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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (7,620)

Search Parameters:
Keywords = mitochondria

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3301 KiB  
Article
Administration of AICAR, an AMPK Activator, Prevents and Reverses Diabetic Polyneuropathy (DPN) by Regulating Mitophagy
by Krish Chandrasekaran, Joungil Choi, Mohammad Salimian, Ahmad F. Hedayat and James W. Russell
Int. J. Mol. Sci. 2025, 26(1), 80; https://doi.org/10.3390/ijms26010080 (registering DOI) - 25 Dec 2024
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes in both Type 1 (T1D) and Type 2 (T2D). While there are no specific medications to prevent or treat DPN, certain strategies can help halt its progression. In T1D, maintaining tight glycemic control [...] Read more.
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes in both Type 1 (T1D) and Type 2 (T2D). While there are no specific medications to prevent or treat DPN, certain strategies can help halt its progression. In T1D, maintaining tight glycemic control through insulin therapy can effectively prevent or delay the onset of DPN. However, in T2D, overall glucose control may only have a moderate impact on DPN, although exercise is clearly beneficial. Unfortunately, optimal exercise may not be feasible for many patients with DPN because of neuropathic foot pain and poor balance. Exercise has several favorable effects on health parameters, including body weight, glycemic control, lipid profile, and blood pressure. We investigated the impact of an exercise mimetic, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), on DPN. AICAR treatment prevented or reversed experimental DPN in mouse models of both T2D and T1D. AICAR in high-fat diet (HFD-fed) mice increased the phosphorylation of AMPK in DRG neuronal extracts, and the ratio of phosphorylated AMPK to total AMPK increased by 3-fold (HFD vs. HFD+AICAR; p < 0.001). Phospho AMP increased the levels of dynamin-related protein 1 (DRP1, a mitochondrial fission marker), increased phosphorylated autophagy activating kinase 1 (ULK1) at Serine-555, and increased microtubule-associated protein light chain 3-II (LC3-II, a marker for autophagosome assembly) by 2-fold. Mitochondria isolated from DRG neurons of HFD-fed had a decrease in ADP-stimulated state 3 respiration (120 ± 20 nmol O2/min in HFD vs. 220 ± 20 nmol O2/min in control diet (CD); p < 0.001. Mitochondria isolated from HFD+AICAR-treated mice had increased state 3 respiration (240 ± 30 nmol O2/min in HFD+AICAR). However, AICAR’s protection in DPN in T2D mice was also mediated by its effects on insulin sensitivity, glucose metabolism, and lipid metabolism. Drugs that enhance AMPK phosphorylation may be beneficial in the treatment of DPN. Full article
(This article belongs to the Special Issue Mitochondrial Metabolism Alterations in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Exercise consumes a large amount of ATP, elevating the AMP-to-ATP ratio. Increased AMP binds to the enzyme AMPK and induces the phosphorylation of AMPK (<a href="#ijms-26-00080-f001" class="html-fig">Figure 1</a>). On the other hand, <b>AICAR directly phosphorylates AMPK</b>. (1) Phosphorylated AMPK activates SIRT1/PGC1-α in the catabolic process of exercise (increased OXPHOS), resulting in decreased body weight, reduced triglycerides, improved HOMA-IR index and lower NEFA levels; (2) phosphorylated AMPK activates PGC-1α co-transcriptional complexes that initiate the overexpression of target genes to promote myogenesis, neurogenesis, and mitochondrial bioenergetics; and (3) phosphorylated AMPK led to phosphorylation of mitochondrial fission factor (MFF), recruitment of dynamin-like protein DRP1 to mitochondria, and activation of ULK1, an upstream kinase in autophagy and mitophagy. Mitochondrial fission allows damaged mitochondria to be selectively degraded through mitophagy pathways. The main points of this paper are boxed in red.</p>
Full article ">Figure 2
<p>Western blot analysis of AMPK and quantification of phospho-AMPK to total AMPK (<a href="#ijms-26-00080-f002" class="html-fig">Figure 2</a>). DRG neurons were isolated from CD (<span class="html-italic">n</span> = 6), CD+AICAR (<span class="html-italic">n</span> = 6), HFD (<span class="html-italic">n</span> = 6), and HFD+AICAR (<span class="html-italic">n</span> = 6) mice, and protein extracts were prepared. Antibodies recognize total AMPK, AMPK phosphorylated at the Thr 176 residue, and beta-actin. The levels of expression were quantified by the intensity of the bands. The ratio of pAMPK to total AMPK was calculated, and the values were analyzed by ANOVA. The significance is denoted by the following <span class="html-italic">p</span>-values: *** <span class="html-italic">p</span> &lt; 0.001; CD or CD+AICARD vs. HFD, +++ <span class="html-italic">p</span> &lt; 0.001; HFD vs. HFD+AICAR.</p>
Full article ">Figure 3
