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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (530)

Search Parameters:
Keywords = meiosis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 2933 KiB  
Article
Therapeutic Potential of Lappula patula Extracts on Germline Development and DNA Damage Responses in C. elegans
by Qinghao Meng, Anna Hu, Weiyu Xiao, Robert P. Borris and Hyun-Min Kim
Pharmaceuticals 2025, 18(1), 89; https://doi.org/10.3390/ph18010089 - 13 Jan 2025
Viewed by 300
Abstract
Background: Lappula patula (L. patula) is a plant with known medicinal properties, and its extracts have shown promise as potential anti-cancer agents. This study aimed to evaluate the nematocidal effects of L. patula extracts and investigate their impact on germline development, DNA [...] Read more.
Background: Lappula patula (L. patula) is a plant with known medicinal properties, and its extracts have shown promise as potential anti-cancer agents. This study aimed to evaluate the nematocidal effects of L. patula extracts and investigate their impact on germline development, DNA damage responses, and apoptosis in Caenorhabditis elegans (C. elegans), a model organism for studying these processes. Methods: C. elegans was exposed to L. patula extracts to assess survival, development, and incidence of male phenotype. Germline abnormalities were examined using microscopy at different developmental stages. The DNA damage response was evaluated through the expression of the atm-1, atl-1 and pCHK-1. Apoptosis was quantified by monitoring cell death during the pachytene stage. LC-MS was used to identify bioactive compounds within the extracts. Results: Exposure to L. patula extracts resulted in a dose-dependent reduction in worm survival and larval developmental progress, with no significant impact on the male incidence. Germline defects were observed, including increased nuclear spacing at premeiotic and pachytene stages, altered number of bivalents during diakinesis. These defects correlated with a significant decrease in brood size. Also, L. patula extracts activated the DNA damage response pathway, marked by increased expression of atm-1 and atl-1. Moreover, the extracts induced apoptosis in the germline in a pCHK-1-independent manner. LC-MS analysis revealed 31 potential anti-tumor compounds, supporting the extract’s cytotoxic properties. Conclusions: Lappula patula extracts exhibit potent nematocidal and cytotoxic properties, suggesting their potential for cancer therapy. The observed DNA damage and apoptosis in C. elegans emphasize the extract’s promising role in anti-cancer drug development. Further studies are needed to explore the therapeutic potential of these compounds in clinical settings. Full article
Show Figures

Figure 1

Figure 1
<p>Dose-dependent nematocidal effects of <span class="html-italic">Lappula patula</span> extracts on <span class="html-italic">C. elegans</span> survival and development, with no impact on OP50 growth. (<b>A</b>) <span class="html-italic">Lappula patula</span> extracts significantly reduced the survival and development of <span class="html-italic">C. elegans</span>. The effects were evaluated by treating worms with different concentrations of <span class="html-italic">L. patula</span> extracts (0.03, 0.3, and 3 µg/mL, indicated by brown, orange, and gray colors, respectively) and monitoring their survival, adult formation, and male (HIM) phenotype over a 48 h period. A clear inverse relationship was observed between the dose of the herbal extract and the survival and adult worm percentages, indicating that higher concentrations of <span class="html-italic">L. patula</span> extract led to a marked decrease in worm viability and maturation. However, the percentage of males did not exhibit a dose-dependent trend, suggesting that the disruption in sex chromosome segregation may not be directly influenced by the dosage of the extract. Statistical significance was assessed using a two-tailed <span class="html-italic">t</span>-test, with ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; and **** <span class="html-italic">p</span> &lt; 0.0001, comparing the control (+DMSO) with the treated samples. (<b>B</b>) To evaluate whether the nematocidal effects of <span class="html-italic">Lappula patula</span> could be attributed to an inhibition of bacterial growth, we assessed the growth of OP50 in the presence of <span class="html-italic">L. patula</span> extract. Over a 24 h incubation period, no significant inhibition of bacterial growth was observed at 0.03 μg/mL of <span class="html-italic">L. patula</span> extract.</p>
Full article ">Figure 2
<p>Disruption of germline development caused by <span class="html-italic">Lappula patula</span> extracts. (<b>A</b>) DAPI-stained nuclei during germline development. Exposure to <span class="html-italic">Lappula patula</span> extracts led to an increase in the gaps between nuclei in the PMT and pachytene, as indicated by the arrows. The distances between adjacent nuclei were greater in the worms treated with the herb extract compared with the control (DMSO). Worms exposed to the herbal extract often exhibited fewer DAPI-stained bivalent bodies during diakinesis, suggesting impaired DNA recombination. Scale bar: 2 µm. (<b>B</b>) Quantification of the increased nuclear spacing in the premeiotic tip (PMT) and pachytene stages shown in the panel. (<b>C</b>) Quantification of crescent-shaped nuclei in the germline. The number of crescent-shaped nuclei per gonad arm is indicated. (<b>D</b>) Quantification of DAPI-stained bivalents in the germline. The percentage of bivalent is indicated. (<b>E</b>) Quantification of germline size. Germline size, as indicated by the length of the PMT, TZ, and pachytene stages, was measured in worms treated with <span class="html-italic">L. patula</span> or <span class="html-italic">O. cornuta</span> extracts. (<b>F</b>) Brood size of <span class="html-italic">Lappula patula</span>-exposed worms. Treatment with <span class="html-italic">Lappula patula</span> extracts led to a notable decrease in the number of offspring produced by hermaphrodite worms over a span of four days. Statistical significance was determined using a two-tailed Mann–Whitney test, as indicated by asterisks. All experiments were performed with <span class="html-italic">C. elegans</span> hermaphrodites, and the data are presented as the mean ± SEM.</p>
Full article ">Figure 3
<p><span class="html-italic">Lappula patula</span> extracts activated the DNA damage response, leading to upregulation of <span class="html-italic">ATM-1</span> and <span class="html-italic">ATL-1</span>, and increased apoptosis but did not induce pCHK-1 foci. (<b>A</b>) Exposure to <span class="html-italic">Lappula patula</span> extracts significantly enhanced the mRNA expression of <span class="html-italic">atm-1</span> and <span class="html-italic">atl-1</span>, two essential proteins involved in the DNA damage response pathway, confirming the activation of this cellular defense mechanism. Both <span class="html-italic">atm-1</span> and <span class="html-italic">atl-1</span> are critical for activating repair pathways such as homologous recombination and cell cycle arrest, and their upregulation in response to extract exposure provides evidence of the activation of these protective mechanisms. This upregulation suggests that the extracts may trigger a cellular reaction to DNA damage, which is necessary to repair the compromised genome. (<b>B</b>) No distinct increase in pCHK-1 foci (arrows) was observed in the germline cells of worms treated with <span class="html-italic">Lappula patula</span> extracts. Despite the elevation of <span class="html-italic">atm-1</span> and <span class="html-italic">atl-1</span> levels, no significant increase in pCHK-1 levels was observed, implying that while the DNA damage response was triggered, the downstream signaling associated with checkpoint activation, particularly the phosphorylation of CHK-1, did not occur as expected. The extract from <span class="html-italic">O.c.</span> was used as a positive control for these experiments, as it has been previously shown to activate the DNA damage response pathway robustly (<span class="html-italic">p</span> = 0.825 in control and <span class="html-italic">L.p.</span>, <span class="html-italic">p</span> = 0.0021 in control and <span class="html-italic">O.c.</span>; scale bar = 2 µm. (<b>C</b>) When examined during the pachytene stage, a significant increase in apoptosis (arrow) was detected in the germline cells exposed to <span class="html-italic">Lappula patula</span> extracts. Statistical significance was determined by a two-tailed Mann–Whitney test, with asterisks denoting <span class="html-italic">p</span> values indicating significant differences between the control and experimental groups. Scale bar = 20 µm.</p>
Full article ">Figure 4
<p>Fragmentation patterns of 31 potential anti-tumor components identified in <span class="html-italic">Lappula patula</span> extracts. Out of 112 identified substances, 31 were found to be potential anti-tumor components. The peaks (hollow arrows) shown represent the breakdown patterns of these 31 components, analyzed using PeakView Analyst TF 1.6 software. The <span class="html-italic">x</span> axis shows the mass-to-charge ratio (<span class="html-italic">m</span>/<span class="html-italic">z</span>), and the <span class="html-italic">y</span> axis shows the intensity.</p>
Full article ">Figure 5
<p><span class="html-italic">Lappula patula</span> extract triggered a DNA damage response, which resulted in disturbances in germline development, enhanced apoptotic cell death, and disruption of the normal meiotic process. These findings suggest that the extract has cytotoxic effects. Furthermore, <span class="html-italic">Lappula patula</span> contains at least 31 compounds known for their potential anti-tumor properties. This presence of bioactive compounds underscores the potential of <span class="html-italic">Lappula patula</span> as a source of natural anti-cancer agents. Given the growing interest in plant-based treatments for cancer, further investigation into the specific compounds and their mechanisms in <span class="html-italic">Lappula patula</span> is essential to fully explore its therapeutic potential.</p>
Full article ">
22 pages, 4818 KiB  
Article
A Comparative Transcriptome Analysis Reveals the Effects of Letrozole and 17α-Methyltestosterone on the Expression of Reproductive-Related Genes in Golden Pompano (Trachinotus ovatus)
by Qi Zhou, Yan Wang, Yuwen Guo, Guangli Li, Tao Wang and Huapu Chen
Fishes 2025, 10(1), 21; https://doi.org/10.3390/fishes10010021 - 6 Jan 2025
Viewed by 314
Abstract
Aromatase inhibitor letrozole (LZ) and 17α-methyltestosterone (MT) are commonly used for sex control breeding to induce female-to-male sex reversal in aquaculture. However, studies on the molecular regulatory mechanisms of LZ and MT are still limited. Here, a comparative transcriptome analysis was conducted on [...] Read more.
Aromatase inhibitor letrozole (LZ) and 17α-methyltestosterone (MT) are commonly used for sex control breeding to induce female-to-male sex reversal in aquaculture. However, studies on the molecular regulatory mechanisms of LZ and MT are still limited. Here, a comparative transcriptome analysis was conducted on the hypothalamus, the gonad, and the liver of golden pompano (Trachinotus ovatus) after 30 days of LZ and MT treatment, which demonstrated significant sexual size dimorphism. Following LZ treatment, the transcriptome data identified 156, 890, and 1286 different expression genes (DEGs) in the hypothalamus, the liver, and the gonad, respectively. After MT treatment, the transcriptome data identified 114, 876, and 950 DEGs in the hypothalamus, the liver, and the gonad, respectively. Functional enrichment analysis revealed that DEGs were involved in oocyte meiosis and steroid-hormone synthesis following LZ treatment. Similarly, the analysis indicated that DEGs were associated with cysteine and methionine metabolism and steroid-hormone synthesis after MT treatment. Further analysis showed that LZ treatment can promote the first meiotic division but inhibit the second meiotic division of the oocyte. MT treatment promotes polyamine synthesis to induce oocyte apoptosis, both of which affect the trajectory of ovarian differentiation in golden pompano. However, LZ and MT have converse effects in sex hormone synthesis. LZ inhibits estrogen metabolism but promotes cholesterol synthesis by regulating sqle, cyp3a40, ugt1a1, and ugt2a1, while MT promotes estrogen metabolism but inhibits the conversion of cholesterol to pregnenolone by regulating cyp19a1, cyp1a, and hsd17b7. This study reveals that both LZ and MT may inhibit the ovarian differentiation and induce masculinization through different regulatory mechanisms, which provides a fundamental insight into the sex control breeding in aquaculture. Full article
Show Figures

