of HDAC8 pertaining to diverse molecular mechanisms. Consequently, deregulation and overexpression of HDAC8 give rise to diseases. HDAC8 is especially involved in various aspects of cancer progression, such as cancer cell proliferation, metastasis, immune evasion, and drug resistance. HDAC8 is also associated with the development of non-cancer diseases such as Cornelia de Lange Syndrome (CdLS), infectious diseases, cardiovascular diseases, pulmonary diseases, and myopathy. Therefore, HDAC8 is an attractive therapeutic target and various HDAC8 selective inhibitors (HDAC8is) have been developed. Here, we address the pathological function of HDAC8 in cancer and other diseases, as well as illustrate several HDAC8is that have shown anti-cancer effects. Full article
(This article belongs to the Special Issue Transcription and Chromatin Dysregulation in Cancer)
Show Figures

Figure 1

Figure 1
<p>Crystal structure of HDAC8. (<b>A</b>) Head-to-head dimeric arrangement of HDAC8 (PDB ID: 2V5W [<a href="#B27-cells-11-03161" class="html-bibr">27</a>]). Two monomers of HDAC8 are shown in light and dark yellow. Zinc and potassium are represented as grey and purple spheres, respectively (<b>B</b>) Liganded form of HDAC8 (PDB ID: 1T67 [<a href="#B15-cells-11-03161" class="html-bibr">15</a>]). L2 loop and MS-344 are colored grey and green, respectively. (<b>C</b>) Catalytic machinery of HDAC8 (PDB ID: 1T64 [<a href="#B15-cells-11-03161" class="html-bibr">15</a>]). Zinc ion is coordinated to three HDAC8 residues and two oxygen atoms of TSA. The zinc coordination shell is indicated by dashed lines. (<b>D</b>) Active site of HDAC8 (PDB ID: 1T64 [<a href="#B15-cells-11-03161" class="html-bibr">15</a>]). Six key residues form the hydrophobic tunnel that occupies two TSA molecules.</p>
Full article ">Figure 2
<p>Schematic representation of human HDAC8 and its regulation. HDAC8 is upregulated by transcription factors and downregulated by miRNA, autophagy, and proteasomal degradation.</p>
Full article ">Figure 3
<p>Schematic representation of histone and non-histone substrates of HDAC8. HDAC8 deacetylates histones and suppresses transcription of target genes. HDAC8 also deacetylates non-histone substrates such as SMC3, α-tubulin, cortactin, HSP20, p53, PKM2, AKT, ERRα, and c-Jun.</p>
Full article ">Figure 4
<p>Schematic representation of pathological functions of HDAC8 in cancer. HDAC8 promotes tumor growth by enhancing tumor cell proliferation and suppressing apoptosis. HDAC8 also enhances metastasis and is involved in drug resistance and immune evasion.</p>
Full article ">
10 pages, 3458 KiB  
Article
Stereological Estimation of Myocardial Fat and Its Associations with Obesity, Epicardial, and Visceral Adipose Tissue
by Pernille Heimdal Holm, Louise Hindsø, Kristine Boisen Olsen and Jytte Banner
Cells 2022, 11(19), 3160; https://doi.org/10.3390/cells11193160 - 8 Oct 2022
Viewed by 1841
Abstract
The normal human heart contains epicardial adipose tissue (EAT) and myocardial fat. The associations between obesity, myocardial fat, visceral adipose tissue (VAT), and cardiovascular disease are not fully understood. The objective of this study was to estimate myocardial fat using stereological methods and [...] Read more.
The normal human heart contains epicardial adipose tissue (EAT) and myocardial fat. The associations between obesity, myocardial fat, visceral adipose tissue (VAT), and cardiovascular disease are not fully understood. The objective of this study was to estimate myocardial fat using stereological methods and investigate its relations with obesity, EAT, and VAT. To establish the EAT volume, 115 deceased individuals were included, and postmortem computed tomography was conducted on their eviscerated hearts. Six samples from the left and right ventricles (LV and RV) of the heart were stereologically examined to calculate the percentage of myocardial fat. Kidney and omental fat were weighed at autopsy, and the waist–hip ratio was calculated. Females had a slightly non-significantly (p = 0.054) larger proportion of RV fat (13.2% ± 4.4) compared to that in men (11.5% ± 2.7). We found a significant positive correlation between body mass index (BMI) and LV myocardial fat (p = 0.033). In the RV, this correlation was only at the borderline of significance (p = 0.052). The EAT volume was positively correlated with both RV and LV myocardial fat. We found no association with the waist–hip ratio (WHR) or the omental or kidney fat as measures of VAT. The myocardial fat was normal, most prominent in the RV, and correlated with the EAT and, partly, BMI. We found no association with VAT. Full article
(This article belongs to the Special Issue Adipose Tissue in Cardiovascular Health)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the exclusion process. PMCT = Postmortem Computed Tomography.</p>
Full article ">Figure 2
<p>(<b>A</b>) Superimage of a whole-slide-scanned image at a resolution of x20. The epicardial adipose tissue (EAT) was excluded in the selection of the region for sampling. (<b>B</b>) Example of a sample ROI from the right ventricle (RV) with a 1/56 grid. (<b>C</b>) Left-ventricle (LV) sample with small areas of myocardial fat. (<b>D</b>) RV sample with extensive myocardial fat.</p>
Full article ">Figure 3
<p>Scatterplots: (<b>A</b>) volume of EAT and RV myocardial fat; (<b>B</b>) volume of EAT and LV myocardial fat; (<b>C</b>) correlation between LV and RV myocardial fat; the combined Pearson correlation coefficient was significant (<span class="html-italic">r</span> = 0.54, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3 Cont.