<p>Western blot analysis of proteins involved in mitophagy in neuronal mitochondria isolated from CD (<span class="html-italic">n</span> = 6), CD+AICAR (<span class="html-italic">n</span> = 6), HFD (<span class="html-italic">n</span> = 6), and HFD+AICAR (<span class="html-italic">n</span> = 6) mice. Western blots were carried out on the protein extracts using anti-LC3, anti-DRP1, anti-phospho (S555) ULK1, and anti-VDAC. The levels of expression were quantified based on the intensity of the bands. The relative ratio was calculated, and the values were analyzed by ANOVA. The significance is denoted by the following <span class="html-italic">p</span>-values: ** <span class="html-italic">p</span> &lt; 0.01 HFD vs. CD or CD+AICAR; +++ <span class="html-italic">p</span> &lt; 0.001 HFD+AICAR vs. HFD.</p>
Full article ">Figure 4
<p>Impaired ADP-stimulated mitochondrial respiration in HFD mice was prevented by AICAR treatment. Oxygen consumption rate (OCR) was measured in the presence of complex I substrates (malate/glutamate) with the subsequent and sequential addition of ADP, oligomycin, and rotenone + antimycin A to mitochondria to measure state 2 (basal), state 3 (ADP-stimulated), state 4<sub>0</sub> (oligomycin-sensitive), and FCCP-induced respiration rates were measured. ADP-stimulated respiration was significantly decreased in neuronal mitochondria from HFD-fed mice compared to CD and CD+AICAR-treated mice. Administration of AICAR to HFD-fed mice significantly increased ADP-stimulated respiration. The respiratory control ratio (RCR) was calculated. *** <span class="html-italic">p</span> &lt; 0.001 HFD vs. CD, CD+AICAR. +++ <span class="html-italic">p</span> &lt; 0.001 HFD vs. HFD+AICAR.</p>
Full article ">Figure 5
<p><b>AICAR prevented HFD-induced neuropathy in C57Bl6 mice (<span class="html-italic">n</span> = 6 to 8 per group).</b> WT C57BL6 mice were randomly assigned to four groups of six to eight mice per group. Group #1: CD; Group #2: CD+AICAR (500 mg/kg); Group #3: HFD; and Group #4: HFD+AICAR (500 mg/kg). Placebo or AICAR was administered for 4 months to CD and HFD mice. Mice were tested for the following parameters: SMNCV (<b>A</b>), TML (<b>B</b>), TSNCV (<b>C</b>), mechanical allodynia (MA) by Von Frey filament paw withdrawal threshold (<b>D</b>), and IENFD of the hind paw (<b>E</b>). Statistical comparisons were made among the 5 groups by ANOVA and post hoc Tukey test. *** <span class="html-italic">p</span> &lt; 0.001; HFD, Group #3 at 4 months compared to all other groups in all the parameters.</p>
Full article ">Figure 6
<p>Two months of AICAR treatment reversed HFD-induced neuropathy in C57BL6 mice (<span class="html-italic">n</span> = 6/group). Three-month-old C57BL6 WT mice were fed with either a control diet (CD) or a high-fat diet (HFD). Baseline NCSs were completed at the beginning of the study. At 2 months, SMNCV, TML, TSNCV, and MA were measured in the mice fed a CD and the mice fed an HFD. After confirming that consumption of the HFD for 2 months induced development of peripheral neuropathy as observed by the changes in the NCSs and MA, AICAR was administered to the CD and HFD mice at a dose of 500 mg/kg for an additional 2 months. The vehicle was administered to CD and HFD mice. Nerve conduction studies were performed 2 months after administration of the AICAR or vehicle. The protocol is described in (<b>A</b>), the results of SMNCV (<b>B</b>), TSNCV (<b>C</b>), TML (<b>D</b>), MA using the Von Frey filament paw withdrawal threshold method (<b>E</b>), and intraepidermal fiber density (<b>F</b>) are shown. Statistical comparisons were made among the three groups with the ANOVA and post hoc Tukey test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001; HFD mice at 2 months compared to 0-month-old HFD and CD mice in all parameters. <sup>++</sup> <span class="html-italic">p</span> &lt; 0.01, HFD+AICAR mice at 4 months compared to HFD at 2 months in all parameters. The administration of AICAR reversed all the deficits of HFD-induced neuropathy. The administration of AICAR to non-diabetic mice had no significant effect. There was no statistically significant difference in CMAP or SNAP amplitudes compared between groups or compared at the start, during, and at the end of the treatment. The baseline (prior to starting any treatment) CMAP sciatic amplitude (mV) was 6.17 ± 0.78; CMAP tail amplitude (mV) was 3.050 ± 0.60, and SNAP tail amplitude (μV) was 51.92 ± 13.08. The CMAP sciatic amplitudes (mV) after 4 months of treatment were as follows: CD: 5.14 ± 0.99 (SEM); CD+AICAR: 5.70 ± 1.68; HFD: 8.31 ± 1.66; HFD+AICAR: 5.09 ± 0.76. The CMAP tail amplitudes (mV) after 4 months of treatment were as follows: CD: 1.24 ± 0.24; CD+AICAR: 2.16 ± 0.41; HFD: 2.96 ± 0.96; HFD+AICAR: 1.87 ± 0.70. The SNAP tail amplitudes (μV) after 4 months of treatment were as follows: CD: 102.48 ± 24.88; CD+AICAR: 126.77 ± 22.88; HFD: 56.21 ± 19.47; HFD+AICAR: 77.375 ± 19.46. There was no statistically significant difference in the CMAP or SNAP amplitudes during the reversal study, compared between groups or compared at the start and end of the treatment. This is as expected because of considerable variability in the CMAP and SNAP amplitudes [<a href="#B26-ijms-26-00080" class="html-bibr">26</a>,<a href="#B27-ijms-26-00080" class="html-bibr">27</a>,<a href="#B28-ijms-26-00080" class="html-bibr">28</a>].</p>
Full article ">Figure 7