Figure 1

Figure 1
<p>The detailed procedure for the treatment of golden pompano by LZ or MT.</p>
Full article ">Figure 2
<p>Analysis of gene expression patterns in hypothalamus tissues of the golden pompano. (<b>A</b>) Number of DEGs. (<b>B</b>) Venn diagrams showing the distribution of DEGs under the influence of LZ and MT. (<b>C</b>,<b>D</b>) Heatmap indicates the expression levels of all DEGs in the Hy-A and the Hy-B groups throughout processing periods.</p>
Full article ">Figure 3
<p>Analysis of gene expression patterns in gonad tissues of the golden pompano. (<b>A</b>) Number of DEGs. (<b>B</b>) Venn diagrams showing the distribution of DEGs under the influence of LZ and MT. (<b>C</b>,<b>D</b>) Heatmap indicates the expression levels of all DEGs in the Hy-A and the Hy-B groups throughout processing periods.</p>
Full article ">Figure 4
<p>Analysis of gene expression patterns in liver tissues of the golden pompano. (<b>A</b>) Number of DEGs. (<b>B</b>) Venn diagrams showing the distribution of DEGs under the influence of LZ and MT. (<b>C</b>,<b>D</b>) Heatmap indicates the expression levels of all DEGs in the Hy-A and the Hy-B groups throughout processing periods.</p>
Full article ">Figure 5
<p>Volcano plots of gene expression differences. Red dots: up-regulation; blue dots: down-regulation; grey dots: normal-regulation. (<b>A</b>) Hypothalamus tissue transcriptome (LZ); (<b>B</b>) hypothalamus tissue transcriptome (MT); (<b>C</b>) ovarian tissue transcriptome (LZ); (<b>D</b>) ovarian tissue transcriptome (MT); (<b>E</b>) liver tissue transcriptome (LZ); (<b>F</b>) liver tissue transcriptome (MT).</p>
Full article ">Figure 6
<p>Functional annotation of DEGs based on GO classification. The horizontal axis indicates GO function and the vertical axis indicates the number of genes with GO function. (<b>A</b>) Pathway enrichment analysis of DEGs in the hypothalamus under the influence of LZ; (<b>B</b>) pathway enrichment analysis of DEGs in the hypothalamus under the influence of MT; (<b>C</b>) pathway enrichment analysis of DEGs in the gonad under the influence of LZ; (<b>D</b>) pathway enrichment analysis of DEGs in the gonad under the influence of MT; (<b>E</b>) pathway enrichment analysis of DEGs in the liver under the influence of LZ; (<b>F</b>) pathway enrichment analysis of DEGs in the liver under the influence of MT.</p>
Full article ">Figure 7
<p>DEGs are significantly enriched in KEGG pathways. (<b>A</b>) LZ affects the pathway enrichment analysis of DEGs in the hypothalamus. (<b>B</b>) Pathway enrichment analysis of DEGs in the MT-influenced hypothalamus. (<b>C</b>) Pathway enrichment analysis of DEGs in the LZ-influenced hypothalamus. (<b>D</b>) Pathway enrichment analysis of DEGs in the gonad under the influence of MT. (<b>E</b>) Pathway enrichment analysis of DEGs in the liver under the influence of LZ. (<b>F</b>) Pathway enrichment analysis of DEGs in the liver under the influence of MT. Pathways and enrichment factors are indicated on the vertical and horizontal axes, respectively. The size of the dots indicates the number of genes and the color indicates the q-value.</p>
Full article ">Figure 8
<p>(<b>A</b>) The expression profiles of 8 hypothalamus genes using qRT-PCR (LZ); (<b>B</b>) the expression profiles of 8 hypothalamus genes using qRT-PCR (MT); (<b>C</b>) the expression profiles of 10 ovarian genes using qRT-PCR (LZ); (<b>D</b>) the expression profiles of 9 ovarian genes using qRT-PCR (MT); (<b>E</b>) validation of the expression profiles of 9 liver genes using qRT-PCR (LZ); (<b>F</b>) validation of the expression profiles of 10 liver genes using qRT-PCR (MT).</p>
Full article ">Figure 9
<p>Reproduction-related DEGs under the influence of LZ.</p>
Full article ">Figure 10
<p>Reproduction-related DEGs under the influence of MT.</p>
Full article ">
19 pages, 10617 KiB  
Article
RNA-Seq and ATAC-Seq Reveal CYP26A1-Mediated Regulation of Retinoic Acid-Induced Meiosis in Chicken Primordial Germ Cells
by Zhaochuan Wang, Jiayi Chen, Jintian Wen, Siyu Zhang, Yantao Li, Jiali Wang and Zhenhui Li
Animals 2025, 15(1), 23; https://doi.org/10.3390/ani15010023 - 25 Dec 2024
Viewed by 441
Abstract
Retinoic acid (RA) plays a critical role in initiating meiosis in primordial germ cells (PGC), yet the specific mechanisms of its interaction with PGC remain unclear. In this study, we used an in vitro feeder-free culture system with chicken PGC as a model [...] Read more.
Retinoic acid (RA) plays a critical role in initiating meiosis in primordial germ cells (PGC), yet the specific mechanisms of its interaction with PGC remain unclear. In this study, we used an in vitro feeder-free culture system with chicken PGC as a model to explore the mechanisms by which RA induces the entry of PGC into meiosis. Results demonstrated that exogenous RA treatment altered the cell cycle distribution of PGC, significantly increasing the proportion of cells in the G1 phase and decreasing those in the G2 phase, suggesting that RA may promote the transition of PGC from proliferation to differentiation. Giemsa staining further revealed that chromosomes in a subset of RA-treated PGC exhibited meiotic characteristics. Through combined RNA-seq and ATAC-seq analyses, we identified that CYP26A1, a gene involved in RA degradation, was significantly upregulated in the RA-treated group, with enhanced accessibility in its chromatin regions. This finding suggests a robust mechanism for self-regulation of RA levels within PGC, indicating that CYP26A1 may play a pivotal role in the degradation of exogenous RA in chicken PGC. This study elucidated the effects of RA on chicken PGC and provided new insights into the role of RA in germ cell differentiation. Full article
(This article belongs to the Collection Current Advances in Poultry Research)
Show Figures

Figure 1

Figure 1
<p>Cultivation and identification of male and female primordial germ cells (PGC): (<b>A</b>) Male and female PGC after separation of chicken embryos; (<b>B</b>) Identification of PGC marker genes, where DF-1 cells were used as a control; (<b>C</b>) Photomicrograph of male and female PGC stained for alkaline phosphatase activity; DF-1 cells as a negative control result; (<b>D</b>) Photomicrograph of male and female PGC stained with Periodic Acid-Schiff (PAS) reagent; DF-1 cells as a negative control result; (<b>E</b>) SSEA1, DAPI, and merged plots of male and female PGC; Secondary antibodies were used as control. (<b>F</b>) EMA1, DAPI, and merged plots of male and female PGC; Secondary antibodies were used as control.</p>
Full article ">Figure 2
<p>Cell cycle analysis of primordial germ cells (PGC). (<b>A</b>) The cell cycle of PGC in the female control group; (<b>B</b>) Cell cycle of PGC in the female RA-treated group; (<b>C</b>) Bar chart of the cell cycle distribution of female PGC; (<b>D</b>) The cell cycle of PGC in the male control group; (<b>E</b>) Cell cycle of PGC in the male RA-treated group; (<b>F</b>) Bar chart of the cell cycle distribution of male PGC.</p>
Full article ">Figure 3
<p>Giemsa-stained image of primordial germ cells (PGC). (<b>A</b>) Morphology of normal PGC; (<b>B</b>) PGC entering the mitotic state; (<b>C</b>) PGC following mitotic; (<b>D</b>) PGC at the leptotene stage of meiosis; (<b>E</b>) PGC at the zygotene stage of meiosis; (<b>F</b>) PGC following meiosis.</p>
Full article ">Figure 4
<p>Sample relationship analysis in RNA-seq. (<b>A</b>) Principal component analysis plot between samples; (<b>B</b>) principal component analysis plot between samples; (<b>C</b>) heatmap of DEG in control and RA-treated groups in male; (<b>D</b>) heatmap of DEG in control and RA-treated groups in female.</p>
Full article ">Figure 5
<p>Analyses of the differentially expressed genes (DEG). (<b>A</b>) Volcano plot of DEG. M1 and F1 represent male and female RA treatment groups; M0 and F0 represent control groups of males and females. The screening threshold for DEG was |log2 FC| ≥ 1 and q value &lt; 0.05; (<b>B</b>) Venn diagram showing intersection of DEG in male and female PGC comparison groups; (<b>C</b>) annotation of GO functions in M1vsM0 group DEG; (<b>D</b>) KEGG pathway enrichment analysis in M1vsM0 group DEG; (<b>E</b>) annotation of GO functions in F1vsF0 group DEG; (<b>F</b>) KEGG pathway enrichment analysis in F1vsF0 group DEG.</p>
Full article ">Figure 6
<p>Analyses of the ATAC-seq. (<b>A</b>) Fragment length distribution map; (<b>B</b>) Principal component analysis plot. FN and MN are female and male control groups; FR and MR represent female and male RA treatment groups; (<b>C</b>) volcano map showing the different peaks between FN and FR; (<b>D</b>) volcano map showing the different peaks between MN and MR; (<b>E</b>) annotation of GO functions in different peaks between FN and FR; (<b>F</b>) KEGG pathway-enrichment analysis in different peaks between FN and FR.</p>
Full article ">Figure 7
<p>RNA-seq and ATAC-seq for combined analysis; (<b>A</b>) intersection of different mRNAs and different peaks corresponding to the target genes, F and M represent female and male comparison groups; (<b>B</b>) peak plots showing the relationship between genes in open regions of chromatin and gene expression, where the blue peak is the control group and the red peak is the RA treatment group; (<b>C</b>) validation of RNA-seq data by qRT-PCR. The left axis represents the relative expression levels determined by qRT-PCR and the right axis represents the FPKM values determined by RNA-seq. All data represent the mean of three biological replicates. The error line represents the standard error of the three replicates, and all data were normalized. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
22 pages, 7498 KiB  
Article
Recycling of Uridylated mRNAs in Starfish Embryos
by Haruka Yamazaki, Megumi Furuichi, Mikoto Katagiri, Rei Kajitani, Takehiko Itoh and Kazuyoshi Chiba
Biomolecules 2024, 14(12), 1610; https://doi.org/10.3390/biom14121610 - 16 Dec 2024
Viewed by 497
Abstract
In eukaryotes, mRNAs with long poly(A) tails are translationally active, but deadenylation and uridylation of these tails generally cause mRNA degradation. However, the fate of uridylated mRNAs that are not degraded quickly remains obscure. Here, using tail-seq and microinjection of the 3′ region [...] Read more.
In eukaryotes, mRNAs with long poly(A) tails are translationally active, but deadenylation and uridylation of these tails generally cause mRNA degradation. However, the fate of uridylated mRNAs that are not degraded quickly remains obscure. Here, using tail-seq and microinjection of the 3′ region of mRNA, we report that some mRNAs in starfish are re-polyadenylated to be translationally active after deadenylation and uridylation. In oocytes, uridylated maternal cyclin B mRNAs are stable without decay, and they are polyadenylated to be translated after hormonal stimulation to resume meiosis, whereas they are deadenylated and re-uridylated at the blastula stage, followed by decay. Similarly, deadenylated and uridylated maternal ribosomal protein mRNAs, Rps29 and Rpl27a, were stable and inactive after hormonal stimulation, but they had been polyadenylated and active before hormonal stimulation. At the morula stage, uridylated maternal ribosomal protein mRNAs were re-polyadenylated, rendering them translationally active. These results indicate that uridylated mRNAs in starfish exist in a poised state, allowing them to be recycled or decayed. Full article
(This article belongs to the Special Issue Gametogenesis and Gamete Interaction, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Cyclin B</span> mRNA uridylation and degradation. (<b>A</b>) Relative mRNA expression levels for <span class="html-italic">cyclin B</span> in starfish oocytes and embryos. Total RNAs were purified at the indicated time and relative quantification of <span class="html-italic">cyclin B</span> mRNA expression was conducted using RT-qPCR (median ± SEM) (<span class="html-italic">n</span> = 3). (<b>B</b>) Experimental scheme for the microinjection of artificially tagged <span class="html-italic">cyclin B</span> mRNA. Tag-labeled <span class="html-italic">cyclin B</span> mRNA was synthesized and injected (red arrowheads) into oocytes at 0.5 h after 1-MA treatment (+1-MA) or into oocytes without the 1-MA treatment (−1-MA). Oocytes (+1-MA) were inseminated before the first polar body formation, and the total RNAs were purified at the indicated time. (<b>C</b>) Tail length measurements for <span class="html-italic">cyclin B</span> mRNA. At the indicated time, the total RNAs were purified, and a TGIRT template-switching reaction was performed (<a href="#app1-biomolecules-14-01610" class="html-app">Supplementary Figure S1B, left panel</a>). RT-PCR was conducted using the 3′ adaptor reverse primer and <span class="html-italic">cyclin B</span>-specific forward primer. The PCR products were then subjected to polyacrylamide gel electrophoresis and visualized using SYBR-Green I staining. The left and right panels show changes in the tail lengths of endogenous <span class="html-italic">cyclin B</span> mRNA and exogenously microinjected <span class="html-italic">cyclin B</span> mRNAs, respectively. (<b>D</b>) Sanger sequencing results of the 3′ terminal region of cDNA from endogenous maternal <span class="html-italic">cyclin B</span> mRNA of oocytes at Pro-I without 1-MA treatment [Endogenous −1-MA], from stimulated oocytes following first polar body formation (1 h after 1-MA treatment) [Endogenous +1-MA 1 h], and from embryos at the blastula stage (12 h after 1-MA treatment) [Endogenous +1-MA 12 h]. The mean number ± SE of uridine residues in the five nucleotides from the 3′ end was 0.0 ± 0.0 for [Endogenous −1-MA], 2.6 ± 0.5 for [Endogenous +1-MA 1 h], and 1.4 ± 0.3 for [Endogenous +1-MA 12 h]. A Tukey HSD test revealed that the mean number in the [Endogenous −1-MA] group was significantly lower than those in the other two groups (<span class="html-italic">p</span> &lt; 0.05). (<b>E</b>) Sequencing results of the 3′ terminal region of exogenous <span class="html-italic">cyclin B</span> mRNA from stimulated oocytes following first polar body formation (1 h after 1-MA treatment) [Exogenous +1-MA] and from embryos at the blastula stage (12 h after 1-MA treatment) [Exogenous −1-MA].</p>
Full article ">Figure 2
<p>Deadenylation, uridylation, and non-canonical poly(A) elongation of the mRNA for ribosomal protein <span class="html-italic">Rps29</span>. (<b>A</b>) Relative levels of mRNA expression for the ribosomal protein in starfish oocytes and embryos. Total RNAs were purified at the indicated time and relative quantification of mRNA expression for the ribosomal protein <span class="html-italic">Rps29</span> was performed using RT-qPCR (median ± SEM) (<span class="html-italic">n</span> = 3). (<b>B</b>) Measurement of the <span class="html-italic">Rps29</span> mRNA tail lengths. Oocytes stimulated with 1-MA were inseminated following second polar body formation. At the indicated time before or after 1-MA stimulation, total RNAs were purified, and adaptor ligation was performed (<a href="#app1-biomolecules-14-01610" class="html-app">Supplementary Figure S1B, right panel</a>). RT-PCR was conducted using the 3′ adaptor reverse primer and <span class="html-italic">Rps29</span>-specific forward primer. The PCR products were then subjected to polyacrylamide gel electrophoresis and visualized using SYBR-Green I staining. Similar results were obtained for three animals. (<b>C</b>) Sequencing results of the 3′ terminal region of cDNA of endogenous maternal <span class="html-italic">Rps29</span> mRNA from oocytes at Pro-I without 1-MA treatment [Endogenous −1-MA], stimulated oocytes after the first polar body formation (1.5 h after 1-MA treatment) [Endogenous +1-MA 1.5 h], and embryos at the blastula stage (12 h after 1-MA treatment) [Endogenous +1-MA 12 h]. The mean number ± SE of uridine residues in the five nucleotides from the 3′ end was 0.0 ± 0.0 for [Endogenous −1-MA] (poly(A) length &gt; 40 residues), 1.9 ± 0.5 for [Endogenous +1-MA 1.5 h] and 0.4 ± 0.2 for [Endogenous +1-MA 12 h] (poly(A) length &gt; 40 residues). A Tukey HSD test revealed that the mean number in the [Endogenous −1-MA] group was significantly lower than the [Endogenous +1-MA 1.5 h] group (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Targeted TAIL-Seq of <span class="html-italic">Rps29</span> mRNA. (<b>A</b>) Distribution of poly(A) tail lengths of <span class="html-italic">Rps29</span> mRNA from oocytes without 1-MA treatment (−1-MA), with 1-MA treatment for 1.5 h (+1-MA 1.5 h), and with 1-MA treatment followed by insemination to obtain blastulae at 12 h. Relative frequencies (Y-axis, %) were calculated by dividing the number of detected reads that have indicated poly(A) lengths by the total number of reads that have poly(A) tails. Frequencies of poly(A) tail length &gt; 40 nucleotides are plotted on the right side. The number of reads is shown in parentheses. (<b>B</b>) Relative frequencies of the most frequent nucleotide with additional modifications at the 3′ end of the <span class="html-italic">Rps29</span> mRNA. Using each <span class="html-italic">Rps29</span> mRNA read, the most frequent nucleotides, such as U, G, and C, were determined. Relative frequencies (Y-axis, %) were calculated by dividing the number of reads with the most frequent nucleotides by the total number of reads with the indicated lengths of the poly(A) tails. mRNAs with tail lengths of ≤40 nucleotides and &gt;40 nucleotides were compared in the oocytes with or without 1-MA and at the embryonic stages of development. No modification, neither poly(A) tail nor additional modifications, were present at the end of the poly(A) tails. ≥2; two or more nucleotides comprised the most frequent nucleotides in the mRNA. (<b>C</b>) Distribution of the relative frequencies of non-A residues in the <span class="html-italic">Rps29</span> mRNA tails from blastulae and unstimulated oocytes. At the indicated position of the tails, the relative frequencies of the non-A residues (Y-axis, %) were calculated by dividing the number of reads carrying non-A residues by the total number of reads. The distribution of frequencies for the non-A residues is shown at the indicated position in the tails from 5′ to 3′ (left panel) and from 3′ to 5′ (right panel). The numbers of reads are shown in parentheses. (<b>D</b>) Relative frequencies of non-A residues in the non-canonical poly(A) tails of <span class="html-italic">Rps29</span> mRNA. The relative frequencies of the non-A residues (Y-axis, %) were calculated by dividing the number of each non-A residue (G, C, and U) in the tails of all reads by the number of tail lengths for all reads that have indicated lengths of poly(A) tails. mRNAs with tail lengths of ≤40 nucleotides and &gt;40 nucleotides are compared at the blastula stage.</p>
Full article ">Figure 4
<p>Deadenylation, uridylation, and non-canonical poly(A) elongation of the mRNA for exogenous ribosomal protein <span class="html-italic">Rps29</span>. (<b>A</b>) Experimental scheme. Tag-labeled <span class="html-italic">Rps29</span> mRNAs carrying long poly(A) tails were injected into oocytes with or without 1-MA treatment. Total RNAs were purified at the indicated times, whereas stimulated oocytes at 1 h and unstimulated oocytes were not inseminated. (<b>B</b>) Measurement of the tail length of endogenous and exogenous <span class="html-italic">Rps29</span> mRNA. RT-PCR was conducted using the 3′ adaptor reverse primer and <span class="html-italic">Rps29</span> mRNA-specific forward primer. The PCR products were subjected to polyacrylamide gel electrophoresis and SYBR-Green I staining. Similar results were obtained for two animals. (<b>C</b>) Sequencing results of exogenous <span class="html-italic">Rps29</span> mRNA from oocytes without 1-MA treatment [−1-MA], oocytes at 1 h after 1-MA treatment [+1-MA 1 h], and embryos at the morula stage (6 h after 1-MA treatment) [+1-MA 6 h]. ‘(30)’ in the sequences indicates ‘AA…AA’ containing 30 nucleotides. (<b>D</b>) Relative frequencies of the most frequent nucleotides in the additional modifications at the 3′ end of exogenous <span class="html-italic">Rps29</span> mRNA. Relative frequencies (Y-axis, %) were calculated by dividing the number of reads carrying the most frequent nucleotides by the total number of reads from oocytes and embryos at the indicated time. No modification, neither poly(A) tail nor additional modifications were present. “≥2”; two or more nucleotides comprised the most frequent nucleotides. (<b>E</b>) Distribution of the relative frequencies of non-A residues in tails of exogenous <span class="html-italic">Rps29</span> mRNA from oocytes and embryos. At the indicated position of the tails, the relative frequencies of the non-A residues (Y-axis, %) were calculated by dividing the number of reads carrying the non-A residues by the total number of reads. The distribution of frequencies of non-A residues is shown at the indicated position in tails from 5′ to 3′ (left panel) and from 3′ to 5′ (right panel). The numbers of reads are shown in parentheses. Yellow, morulae (5 h after 1-MA treatment). Red, blastulae (11 h after 1-MA treatment). Purple, Pro-I oocytes without 1-MA stimulation (2 h after injection). Blue, Pro-I oocytes without 1-MA stimulation (11 h after injection). (<b>F</b>) Relative frequencies of non-A residues in poly(A) tails of exogenous <span class="html-italic">Rps29</span> mRNA. The relative frequencies of the non-A residues (Y-axis, %) were calculated by dividing the number of each non-A residue (G, C, and U) in the tails of all reads by the number of tail lengths of all reads.</p>
Full article ">Figure 5
<p>Deadenylation, uridylation, and non-canonical poly(A) elongation of the mRNA for exogenous ribosomal protein ∆ PAS <span class="html-italic">Rps29</span>. (<b>A</b>) Experimental scheme. Tag-labeled wild-type and ∆ PAS <span class="html-italic">Rps29</span> mRNAs were injected into oocytes. At the indicated time, the total RNA was purified. (<b>B</b>) Measurement of the <span class="html-italic">Rps29</span> mRNA tail length. Total RNA was subjected to a TGIRT template-switching reaction. RT-PCR was conducted using the 3′ adaptor reverse primer and Tag-specific forward primer. The PCR products were then subjected to polyacrylamide electrophoresis and visualized using SYBR-Green I staining. The left and right panels show the changes in tail lengths for the exogenous wild-type and ∆ PAS <span class="html-italic">Rps29</span> mRNAs, respectively. Similar results were obtained for two animals. (<b>C</b>) Sequencing results of the 3′ terminal region of exogenous ∆ PAS <span class="html-italic">Rps29</span> mRNA purified from oocytes at Pro-I without 1-MA treatment at 2 h following injection of the mRNA [−1-MA 2 h], stimulated oocytes at 2 h following 1-MA treatment [+1-MA 2 h], and blastulae (11 h after 1-MA treatment) [+1-MA 11 h].</p>
Full article ">Figure 6
<p>Translational activities of the non-canonical poly(A) tails. (<b>A</b>) Experimental scheme. Tag(SGK)-labeled 40S <span class="html-italic">Rps29</span> mRNAs with canonical long poly(A) tails, short poly(A) tails, and non-canonical long and short poly(A) tails were synthesized and injected into oocytes without the 1-MA treatment. The table shows the number of A, T, G, and C nucleotides in the tails. (<b>B</b>) Western blotting of oocytes injected with mRNA carrying an <span class="html-italic">SGK</span> and non-canonical poly(A) tails of the 40S <span class="html-italic">Rps29</span> mRNA. At the indicated time after the injection of exogenous mRNA, oocytes were treated with a sample buffer, followed by polyacrylamide gel electrophoresis and Western blotting using an anti-SGK antibody. Arrowheads (endogenous) indicate endogenous SGK in oocytes. Similar results were obtained for 3 animals. (<b>C</b>) Experimental scheme. Reporter luciferase mRNA between the 5′ and 3′ UTR of 40S <span class="html-italic">Rps29</span> with a canonical long poly(A) tail was injected into oocytes with or without the 1-MA treatment. D, E. 1-MA-stimulated oocytes were inseminated after GVBD to start embryonic development and used to determine the reporter activities (<b>D</b>) (mean ± SE) (<span class="html-italic">n</span> = 3) and the length of poly(A) tails (<b>E</b>). To calculate the relative activity of translation, the luciferase activity of the unstimulated oocytes at 3 h after injection of the reporter mRNA was considered to be 100%, and Student’s t-test was used to determine the significance between the results observed after 3 and 11 h (<b>D</b>). Similar results were obtained for two animal models (<b>D</b>,<b>E</b>).</p>
Full article ">Figure 7
<p>Summary of the findings and proposed model. (<b>A</b>) Maternal mRNA deadenylation, uridylation, and non-canonical poly(A) elongation in starfish oocytes and embryos. Upper panel: Before hormonal stimulation of 1-MA, oocytes at Pro-I contain maternal ribosomal protein mRNAs (<span class="html-italic">Rps29</span> and <span class="html-italic">Rpl27a</span>) carrying long poly(A) tails and <span class="html-italic">cyclin B</span> mRNAs with uridylated short poly(A) tails. Following the resumption of meiosis of oocytes undergoing nuclear division of germinal vesicle breakdown, long poly(A) tails of ribosomal protein mRNAs are deadenylated and uridylated (newly added nucleotides are shown in larger font size). Some uridine residues of <span class="html-italic">cyclin B</span> mRNA are trimmed [<a href="#B26-biomolecules-14-01610" class="html-bibr">26</a>], followed by poly(A) elongation. After fertilization, uridylated short poly(A) tails of ribosomal proteins in morulae are re-elongated, forming non-canonical poly(A) tails. At the blastula stage, <span class="html-italic">cyclin B</span> mRNAs are deadenylated and uridylated. They are then degraded before gastrulation. Lower panel: Upper panel summarization. The arrow numbers correspond to those in the upper panel. Uridylated mRNAs are stable but inactive for translation. Canonical or non-canonical polyadenylation renders inactive mRNAs stably active for translation. Alternatively, uridylated mRNAs become unstable and inactive, followed by decay, as observed in other animals. (<b>B</b>) Proposed model for uridylation in starfish. Uridylated mRNAs are required to determine mRNA fate: destruction or recycling.</p>
Full article ">
22 pages, 1509 KiB  
Review
Mechanisms, Machinery, and Dynamics of Chromosome Segregation in Zea mays
by Marissa E. Duffy, Michael Ngaw, Shayna E. Polsky, Abby E. Marzec, Sean S. Zhang, Owen R. Dzierzgowski and Natalie J. Nannas
Genes 2024, 15(12), 1606; https://doi.org/10.3390/genes15121606 - 16 Dec 2024
Viewed by 960
Abstract
Zea mays (maize) is both an agronomically important crop and a powerful genetic model system with an extensive molecular toolkit and genomic resources. With these tools, maize is an optimal system for cytogenetic study, particularly in the investigation of chromosome segregation. Here, we [...] Read more.
Zea mays (maize) is both an agronomically important crop and a powerful genetic model system with an extensive molecular toolkit and genomic resources. With these tools, maize is an optimal system for cytogenetic study, particularly in the investigation of chromosome segregation. Here, we review the advances made in maize chromosome segregation, specifically in the regulation and dynamic assembly of the mitotic and meiotic spindle, the inheritance and mechanisms of the abnormal chromosome variant Ab10, the regulation of chromosome–spindle interactions via the spindle assembly checkpoint, and the function of kinetochore proteins that bridge chromosomes and spindles. In this review, we discuss these processes in a species-specific context including features that are both conserved and unique to Z. mays. Additionally, we highlight new protein structure prediction tools and make use of these tools to identify several novel kinetochore and spindle assembly checkpoint proteins in Z. mays. Full article
(This article belongs to the Special Issue Maize Molecular Genetics and Functional Genomics in 2024)
Show Figures