<p>Scatterplots: (<b>A</b>) volume of EAT and RV myocardial fat; (<b>B</b>) volume of EAT and LV myocardial fat; (<b>C</b>) correlation between LV and RV myocardial fat; the combined Pearson correlation coefficient was significant (<span class="html-italic">r</span> = 0.54, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
11 pages, 606 KiB  
Review
Cell-Enriched Lipotransfer (CELT) Improves Tissue Regeneration and Rejuvenation without Substantial Manipulation of the Adipose Tissue Graft
by Lukas Prantl, Andreas Eigenberger, Ruben Reinhard, Andreas Siegmund, Kerstin Heumann and Oliver Felthaus
Cells 2022, 11(19), 3159; https://doi.org/10.3390/cells11193159 - 8 Oct 2022
Cited by 6 | Viewed by 2180
Abstract
The good availability and the large content of adult stem cells in adipose tissue has made it one of the most interesting tissues in regenerative medicine. Although lipofilling is one of the most frequent procedures in plastic surgery, the method still struggles with [...] Read more.
The good availability and the large content of adult stem cells in adipose tissue has made it one of the most interesting tissues in regenerative medicine. Although lipofilling is one of the most frequent procedures in plastic surgery, the method still struggles with high absorption rates and volume losses of up to 70%. Therefore, many efforts have been made to optimize liposuction and to process the harvested tissue in order to increase fat graft retention. Because of their immunomodulatory properties, their cytokine secretory activity, and their differentiation potential, enrichment with adipose tissue-derived stem cells was identified as a promising tool to promote transplant survival. Here, we review the important parameters for lipofilling optimization. Finally, we present a new method for the enrichment of lipoaspirate with adipose tissue-derived stem cells and discuss the parameters that contribute to fat graft survival. Full article
(This article belongs to the Special Issue Regeneration of Tissue with Mesenchymal Stem Cells)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of macroscopic lipoaspirate morphology after each of the four major steps for tissue processing with the CELT protocol. (<b>A</b>) After it is allowed to sediment under gravitational forces, a syringe is filled with lipoaspirate (illustrated in orange). (<b>B</b>) After the first centrifugation, a large aqueous phase settles at the bottom of the syringe (illustrated in red) and a small oily phase separates at the top of the syringe (illustrated in yellow). The size of the aqueous phase varies depending on the time the tissue is allowed to sediment. The size of the oily phase varies depending on the shear stress the harvesting method has exerted. (<b>C</b>) After the aqueous and oily phases have been discarded, shear-force mechanical processing via intersyringe processing can occur. (<b>D</b>) After mechanical processing and a second centrifugation, a small aqueous and a large oily phases separate from the tissue at the syringe’s top and bottom, respectively. (<b>E</b>) After the aqueous and oily phases have been discarded, a stem cell-enriched lipograft tissue is ready for clinical application.</p>
Full article ">
3 pages, 226 KiB  
Editorial
Mesenchymal Stem/Stromal Cells as a Therapeutic Tool in Cell-Based Therapy and Regenerative Medicine: An Introduction Expertise to the Topical Collection
by Makram Merimi, Hassan Fahmi, Joery De Kock, Charline Beguin, Arsène Burny, Guido Moll, Alessandro Poggi and Mehdi Najar
Cells 2022, 11(19), 3158; https://doi.org/10.3390/cells11193158 - 8 Oct 2022
Cited by 7 | Viewed by 1781
Abstract
We are pleased to present this opening editorial, introducing our topical collection, “The New Era of Mesenchymal Stromal/Stem Cell Functional Application: State of the Art, Therapeutic Challenges and Future Directions” [...] Full article
26 pages, 940 KiB  
Review
Left Atrial Myocardium in Arterial Hypertension
by Jens Kockskämper and Florentina Pluteanu
Cells 2022, 11(19), 3157; https://doi.org/10.3390/cells11193157 - 8 Oct 2022
Cited by 17 | Viewed by 3836
Abstract
Arterial hypertension affects ≈ 1 billion people worldwide. It is associated with increased morbidity and mortality and responsible for millions of deaths each year. Hypertension mediates damage of target organs including the heart. In addition to eliciting left ventricular hypertrophy, dysfunction and heart [...] Read more.
Arterial hypertension affects ≈ 1 billion people worldwide. It is associated with increased morbidity and mortality and responsible for millions of deaths each year. Hypertension mediates damage of target organs including the heart. In addition to eliciting left ventricular hypertrophy, dysfunction and heart failure, hypertension also causes left atrial remodeling that may culminate in atrial contractile dysfunction and atrial fibrillation. Here, we will summarize data on the various aspects of left atrial remodeling in (essential) hypertension gathered from studies on patients with hypertension and from spontaneously hypertensive rats, an animal model that closely mimics cardiac remodeling in human hypertension. Analyzing the timeline of remodeling processes, i.e., distinguishing between alterations occurring in prehypertension, in early hypertension and during advanced hypertensive heart disease, we will derive the potential mechanisms underlying left atrial remodeling in (essential) hypertension. Finally, we will discuss the consequences of these remodeling processes for atrial and ventricular function. The data imply that left atrial remodeling is multifactorial, starts early in hypertension and is an important contributor to the progression of hypertensive heart disease, including the development of atrial fibrillation and heart failure. Full article
(This article belongs to the Special Issue Molecular Biology of Atrial Myocardium)
Show Figures

Figure 1

Figure 1
<p>Risk factors for, initiating mechanisms of, LA remodeling in, and consequences of essential hypertension.</p>
Full article ">
17 pages, 3070 KiB  
Article
Calcium-Related Genes Predicting Outcomes and Serving as Therapeutic Targets in Endometrial Cancer
by Ting Huang, Xuan Feng, Jiaqi Wang, Jingyi Zhou and Jianliu Wang
Cells 2022, 11(19), 3156; https://doi.org/10.3390/cells11193156 - 8 Oct 2022
Cited by 3 | Viewed by 2429
Abstract
Endometrial cancer (EC) is the most common gynecologic cancer with increasing incidence. The dysregulation of intracellular calcium plays a crucial role in cancer progression. However, the relationship between calcium-related genes and prognosis remains unclear. In this study, we aimed to establish a risk [...] Read more.