<p>AICAR reverses STZ-induced neuropathy in C57BL6 mice (<span class="html-italic">n</span> = 6/group). Three-month-old C57BL6 WT and STZ-induced diabetic mice were purchased from Jackson Labs. The mice were fed with a control diet for a month. Measurement of NCSs and MA after a month showed that the STZ mice had developed neuropathy. Some of the STZ mice were then administered AICAR at a dose of 500 mg/kg for an additional 2 months. NCSs were performed at 5 and 6 months of age, namely after 2 and 3 months of STZ-induced diabetes or after 1 or 2 months of AICAR treatment. The results are shown as follows: SMNCV (<b>A</b>), TSNCV (<b>B</b>), TML (<b>C</b>), and MA (<b>D</b>) using the Von Frey filament paw withdrawal threshold method. Statistical comparisons were made among the three groups with the ANOVA and post hoc Tukey test. *** <span class="html-italic">p</span> &lt; 0.001; STZ at experimental time 3 months compared to 1 month in all parameters. <sup>+++</sup> <span class="html-italic">p</span> &lt; 0.001, STZ+AICAR at experimental time 3 months compared to STZ at 1 month in all parameters. Thus, administration of AICAR reversed all the peripheral neuropathy deficits of STZ mice.</p>
Full article ">
16 pages, 4153 KiB  
Article
Metabolic Activity in Human Intermuscular Adipose Tissue Directs the Response of Resident PPARγ+ Macrophages to Fatty Acids
by Xiaoying Chen, Sebastian Ludger Schubert, Aline Müller, Miguel Pishnamaz, Frank Hildebrand and Mahtab Nourbakhsh
Biomedicines 2025, 13(1), 10; https://doi.org/10.3390/biomedicines13010010 (registering DOI) - 25 Dec 2024
Abstract
Background/Objectives: Peroxisome proliferator-activated receptor gamma (PPARγ) is a fatty acid-binding transcription activator of the adipokine chemerin. The key role of PPARγ in adipogenesis was established by reports on adipose tissue-resident macrophages that express PPARγ. The present study examined PPARγ+ macrophages in [...] Read more.
Background/Objectives: Peroxisome proliferator-activated receptor gamma (PPARγ) is a fatty acid-binding transcription activator of the adipokine chemerin. The key role of PPARγ in adipogenesis was established by reports on adipose tissue-resident macrophages that express PPARγ. The present study examined PPARγ+ macrophages in human skeletal muscle tissues, their response to fatty acid (FA) species, and their correlations with age, obesity, adipokine expression, and an abundance of other macrophage phenotypes. Methods: An ex vivo human skeletal muscle model with surgical specimens that were maintained without or with FAs for up to 11 days was utilized. Immunofluorescence analysis was used to detect macrophage phenotypes and mitochondrial activity. Preconfigured arrays were used to detect the expression of 34 different adipokines and chemokines. Results: Data from 14 adults revealed that PPARγ+ macrophages exclusively reside in intermuscular adipose tissue (IMAT), and their abundance correlates with the metabolic status of surrounding adipocytes during tissue maintenance in vitro for 9–11 days. Elevated fatty acid levels lead to significant increases in PPARγ+ populations, which are correlated with the donor’s body mass index (BMI). Conclusions: PPARγ+ macrophages represent a distinctly specialized population of regulatory cells that reside within human IMATs in accordance with their metabolic status. Thus, future in-depth studies on IMAT-resident PPARγ+ macrophage action mechanisms will elucidate the role of skeletal muscle in the pathogenesis of human metabolic dysfunction. Full article
(This article belongs to the Special Issue The Role of Chemerin in Human Disease2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Representative images of skeletal muscle tissue (HE) and IMATs (IF). All images were obtained from Participant P6. (<b>a</b>) HE image showing the human skeletal muscle tissue comprising the areas of SMFs (red arrowhead) and IMATs (green arrowhead). The scale bar (lower right) indicates 1000 µm. (<b>b</b>) IF images were obtained after costaining with DAPI and secondary antibodies as negative controls (negative-488 or 594). The scale bars (upper left) indicate 50 µm. (<b>c</b>–<b>i</b>) IF images of IMATs after costaining with primary antibodies against designated human markers (white, lower left) and the corresponding secondary antibodies and DAPI. The small panels on the left side represent magnified single-cell images labeled with dashed line circles in larger images using IgG488 (green), IgG594 (red), and DAPI (blue) filters. DAPI and IgG594 or DAPI and IgG488 were merged (Merge) to determine the specificity of the detected signals. The white arrowheads indicate verified positive macrophages. The scale bars (upper left) indicate 50 µm.</p>
Full article ">Figure 2