Figure 1

Figure 1
<p>Chromosome segregation machinery. The segregation machinery that separates chromosomes in mitosis and meiosis is the spindle, a microtubule-based structure. (<b>A</b>) Spindle microtubules attach to kinetochores, multi-protein structures that assemble on centromeres. The dashed box shows a zoomed in image of a kinetochore and includes the proteins identified in <span class="html-italic">Z. mays</span> (kinetochore proteins NDC80, MIS12, KNL1, CENPC and CENH3, and spindle checkpoint proteins MAD2, BUB1, and BUB3 that localize on the outer kinetochore). Replicated chromosomes contain two sister chromatids held together by cohesin until cleavage in mitotic anaphase or meiotic anaphase II. (<b>B</b>) In meiosis I, the spindle segregates homologous chromosomes, and in meiosis II and mitosis, the spindle segregates sister chromatids after cohesin is degraded. The dashed line indicates the plane of cell division.</p>
Full article ">Figure 2
<p><span class="html-italic">Zea mays</span> spindles. Live images of <span class="html-italic">Z. mays</span> spindles can be acquired due to the development of fluorescently tagged tubulin. In all images, B-tubulin was tagged with a florescent protein to facilitate imaging. (<b>A</b>) Maize mitotic spindle; microtubules are shown in white (image courtesy of Carolyn G. Rasmussen). (<b>B</b>) Maize meiosis I spindle; microtubules are shown in white. (<b>C</b>) Maize meiosis II spindles, microtubules are shown in white. There are two cells, each containing a meiotic spindle. (<b>D</b>) Time course of meiosis I spindle assembly (0–20 min), alignment of chromosomes on the metaphase I spindle (30 min), and anaphase I segregation of chromosomes (35–45 min). Microtubules are shown in green and chromosomes (labelled with SYTO12 DNA stain) are shown in pink. At time point 0 min, the nuclear envelope is still intact and microtubules can be seen encircling the nuclear membrane. At time point 45 min, the spindle has disassembled and the remaining microtubule structure between the separated chromosomes is the phragmoplast.</p>
Full article ">Figure 3
<p>Abnormal chromosome 10 (Ab10) structure and function in maize meiotic drive. (<b>A</b>) Ab10 differs from N10 due to the presence of heterochromatic repetitive DNA sequences, TR-1 (shown in red), and knob180 (shown in green). Ab10 also contains genes that code for the TRKIN and KINDR kinesins that associate with TR-1 and knob180 sequences, respectively. (<b>B</b>) The kinesins interact with the TR-1 and knob180 sequence to create neocentromere function capable of binding microtubules and pulling Ab10 chromosomes preferentially toward the developing egg cell. This neocentromere activity is functional before kinetochores are operational on centromeres, thus giving Ab10 chromosomes an advantage due to early interaction with spindle microtubules. (<b>C</b>) There are three distinct Ab10 types (type I, II, III) that can be cytologically distinguished from N10 due to the presence of TR-1 and knob180 sequences. K10L2 is another chromosome 10 variant that is unique from both N10 and Ab10. All variants show differing rates of meiotic drive, likely due to the difference the amount and location of knob DNA sequences.</p>
Full article ">
17 pages, 6800 KiB  
Article
Deletion of ddx4 Ovary-Specific Transcript Causes Dysfunction of Meiosis and Derepress of DNA Transposons in Zebrafish Ovaries
by Yuanyuan Chen, Xing Lin, Jing Dai, Yifan Bai, Fei Liu and Daji Luo
Biology 2024, 13(12), 1055; https://doi.org/10.3390/biology13121055 - 16 Dec 2024
Viewed by 703
Abstract
Alternative splicing of ddx4 (DEAD-box helicase 4), a key germline marker gene, has been reported to generate sex-specific transcripts in zebrafish gonads. The biological functions and regulatory mechanisms of the ddx4 ovary-specific transcript (ddx4-L) during oogenesis remain unclear. In this study, [...] Read more.
Alternative splicing of ddx4 (DEAD-box helicase 4), a key germline marker gene, has been reported to generate sex-specific transcripts in zebrafish gonads. The biological functions and regulatory mechanisms of the ddx4 ovary-specific transcript (ddx4-L) during oogenesis remain unclear. In this study, we found that ddx4-L mutants, in which ddx4-L was specifically deleted, had enlarged ovaries but laid fewer eggs, along with having a lower fertilization rate compared to WT controls. RNA-seq analysis was performed to detect the changes in gene expression between WT and ddx4-L mutant ovaries. A total of 524 upregulated and 610 downregulated DEGs were identified. GO and GSEA enrichment analyses showed that genes involved in fertilization and reproduction biological processes were significantly downregulated. More specifically, we observed a remarkable reduction in Sycp1, a core component of synaptonemal complex, in ddx4-L mutant ovaries at both the mRNA and protein levels. In addition, the expressions of transposon elements, as well as the events of alternative splicing, alternative polyadenylation, and RNA editing, were analyzed based on the RNA-seq data. We found that the deletion of ddx4-L resulted in derepression of DNA transposons in zebrafish ovaries, possibly causing genome instability. In conclusion, our work demonstrates that the ovary-specific ddx4 transcript plays important roles in oocyte meiosis and DNA transposon repression, which extends our understanding of the biological functions and regulatory mechanisms of sex-specific alternative splicing in zebrafish oogenesis and reproduction. Full article
(This article belongs to the Special Issue The Biology of Animal Reproduction)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p><span class="html-italic">ddx4-L</span> knockout causes reduced fecundity of female zebrafish. (<b>A</b>) Alternative splicing patterns of <span class="html-italic">ddx4</span> in WT and ddx4-E4Δ zebrafish ovaries are shown. (<b>B</b>) Abdominal morphology and secondary sexual characteristics of WT and ddx4-E4Δ zebrafish at 9 mpf. The insets show the pectoral fin of zebrafish. Red dotted frames indicate the position of ovaries. n = 7. Scale bars: 2000 μm. (<b>C</b>) Overall ovarian morphology of WT and ddx4-E4Δ zebrafish at 9 mpf. The red arrows indicate examples of mature eggs, and the blue arrows indicate examples of immature eggs. n = 7. Scale bars: 500 μm. (<b>D</b>) Hematoxylin and eosin (HE) staining of WT and ddx4-E4Δ ovarian sections at 9 mpf. The green arrows indicate early-stage oocytes, which have large, spherical nuclei stained blue or purple due to the high affinity of hematoxylin. n = 7. Scale bars: 200 μm. (<b>E</b>) Ovarian weights of WT and ddx4-E4Δ zebrafish at 9 mpf are shown. n = 7. (<b>F</b>) GSI of WT and ddx4-E4Δ females at 9 mpf are shown. n = 7. (<b>G</b>) Embryos produced by WT and ddx4-E4Δ zebrafish are shown. The arrows indicate the unfertilized embryos. (<b>H</b>) Fertilization rates of spawning eggs were quantified for WT and ddx4-E4Δ zebrafish. (<b>I</b>) The number of eggs produced by a single female during a spawning cycle was quantified. The ddx4-E4Δ females laid fewer eggs than WT controls. n = 6.</p>
Full article ">Figure 2
<p>RNA sequencing of WT and ddx4-E4Δ zebrafish ovaries. (<b>A</b>) The workflow of RNA sequencing and bioinformatics analysis. Three biological replicates were employed in each experimental group. (<b>B</b>) Quality control of the RNA-seq raw data. The mapping rate of each sample is shown. (<b>C</b>) PCA analysis of WT and ddx4-E4Δ samples. (<b>D</b>) Correlation heatmap of WT and ddx4-E4Δ samples.</p>
Full article ">Figure 3
<p>Identification and functional enrichment analysis of DEGs between WT and ddx4-E4Δ zebrafish ovaries. (<b>A</b>) The volcano plot shows the 524 upregulated and 610 downregulated genes in ddx4-E4Δ ovaries. (<b>B</b>) The heatmap shows the expression pattern of DEGs between WT and ddx4-E4Δ ovaries. (<b>C</b>) Enriched GO terms in all, upregulated, and downregulated DEGs, respectively. (<b>D</b>) GSEA analysis shows the global upregulation of genes involved in reproduction biological process. (<b>E</b>) The expression pattern of upregulated genes in the reproduction biological process term. (<b>F</b>) The expression pattern of downregulated genes in the reproduction biological process term. The purple box indicates the significantly down-regulated meiosis-related gene <span class="html-italic">sycp1</span>.</p>
Full article ">Figure 4
<p><span class="html-italic">ddx4-L</span> knockout leads to significant downregulation of <span class="html-italic">sycp1</span> at mRNA and protein levels. (<b>A</b>) Expression levels of several meiosis-related genes in WT and ddx4-E4Δ ovaries based on the RNA-seq data. The FPKM values are shown. n = 3. (<b>B</b>) qPCR validation of the mRNA levels of meiosis-related genes in WT and ddx4-E4Δ ovaries at 6 mpf. n = 3. (<b>C</b>) Detection of Sycp1 protein levels in WT and ddx4-E4Δ ovaries at 6 mpf by Western blot assay. RNA-seq tracks of <span class="html-italic">sycp1</span> gene in WT and ddx4-E4Δ ovaries are shown in the top panel. Original western blot images are shown in <a href="#app1-biology-13-01055" class="html-app">Figure S1A</a>. (<b>D</b>) Quantitative analysis of the bands in (<b>C</b>) shows the changes in Sycp1 protein levels in ddx4-E4Δ ovaries. n = 3.</p>
Full article ">Figure 5
<p>Identification of TEs and quantitative analysis of TE expression in WT and ddx4-E4Δ zebrafish ovaries. (<b>A</b>) The percentages of RNA-seq reads belonging to TEs in WT and ddx4-E4Δ ovaries. (<b>B</b>) Count numbers of different subcategories of TEs identified in WT and ddx4-E4Δ ovaries by RNA-seq. (<b>C</b>) Count numbers of DNA transposon subclasses in WT and ddx4-E4Δ ovaries.</p>
Full article ">Figure 6
<p>Identification of differential alternative splicing events between WT and ddx4-E4Δ zebrafish ovaries. (<b>A</b>) Five types of alternative splicing events analyzed in this study are shown. (<b>B</b>) The numbers of DASEs for each AS type between WT and ddx4-E4Δ ovaries are shown. (<b>C</b>) RNA-seq tracks of <span class="html-italic">aak1a</span>, <span class="html-italic">ndufv3</span>, and <span class="html-italic">sh3d21</span> are shown. The alternative exons are indicated with boxes. (<b>D</b>) RT-PCR validation of the three selected DASEs. Original gel images are shown in <a href="#app1-biology-13-01055" class="html-app">Figure S1B</a>. (<b>E</b>) The differentially spliced exons affected by <span class="html-italic">ddx4-L</span> knockout have relatively shorter length, higher GC contents, similar 5′ splicing sites, and stronger 3′ splicing sites.</p>
Full article ">Figure 7
<p>Identification of APA events between WT and ddx4-E4Δ zebrafish ovaries. (<b>A</b>) A plot of PDUI score of each gene in WT and ddx4-E4Δ groups. Grey dots indicate APA events showing no difference between WT and ddx4-E4Δ ovaries. (<b>B</b>) A volcano plot denoting 3′UTR-shorterned (blue) and -lengthened (red) gene hits. Grey dots indicate APA events showing no difference between WT and ddx4-E4Δ ovaries. (<b>C</b>) Heatmap of proximal and distal PAS usage. (<b>D</b>) Expression profile of key APA factors. (<b>E</b>) Log-fold change in gene expression is plotted against ΔPDUI for 3′UTR-altered genes. Grey dots indicate differential APA events that do not affect gene expression.</p>
Full article ">Figure 8
<p>Identification of RNA editing events in WT and ddx4-E4Δ zebrafish ovaries. (<b>A</b>) Percentages of A-to-I, C-to-T, and other types of RNA editing events. (<b>B</b>) Proportion of A-to-I RNA editing events. (<b>C</b>) Distribution of RNA editing events on each chromosome. The RNA editing levels are shown with red bars. (<b>D</b>) The number of RNA editing sites on unite chromosome length of 25 chromosomes. (<b>E</b>) Distribution of the RNA editing sites in different regions of genes in WT and ddx4-E4Δ ovaries. (<b>F</b>) Functional consequences of the RNA editing sites located in CDSs.</p>
Full article ">
17 pages, 2478 KiB  
Review
Genetic Abnormalities of Oocyte Maturation: Mechanisms and Clinical Implications
by Giorgio Maria Baldini, Daniele Ferri, Antonio Malvasi, Antonio Simone Laganà, Antonella Vimercati, Miriam Dellino, Domenico Baldini and Giuseppe Trojano
Int. J. Mol. Sci. 2024, 25(23), 13002; https://doi.org/10.3390/ijms252313002 - 3 Dec 2024
Viewed by 2451
Abstract
Genetic anomalies in oocyte maturation present significant fertility and embryonic development challenges. This review explores the intricate mechanisms of nuclear and cytoplasmic maturation, emphasizing the genetic and molecular factors contributing to oocyte quality and competence. Chromosomal mutations, errors in segregation, genetic mutations in [...] Read more.
Genetic anomalies in oocyte maturation present significant fertility and embryonic development challenges. This review explores the intricate mechanisms of nuclear and cytoplasmic maturation, emphasizing the genetic and molecular factors contributing to oocyte quality and competence. Chromosomal mutations, errors in segregation, genetic mutations in signaling pathways and meiosis-related genes, and epigenetic alterations are discussed as critical contributors to oocyte maturation defects. The role of mitochondrial defects, maternal mRNA dysregulation, and critical proteins such as NLRP14 and BMP6 are highlighted. Understanding these genetic factors is crucial for improving diagnostic approaches and therapeutic interventions in reproductive medicine, particularly for couples encountering recurrent in vitro fertilization failures. This review will explore how specific genetic mutations impact fertility treatments and reproductive success by examining the intricate oocyte maturation process. We will focus on genetic abnormalities that may disrupt the oocyte maturation pathway, discussing the underlying mechanisms involved and considering their potential clinical implications for enhancing fertility outcomes. Full article
(This article belongs to the Special Issue Gynecologic Diseases: From Molecular Basis to Therapy)
Show Figures

Figure 1

Figure 1
<p>Illustration showing the structure of an oocyte surrounded by corona radiata and cumulus oophorus cells, highlighting critical genes involved in oocyte maturation and fertility, with their chromosomal locations.</p>
Full article ">Figure 2
<p>Epigenetics mechanism in oocyte defect. Mechanism of methylation through DNA alterations and epigenetic damage via histones on accessible genes.</p>
Full article ">Figure 3
<p>Mitochondrial and mtDNA defect, decrease in oocyte quality, through alterations of some genes by ROS.</p>
Full article ">
31 pages, 4693 KiB  
Review
Decoding the Nucleolar Role in Meiotic Recombination and Cell Cycle Control: Insights into Cdc14 Function
by Paula Alonso-Ramos and Jesús A. Carballo
Int. J. Mol. Sci. 2024, 25(23), 12861; https://doi.org/10.3390/ijms252312861 - 29 Nov 2024
Viewed by 745
Abstract
The cell cycle, essential for growth, reproduction, and genetic stability, is regulated by a complex network of cyclins, Cyclin-Dependent Kinases (CDKs), phosphatases, and checkpoints that ensure accurate cell division. CDKs and phosphatases are crucial for controlling cell cycle progression, with CDKs promoting it [...] Read more.
The cell cycle, essential for growth, reproduction, and genetic stability, is regulated by a complex network of cyclins, Cyclin-Dependent Kinases (CDKs), phosphatases, and checkpoints that ensure accurate cell division. CDKs and phosphatases are crucial for controlling cell cycle progression, with CDKs promoting it and phosphatases counteracting their activity to maintain balance. The nucleolus, as a biomolecular condensate, plays a key regulatory role by serving as a hub for ribosome biogenesis and the sequestration and release of various cell cycle regulators. This phase separation characteristic of the nucleolus is vital for the specific and timely release of Cdc14, required for most essential functions of phosphatase in the cell cycle. While mitosis distributes chromosomes to daughter cells, meiosis is a specialized division process that produces gametes and introduces genetic diversity. Central to meiosis is meiotic recombination, which enhances genetic diversity by generating crossover and non-crossover products. This process begins with the introduction of double-strand breaks, which are then processed by numerous repair enzymes. Meiotic recombination and progression are regulated by proteins and feedback mechanisms. CDKs and polo-like kinase Cdc5 drive recombination through positive feedback, while phosphatases like Cdc14 are crucial for activating Yen1, a Holliday junction resolvase involved in repairing unresolved recombination intermediates in both mitosis and meiosis. Cdc14 is released from the nucleolus in a regulated manner, especially during the transition between meiosis I and II, where it helps inactivate CDK activity and promote proper chromosome segregation. This review integrates current knowledge, providing a synthesis of these interconnected processes and an overview of the mechanisms governing cell cycle regulation and meiotic recombination. Full article
(This article belongs to the Special Issue Cell Division: A Focus on Molecular Mechanisms)
Show Figures