Endometrial cancer (EC) is the most common gynecologic cancer with increasing incidence. The dysregulation of intracellular calcium plays a crucial role in cancer progression. However, the relationship between calcium-related genes and prognosis remains unclear. In this study, we aimed to establish a risk model based on calcium-related genes for prognosis prediction in patients with EC. The TCGA-total set was divided into a training set and a testing set (1:1). The four-gene prognostic signature (CACNA2D1, SLC8A1, TRPM4 and CCL2) was established and classified all EC patients into a low-risk or high-risk group. This model was validated in both the testing dataset and the total set. The EC patients with high RiskScores showed significantly shorter overall survival than those with low RiskScores, and this trend was consistent among most subgroups. Moreover, an enrichment analysis confirmed that calcium-related and estrogen-response signalings were significantly enriched in the high-risk group. The knockdown of CACNA2D1 by siRNA or its blocker, amlodipine (AM) inhibited cell proliferation and induced cycle arrest in vitro. The calcium channel blocker AM inhibited cell proliferation and induced cycle arrest in vitro. AM also showed marked tumor inhibition effects in vivo. In summary, the prognostic model constructed by four calcium-related genes can reliably predict the outcomes of EC patients, and a calcium channel blocker, AM, has significant potential for EC treatment. Full article
(This article belongs to the Special Issue Advances in Calcium Signaling)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Prognostic analysis of differentially expressed calcium-related genes and the construction of a risk score signature. (<b>A</b>) Venn diagram to identify the common genes of calcium-related genes and DEGs. (<b>B</b>) Volcano plot for DEGs. A total of 158 calcium-related DEGs are highlighted in boxes. (<b>C</b>,<b>D</b>) Visualization of the protein–protein interaction (PPI) network formed by all 158 genes. The MCODE complexes are colored according to their clustering. (<b>E</b>) Significant genes of each MCODE complex are labeled and visualized. (<b>F</b>) Forest plots showing the results of the univariate Cox regression analysis of 6 prognostic genes. (<b>G</b>,<b>H</b>) The cvfit and lambda curves in LASSO regression. (<b>I</b>) The coefficients of four genes. (<b>J</b>) Forest plots showing the results of the multivariate Cox regression analysis of 4 prognostic genes.</p>
Full article ">Figure 2
<p>Validation of the calcium-related signature model in the training cohort, testing cohort and total cohort. (<b>A</b>–<b>C</b>) Risk curves, survival statuses and gene expression levels for high- and low-risk groups in the TCGA-training set, TCGA-test set and TCGA-total set. (<b>D</b>–<b>F</b>) ROC curves showing the potential of the prognostic 4-gene calcium-related gene model in predicting 1-, 3- and 5-year OS in the TCGA-training, TCGA-testing and TCGA-total sets. (<b>G</b>–<b>I</b>) Kaplan–Meier curves for survival status and survival time in the TCGA-training, TCGA-testing and TCGA-total sets. (<b>J</b>–<b>L</b>) The nomograms to predict the 1-year, 3-year and 5-year overall survival rates of EC patients in the TCGA-training, TCGA-testing and TCGA-total sets. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Correlation analysis between the prognostic signature and different clinicopathological characteristics in the TCGA-total cohort. (<b>A</b>–<b>D</b>) The histogram depicting the significant differences in the RiskScores in EC patients stratified by age, histopathological type, stage, and grade. (<b>E</b>,<b>F</b>) Subgroup analysis of Kaplan–Meier curves in different ages ≤60 and &gt;60. (<b>G</b>,<b>H</b>) Subgroup analysis of Kaplan–Meier curves in different histopathological types. (<b>I</b>,<b>J</b>) Subgroup analysis of Kaplan–Meier curves in different stages I–II and III–IV. (<b>K</b>,<b>L</b>) Subgroup analysis of Kaplan–Meier curves in different grades 1–2 and 3. * <span class="html-italic">p</span> &lt; 0.05, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Enrichment analysis of the high-risk group and low-risk group based on the 4-gene prognostic signature. (<b>A</b>) Volcano map of DEGs between the high-risk and low-risk groups. (<b>B</b>–<b>D</b>) GSEA against Hallmark showing significant enrichment of xenobiotic metabolism, estrogen response early and estrogen response late, respectively, in the low-risk endometrial cancer patients. (<b>E</b>) KEGG and GO analyses showing that calcium-related biological processes were significantly enriched.</p>
Full article ">Figure 5
<p>Silencing <span class="html-italic">CACNA2D1</span> inhibited the proliferation and migration of EC cells. (<b>A</b>) Representative images of Western blotting of endogenous expression of CACNA2D1 in eight endometrial cancer cell lines. (<b>B</b>) Immunofluorescence staining of CACNA2D1 and phalloidin in Ishikawa (<b>upper</b>) and HEC-108 (<b>lower</b>) cells. Nuclei were stained with DAPI. Scale bar, 25 µm (Magnification: 40×). (<b>C</b>,<b>D</b>) Transfection efficiency of <span class="html-italic">CACNA2D1</span> via small interfering RNA in Ishikawa (<b>C</b>) and HEC-108 cells (<b>D</b>). (<b>E</b>,<b>F</b>) CCK8 assay showing the effect of CACNA2D1 silencing on Ishikawa (<b>E</b>) and HEC-108 (<b>F</b>) cell proliferation. (<b>G</b>) EdU staining showing the effect of <span class="html-italic">CACNA2D1</span> silencing on Ishikawa and HEC-108 cells. (<b>H</b>,<b>I</b>) The statistical data of the EdU-positive rate after interfering with <span class="html-italic">CACNA2D1</span>. (<b>J</b>) Transwell assay detecting the effect of CACNA2D1 silencing on Ishikawa and HEC-108 cells. (<b>K</b>,<b>L</b>) The statistical data of the migrating cells after interfering with <span class="html-italic">CACNA2D1</span>. Data are shown as the mean ± SD. T-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001. These experiments were conducted in triplicate independently with similar results.</p>
Full article ">Figure 6