<p>PPARγ<sup>+</sup> macrophages exclusively reside in IMATs. (<b>a</b>) Representative images from a skeletal muscle tissue slice from P2 after IF staining using a primary antibody against PPARγ, IgG488-labeled secondary antibody, and DAPI. The image of brightfield microscopy (middle panel) comprises skeletal muscle fibers (left) and intermuscular adipose tissue with adipocytes (right). Magnified IF images show the labeled areas of skeletal muscle fibers (upper left panel) and intermuscular adipose tissue (lower right panel) exposing a PPARγ<sup>+</sup> macrophage (white arrowhead), respectively. (<b>b</b>) The diagram shows the mean number of PPARγ<sup>+</sup> macrophages (<span class="html-italic">y</span>-axis) in the IMAT and SMF fields of 0.24 mm<sup>2</sup> (<span class="html-italic">x</span>-axis) in donor tissue samples (n = 14). (<b>c</b>) The diagram shows the mean number of PPARγ<sup>+</sup> macrophages (<span class="html-italic">y</span>-axis) relative to 1 mm<sup>2</sup> of IMATs (left <span class="html-italic">y</span>-axis) or relative to the number of adipocytes in 1 mm<sup>2</sup> of IMATs in donor tissue samples (n = 14). The Mann–Whitney test was used to assess the significance of differences in the number of PPARγ<sup>+</sup> macrophages between SMFs and IMATs. <span class="html-italic">p</span> ≤ 0.0001 (****).</p>
Full article ">Figure 3
<p>The numbers of CD80<sup>+</sup> and CD11c<sup>+</sup> macrophages correlate with adipocyte VDAC1 expression in the IMATs of donor samples. Pearson correlation analyses were employed to determine the relationships between the mean VDAC1 expression levels and the mean numbers of CD80<sup>+</sup> (<b>a</b>) and CD11c<sup>+</sup> (<b>b</b>) macrophages in 0.24 mm<sup>2</sup> of IMATs from the donors (n = 14). The correlation coefficients (r) and significance levels (<span class="html-italic">p</span>) for the relationships are presented at the top right of each diagram.</p>
Full article ">Figure 4
<p>The expression levels of IL-23 and IL-31 correlate with adipocyte VDAC1 expression in the IMATs of donor samples. Spearman’s rank correlation analyses were employed to determine the relationships between mean VDAC1 and IL-23 (<b>a</b>) or IL-31 (<b>b</b>) expression levels (n = 12). The correlation coefficients (r) and significance levels (<span class="html-italic">p</span>) for the relationships are presented at the top right of each diagram.</p>
Full article ">Figure 5
<p>Dynamics of the PPARγ<sup>+</sup> macrophage population in IMATs during maintenance in vitro. (<b>a</b>) The diagram shows the mean number of PPARγ<sup>+</sup> macrophages (<span class="html-italic">y</span>-axis) in 0.24 mm<sup>2</sup> of IMAT from all participants (n = 14) before (pre, white bar) and after (post, gray bars) tissue maintenance in vitro. (<b>b</b>) The diagram shows the mean expression of VDAC1 and COXIV (<span class="html-italic">y</span>-axis) in 0.24 mm<sup>2</sup> of IMAT from all participants (n = 14) before (pre, white bars) and after (post, gray bars) tissue maintenance in vitro. A paired t-test or Wilcoxon signed-rank test was applied to evaluate the significance of differences before and after cultivation. <span class="html-italic">p</span> ≤ 0.01 (**). (<b>c</b>,<b>d</b>) Spearman’s rank correlation analyses were applied to determine the relationships between the mean number of PPARγ<sup>+</sup> macrophages and the mean number of CD163<sup>+</sup> (<b>c</b>) or the expression level of COXIV (<b>d</b>) in 0.24 mm<sup>2</sup> of IMAT from all donors (n = 14). The correlation coefficients (r) and significance levels (<span class="html-italic">p</span>) for the relationships are presented at the top right of each diagram.</p>
Full article ">Figure 6
<p>Dynamics of the PPARγ<sup>+</sup> macrophage population in IMATs in response to S-FAs and U-FAs during maintenance in vitro. (<b>a</b>) The diagram shows the relative fold change in PPARγ<sup>+</sup> macrophage numbers (<span class="html-italic">y</span>-axis) in 0.24 mm<sup>2</sup> of IMAT from all participants (n = 14) in response to U-FA or S-FAs before (<span class="html-italic">x</span>-axis) in vitro culture. (<b>b</b>) The diagram shows the relative fold change in the expression of VDAC1 (<span class="html-italic">y</span>-axis) in 0.24 mm<sup>2</sup> of IMAT from all participants (n = 14) in response to U-FA or S-FAs before (<span class="html-italic">x</span>-axis) in vitro culture. One-sample <span class="html-italic">t</span>-tests or Wilcoxon signed-rank tests were used to assess the significance of differences before and after cultivation. <span class="html-italic">p</span> ≤ 0.05 (*). (<b>c</b>,<b>d</b>) Pearson correlation and Spearman’s rank correlation analyses were employed to determine the relationships between the S-FA-mediated relative fold change in the number of PPARγ<sup>+</sup> macrophages ((<b>c</b>), <span class="html-italic">y</span>-axis) or the relative fold change in the expression of VDAC1 ((<b>d</b>), <span class="html-italic">y</span>-axis) and donor BMI (n = 14). The correlation coefficients (r) and significance levels (<span class="html-italic">p</span>) for the relationships are presented at the top right of each diagram.</p>
Full article ">
32 pages, 1556 KiB  
Review
Reactive Oxygen Species, a Potential Therapeutic Target for Vascular Dementia
by Amanda Louise White, Grant M. Talkington, Blake Ouvrier, Saifudeen Ismael, Rebecca J. Solch-Ottaiano and Gregory Bix
Biomolecules 2025, 15(1), 6; https://doi.org/10.3390/biom15010006 (registering DOI) - 25 Dec 2024
Abstract
Vascular dementia (VaD) is a progressive neurodegenerative condition prevalent among elderly adults marked by cognitive decline resulting from injured and/or improperly functioning cerebrovasculature with resultant disruptions in cerebral blood flow. Currently, VaD has no specific therapeutics and the exact pathobiology is still being [...] Read more.