Figure 1

Figure 1
<p>Cell cycle in eukaryotic cells. The four phases of the cell cycle, G<sub>1</sub>, S, G<sub>2</sub>, and M, and the main functions that take place in them are represented. The main checkpoints are also shown, as well as the main cyclins that act during the cycle.</p>
Full article ">Figure 2
<p>Diagram of the phases of meiosis and their regulation in <span class="html-italic">S. cerevisiae</span>. The nucleolus is represented in pink, the nucleus is in blue, and the SC is represented by its Zip1 component in yellow. The SPBs are represented by a blue square and the tubulin by a yellow rectangle. Arrows represent promotion of processes, while T-bars indicate prevention or inhibition.</p>
Full article ">Figure 3
<p>Variation in CDK activity levels of both S phase CDKs (S-CDKs) and M phase CDKs (M-CDKs). The arrows indicate the progression of meiosis, together with the cyclins that are acting at each point in time.</p>
Full article ">Figure 4
<p>Diagram of the structure of the synaptonemal complex.</p>
Full article ">Figure 5
<p>Models of nucleolus organization: (<b>A</b>) organization of the nucleolus of an <span class="html-italic">S. cerevisiae</span>; (<b>B</b>) organization of the nucleolus of a eukaryotic cell. The three subcompartments of the nucleolus are depicted: the fibrillar center, the dense fibrillar component, and the granular component.</p>
Full article ">Figure 6
<p>Schematic of the three models of organization of the membrane-less subcompartments. The nucleolus can be explained as a combination of the PPPS and LLPS model.</p>
Full article ">Figure 7
<p>Schematic representation of the recombination process in <span class="html-italic">S. cerevisiae</span>.</p>
Full article ">Figure 8
<p>Domains and structure of Cdc14 phosphatase. (<b>A</b>) Schematic representation of the structure of Cdc14, with its main domains and binding sites. The phosphatase domain, the NES (nuclear export signal) and NLS (nuclear localization signal) region, and the 408,494,517 phosphorylation sites are shown. (<b>B</b>) Alphafold prediction model for Cdc14. Colors indicate the different functional motifs described in (<b>A</b>).</p>
Full article ">Figure 9
<p>Role of Cdc14 in mitosis. The FEAR network initiates the early release of Cdc14, followed by the MEN pathway, which regulates its final release, enabling the dephosphorylation events crucial for completing mitosis. The large blue circle represents the nucleolus, with colors indicating key proteins involved in repressing Cdc14 release. The figure illustrates the two-wave pathway required for Cdc14 release, showing how the first wave also activates the MEN network. The right part displays how Cdc14, through Cdh1, Sic1, and Swi5, inactivates CDKs. The bottom of the figure shows how different mitotic phases correspond with the two waves of Cdc14 release.</p>
Full article ">Figure 10
<p>Schematic of Cdc14 regulation during mitosis via the FEAR and MEN pathways. The figure shows the early, partial release of Cdc14 from the nucleolus (blue circle) by the FEAR network, followed by its complete release through the MEN pathway. Proteins are depicted as circles, while proteins requiring a cofactor are represented as squares divided into two binding parts. The figure also illustrates CDK phosphorylation stages during Cdc14 release and the active or inactive states of Cdc14 throughout different mitotic phases.</p>
Full article ">Figure 11
<p>Schematic representation of Cdc14 release during the first and second meiotic divisions. The figure shows the key regulators and inhibitors required for Cdc14 release in meiosis I (top panel), along with variations in cyclin levels. Arrows indicate increases or decreases in cyclin levels, with multiple arrows representing a stronger effect. An oblique arrow pointing up denotes a progressive increase in cyclin levels, while the “=” symbol indicates no change. These adjustments lead to a downregulation of CDK activity. In meiosis II (bottom panel), the role of the FEAR pathway in Cdc14 release is uncertain (noted as FEAR??). Cyclin level changes are similarly depicted, illustrating how they contribute to CDK activity downregulation.</p>
Full article ">
12 pages, 2210 KiB  
Article
Clinical and Cytogenetic Impact of Maternal Balanced Double Translocation: A Familial Case of 15q11.2 Microduplication and Microdeletion Syndromes with Genetic Counselling Implications
by Daniela Koeller R. Vieira, Ingrid Bendas Feres Lima, Carla Rosenberg, Carlos Roberto da Fonseca, Leonardo Henrique Ferreira Gomes, Letícia da Cunha Guida, Patrícia Camacho Mazzonetto, Juan Llerena and Elenice Ferreira Bastos
Genes 2024, 15(12), 1546; https://doi.org/10.3390/genes15121546 - 29 Nov 2024
Viewed by 824
Abstract
Background: Balanced chromosomal translocations occur in approximately 0.16 to 0.20% of live births. While most carriers are phenotypically normal, they are at risk of generating unbalanced gametes during meiosis, leading to genetic anomalies such as aneuploidies, deletions, duplications, and gene disruptions. These anomalies [...] Read more.
Background: Balanced chromosomal translocations occur in approximately 0.16 to 0.20% of live births. While most carriers are phenotypically normal, they are at risk of generating unbalanced gametes during meiosis, leading to genetic anomalies such as aneuploidies, deletions, duplications, and gene disruptions. These anomalies can result in spontaneous abortions or congenital anomalies, including neurodevelopmental disorders. Complex chromosomal rearrangements (CCRs) involving more than two chromosomes are rare but further increase the probability of producing unbalanced gametes. Neurodevelopmental disorders such as Angelman syndrome (AS) and duplication 15q11q13 syndrome (Dup15q) are associated with such chromosomal abnormalities. Methods: This study describes a family with a de novo maternal balanced double translocation involving chromosomes 13, 19, and 15, resulting in two offspring with unbalanced chromosomal abnormalities. Cytogenetic evaluations were performed using GTG banding, fluorescence in situ hybridization (FISH), and low-pass whole-genome sequencing (LP-WGS). Methylation analysis was conducted using methylation-sensitive high-resolution melting (MS-HRM) to diagnose Angelman syndrome. Results: The cytogenetic and molecular analyses identified an 8.9 Mb duplication in 15q11.2q13.3 in one child, and an 8.9 Mb deletion in the same region in the second child. Both abnormalities affected critical neurodevelopmental genes, such as SNRPN. FISH and MS-HRM confirmed the chromosomal imbalances and the diagnosis of Angelman syndrome in the second child. The maternal balanced translocation was found to be cryptic, contributing to the complex inheritance pattern. Conclusion: This case highlights the importance of using multiple genetic platforms to uncover complex chromosomal rearrangements and their impact on neurodevelopmental disorders. The findings underscore the need for thorough genetic counseling, especially in families with such rare chromosomal alterations, to manage reproductive outcomes and neurodevelopmental risks. Full article
(This article belongs to the Section Genetic Diagnosis)
Show Figures