<p>AM inhibited EC cell proliferation both in vitro and in vivo. (<b>A</b>) A CCK-8 assay was used to detect the effect of AMs on the proliferation of Ishikawa cells. (<b>B</b>) EdU staining showing the effect of AMs on Ishikawa cells. (<b>C</b>) The statistical data of the EdU-positive rate after treatment with AMs. (<b>D</b>) Cell apoptosis rate was detected by flow cytometry using Annexin-V/PI staining. (<b>E</b>) Representative images of tumor samples (<span class="html-italic">n</span> = 6). (<b>F</b>,<b>G</b>) The statistical data of tumor weight are shown as the mean ± SD. One-way ANOVA. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>H</b>) H&amp;E and PCNA staining of sections from mice treated with AMs (0, 15, 20 mg/kg). Data are shown as the mean ± SD. (<b>C</b>) <span class="html-italic">t</span>-Test. (<b>E</b>) One-way ANOVA. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001. These experiments were conducted in triplicate independently with similar results.</p>
Full article ">
20 pages, 1078 KiB  
Review
Nasal Microbiome and Its Interaction with the Host in Childhood Asthma
by Yao Zeng and Jessie Qiaoyi Liang
Cells 2022, 11(19), 3155; https://doi.org/10.3390/cells11193155 - 7 Oct 2022
Cited by 6 | Viewed by 3131
Abstract
Childhood asthma is a major chronic non-communicable disease in infants and children, often triggered by respiratory tract infections. The nasal cavity is a reservoir for a broad variety of commensal microbes and potential pathogens associated with respiratory illnesses including asthma. A healthy nasal [...] Read more.
Childhood asthma is a major chronic non-communicable disease in infants and children, often triggered by respiratory tract infections. The nasal cavity is a reservoir for a broad variety of commensal microbes and potential pathogens associated with respiratory illnesses including asthma. A healthy nasal microenvironment has protective effects against respiratory tract infections. The first microbial colonisation in the nasal region is initiated immediately after birth. Subsequently, colonisation by nasal microbiota during infancy plays important roles in rapidly establishing immune homeostasis and the development and maturation of the immune system. Dysbiosis of microbiota residing in the mucosal surfaces, such as the nasopharynx and guts, triggers immune modulation, severe infection, and exacerbation events. Nasal microbiome dysbiosis is related to the onset of symptomatic infections. Dynamic interactions between viral infections and the nasal microbiota in early life affect the later development of respiratory infections. In this review, we summarise the existing findings related to nasal microbiota colonisation, dynamic variations, and host–microbiome interactions in childhood health and respiratory illness with a particular examination of asthma. We also discuss our current understanding of biases produced by environmental factors and technical concerns, the importance of standardised research methods, and microbiome modification for the prevention or treatment of childhood asthma. This review lays the groundwork for paying attention to an essential but less emphasized topic and improves the understanding of the overall composition, dynamic changes, and influence of the nasal microbiome associated with childhood asthma. Full article
(This article belongs to the Special Issue The Multifaceted Microbiome in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Variations in nasal microbiota and species-specific responses of the airway mucosa and metabolic pathways induced by microbiota colonisation. Changes in representative nasal microbiota, such as higher abundances of Oxalobacteraceae, Aerococcaceae, and Alloiococcus (↑) and lower abundances of Corynebacteriaceae and Staphylococcaceae (↓) [<a href="#B28-cells-11-03155" class="html-bibr">28</a>], are involved in the development of early-onset wheezing in infants. Higher abundances of Staphylococcus and Streptococcus (↑) are more prevalent in asthmatic children than in healthy controls [<a href="#B55-cells-11-03155" class="html-bibr">55</a>]. Opportunistic pathogens that colonise the mucosal layer are associated with host inflammatory immune responses (IL-1β, TNF-α, MIP-1β, IL-17, CCL20, IL1A, IRAK2, IL-8, IL-33), apoptosis signals (TNF and C8orf4), and epithelial damage (LDH) [<a href="#B56-cells-11-03155" class="html-bibr">56</a>,<a href="#B59-cells-11-03155" class="html-bibr">59</a>,<a href="#B60-cells-11-03155" class="html-bibr">60</a>,<a href="#B62-cells-11-03155" class="html-bibr">62</a>]. Host IL-17 signalling can significantly restructure the nasal microbiome and successful resistance to pathogenic Proteobacteria colonisation [<a href="#B64-cells-11-03155" class="html-bibr">64</a>]. <span class="html-italic">p</span>-Cresol sulphate, a metabolite of L-tyrosine used by gut microbiota, can protect the host against allergic airway inflammation by reducing CCL20 [<a href="#B102-cells-11-03155" class="html-bibr">102</a>]. The different expression levels of functional genes from specific microbiota (<span class="html-italic">B. longum</span>, <span class="html-italic">E. coli</span>, <span class="html-italic">V. parvula</span>, and <span class="html-italic">B. caccae</span>) between asthmatic and non-asthmatic children significantly influence metabolic pathways [<a href="#B68-cells-11-03155" class="html-bibr">68</a>].</p>
Full article ">
15 pages, 3593 KiB  
Article
A Novel Homozygous Founder Variant of RTN4IP1 in Two Consanguineous Saudi Families
by Mazhor Aldosary, Maysoon Alsagob, Hanan AlQudairy, Ana C. González-Álvarez, Stefan T. Arold, Mohammad Anas Dababo, Omar A. Alharbi, Rawan Almass, AlBandary AlBakheet, Dalia AlSarar, Alya Qari, Mysoon M. Al-Ansari, Monika Oláhová, Saif A. Al-Shahrani, Moeenaldeen AlSayed, Dilek Colak, Robert W. Taylor, Mohammed AlOwain and Namik Kaya
Cells 2022, 11(19), 3154; https://doi.org/10.3390/cells11193154 - 7 Oct 2022
Cited by 1 | Viewed by 2074
Abstract
The genetic architecture of mitochondrial disease continues to expand and currently exceeds more than 350 disease-causing genes. Bi-allelic variants in RTN4IP1, also known as Optic Atrophy-10 (OPA10), lead to early-onset recessive optic neuropathy, atrophy, and encephalopathy in the afflicted patients. The gene [...] Read more.