Vascular dementia (VaD) is a progressive neurodegenerative condition prevalent among elderly adults marked by cognitive decline resulting from injured and/or improperly functioning cerebrovasculature with resultant disruptions in cerebral blood flow. Currently, VaD has no specific therapeutics and the exact pathobiology is still being investigated. VaD has been shown to develop when reactive oxygen species (ROS) form from damaged targets at different levels of organization—mitochondria, endothelial cells, or cerebrovasculature. In this review, we highlight how specific ROS molecules may be important in the development of VaD and how they can be targeted as a potential therapeutic for VaD. Full article
Show Figures

Figure 1

Figure 1
<p>Overview of the possible mechanistic link of primary reactive oxygen species in the development of vascular dementia adverse events in the brain (stroke, eNOS uncoupling, and electron leak from the ETC) which lead to the production of ROS that contribute to much of the common pathological damage seen in vascular dementia. eNOS; endothelial nitric oxide synthase. ETC; electron transport chain. H<sub>2</sub>O<sub>2</sub>; hydrogen peroxide. ONOO−; peroxynitrite. O<sub>2</sub>−; superoxide. VaD; vascular dementia.</p>
Full article ">Figure 2
<p>Summary of how brain-mitochondrial-sourced hydrogen peroxide contributions to vascular dementia electron leakage from the electron transport chain (ETC) in the presence of superoxide (O<sub>2</sub>−) lead to the production of hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>). H<sub>2</sub>O<sub>2</sub> can then modify protein and nucleotide structures, leading to various damages. H<sub>2</sub>O<sub>2</sub> can also oxidize LDL, leading to increases in inflammation. The combined effects of high levels of H<sub>2</sub>O<sub>2</sub> promote mitochondrial dysfunction and ultimately result in VaD.</p>
Full article ">Figure 3
<p>Summary of peroxynitrite contributions to vascular dementia in abnormal brain endothelial cells. The reaction between nitric oxide (NO) and superoxide (O<sub>2</sub>−) from eNOS uncoupling leads to the production of peroxynitrite (ONOO−). ONOO− is then able to modify neighboring structures, promote eNOS uncoupling, and inhibit glutathione, preventing its antioxidant activity. Various ONOO− induced damage causes endothelial dysfunction and increases in NOX, and ultimately leads to VaD.</p>
Full article ">Figure 4
<p>Summary of superoxide contributions to vascular dementia in abnormal cerebrovasculature. An ischemic event can lead to the upregulation of NOX enzymes and the production of superoxide (O<sub>2</sub>−) and hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>). O<sub>2</sub>− can promote inflammation and BBB damage. H<sub>2</sub>O<sub>2</sub> can prolong the ischemic state, leading to further injury. Ultimately, the damage induced by O<sub>2</sub>− and H<sub>2</sub>O<sub>2</sub> leads to VaD.</p>
Full article ">
19 pages, 8543 KiB  
Article
ATAD1 Regulates Neuronal Development and Synapse Formation Through Tuning Mitochondrial Function
by Hao-Hao Yan, Jia-Jia He, Chuanhai Fu, Jia-Hui Chen and Ai-Hui Tang
Int. J. Mol. Sci. 2025, 26(1), 44; https://doi.org/10.3390/ijms26010044 (registering DOI) - 24 Dec 2024
Abstract
Mitochondrial function is essential for synaptic function. ATAD1, an AAA+ protease involved in mitochondrial quality control, governs fission–fusion dynamics within the organelle. However, the distribution and functional role of ATAD1 in neurons remain poorly understood. In this study, we demonstrate that ATAD1 is [...] Read more.