Figure 1

Figure 1
<p>Duplication and deletion of 8.9 Mb on 15q11.2q13.3 identified by LP-WGS. (<b>A</b>). Proband duplication characterizing 15q11q13 microduplication syndrome. (<b>B</b>) Proband’s sister deletion characterizing Angelman syndrome.</p>
Full article ">Figure 2
<p>Methylation pattern of proband’s sister and proband, analyzed by MS-HRM. Amplifications plot related to normal (<b>A</b>), and Angelman syndrome (<b>B</b>). Normalized graphs (<b>C</b>,<b>D</b>) show initial fluorescence, where all products are double-stranded and bound to the maximum amount of dye. Normal patients present fluorescence drops corresponding to both paternal and maternal alleles (<b>B</b>). As the temperature rose, PCR products dissociated, releasing the dye and decreasing the fluorescent signal. The temperature differences between paternal and maternal alleles are attributed to the CpG binding chemistry. Methylated cytosines are nonreactive to bisulfite conversion while nonmethylated cytosines convert to uracil. CpG-rich regions require higher dissociation temperatures. The melting temperature detected for methylated maternal allele was 84.07 °C, and 79.5 °C for the nonmethylated paternal allele. Derivative graphs (<b>E</b>,<b>F</b>) illustrate the melting peak of each allele. The normal patient in derivative graphs (<b>E</b>) presents two peaks corresponding to the unmethylated (79.5 °C) and methylated (84.07 °C) alleles. The absence of maternal allele confirms Angelman syndrome (<b>D</b>,<b>F</b>). The different colors indicate experimental triplicates.</p>
Full article ">Figure 3
<p>(<b>A</b>) Proband GTG analysis—Karyotype: 46,XX. (<b>B</b>) The FISH technique using Prader–Willi/Angelman region probe (<span class="html-italic">SNRPN</span>) demonstrating three red signals corresponding to 15q11.2 region in the proband. The arrow indicates the third red signal, confirming triplicate of the 15q11.2 region. (<b>C</b>) Proband´s sister analysis by GTG banding—Karyotype: 46,XX, t(13;19)(q22;p13.1). (<b>D</b>) The FISH analysis using Prader–Willi/Angelman region probe <span class="html-italic">(SNRPN</span>) demonstrating deletion of 15q11.2 region in proband’s sister.</p>
Full article ">Figure 4
<p>(<b>A</b>) Mother’s karyotype by GTG banding—46,XX, t(13;19)(q22;p13.1) (<b>B</b>) Partial mother’s karyotype demonstrating translocation t(13;19) with breakpoints (arrows) (<b>C</b>) Mother’s FISH analysis using Prader–Willi/Angelman region probe (SNRPN) and chromosome 13 centromeric probe (D13Z1) showing a balanced translocation between the other homologue of chromosome 13 with one of chromosomes 15 (.ish t(13;15)(q12?;q12?) identified two green signals (D13Z1 and 15qter) at the same chromosome (derivative chromosome 13), a normal chromosome 15 (both signals red and green), and a derivative chromosome 15 showing only the red signal (15q11.2). The chromosome marked only with centromeric probe (D13Z1) is the derivative of t(13;19).</p>
Full article ">Figure 5
<p>Three-generation pedigree. Shaded symbols denote affected individuals (III-4 = case 1 and III-5 = case 2), and stripe-patterned symbols indicate carriers of balanced translocations. Individuals I-2, I-3, II-3, II-6, and III-2 have a normal karyotype and/or FISH findings, with III-2 clinically diagnosed with Marfan syndrome and a variant of uncertain significance at seq[GRCh38] 15q13.3(31739865_32239864)x3, which is likely benign. The proband is marked with an arrow.</p>
Full article ">
16 pages, 9375 KiB  
Article
The Loss of Tafazzin Transacetylase Activity Is Sufficient to Drive Testicular Infertility
by Paige L. Snider, Elizabeth A. Sierra Potchanant, Catalina Matias, Donna M. Edwards, Jeffrey J. Brault and Simon J. Conway
J. Dev. Biol. 2024, 12(4), 32; https://doi.org/10.3390/jdb12040032 - 26 Nov 2024
Viewed by 688
Abstract
Barth syndrome (BTHS) is a rare, infantile-onset, X-linked mitochondriopathy exhibiting a variable presentation of failure to thrive, growth insufficiency, skeletal myopathy, neutropenia, and heart anomalies due to mitochondrial dysfunction secondary to inherited TAFAZZIN transacetylase mutations. Although not reported in BTHS patients, male infertility [...] Read more.
Barth syndrome (BTHS) is a rare, infantile-onset, X-linked mitochondriopathy exhibiting a variable presentation of failure to thrive, growth insufficiency, skeletal myopathy, neutropenia, and heart anomalies due to mitochondrial dysfunction secondary to inherited TAFAZZIN transacetylase mutations. Although not reported in BTHS patients, male infertility is observed in several Tafazzin (Taz) mouse alleles and in a Drosophila mutant. Herein, we examined the male infertility phenotype in a BTHS-patient-derived D75H point-mutant knockin mouse (TazPM) allele that expresses a mutant protein lacking transacetylase activity. Neonatal and adult TazPM testes were hypoplastic, and their epididymis lacked sperm. Histology and biomarker analysis revealed TazPM spermatogenesis is arrested prior to sexual maturation due to an inability to undergo meiosis and the generation of haploid spermatids. Moreover, TazPM testicular mitochondria were found to be structurally abnormal, and there was an elevation of p53-dependent apoptosis within TazPM seminiferous tubules. Immunoblot analysis revealed that TazPM gamete genome integrity was compromised, and both histone γ-H2Ax and Nucleoside diphosphate kinase-5 protein expression were absent in juvenile TazPM testes when compared to controls. We demonstrate that Taz-mediated transacetylase activity is required within mitochondria for normal spermatogenesis, and its absence results in meiotic arrest. We hypothesize that elevated TazPM spermatogonial apoptosis causes azoospermia and complete infertility. Full article
(This article belongs to the Special Issue Feature Papers in Journal of Developmental Biology 2025)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Tafazzin</span> mRNA expression during postnatal <span class="html-italic">wt</span> mouse testis growth. (<b>A</b>) Quantitative triplicate PCR revealed <span class="html-italic">Taz</span> levels are robust in newborn, P5, P10 and P16 testes (averaging ~3.67-fold), but is expressed at basal levels in P21, P28 and adult (4-month) testes. <span class="html-italic">Taz</span> expression was compared to housekeeping gene <span class="html-italic">Ppia</span>. qPCR data are presented as a logarithmic plot of relative expression, where a value of 1 indicates no difference in 4-month-old adult testes and values &lt; 1 indicate reduced and &gt;1 indicate increased expression. The Y-axis is the relative fold difference, and error bars represent SD. (<b>B</b>–<b>E</b>) Non-radioactive in situ hybridization detection of <span class="html-italic">Taz</span> (purple precipitate) in staged postnatal mouse testis sections revealed punctate robust expression within nascent seminiferous tubules at P5 (<b>B</b>), P16 (<b>C</b>) and P21 (<b>D</b>), but only low-level expression in P28 seminiferous tubules (<b>E</b>). <span class="html-italic">Taz</span> signal was only observed when sections were hybridized with the <span class="html-italic">Taz</span> anti-sense probe, confirming signal specificity. Serial sections were examined for comparable spatiotemporal patterns in at least three consecutive serial sections/stage. Scale bars (<b>B</b>–<b>E</b>) = 50 μm.</p>
Full article ">Figure 2
<p>Characterization of <span class="html-italic">Taz<sup>PM</sup></span> testis phenotype. (<b>A</b>) Adult 4-month testis/body weight ratio in <span class="html-italic">wt</span> (black, n = 4) verses <span class="html-italic">Taz<sup>PM</sup></span> knockin (red, n = 6) mice. (<b>B</b>–<b>G</b>) Representative images of whole <span class="html-italic">wt</span> (<b>B</b>,<b>D</b>,<b>F</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>C</b>,<b>E</b>,<b>G</b>) 4-month (<b>B</b>,<b>C</b>), P17 (<b>D</b>,<b>E</b>) and P5 (<b>F</b>,<b>G</b>) littermate testes. (<b>H</b>–<b>O</b>) Hematoxylin and eosin staining of <span class="html-italic">wt</span> (<b>H</b>,<b>J</b>,<b>L</b>,<b>N</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>I</b>,<b>K</b>,<b>M</b>,<b>O</b>) testes (<b>H</b>,<b>I</b>) and epididymis (<b>J</b>,<b>K</b>) from 4-month-old mice, as well as P28 (<b>L</b>,<b>M</b>) and P17 (<b>N</b>,<b>O</b>) testis sections. (<b>P</b>,<b>Q</b>) Analysis of 4-month epidydimal <span class="html-italic">wt</span> and <span class="html-italic">Taz<sup>PM</sup></span> content numbers ((<b>P</b>), n = 7/genotype) and resultant staining of sperm head nuclei DNA chromatin (blue DAPI-positive), mitochondria-rich midpiece (green MitoTracker-positive) and tail (red actin-positive) within contents. (<b>R</b>,<b>S</b>) Oil red-O staining of lipid deposition in P28 <span class="html-italic">wt</span> (<b>R</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>S</b>) testes. Scale bars (<b>B</b>,<b>C</b>,<b>E</b>) = 1 mm; (<b>D</b>,<b>F</b>,<b>G</b>) = 500 μm; (<b>H</b>,<b>I</b>,<b>K</b>,<b>R</b>,<b>S</b>) = 20 μm; (<b>J</b>,<b>K</b>) = 50 μm; (<b>L</b>,<b>M</b>,<b>N</b>,<b>O</b>,<b>Q</b>) = 10 μm. Statistical significance set at **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Hook1 and Dazl biomarker analysis. (<b>A</b>–<b>F</b>) Immunostaining using Hook1 antibody (signal is brown DAB precipitate) expression in 4-month (<b>A</b>,<b>B</b>), P17 (<b>C</b>,<b>D</b>) and P5 (<b>E</b>,<b>F</b>) <span class="html-italic">wt</span> (<b>A</b>,<b>C</b>,<b>E</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>B</b>,<b>D</b>,<b>F</b>) testis sections. (<b>G</b>–<b>L</b>) Immunostaining using Dazl antibody (signal is brown DAB precipitate) expression in 4-month (<b>G</b>,<b>H</b>), P17 (<b>I</b>,<b>J</b>) and P5 (<b>K</b>,<b>L</b>) <span class="html-italic">wt</span> (<b>G</b>,<b>I</b>,<b>K</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>H</b>,<b>J</b>,<b>L</b>) testis sections. Scale bars (<b>A</b>,<b>B</b>,<b>E</b>,<b>F</b>,<b>K</b>,<b>L</b>) = 20 μm; (<b>C</b>,<b>D</b>,<b>G</b>,<b>H</b>,<b>I</b>,<b>J</b>) = 50 μm.</p>
Full article ">Figure 4
<p>Molecular analysis of <span class="html-italic">Taz<sup>PM</sup></span> testis phenotype. (<b>A</b>,<b>B</b>) Western evaluation of Taz and Vdac protein levels in triplicate <span class="html-italic">wt</span> and <span class="html-italic">Taz<sup>PM</sup></span> P28 testes ((<b>A</b>), n = 3 per genotype). Both mitochondria Hsp70 and housekeeping Gapdh were used as independent loading controls, with Taz; Gapdh quantification and statistical analysis (via two-tailed t-test) shown (<b>B</b>). The Y-axis is the relative fold difference. (<b>C</b>,<b>D</b>) Representative electron microscopy images showing P28 <span class="html-italic">wt</span> normal (<b>C</b>) and <span class="html-italic">Taz<sup>PM</sup></span> swollen/abnormal (<b>D</b>) mitochondrial morphology (n = 4/genotype). (<b>E</b>) Ultra-performance liquid chromatography analysis (n = 5/genotype) revealed that ATP, ADP, AMP, and NAD<sup>+</sup> levels as well as ATP/ADP and ATP/AMP ratios remain unchanged, but total adenine nucleotides (Ad Sum) are reduced (<span class="html-italic">p</span> = 0.05) in <span class="html-italic">Taz<sup>PM</sup></span> (red) verses <span class="html-italic">wt</span> (black) testes (n = 5/genotype). Scale bars (<b>C</b>,<b>D</b>) = 500 nm.</p>
Full article ">Figure 5
<p>Spermatogenic failure in <span class="html-italic">Taz<sup>PM</sup></span> testes. (<b>A</b>–<b>D</b>) Non-radioactive in situ hybridization detection of <span class="html-italic">Catsper1</span> (<b>A</b>,<b>B</b>) and <span class="html-italic">Catsper4</span> (<b>C</b>,<b>D</b>) mRNA in immature P28 <span class="html-italic">wt</span> (<b>A</b>,<b>C</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>B</b>,<b>D</b>) testis sections. (<b>E</b>–<b>H</b>) <span class="html-italic">Cracd</span> mRNA in P28 (<b>E</b>,<b>F</b>) and P17 (<b>G</b>,<b>H</b>) <span class="html-italic">wt</span> (<b>E</b>,<b>G</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>F</b>,<b>H</b>) testis sections. (<b>I</b>–<b>L</b>) <span class="html-italic">Nme5</span> mRNA in P28 (<b>I</b>,<b>J</b>) and P17 (<b>K</b>,<b>L</b>) <span class="html-italic">wt</span> (<b>I</b>,<b>K</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>J</b>,<b>L</b>) testis sections. Scale bars (<b>A</b>–<b>J</b>) = 20 μm; (<b>K</b>,<b>L</b>) = 10 μm.</p>
Full article ">Figure 6
<p><span class="html-italic">Taz<sup>PM</sup></span> testes exhibit persistent p53-associated apoptosis. (<b>A</b>–<b>F</b>) Immunohistological detection of phosphohistone H3 cell proliferation marker (punctate brown DAD-positivity) in 4-month (<b>A</b>,<b>B</b>), P28 (<b>C</b>,<b>D</b>) and P17 (<b>E</b>,<b>F</b>) <span class="html-italic">wt</span> (<b>A</b>,<b>C</b>,<b>E</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>B</b>,<b>D</b>,<b>F</b>) testis sections. (<b>G</b>–<b>L</b>) TUNEL apoptosis marker expression (punctate brown DAD-positivity) in methyl green counterstained P28 (<b>G</b>,<b>H</b>), P17 (<b>I</b>,<b>J</b>) and P5 (<b>K</b>,<b>L</b>) <span class="html-italic">wt</span> (<b>G</b>,<b>I</b>,<b>K</b>) and <span class="html-italic">Taz<sup>PM</sup></span> (<b>H</b>,<b>J</b>,<b>L</b>) testis sections. (<b>M</b>,<b>N</b>). Western analysis of total p53, phospho p53<sup>S392</sup>, Sirt1, γ-H2Ax (both upper ubiquitinated and lower phospho γ-H2Ax isoforms) and Nme5 protein levels was performed in triplicate for <span class="html-italic">wt</span> and <span class="html-italic">Taz<sup>PM</sup></span> P28 testes ((<b>M</b>), n = 3 per genotype). Housekeeping Gapdh was used as the loading control, with quantification and statistical analysis (via two-tailed <span class="html-italic">t</span>-tests for all except Sirt1/Nme5 which used one-tailed t-tests due to <span class="html-italic">wt</span> variability) shown (<b>N</b>). Scale bars (<b>A</b>–<b>J</b>) = 50 μm; (<b>K</b>,<b>L</b>) = 20 μm.</p>
Full article ">
30 pages, 2198 KiB  
Article
Transcriptomic Response of the Ovarian Follicle Complex in Post-Vitellogenic Rainbow Trout to 17α,20β-Dihdroxy-4-pregnen-3-one In Vitro
by Hao Ma, Guangtu Gao, Yniv Palti, Vibha Tripathi, Jill E. Birkett and Gregory M. Weber
Int. J. Mol. Sci. 2024, 25(23), 12683; https://doi.org/10.3390/ijms252312683 - 26 Nov 2024
Viewed by 499
Abstract
Gonadotropins and progestins are the primary regulators of follicle maturation and ovulation in fish, and they require complex communication among the oocyte and somatic cells of the follicle. The major progestin and the maturation-inducing hormone in salmonids is 17α,20β-dihdroxy-4-pregnen-3-one (17,20βP), and traditional nuclear [...] Read more.
Gonadotropins and progestins are the primary regulators of follicle maturation and ovulation in fish, and they require complex communication among the oocyte and somatic cells of the follicle. The major progestin and the maturation-inducing hormone in salmonids is 17α,20β-dihdroxy-4-pregnen-3-one (17,20βP), and traditional nuclear receptors and membrane steroid receptors for the progestin have been identified within the follicle. Herein, RNA-seq was used to conduct a comprehensive survey of changes in gene expression throughout the intact follicle in response to in vitro treatment with these hormones to provide a foundation for understanding the coordination of their actions in regulating follicle maturation and preparation for ovulation. A total of 5292 differentially expressed genes were identified from our transcriptome sequencing datasets comparing four treatments: fresh tissue; untreated control; 17,20βP-treated; and salmon pituitary homogenate-treated follicles. Extensive overlap in affected genes suggests many gonadotropin actions leading to the acquisition of maturational and ovulatory competence are mediated in part by gonadotropin induction of 17,20βP synthesis. KEGG analysis identified signaling pathways, including MAPK, TGFβ, FoxO, and Wnt signaling pathways, among the most significantly enriched pathways altered by 17,20βP treatment, suggesting pervasive influences of 17,20βP on actions of other endocrine and paracrine factors in the follicle complex. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Venn diagram showing the distribution of all mapped and differentially expressed genes: (<b>A</b>) Number of transcripts mapped to rainbow trout reference transcriptome for fresh tissue, untreated control, MIH treated, and SPH treated samples. (<b>B</b>) Venn diagram depicting commonalities of differentially expressed transcripts identified by both DESeq2 and edgeR programs for MIH_control, SPH_control, and Fresh_control comparisons (FDR &lt; 0.05).</p>