The genetic architecture of mitochondrial disease continues to expand and currently exceeds more than 350 disease-causing genes. Bi-allelic variants in RTN4IP1, also known as Optic Atrophy-10 (OPA10), lead to early-onset recessive optic neuropathy, atrophy, and encephalopathy in the afflicted patients. The gene is known to encode a mitochondrial ubiquinol oxidoreductase that interacts with reticulon 4 and is thought to be a mitochondrial antioxidant NADPH oxidoreductase. Here, we describe two unrelated consanguineous families from the northern region of Saudi Arabia harboring a missense variant (RTN4IP1:NM_032730.5; c.475G<T, p.Val159Phe) in the gene. Clinically affected individuals presented with intellectual disability, encephalopathy, ataxia, optic atrophy, and seizures. Based on whole exome sequencing and confirmatory Sanger sequencing, the variant was fully segregated with the phenotype in the families, absent among large ethnically matching controls as well as numerous in-house exomes, and predicted to be pathogenic by different in silico classifiers. Structural modeling and immunoblot analyses strongly indicated this variant to be pathogenic. Since the families belong to one of the tribal inhabitants of Saudi Arabia, we postulate that the variant is likely to be a founder. We provide the estimated age of the variant and present data confirming the disease-causality of this founder variant. Full article
(This article belongs to the Section Intracellular and Plasma Membranes)
Show Figures

Figure 1

Figure 1
<p>Histochemical and immunohistochemical analyses of muscle biopsy. (<b>A</b>) Gomori trichrome staining shows prominent subsarcolemmal accumulation of mitochondria in most fibers with ragged red-like fibers. (<b>B</b>) COX staining shows no COX-negative fibers, but there is marked subsarcolemmal staining. (<b>C</b>) SDH staining shows marked subsarcolemmal staining. (<b>D</b>) Oil red staining indicates moderate increase in lipid stores.</p>
Full article ">Figure 2
<p>Brain MRI results. Selected MRI sequences and planes showing optic chiasm atrophy (white/red/black arrows) in patient 1 (image (<b>1B</b>) coronal CISS), Patient 2 (image (<b>1C</b>) coronal T2), Patient 4 (image (<b>1E</b>) sagittal T1), and Patient 5 (image (<b>1F</b>) coronal T2). Other selected images also show bilateral optic nerve atrophy (white arrows) in patient 1 (image (<b>1A</b>) coronal T2), Patient 3 (image (<b>1D</b>) axial T2), and Patient 6 (image (<b>1G</b>) axial T2; image (<b>1H</b>) coronal T2).</p>
Full article ">Figure 3
<p>Brain MRS findings of the patients. Multiple single-voxel MRS studies showed small lactate doublets at 1.3 ppm (white arrows) in Patient 1 (image (<b>2A</b>)), Patient 3 (image (<b>2B</b>)), Patient 5 (image (<b>2D</b>)), and Patient 6 (image (<b>2E</b>)). There is no obvious lactate doublet seen in Patient 4 (image (<b>2C</b>)). An MRS study was not performed for Patient 2.</p>
Full article ">Figure 4
<p>Genetic analysis results. (<b>A</b>) Pedigree of the Family 1. Affected individuals are labeled with black-colored (filled) symbols. Carriers are represented with half-filled symbols; squares represent males, circles represent females, and double line indicates consanguinity. (<b>B</b>) Pedigree for the family 2. Brief Sanger sequencing results of the c.475 G&gt;T variant under the pedigrees show the results for the Families. (<b>C</b>) Homozygosity results for all the affected individuals in both families. The patients are aligned with the normal individuals in both families. The shared ROH block is shown in the red bracket on chromosome 6 where RTN4IP1 is located. Numbers 1-6 indicate family members from Family 1, individuals 1 to Family 2, and individual 3, (F1-II-1, F1-II-2, F1-II-3, F2-II-1, F2-II-2, and F2-II-3), respectively. (<b>D</b>) Protein sequence alignment of RTN4IP1, the blue arrow shows the affected Valine amino acids at position 159 by the novel variant. The alignment showed that the amino acid in this region is highly conserved among different species.</p>
Full article ">Figure 5
<p>Structural analysis and immunoblotting experiment results. (<b>A</b>) The location of the variant within the 3-D crystal structure of RTN4IP1 (PDB ID: 2VN8). RTN4IP1 forms a dimer (green and blue chains). Val159 is shown as a red stick model. The cofactor NADPH is shown as a stick model with carbon atoms in white. The zoom view shows the effect of the p.Val159Phe substitution. Clashes of the Phenylalanine side chain (yellow) are shown as red discs. Key side chains in the vicinity are highlighted as stick models. (<b>B</b>) Immunoblotting analysis results. The Western blotting analysis revealed significantly decreased RTN4IP1 protein levels in the patient’s extracts from cultured fibroblasts in comparison to controls. A polyclonal antibody was targeted against the protein, and beta-actin was used as a control-loading marker. (<b>C</b>) Schematic presentation of <span class="html-italic">RTN4IP1</span>. The top panel shows localization of all previously reported variants (black) and the newly identified novel variant (red) distributed among the nine exons (blue). The bottom panel shows both the alcohol dehydrogenase domain (ADH_N) and the zinc-binding motif (ADH_Zinc).</p>