Mitochondrial function is essential for synaptic function. ATAD1, an AAA+ protease involved in mitochondrial quality control, governs fission–fusion dynamics within the organelle. However, the distribution and functional role of ATAD1 in neurons remain poorly understood. In this study, we demonstrate that ATAD1 is primarily localized to mitochondria in dendrites and, to a lesser extent, in spines in cultured hippocampal neurons. We found that ATAD1 deficiency disrupts the mitochondrial fission–fusion balance, resulting in mitochondrial fragmentation. This deficiency also impairs dendritic branching, hinders dendritic spine maturation, and reduces glutamatergic synaptic transmission in hippocampal neuron. To further investigate the underlying mechanism, we employed an ATP hydrolysis-deficient mutant of ATAD1 to rescue the neuronal deficits associated with ATAD1 loss. We discovered that the synaptic deficits are independent of the mitochondrial morphology changes but rely on its ATP hydrolysis. Furthermore, we show that ATAD1 loss leads to impaired mitochondrial function, including decreased ATP production, impaired membrane potential, and elevated oxidative stress. In conclusion, our results provide evidence that ATAD1 is crucial for maintaining mitochondrial function and regulating neurodevelopment and synaptic function. Full article
(This article belongs to the Special Issue New Insights into Mitochondria in Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>ATAD1 colocalizes with mitochondria. (<b>A</b>) Representative images of PSD-95 (green) and ATAD1 staining (red). Boxed regions are enlarged in inserts. Scale bar 10 μm. (<b>B</b>) Quantification of PSD-95-ATAD1 colocalization using local correlation (<span class="html-italic">n</span> = 9 cells/3 cultures). (<b>C</b>) Representative images of GABA<sub>A</sub>R (green) and ATAD1 staining (red). Boxed regions are enlarged in inserts. Scale bar 10 μm. (<b>D</b>) Quantification of GABA<sub>A</sub>R-ATAD1 colocalization using local correlation (<span class="html-italic">n</span> = 4 cells/2 cultures). (<b>E</b>) Representative images of neuron morphology and ATAD1 distribution in cultured rat hippocampal neurons co-expressing oe-ATAD1 (green) and mCherry (red). Boxed regions are enlarged in inserts. Scale bar 10 μm. (<b>F</b>) Correlation between ATAD1 length and dendrite length (<span class="html-italic">n</span> = 13 cells/3 cultures). (<b>G</b>) Quantification of the percent of ATAD1-containing spines along dendrites (<span class="html-italic">n</span> = 13 cells/3 cultures). (<b>H</b>) Representative colocalization of ATAD1 (green) and mitochondrial (red) in cultured neurons co-expressing oe-ATAD1 and mito-Dsred. Boxed regions are enlarged in inserts. Scale bar 10 μm. (<b>I</b>) Colocalization of ATAD1 and mito-Dsred in cultured rat hippocampal neurons. (<b>J</b>) Pearson coefficient of ATAD1 and mito-Dsred (<span class="html-italic">n</span> = 9 cells/3 cultures).</p>
Full article ">Figure 2
<p>ATAD1 deficiency leads to mitochondrial fragmentation in neurons. (<b>A</b>) Representative images of mitochondrial morphology in shCon-expressing, shATAD1-expressing or oe-ATAD1-expressing neurons. Scale bar 5 μm. Boxed regions are enlarged in inserts. Scale bar 5 μm. (<b>B</b>) Quantification of individual mitochondrial length along dendrites (<span class="html-italic">n</span> = 10–17 cells/3–5 cultures, <span class="html-italic">p</span> &lt; 0.0001). (<b>C</b>,<b>D</b>) The correlation between number of mitochondria and total dendrite length (<b>C</b>), and quantification of the mitochondrial density (<b>D</b>) (<span class="html-italic">n</span> = 10–17 cells/3–5 cultures, <span class="html-italic">p</span> &lt; 0.0001) along dendrites. (<b>E</b>,<b>F</b>) The correlation between total mitochondrial length and total dendrite length (<b>E</b>), and relative length ratio (<b>F</b>) (<span class="html-italic">n</span> = 10–17 cells/3–5 cultures, <span class="html-italic">p</span> = 0.2797). Significance was assessed by one-way ANOVA measures (<b>D</b>), Welch ANOVA (<b>F</b>), or Kruskal–Wallis test (<b>B</b>). All data are presented as the mean ± s.e.m. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001; ns., not significant.</p>
Full article ">Figure 3
<p>ATAD1 deficiency leads to impaired neurodevelopment in cultured hippocampal neurons. (<b>A</b>) Schematic timeline for transfection experimental design of cultured rat hippocampal neurons. (<b>B</b>) Representative images of neuronal morphology and dendritic branching in shCon-expressing, shATAD1-expressing, or oe-ATAD1-expressing neurons in DIV10. Scale bar 100 μm. (<b>C</b>,<b>D</b>) Quantification of primary dendrites number and dendritic intersection number in neurons expressing shCon, shATAD1 or oe-ATAD1 at DIV10 (<span class="html-italic">n</span> = 10–16 cells/3 cultures, (<b>C</b>), <span class="html-italic">p</span> = 0.001, (<b>D</b>), <span class="html-italic">p</span> = 0.001). Significance was assessed by one-way ANOVA measures (<b>C</b>), or two-way ANOVA measures (<b>D</b>). All data are presented as the mean ± s.e.m. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>ATAD1 knockdown reduces neuronal maturity in cultured hippocampal neurons. (<b>A</b>) Schematic timeline for transfection experimental design in cultured rat hippocampal neurons. (<b>B</b>) Representative images of neuronal morphology and dendritic spines in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1. Scale bar 5 μm. Boxed regions are enlarged in inserts. Scale bar 5 μm. (<b>C</b>) Quantification of neuronal dendritic spines density in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1. (<span class="html-italic">n</span> = 11–16 cells/3–5 cultures, <span class="html-italic">p</span> &lt; 0.0001). (<b>D</b>–<b>G</b>) Quantification of mushroom density (<b>D</b>), stubby density (<b>E</b>), thin density (<b>F</b>) and filopodia density (<b>G</b>) in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1 (<span class="html-italic">n</span> = 11–16 cells/3–5 cultures, (<b>D</b>), <span class="html-italic">p</span> &lt; 0.0001, (<b>E</b>), <span class="html-italic">p</span> &lt; 0.0001, (<b>F</b>), <span class="html-italic">p</span> = 0.0117, (<b>G</b>), <span class="html-italic">p</span> = 0.2664). (<b>H</b>–<b>J</b>) Quantification of distribution of spine morphologies in cultured rat hippocampal neurons expressing shCon (<b>H</b>), shATAD1 (<b>I</b>) or oe-ATAD1 (<b>J</b>) (<span class="html-italic">n</span> = 11–16 cells/3–5 cultures). Significance was assessed by one-way ANOVA measures (<b>C</b>,<b>D</b>,<b>F</b>,<b>G</b>), or Kruskal–Wallis test (<b>E</b>). All data are presented as the mean ± s.e.m. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>ATAD1 knockdown reduces synaptic transmission in cultured hippocampal neurons. (<b>A</b>) Representative images of PSD-95 staining (red) in cultured hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1. Scale bar 1 μm. (<b>B</b>) Quantification of PSD-95 density along dendrites in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1 (<span class="html-italic">n</span> = 9–13 cells/3 cultures, <span class="html-italic">p</span> &lt; 0.0001). (<b>C</b>) Quantification of PSD-95 indensity (right) and cumulative probability curve (left) in expressing shCon, shATAD1 or oe-ATAD1 (<span class="html-italic">n</span> = 9–13 cells/3 cultures, left, shCon VS. shATAD1 **** <span class="html-italic">p</span> &lt; 0.0001, shCon VS. oe-ATAD1, **** <span class="html-italic">p</span> &lt; 0.0001, right, <span class="html-italic">p</span> &lt; 0.0001). (<b>D</b>) Representative traces of mEPSC in neurons expressing shCon, shATAD1 or oe-ATAD1. (<b>E</b>) Quantification of mEPSC amplitude (right) and cumulative probability curve (left) in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1 (<span class="html-italic">n</span> = 12–21 cells/3 cultures, left, shCon VS. shATAD1, * <span class="html-italic">p</span> &lt; 0.0301, shCon VS. oe-ATAD1, * <span class="html-italic">p</span> = 0.0497, right, <span class="html-italic">p</span> &lt; 0.0001). (<b>F</b>) Quantification of mEPSC frequency in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1 (<span class="html-italic">n</span> = 12–21 cells/3 cultures, <span class="html-italic">p</span> = 0.0004). (<b>G</b>) Quantification of mEPSC inter-event interval (right) and cumulative probability curve (left) in cultured rat hippocampal neurons expressing shCon, shATAD1 or oe-ATAD1. (<span class="html-italic">n</span> = 12–21 cells/3 cultures, left, shCon VS. shATAD1, **** <span class="html-italic">p</span> &lt; 0.0001; shCon VS. oe-ATAD1, **** <span class="html-italic">p</span> &lt; 0.0001, right, <span class="html-italic">p</span> &lt; 0.0001). Significance was assessed by one-way ANOVA measures (<b>B</b>), Kruskal-Wallis test (<b>C</b>,<b>E</b>–<b>G</b>), or K-S test (<b>C</b>,<b>E</b>,<b>G</b>). All data are presented as the mean ± s.e.m. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>The hydrolysis site mutant of ATAD1 is unable to rescue the reduction in synaptic density. (<b>A</b>) Representative images of spine (Green) and mitochondrial morphology (red) in shCon-expressing, E193Q::shATAD1-expressing (E193Q and shATAD1-coexpressing) or shATAD1-expressing neurons. Scale bar 5 μm. (<b>B</b>–<b>D</b>) Quantification of individual mitochondrial length (<b>B</b>), mitochondrial density (<b>C</b>) and relative length ratio (<b>E</b>) (<span class="html-italic">n</span> = 12–14 cells/3 cultures, (<b>B</b>), <span class="html-italic">p</span> = 0.0154, (<b>C</b>), <span class="html-italic">p</span> = 0.0017, (<b>D</b>), <span class="html-italic">p</span> = 0.0887). (<b>E</b>) Quantification of neuronal dendritic spines density in cultured rat hippocampal neurons expressing shCon-expressing, E193Q::shATAD1-expressing or shATAD1-expressing (<span class="html-italic">n</span> = 12–14 cells/3 cultures, <span class="html-italic">p</span> = 0.0018). Significance was assessed by one-way ANOVA measures (<b>B</b>–<b>E</b>). All data are presented as the mean ± s.e.m. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ns., not significant.</p>
Full article ">Figure 7