Full article ">Figure 2
<p>Upregulated and downregulated differentially expressed genes identified by both DESeq2 and edgeR programs for MIH_control (M_C), SPH_control (S_C), and Fresh_control (F_C) comparisons (FDR &lt; 0.05).</p>
Full article ">Figure 3
<p>Bubble chart showing enriched KEGG pathways for differentially expressed genes in treatment comparisons. The color and size of the bubbles indicate the range of the <span class="html-italic">p</span>-value and the number of DEGs in each pathway.</p>
Full article ">Figure 4
<p>A comparison of 13 genes expressed in fresh, MIH, and SPH treated samples quantified by RT-qPCR and RNA-seq.:(<b>A</b>) RT-qPCR quantification of the selected genes. Gene expression value was normalized by <span class="html-italic">ef1a</span> and further compared to control as Log2 fold change. (<b>B</b>) Log2 fold change in RNA sequencing data was generated by DESeq2. Bars represent means ± SEM.</p>
Full article ">
20 pages, 8899 KiB  
Article
B Chromosome Transcriptional Inactivation in the Spermatogenesis of the Grasshopper Eyprepocnemis plorans
by Juan Luis Santos, María Teresa Parra, Sara Arévalo, Andrea Guajardo-Grence, Jesús Page, José Ángel Suja, Carlos García de la Vega and Alberto Viera
Genes 2024, 15(12), 1512; https://doi.org/10.3390/genes15121512 - 25 Nov 2024
Viewed by 575
Abstract
Background/Objectives: We analyzed the relationship between synapsis, recombination, and transcription during the spermatogenesis of the grasshopper Eyprepocnemis plorans carrying B chromosomes (type B1). Methods: The progression of synapsis was interpreted according to the dynamics of the cohesin subunit SMC3 axes. DNA double-strand breaks [...] Read more.
Background/Objectives: We analyzed the relationship between synapsis, recombination, and transcription during the spermatogenesis of the grasshopper Eyprepocnemis plorans carrying B chromosomes (type B1). Methods: The progression of synapsis was interpreted according to the dynamics of the cohesin subunit SMC3 axes. DNA double-strand breaks were revealed by RAD51 immunolabeling, while transcriptional activity was determined by the presence of RNA polymerase II phosphorylated at serine 2 (pRNApol II) immunolabeling. The two repressive epigenetic modifications, histone H3 methylated at lysine 9 (H3K9me3) and histone H2AX phosphorylated at serine 139 (γ-H2AX), were employed to reveal transcriptional inactivity. Results: During prophase I, spermatocytes with one B1 chromosome showed overall transcription except in the regions occupied by both the X and the B1 chromosomes. This transcriptional inactivity was accompanied by the accumulation of repressive epigenetic modifications. When two B1 chromosomes were present, they could appear as a fully synapsed monochiasmatic bivalent, showing intense H3K9me3 labeling and absence of pRNApol II, while γ-H2AX labeling was similar to that shown by the autosomes. Conclusions: According to our results, B1 transcriptional inactivation was triggered in spermatogonia, long before the beginning of meiosis, and was accompanied by H3K9me3 heterochromatinization that was maintained throughout spermatogenesis. Moreover, when two B1 were present, the transcriptional inactivation did not preclude synapsis and recombination achievement by these chromosomes. Full article
(This article belongs to the Section Cytogenomics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Dynamics of the B1 univalent and bivalent revealed by SMC3 immunolabeling. Prophase I spread spermatocytes from individuals with a B1 univalent (<b>A</b>–<b>E</b>) or a B1 bivalent (<b>F</b>–<b>J</b>). The positions of the sex chromosome (X), the B1 univalent (B), the B1 bivalent (B<sub>II</sub>) and of autosomal bivalents (Au) are indicated. Nuclei are counterstained with DAPI (blue). (<b>A</b>) At leptotene, SMC3 axes (white) are present all along the chromosomes. Thus, the identification of the X and B1 chromosomes was impeded by the imbrication of the SMC3 axes. (<b>B</b>) During zygotene, SMC3 autosomal axes were gradually paired into thicker filaments except at the X and B1 chromosomes. (<b>C</b>) At pachytene, autosomal bivalents displayed fully paired SMC3 axes, whereas a single thin SMC3 axis was observed in the X and B1 chromosomes. (<b>D</b>) 300% magnification of the nuclear region occupied by the X and B1 chromosomes. (<b>E</b>) Electron microscopy microphotograph of a pachytene spermatocyte showing the unsynapsed X and B1 chromosomes. (<b>F</b>) Leptotene spermatocytes with thin SMC3 axes all over the nucleus. (<b>G</b>) During zygotene, the SMC3 axes of the B1 chromosomes were gradually paired, as in the case of autosomes. (<b>H</b>) Pachytene spermatocyte showing fully paired SMC3 axes all along the autosomal bivalents, whereas pairing delay was observable in one of the ends of the B1 bivalent. (<b>I</b>) 300% magnifications of selected B1 bivalents with unpaired ends (red arrowheads), or with complete paired SMC3 axes as the autosomes (Au). In contrast, the X univalent exhibited a single unpaired SMC3 axis. (<b>J</b>) Electron microscopy microphotograph of a pachytene spermatocyte with fully synapsed B1 bivalent. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 2
<p>Meiotic reciprocal recombination in B1 chromosomes. Double immunolabeling of SMC3 (white) and RAD51 (red) in squashed spermatocytes counterstained with DAPI (blue) in individuals without B1 chromosomes (0B) and bearing one B1 (1B) or two B1 chromosomes (2B). The positions of the sex chromosome (X), the B1 univalent (B), the B1 bivalent (B<sub>II</sub>) and autosomal bivalents (Au) are indicated. Note that in prophase I stages, the X and B1 chromosomes are heteropycnotic positive chromatin bodies after DAPI staining. (<b>A</b>) Pachytene spermatocyte of a 0B male. RAD51 foci are visible along paired SMC3 axes of autosomes, but not over the X chromosome. (<b>B</b>) Pachytene spermatocyte of a 1B male. RAD51 foci are located over the fully paired SMC3 axes of autosomes but not over the unpaired SMC3 axes of the X and B1 chromosomes. (<b>C</b>) 300% magnification of the nuclear region occupied by the X and B1 chromosomes (yellow arrowheads signal RAD51 focus on an autosomal bivalent). (<b>D</b>) Zygotene spermatocyte of a 2B male. RAD51 foci were found over paired and unpaired SMC3 axes of autosomal bivalents and the B1 bivalent. (<b>E</b>) 300% magnification of the nuclear region occupied by the X chromosome and the B1 bivalent. Yellow and green arrowheads signal RAD51 foci located on autosomal and B1 bivalents, respectively. (<b>F</b>) Early/mid pachytene spermatocyte of a 2B male. Autosomal and B1 bivalents displayed RAD51 foci over their paired SMC3 axes. (<b>G</b>) 300% magnification of the nuclear region occupied by the X chromosome and the B1 bivalent. Yellow and green arrowheads signal RAD51 foci located on autosome and B bivalents, respectively. (<b>H</b>) Metaphase I spermatocyte of a 2B1 male. SMC3 appeared at the interchromatid domain of autosomal and B1 bivalents and was interrupted at chiasma sites. (<b>I</b>) 300% magnification of the X chromosome and the B1 bivalent. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 3
<p>Transcriptional activity of B1 chromosomes during prophase I stages. Double immunolabelings of SMC3 (white) and pRNApol II (red) in prophase I spread spermatocytes counterstained with DAPI (blue) from individuals with one B1 (<b>A</b>–<b>D</b>) or two B1 chromosomes (<b>E</b>–<b>H</b>). The positions of the sex chromosome (X), the B1 univalent (B) and the B1 bivalent (B<sub>II</sub>) are indicated, and in (<b>B</b>–<b>D</b>,<b>F</b>–<b>H</b>), their positions are outlined. (<b>A</b>) In leptotene, pRNApol II labeling appeared spread throughout most of the nucleus although reduced at some regions (yellow arrowheads). (<b>B</b>–<b>D</b>) During zygotene (<b>B</b>,<b>C</b>) and pachytene (<b>D</b>), pRNApol II labeling covered most of the autosomal chromatin irrespectively of their pairing status. The labeling was clearly reduced only in particular regions possibly corresponding to chromosome ends (yellow arrowheads). pRNApol II labeling is absent in the regions occupied by both the X and the B1 univalents. (<b>E</b>,<b>F</b>) The pattern of the pRNApol II labeling in individuals with two B1 chromosomes is identical to that described above. Significantly, the region occupied by the B1s does not present any signal from leptotene up to pachytene. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 4
<p>H3K9me3 labeling during prophase I stages. Double immunolabelings of SMC3 (white) and H3K9me3 (green) in prophase I spread spermatocytes from individuals with one (<b>A</b>–<b>D</b>) and two B1 chromosomes (<b>E</b>–<b>H</b>). The positions of the sex chromosome (X), the B1 univalent (B) and the B1 bivalent (B<sub>II</sub>) are indicated. (<b>A</b>) At leptotene, an intense H3K9me3 signal was visible in a particular nuclear region and discrete accumulations appeared in the rest of the nucleus. (<b>B</b>–<b>D</b>) From zygotene (<b>B</b>,<b>C</b>) and up to pachytene (<b>D</b>), the large H3K9me3 labeling was maintained and was located in the regions occupied by the X and B1 chromosomes. Additional H3K9me3 signals were positioned at some ends of the SMC3 axes of autosomal bivalents (yellow arrowheads). (<b>E</b>–<b>H</b>) In individuals carrying two B1 chromosomes, the H3K9me3 labeling was identical to that described above. The large signal corresponded to the positions of the X chromosome and the B1 bivalent. Moreover, some ends of autosome SMC3 axes presented bright foci (yellow arrowheads). Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 5
<p>γ-H2AX labeling during prophase I stages. Double immunolabelings of SMC3 (white) and γ-H2AX (purple) in prophase I spread spermatocytes from individuals with one B1 (<b>A</b>–<b>D</b>) and two B1 chromosomes (<b>E</b>–<b>K</b>). The positions of the sex chromosome (X), the B1 univalent (B), the B1 bivalent (B<sub>II</sub>) and autosomes (Au) are indicated. (<b>A</b>) γ-H2AX labeling became visible at leptotene as discrete flares over the unpaired SMC3 axes. (<b>B</b>) At early zygotene, γ-H2AX labeling was only observable over the yet unpaired SMC3 axes of the autosomal bivalents. Both the X and B1 chromosomes did not exhibit γ-H2AX signal. (<b>C</b>) By late zygotene, as DSBs repair proceeded, γ-H2AX was restricted to the last unpaired regions of the SMC3 autosomal axes and covered the unpaired SMC3 axes of the X and B1. (<b>D</b>) In early/mid pachytene, discrete γ-H2AX foci were positioned over the trajectories of the fully paired autosomal SMC3 axes. In contrast, the X and B1 univalents presented γ-H2AX ribbons covering their single SMC3 axes. (<b>E</b>) In individuals with two B1 chromosomes, γ-H2AX labeling become visible at leptotene. (<b>F</b>) By early zygotene, γ-H2AX labeling spread over the nucleus, covering unpaired autosomal SMC3 axes. (<b>G</b>) At late zygotene, γ-H2AX distribution was limited to the last unpaired regions of the SMC3 axes of autosomes and the B1 bivalent and over the single unpaired axis of the X chromosome. (<b>H</b>,<b>I</b>) Early and mid-pachytene spermatocytes presented γ-H2AX labeling as foci over paired SMC3 axes at autosomal bivalents and as a more continuous ribbon over the unpaired SMC3 axis of the X chromosome. (<b>J</b>,<b>K</b>) 300% magnifications of the nuclear region occupied by the X and B1 bivalent during early (<b>J</b>) and mid-pachytene (<b>K</b>). The B1 bivalent appeared labeled by γ-H2AX over the unpaired ends of the SMC3 axes (yellow arrowheads in (<b>J</b>)). By late pachytene (<b>K</b>), the B1 bivalent was fully paired and there was no γ-H2AX labeling. Bar in (<b>A</b>), 10 µm.</p>
Full article ">Figure 6
<p>Transcriptional activity in <span class="html-italic">E. plorans</span> spermatogonial cells<b>.</b> Double immunolabelings of H3K9me3 (green) with either γ-H2AX (purple) or pRNApol II (red) in squashed spermatogonia counterstained with DAPI (blue) from males without (0B), with one (1B) or two (2B) B1 chromosomes<b>.</b> The position of the sex chromosome (X) is indicated. (<b>A</b>,<b>B</b>) Spermatogonia from 0B individuals presented H3K9me3 accumulated at the X chromosome and at certain nuclear regions (white arrowheads). γ-H2AX was not detected and pRNApol II rendered faint labeling over the nucleus, except at those regions labeled with H3K9me3 (white arrowheads). (<b>C</b>–<b>F</b>) In nuclei of 1B and 2B individuals, H3K9me3 labeled several autosomal regions (white arrowheads), and the X chromosome, but γ-H2AX was not detected. Moreover, H3K9me3 labeling corresponded with regions of reduced pRNApol II labeling. (<b>G</b>) Scattered dot plot of the corrected total nuclear fluorescence of H3K9me3 immunolabeling in spermatogonia from individuals presenting 0, 1 or 2 B1 chromosomes. For each class, the individual data (n = 18) and median with 95% CI are depicted. Statistical significance was assessed using an ANOVA test (<span class="html-italic">p</span> &lt; 0.0001; ***) and Tukey’s multiple comparisons test. Bar in (<b>A</b>), 10 µm.</p>
Full article ">
11 pages, 3550 KiB  
Article
Comparison of Nucleosome Landscapes Between Porcine Embryonic Fibroblasts and GV Oocytes
by Minjun Zhao, Shunran Zhao, Zhaoqi Pang, Chunhui Jia and Chenyu Tao
Animals 2024, 14(23), 3392; https://doi.org/10.3390/ani14233392 - 25 Nov 2024
Viewed by 459
Abstract
(1) Background: Nucleosomes represent the essential structural units of chromatin and serve as key regulators of cell function and gene expression. Oocytes in the germinal vesicle (GV) stage will later undergo meiosis and become haploid cells ready for fertilization, while somatic cells undergo [...] Read more.
(1) Background: Nucleosomes represent the essential structural units of chromatin and serve as key regulators of cell function and gene expression. Oocytes in the germinal vesicle (GV) stage will later undergo meiosis and become haploid cells ready for fertilization, while somatic cells undergo mitosis after DNA replication. (2) Purpose: To furnish theoretical insights and data that support the process of cell reprogramming after nuclear transplantation. (3) Methods: We compared the nucleosome occupancy, distribution, and transcription of genes between two types of cells: fully grown GV oocytes from big follicles (BF) and somatic cells (porcine embryonic fibroblast, PEF). (4) Results: The nucleosome occupancy in the promoter of BF was 4.85%, which was significantly higher than that of 3.3% in PEF (p < 0.05), and the nucleosome distribution showed a noticeable increase surrounding transcriptional start sites (TSSs) in BF. Next, we reanalyzed the currently published transcriptome of fully grown GV oocytes and PEF, and a total of 51 genes in BF and 80 genes in PEF were identified as being uniquely expressed. The nucleosome distribution around gene TSSs correlated with expression levels in somatic cells, yet the results in BF differed from those in PEF. (5) Conclusion: This study uncovers the dynamic nature and significance of nucleosome positioning and chromatin organization across various cell types, providing a basis for nuclear transplantation. Full article
(This article belongs to the Special Issue Cutting-Edge Breakthroughs in Animal Reproductive Endocrinology)
Show Figures