Full article ">
20 pages, 2269 KiB  
Review
TFEB; Beyond Its Role as an Autophagy and Lysosomes Regulator
by Berenice Franco-Juárez, Cristina Coronel-Cruz, Beatriz Hernández-Ochoa, Saúl Gómez-Manzo, Noemi Cárdenas-Rodríguez, Roberto Arreguin-Espinosa, Cindy Bandala, Luis Miguel Canseco-Ávila and Daniel Ortega-Cuellar
Cells 2022, 11(19), 3153; https://doi.org/10.3390/cells11193153 - 7 Oct 2022
Cited by 48 | Viewed by 9407
Abstract
Transcription factor EB (TFEB) is considered the master transcriptional regulator of autophagy and lysosomal biogenesis, which regulates target gene expression through binding to CLEAR motifs. TFEB dysregulation has been linked to the development of numerous pathological conditions; however, several other lines of evidence [...] Read more.
Transcription factor EB (TFEB) is considered the master transcriptional regulator of autophagy and lysosomal biogenesis, which regulates target gene expression through binding to CLEAR motifs. TFEB dysregulation has been linked to the development of numerous pathological conditions; however, several other lines of evidence show that TFEB might be a point of convergence of diverse signaling pathways and might therefore modulate other important biological processes such as cellular senescence, DNA repair, ER stress, carbohydrates, and lipid metabolism and WNT signaling-related processes. The regulation of TFEB occurs predominantly at the post-translational level, including phosphorylation, acetylation, SUMOylating, PARsylation, and glycosylation. It is noteworthy that TFEB activation is context-dependent; therefore, its regulation is subjected to coordinated mechanisms that respond not only to nutrient fluctuations but also to stress cell programs to ensure proper cell homeostasis and organismal health. In this review, we provide updated insights into novel post-translational modifications that regulate TFEB activity and give an overview of TFEB beyond its widely known role in autophagy and the lysosomal pathway, thus opening the possibility of considering TFEB as a potential therapeutic target. Full article
Show Figures

Figure 1

Figure 1
<p>The TFEB activity is regulated by post-translational modifications. TFEB is mainly phosphorylated to avoid its nuclear translocation; however, depending on the cellular context, TFEB can be dephosphorylated, acetylated, glycosylated, or PARsylated.</p>
Full article ">Figure 2
<p>The TFEB activation can influence the induction of senescence. Decreased nuclear TFEB localization may be one of many causes of senescence; however, restoring TFEB nuclear activity could contribute to avoiding cellular senescence, perhaps through the induction of autophagy or DNA repair genes.</p>
Full article ">Figure 3
<p>The TFEB regulates the expression of several regulators of the ER stress response. ER stress, triggered by several compounds, promotes the nuclear localization of TFEB in a PERK-, ATF6-, IRE1α-, and calcineurin-dependent manner that consequently induces the transcription of UPR-related genes to restore ER homeostasis and support cell survival or apoptosis, if the stress is persistent.</p>
Full article ">Figure 4
<p>The TFEB regulates genes involved in carbohydrate metabolism, lipid metabolism, and insulin signaling. Once active, TFEB may promote energy utilization through the activation of several related genes. Some of the proposed metabolic processes regulated by TFEB are shown, together with the relevant substrates.</p>
Full article ">Figure 5
<p>The TFEB is expressed in several tissues, including the liver, intestine, and white adipose tissue. In these cell types, in response to several stimuli, TFEB is activated and induces the expression of several genes, as indicated in the figure. In the liver, cholesterol accumulation causes lysosomal stress, which consequently induces TFEB nuclear translocation, activating the expression of cholesterol 7α-hydroxylase (CYP7A1) to produce hepatic bile acid synthesis that, in the intestine cells, promotes the production of the hormone fibroblast growth factor-15/19 (FGF15/19), and this consequently activates both the activate orphan nuclear receptor, the small heterodimer partner (SHP/NR0B2) and TFEB, to activates genes (Ulk1 and Atgl) essential for lipophagy. TFEB activation by overexpression or the occurrence of lysosomal stress in fat-resident macrophages protects against diet-induced weight gain and adiposity through the induction of growth differentiation factor 15 (GDF15)-enhanced adipose lipid catabolism, and it reduces inflammation.</p>
Full article ">
17 pages, 8208 KiB  
Article
Immunomodulatory Effect and Bone Homeostasis Regulation in Osteoblasts Differentiated from hADMSCs via the PD-1/PD-L1 Axis
by Seung-Cheol Lee, Min Kyoung Shin, Bo-Young Jang, Seung-Ho Lee, Min Kim and Jung-Suk Sung
Cells 2022, 11(19), 3152; https://doi.org/10.3390/cells11193152 - 7 Oct 2022
Cited by 3 | Viewed by 2386
Abstract
Human mesenchymal stem cells (hMSCs) are promising candidates for stem cell therapy and are known to secrete programmed death-1 (PD-1) ligand 1 (PD-L1) regulating T cell-mediated immunosuppression. Given the limitations of current stem cell therapy approaches, improvements in immunomodulatory capacity and stem cell [...] Read more.