Figure 1

Figure 1
<p>Global nucleosome occupancy of PEFs and BFs. (<b>A</b>) Bar plot showing the comparison ratio of the functional element distribution between PEFs and BFs. (<b>B</b>) A box plot illustrating the correlation between nucleosome occupancy and GC content. The blue box represents BFs, while the red box corresponds to PEFs.</p>
Full article ">Figure 2
<p>Nucleosome occupancy comparison between PEFs and BFs. (<b>A</b>) A genome-wide comparison of nucleosome occupancy between PEFs and BFs was performed. The color scheme represents the shifts in nucleosome occupancy for every 10 kb region between the two samples: bright red highlights a significant ≥1.5-fold increase in PEF nucleosome occupancy, while deep green marks a pronounced ≤1.5-fold decrease; gray indicates no changes in nucleosome occupancy; and white indicates regions with no enrichment. (<b>B</b>) A differential analysis of nucleosome occupancy between PEFs and BFs was conducted. Red highlights a dramatic ≥2-fold increase in nucleosome occupancy in BF, whereas blue reveals a notable ≤2-fold decrease in nucleosome occupancy in SF. (<b>C</b>–<b>E</b>) Average profile of MNase peaks surrounding the TSS region. Comparison of MNase peak distributions between BFs (<b>C</b>), PEFs (<b>D</b>), and merge (<b>E</b>).</p>
Full article ">Figure 3
<p>Average profile plot showing nucleosome occupancy in regions containing dynamic nucleosomes. (<b>A</b>,<b>B</b>) Nucleosomes showing an increase in PEF fuzziness (<b>A</b>) and an occupancy decrease (<b>B</b>).</p>
Full article ">Figure 4
<p>The RNA-seq analysis of BFs and PEFs. (<b>A</b>) Pearson correlations among the replicate samples of BFs and PEFs. (<b>B</b>,<b>C</b>) The GO functional annotation of uniquely expressed genes in BFs (B) and PEFs (<b>C</b>). The <span class="html-italic">x</span>-axis shows the three categories and the detailed GO terms, while the <span class="html-italic">y</span>-axis shows the number of genes. (<b>D</b>,<b>E</b>) The q-PCR validation of the RNA-seq data. ZP3, ZP4, OOEP, SYCN, and DAPP5 in BFs (<b>D</b>) and THBS1, ACTA2, SPARC, FN1, and COL1A1 in PEFs (FC, fold change) (<b>E</b>).</p>
Full article ">Figure 5
<p>The patterns of nucleosome occupancy around TSSs reveal striking differences among the top 5% most expressed genes (red), silent genes (green), and the remaining genes (blue) in both BFs (<b>A</b>) and PEFs (<b>B</b>).</p>
Full article ">
23 pages, 4889 KiB  
Article
Genome-Wide Association Study and Phenotype Prediction of Reproductive Traits in Large White Pigs
by Hao Zhang, Shiqian Bao, Xiaona Zhao, Yangfan Bai, Yangcheng Lv, Pengfei Gao, Fuzhong Li and Wuping Zhang
Animals 2024, 14(23), 3348; https://doi.org/10.3390/ani14233348 - 21 Nov 2024
Viewed by 829
Abstract
In a study involving 385 Large White pigs, a genome-wide association study (GWAS) was conducted to investigate reproductive traits, specifically the number of healthy litters (NHs) and the number of weaned litters (NWs). Several SNP loci, including ALGA0098819, ALGA0037969, and H3GA0032302, were significantly [...] Read more.
In a study involving 385 Large White pigs, a genome-wide association study (GWAS) was conducted to investigate reproductive traits, specifically the number of healthy litters (NHs) and the number of weaned litters (NWs). Several SNP loci, including ALGA0098819, ALGA0037969, and H3GA0032302, were significantly associated with these traits. In the combined-parity analysis, candidate genes, such as BLVRA, STK17A, PSMA2, and C7orf25, were identified. GO and KEGG pathway enrichment analyses revealed that these genes are involved in key biological processes, including organic synthesis, the regulation of sperm activity, spermatogenesis, and meiosis. In the by-parity analysis, the PLCXD3 gene was significantly associated with the NW trait in the second and fourth parities, while RNASEH1, PYM1, and SEPTIN9 were linked to cell proliferation, DNA repair, and metabolism, suggesting their potential role in regulating reproductive traits. These findings provide new molecular markers for the genetic study of reproductive traits in Large White pigs. For the phenotypic prediction of NH and NW traits, several machine learning models (GBDT, RF, LightGBM, and Adaboost.R2), as well as traditional models (GBLUP, BRR, and BL), were evaluated using SNP data in varying proportions. After PCA processing, the GBDT model achieved the highest PCC for NH (0.141), while LightGBM reached the highest PCC for NW (0.146). The MAE, MSE, and RMSE results showed that the traditional models exhibited stable error rates, while the machine learning models performed comparatively better across the different SNP ratios. Overall, PCA processing provided some improvement in the predictive performance of all of the models, though the overall increase in accuracy was limited. Full article
(This article belongs to the Section Pigs)
Show Figures

Figure 1

Figure 1
<p>Phenotypic frequency distribution of two traits. (<b>A</b>) Distribution of the NH trait, showing the mean and standard deviation values. (<b>B</b>) Distribution of the NW trait, showing the mean and standard deviation values.</p>
Full article ">Figure 2
<p>Distribution of genome-wide SNPs across chromosomes.</p>
Full article ">Figure 3
<p>Manhattan plot and Q-Q plot based on two reproductive traits under combined parities. (<b>A</b>) Manhattan plot of the NH trait, displaying the genomic positions of SNPs on the <span class="html-italic">x</span>-axis and their corresponding −log10(<span class="html-italic">p</span>-values) on the <span class="html-italic">y</span>-axis. Significant SNPs are indicated by green dots. (<b>B</b>) Q-Q plot of the NH trait, illustrating the relationship between observed and expected −log10(<span class="html-italic">p</span>-values). The blue area represents the confidence interval, while the red dashed line represents the expected values under the null hypothesis. (<b>C</b>) Manhattan plot of the NW trait, displaying the genomic positions of SNPs on the <span class="html-italic">x</span>-axis and their corresponding −log10(<span class="html-italic">p</span>-values) on the <span class="html-italic">y</span>-axis. Significant SNPs are indicated by green dots. (<b>D</b>) Q-Q plot of the NW trait, illustrating the relationship between observed and expected −log10(<span class="html-italic">p</span>-values). The blue area represents the confidence interval, while the red dashed line represents the expected values under the null hypothesis. Note: Green dots in the Manhattan plot indicate significant SNPs. The horizontal axis represents the position of the marker on the pig chromosome, while the vertical axis represents the <math display="inline"><semantics> <mrow> <mo>−</mo> <msub> <mrow> <mi>log</mi> </mrow> <mrow> <mn>10</mn> </mrow> </msub> </mrow> </semantics></math>(<span class="html-italic">p</span>-value) of the marker–trait association. The red, dashed line indicates the significance threshold criteria set in this study. The quantile–quantile (Q-Q) plot shows the relationship between the expected and the observed <span class="html-italic">p</span>-values. The horizontal axis represents the negative logarithm of the expected <span class="html-italic">p</span>-value, while the vertical axis represents the negative logarithm of the observed <span class="html-italic">p</span>-value for each SNP. The red, dashed line represents the expected relationship under the null hypothesis.</p>
Full article ">Figure 4
<p>Manhattan plot and quantile–quantile plot based on different parity NH traits. The Manhattan plots (<b>left</b>) display the genomic positions of SNPs (<span class="html-italic">x</span>-axis) and their corresponding −log10(<span class="html-italic">p</span>-values) (<span class="html-italic">y</span>-axis). The Q-Q plots (<b>right</b>) show the relationship between observed and expected −log10(<span class="html-italic">p</span>-values). In the Q-Q plots, the blue area represents the confidence interval under the null hypothesis, while the red dashed line represents the expected values under perfect correlation between observed and expected <span class="html-italic">p</span>-values.</p>
Full article ">Figure 5
<p>Manhattan plot and quantile–quantile plot based on different parity NW traits. The Manhattan plots (<b>left</b>) display the genomic positions of SNPs (<span class="html-italic">x</span>-axis) and their corresponding −log10(<span class="html-italic">p</span>-values) (<span class="html-italic">y</span>-axis). The Q-Q plots (<b>right</b>) show the relationship between observed and expected −log10(<span class="html-italic">p</span>-values). In the Q-Q plots, the blue area represents the confidence interval under the null hypothesis, while the red dashed line represents the expected values under perfect correlation between observed and expected <span class="html-italic">p</span>-values.</p>
Full article ">Figure 6
<p>The GO enrichment analysis of significant SNPs for the NH trait with combined parities.</p>
Full article ">Figure 7
<p>The GO enrichment analysis of significant SNPs for the NW trait with combined parities.</p>
Full article ">Figure 8
<p>Comparison of model performance using the PCA method for predicting two traits. (<b>A</b>) Correlation coefficient (PCC) of different models (GBLUP, BL, BRR, LightGBM, RF, GBDT, and AdaBoost.R2) for NH and NW traits. (<b>B</b>) Mean absolute error (MAE) of the models for NH and NW traits. (<b>C</b>) Mean squared error (MSE) of the models for NH and NW traits. (<b>D</b>) Root mean squared error (RMSE) of the models for NH and NW traits.</p>
Full article ">
20 pages, 5198 KiB  
Article
Multi-Omics Approaches Uncovered Critical mRNA–miRNA–lncRNA Networks Regulating Multiple Birth Traits in Goat Ovaries
by Weibing Lv, Ren An, Xinmiao Li, Zengdi Zhang, Wanma Geri, Xianrong Xiong, Shi Yin, Wei Fu, Wei Liu, Yaqiu Lin, Jian Li and Yan Xiong
Int. J. Mol. Sci. 2024, 25(22), 12466; https://doi.org/10.3390/ijms252212466 - 20 Nov 2024
Viewed by 818
Abstract
The goat breeding industry on the Tibetan Plateau faces strong selection pressure to enhance fertility. Consequently, there is an urgent need to develop goat lines with higher fertility and adaptability. The ovary, as a key organ determining reproductive performance, is regulated by a [...] Read more.
The goat breeding industry on the Tibetan Plateau faces strong selection pressure to enhance fertility. Consequently, there is an urgent need to develop goat lines with higher fertility and adaptability. The ovary, as a key organ determining reproductive performance, is regulated by a complex transcriptional network involving numerous protein-coding and non-coding genes. However, the molecular mechanisms of the key mRNA–miRNA–lncRNA regulatory network in goat ovaries remain largely unknown. This study focused on the histology and differential mRNA/miRNA/lncRNA between Chuanzhong black goat (CBG, high productivity, multiple births) and Tibetan goat (TG, strong adaptability, single birth) ovaries. Histomorphological analysis showed that the medulla proportion in CBG ovaries was significantly reduced compared to TG. RNA-Seq and small RNA-Seq analysis identified 1218 differentially expressed (DE) mRNAs, 100 DE miRNAs, and 326 DE lncRNAs, which were mainly enriched in ovarian steroidogenesis, oocyte meiosis, biosynthesis of amino acids and protein digestion, and absorption signaling pathways. Additionally, five key mRNA–miRNA–lncRNA interaction networks regulating goat reproductive performance were identified, including TCL1B–novel68_mature–ENSCHIT00000010023, AKAP6–novel475_mature–ENSCHIT00000003176, GLI2–novel68_mature–XR_001919123.1, ITGB5–novel65_star–TCONS_00013850, and VWA2–novel71_mature–XR_001919911.1. Further analyses showed that these networks mainly affected ovarian function and reproductive performance by regulating biological processes such as germ cell development and oocyte development, which also affected the plateau adaptive capacity of the ovary by participating in the individual immune and metabolic capacities. In conclusion, we identified numerous mRNA–miRNA–lncRNA interaction networks involved in regulating ovarian function and reproductive performance in goats. This discovery offers new insights into the molecular breeding of Tibetan Plateau goats and provides a theoretical foundation for developing new goat lines with high reproductive capacity and strong adaptability to the plateau environment. Full article
Show Figures

Figure 1

Figure 1
<p>Morphological and statistical analysis of ovarian tissues. (<b>A</b>,<b>B</b>) H&amp;E staining image of the maximum cross-section of TG (<b>A</b>) and CBG (B) ovaries, scale bar = 1000 μm. (<b>C</b>) The number of follicles in ovarian tissue by statistical analysis. (<b>D</b>) The total area of the maximum cross-section of ovaries. (<b>E</b>,<b>F</b>) The area (<b>E</b>) and proportion (<b>F</b>) of medullary in the maximum cross-section of ovaries. (<b>G</b>) Relative expression levels of reproduction-related marker genes in ovarian tissues. Note: ** represents <span class="html-italic">p</span> &lt; 0.01, * represents <span class="html-italic">p</span> &lt; 0.05, and ns represents <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 2
<p>DE mRNA screening and functional enrichment analysis between CBG and TG. (<b>A</b>) FPKM density distribution curve. (<b>B</b>) PCA plot. (<b>C</b>) Volcano plot of DE mRNAs. The <span class="html-italic">X</span>-axis was log<sub>2</sub>FoldChange and the <span class="html-italic">Y</span>-axis was −log<sub>10</sub>pValue. (<b>D</b>) Bar chart of DE mRNAs statistics. The <span class="html-italic">x</span>-axis represents the comparison groups, and the y-axis represents the number of differential genes in each group. (<b>E</b>) RT-qPCR validation of DE mRNAs. (<b>F</b>,<b>G</b>) GO enrichment analysis for up-regulated mRNAs (<b>F</b>) and (<b>G</b>) down-regulated mRNAs. The <span class="html-italic">x</span>-axis represented GO term names, and the <span class="html-italic">y</span>-axis represented −log<sub>10</sub>pValue. (<b>H</b>,<b>I</b>) KEGG enrichment analysis for up-regulated mRNAs (<b>H</b>) and (<b>I</b>) down-regulated mRNAs. The <span class="html-italic">X</span>-axis represented the enrichment score.</p>
Full article ">Figure 3
<p>DE miRNA screening and functional enrichment analysis. (<b>A</b>) Length distribution of miRNAs. (<b>B</b>) PCA plot. (<b>C</b>) Volcano plot of DE miRNAs. (<b>D</b>) Histogram of DE miRNAs number in these two groups. (<b>E</b>) RT-qPCR validation of DE miRNAs. (<b>F</b>,<b>G</b>) Top 10 bar plots of GO enrichment analysis for target genes of up-regulated miRNAs (<b>F</b>) and down-regulated miRNAs (<b>G</b>). The <span class="html-italic">Y</span>-axis represented GO terms and the <span class="html-italic">X</span>-axis was −log<sub>10</sub>pValue. (<b>H</b>,<b>I</b>) KEGG enrichment analysis for target genes of up-regulated miRNAs (<b>H</b>) and down-regulated miRNAs (<b>I</b>). The <span class="html-italic">X</span>-axis was the enrichment score.</p>
Full article ">Figure 4
<p>DE lncRNA screening and functional enrichment analysis in the CBG and BG. (<b>A</b>) Venn diagram of coding potential prediction results for candidate lncRNAs. (<b>B</b>) FPKM density distribution curve. (<b>C</b>) PCA plot. (<b>D</b>) Volcano plot of DE lncRNAs expression. (<b>E</b>) Histogram of DE lncRNAs statistics. (<b>F</b>) RT-qPCR validation of DE lncRNAs. (<b>G</b>,<b>H</b>) GO enrichment analysis for up-regulated lncRNAs (<b>G</b>) and down-regulated lncRNAs (<b>H</b>). The <span class="html-italic">X</span>-axis represented GO term names; The <span class="html-italic">Y</span>-axis represented −log<sub>10</sub>pValue. (<b>I</b>,<b>J</b>) KEGG enrichment analysis for up-regulated lncRNAs (<b>I</b>) and down-regulated lncRNAs (<b>J</b>). The <span class="html-italic">X</span>-axis represented the enrichment score, with larger bubbles indicating more DE lncRNAs, and bubble color ranging between purple, blue, green and red, with smaller <span class="html-italic">p</span> values indicating higher significance. G: GO:0043154: negative regulation of cysteine-type endopeptidase activity. GO:2000480: negative regulation of cytokine-mediated signaling pathway. H: GO:0000981: DNA-binding transcription factor activity, RNA polymerase II-specific. GO:0001077: proximal promoter DNA-binding transcription activator activity, RNA polymerase II-specific. GO:0000978: RNA polymerase II cis-regulatory region sequence-specific DNA binding.</p>
Full article ">Figure 5
<p>Co-expression analysis of lncRNAs and mRNAs. (<b>A</b>) GO enrichment analysis of co-expressed DE genes with lncRNAs (top 10). The <span class="html-italic">X</span>-axis represented GO terms, and the <span class="html-italic">Y</span>-axis represented the number of lncRNAs enriched. (<b>B</b>) KEGG enrichment bubble chart of co-expressed DE genes with lncRNAs (top 10). The <span class="html-italic">X</span>-axis represented the enrichment score, with larger bubbles indicating more DE genes in the term, and bubble color ranging from gray to red, reflecting decreasing <span class="html-italic">p</span> -values and increasing significance. CP: Cellular Processes; EIP: Environmental Information Processing; GIP: Genetic Information Processing; HD: Human Diseases; Meta.: Metabolism; OS: Organismal Systems. (<b>C</b>) Analysis of lncRNA trans-acting target genes. The red circles represented lncRNAs, green inverted triangles represented genes, and node size indicated quantity. (<b>D</b>) Analysis of lncRNA cis-acting target genes. The left and right sides of the <span class="html-italic">y</span>-axis represented mRNA and lncRNA, respectively. The <span class="html-italic">x</span>-axis indicated the distance between mRNA and lncRNA, with negative values indicating upstream and positive values indicating downstream. Identical lncRNAs were represented by the same color bar chart. Note: ** represents <span class="html-italic">p</span> &lt; 0.01, * represents <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>CeRNA interaction network analysis. (<b>A</b>) Co-expression network analysis of miRNAs and mRNAs. (<b>B</b>) Regulatory network analysis of mRNAs and lncRNAs. (<b>C</b>) Regulatory network analysis of miRNAs and lncRNAs. (<b>D</b>) Regulatory network analysis of mRNAs, miRNAs, and lncRNAs. Red circles represented mRNAs, green triangles represented miRNAs, and blue rounded rectangles represented lncRNAs. The size of the shapes indicated the quantity. (<b>E</b>) GO enrichment analysis of mRNAs in ceRNA. The <span class="html-italic">Y</span>-axis represented GO terms, and the <span class="html-italic">X</span>-axis represented the enrichment score. (<b>F</b>) KEGG enrichment analysis of mRNAs in ceRNA. The <span class="html-italic">X</span>-axis represented the enrichment score, and the <span class="html-italic">Y</span>-axis represented enriched pathways.</p>
Full article ">Figure 7
<p>Key mRNA–miRNA–lncRNA regulating goat lambing traits and plateau adaptability.</p>
Full article ">
Back to TopTop