Human mesenchymal stem cells (hMSCs) are promising candidates for stem cell therapy and are known to secrete programmed death-1 (PD-1) ligand 1 (PD-L1) regulating T cell-mediated immunosuppression. Given the limitations of current stem cell therapy approaches, improvements in immunomodulatory capacity and stem cell differentiation efficacy are needed. In this study, we propose novel strategies to overcome the challenges that remain in hMSC-mediated bone regeneration. We found that PD-1 is highly expressed in osteoblasts, and the PD-1/PD-L1 axis mediated the decreased proinflammatory cytokine expressions in differentiated osteoblasts cocultured with human adipose derived mesenchymal stem cells (hADMSCs). Moreover, the decrease was attenuated by PD-1/PD-L1 pathway inhibition. Osteogenic properties including osteogenic gene expression and calcium deposits were increased in osteoblasts cocultured with hADMSCs compared with those that were monocultured. Osteoblasts treated with PD-L1 and exosomes from hADMSCs also exhibited enhanced osteogenic properties, including calcium deposits and osteogenic gene expression. In our cocultured system that mimics the physiological conditions of the bone matrix, the PD-1/PD-L1 axis mediated the increased expression of osteogenic genes, thereby enhancing the osteogenic properties, while the calcium deposits of osteoblasts were maintained. Our results provide the therapeutic potentials and novel roles of the PD-1/PD-L1 axis in bone matrix for modulating the bone properties and immunosuppressive potentials that can aid in the prevention of bone diseases via maintaining bone homeostasis. Full article
Show Figures

Figure 1

Figure 1
<p>Relative PD-1 expression levels in differentiated cells derived from hADMSCs. (<bold>A</bold>) Relative mRNA expression level of <italic>PD-1</italic> in adipocytes (Ad) and (<bold>B</bold>) osteoblasts (Os) compared with hADMSCs (human adipose-derived mesenchymal stem cells). (<bold>C</bold>) Variance in the expression level of PD-1 in differentiated cells. <italic>N</italic> = 3 trials per sample and control. (<bold>D</bold>) Cells were stained with PD-1 (green); DAPI was used to stain the nuclei (blue). (<bold>E</bold>) Relative fluorescence intensity of PD-1 receptor on cell surface of differentiated cells. <italic>N</italic> = 5 trials per sample and control. Data are presented as mean ± SEM. *** <italic>p</italic> &lt; 0.001, compared with the hADMSCs and Ad group. hADMSCs: human adipose-derived mesenchymal stem cells; Ad: adipocytes; Os: osteoblasts.</p>
Full article ">Figure 2
<p>Regulation of the expression levels of proinflammatory cytokines in differentiated cells cocultured with hADMSCs. (<bold>A</bold>) Differentiated adipocytes were cocultured with hADMSCs. Relative mRNA expression levels of (<bold>B</bold>) <italic>TNF-α</italic>, (<bold>C</bold>) <italic>IL-1β</italic>, and (<bold>D</bold>) <italic>IL-6</italic>. (<bold>E</bold>) Differentiated osteoblasts were cocultured with hADMSCs. Relative mRNA expression levels of (<bold>F</bold>) <italic>TNF-α</italic>, (<bold>G</bold>) <italic>IL-1β</italic>, and (<bold>H</bold>) <italic>IL-6</italic>. <italic>N</italic> = 3 trials per sample and control. Data are presented as mean ± SEM. * <italic>p</italic> &lt; 0.05 and *** <italic>p</italic> &lt; 0.001, compared with the Os group. hADMSCs: human adipose-derived mesenchymal stem cells; Ad: adipocytes; coAd: cocultured adipocytes; Os: osteoblasts; coOS: cocultured osteoblasts.</p>
Full article ">Figure 3
<p>Attenuation of proinflammatory cytokine expression by PD-1/PD-L1 axis inhibition in differentiated cells cocultured with hADMSCs. (<bold>A</bold>) PD-1/PD-L1 interaction was inhibited in differentiated adipocytes cocultured with hADMSCs. Relative mRNA expression levels of (<bold>B</bold>) <italic>TNF-α</italic>, (<bold>C</bold>) <italic>IL-1β</italic>, and (<bold>D</bold>) <italic>IL-6</italic>. (<bold>E</bold>) PD-1/PD-L1 interaction was inhibited in differentiated osteoblasts cocultured with hADMSCs. Relative mRNA expression levels of (<bold>F</bold>) <italic>TNF-α</italic>, (<bold>G</bold>) <italic>IL-1β</italic>, and (<bold>H</bold>) <italic>IL-6</italic>. <italic>N</italic> = 3 trials per sample and control. Data are presented as mean ± SEM. hADMSCs: human adipose-derived mesenchymal stem cells; Ad: adipocytes; coAd: cocultured adipocytes; Os: osteoblasts; coOs: cocultured osteoblasts; B: BMS 202 (10 µM).</p>
Full article ">Figure 4
<p>Effects of the PD-1/PD-L1 axis on osteogenic properties in osteoblasts cocultured with hADMSCs. (<bold>A</bold>) Expression levels of osteogenic marker proteins in monocultured or cocultured osteoblasts for 3 days. Relative expression levels of (<bold>B</bold>) ALP, (<bold>C</bold>) OCN, and (<bold>D</bold>) RUNX2 in cocultured osteoblasts for 3 days compared with monocultured osteoblasts. (<bold>E</bold>) Expression levels of osteogenic marker proteins in monocultured or cocultured osteoblasts for 7 days. Relative expression levels of (<bold>F</bold>) ALP, (<bold>G</bold>) OCN, and (<bold>H</bold>) RUNX2 in cocultured osteoblasts for 7 days compared with monocultured osteoblasts. <italic>N</italic> = 3 trials per sample and control. (<bold>I</bold>) Enhancement of matrix mineralization in cocultured osteoblasts and attenuation via inhibition of PD-1/PD-L1 interaction. (<bold>J</bold>) Stained calcium deposits were dissolved and quantified. <italic>N</italic> = 5 trials per sample and control. Data are presented as mean ± SEM. *** <italic>p</italic> &lt; 0.001, compared with the Os group and <sup>###</sup> <italic>p</italic> &lt; 0.001, compared with the coOs group. Os: osteoblasts; coOs: cocultured osteoblasts; B: BMS 202 (10 µM).</p>
Full article ">Figure 5
<p>Regulatory effects of exosomal PD-L1 on bone matrix formation in osteoblasts. (<bold>A</bold>) Regulatory effects on the enhancement of matrix mineralization in PD-L1- and exosome-treated osteoblasts. (<bold>B</bold>) Stained calcium deposits were dissolved and quantified. <italic>N</italic> = 5 trials per sample and control. (<bold>C</bold>) Expression levels of proteins mediating the osteogenic properties upon PD-L1 and exosome treatment. Relative expression levels of (<bold>D</bold>) COL1A1, (<bold>E</bold>) PDLIM3, and (<bold>F</bold>) RUNX2 in osteoblasts. <italic>N</italic> = 3 trials per sample and control. (<bold>G</bold>) Attenuation of the enhanced matrix mineralization in PD-L1- and exosome-treated osteoblasts via blocking of PD-1/PD-L1 interaction. (<bold>H</bold>) Stained calcium deposits were dissolved and quantified. <italic>N</italic> = 5 trials per sample and control. (<bold>I</bold>) Expression levels of proteins mediating the osteogenic properties upon the inhibition of PD-1/PD-L1 interaction in PD-L1- and exosome-treated osteoblasts. Relative expression levels of (<bold>J</bold>) PDLIM3 and (<bold>K</bold>) RUNX2 in osteoblasts. <italic>N</italic> = 5 trials per sample and control. Data are presented as mean ± SEM. ** <italic>p</italic> &lt; 0.01, and *** <italic>p</italic> &lt; 0.001, compared with the Os and Os B group. Os: osteoblasts; PD-L1: PD-L1 (500 ng/mL); Exo: exosome (10 µg/mL, 2.5 × 10<sup>8</sup> particles/mL); B: BMS 202 (10 µM).</p>
Full article ">Figure 6
<p>Modulatory effect of exosomal PD-L1 on bone matrix formation in cocultured environment. (<bold>A</bold>) Osteoblasts and osteoclasts were cocultured to mimic the bone metabolism under physiological conditions. (<bold>B</bold>) Maintenance of the bone homeostasis in PD-L1- and exosome-treated cocultured osteoblasts. (<bold>C</bold>) Stained calcium deposits were dissolved and quantified. <italic>N</italic> = 5 trials per sample and control. (<bold>D</bold>) Expression levels of proteins mediating the osteogenic properties upon PD-L1 and exosome treatment in cocultured osteoblasts. Relative expression levels of (<bold>E</bold>) COL1A1, (<bold>F</bold>) PDLIM3, and (<bold>G</bold>) RUNX2. <italic>N</italic> = 3 trials per sample and control. Relative mRNA expressions of osteogenic markers (<bold>H</bold>) <italic>ALP</italic>, (<bold>I</bold>) <italic>OCN</italic>, and (<bold>J</bold>) <italic>RUNX2</italic>. <italic>N</italic> = 3 trials per sample and control. Data are presented as mean ± SEM. * <italic>p</italic> &lt; 0.05, ** <italic>p</italic> &lt; 0.01, and *** <italic>p</italic> &lt; 0.001, compared with the Os group. Os: osteoblasts (cocultured); PD-L1: PD-L1 (500 ng/mL); Exo: exosome (10 µg/mL, 2.5 × 10<sup>8</sup> particles/mL).</p>
Full article ">Figure 7
<p>Attenuation of osteogenic properties in cocultured osteoblasts via inhibition of PD-1/PD-L1 interaction. (<bold>A</bold>) PD-1/PD-L1 interaction was inhibited in the cocultured environment. (<bold>B</bold>) Maintenance of bone mineralization in cocultured osteoblasts. (<bold>C</bold>) Stained calcium deposits were dissolved and quantified. <italic>N</italic> = 5 trials per sample and control. (<bold>D</bold>) Attenuation of the expression levels of proteins mediating osteogenic properties upon inhibition of PD-1/PD-L1 interaction. Relative expression levels of (<bold>E</bold>) COL1A1, (<bold>F</bold>) PDLIM3, and (<bold>G</bold>) RUNX2. <italic>N</italic> = 3 trials per sample and control. Data are presented as mean ± SEM. ** <italic>p</italic> &lt; 0.01, and *** <italic>p</italic> &lt; 0.001, compared with the Os B group. Os: osteoblasts (cocultured); PD-L1: PD-L1 (500 ng/mL); Exo: exosome (10 µg/mL, 2.5 × 10<sup>8</sup> particles/mL); B: BMS 202 (10 µM).</p>
Full article ">
Previous Issue
Back to TopTop