<p>ATAD1 deficiency disrupts mitochondrial function in cultured hippocampal neurons. (<b>A</b>) Representative images of DIOC6 staining (green, labeling mitochondrial membrane potential, ΔΨ) in shCon-expressing-control, shATAD1-expressing and oe-ATAD1-expressing neuron. Scale bar 1 μm. (<b>B</b>) Quantification of DIOC6 fluorescence intensity in shCon-expressing-control, shATAD1-expressing and oe-ATAD1-expressing neurons (<span class="html-italic">n</span> = 41–57 mitochondria /3 cultures, <span class="html-italic">p</span> &lt; 0.0001). (<b>C</b>) Representative images of DHR123 staining (green, labeling mitochondrial ROS) in shCon-expressing-control, shATAD1-expressing and oe-ATAD1-expressing neurons. Scale bar 1 μm. (<b>D</b>) Quantification of DHR123 fluorescence intensity (<span class="html-italic">n</span> = 41–57 mitochondria/3 cultures, <span class="html-italic">p</span> &lt; 0.0001). (<b>E</b>) Representative images of ATP-Red1 staining (red, labeling mitochondrial ATP) in shCon-expressing-control, shATAD1-expressing and oe-ATAD1-expressing neuron. Scale bar 1 μm. (<b>F</b>) Quantification of ATP-Red1 fluorescence intensity (<span class="html-italic">n</span> = 39–47 mitochondria/3 cultures; <span class="html-italic">p</span> &lt; 0.0001). Significance was assessed by Kruskal–Wallis test (<b>B</b>,<b>D</b>,<b>F</b>). All data are presented as the mean ± s.e.m. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
17 pages, 4554 KiB  
Article
Cytotoxic Activity of Curcumin- and Resveratrol-Loaded Core–Shell Systems in Resistant and Sensitive Human Ovarian Cancer Cells
by Joanna Weżgowiec, Zofia Łapińska, Łukasz Lamch, Anna Szewczyk, Jolanta Saczko, Julita Kulbacka, Mieszko Więckiewicz and Kazimiera A. Wilk
Int. J. Mol. Sci. 2025, 26(1), 41; https://doi.org/10.3390/ijms26010041 - 24 Dec 2024
Abstract
Due to the high mortality rate of ovarian cancer, there is a need to find novel strategies to improve current treatment modalities. Natural compounds offer great potential in this field but also require the careful design of systems for their delivery to cancer [...] Read more.
Due to the high mortality rate of ovarian cancer, there is a need to find novel strategies to improve current treatment modalities. Natural compounds offer great potential in this field but also require the careful design of systems for their delivery to cancer cells. Our study explored the anticancer effects of novel resveratrol (RSV)- and curcumin (CUR)-loaded core–shell nanoparticles in human ovarian cancer cells. We evaluated the in vitro cytotoxicity of various nanocarriers (CUR 1-3, RSV I-III) delivered to MDAH-2774 and SKOV-3 cells in comparison to free RVS and CUR after 24 h and 72 h treatment. A two-way ANOVA was applied to compare the results of the MTT assay. Confocal laser scanning microscopy was employed to visualize cellular uptake and mitochondrial localization. Our findings revealed that the cytotoxicity of the core–shell nanoparticles with RSV was not significant, but the systems loaded with CUR effectively decreased the viability of cells. The MDAH-2774 cell line was more sensitive to the treatment than SKOV-3. The enhanced cellular uptake of CUR delivered by core–shell systems and its colocalization with mitochondria were demonstrated. Further research focused on the detailed biological effects of the most effective systems (CUR 2 and CUR 3) should be conducted to provide detailed insights. These findings highlight the promising role of CUR-loaded nanoparticles in ovarian cancer treatment. Full article
(This article belongs to the Special Issue Targeted Therapies and Molecular Methods in Cancer, 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>The cytotoxicity of free resveratrol (<b>A</b>,<b>E</b>) and resveratrol loaded in the core–shell systems (<b>B</b>–<b>D</b>,<b>F</b>–<b>H</b>), evaluated using an MTT assay based on the mitochondrial activity after 24 h (<b>A</b>–<b>D</b>) and 72 h (<b>E</b>–<b>H</b>) in SKOV-3 and MDAH-2774 ovarian cancer cell lines; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>The cytotoxicity of free curcumin (<b>A</b>,<b>E</b>) and curcumin loaded in the core–shell systems (<b>B</b>–<b>D</b>,<b>F</b>–<b>H</b>), evaluated using MTT assay based on the mitochondrial activity after 24 h (<b>A</b>–<b>D</b>) and 72 h (<b>E</b>–<b>H</b>) in SKOV-3 and MDAH-2774 ovarian cancer cell lines; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Confocal laser scanning microscopy (CLSM) visualization of mitochondria (in red) after 24 h of incubation of SKOV-3 and MDAH-2774 ovarian cancer cells with free resveratrol and resveratrol loaded in the core–shell systems.</p>
Full article ">Figure 4
<p>Confocal laser scanning microscopy (CLSM) visualization of mitochondria (in red, MT633) and intracellular localization of CUR (in green, AF430) after 24 h of incubation of SKOV-3 and MDAH-2774 ovarian cancer cells with free curcumin and curcumin loaded in the core–shell systems; graphs present mean intensity of fluorescence emitted from CUR.</p>
Full article ">
24 pages, 9420 KiB  
Article
Changes in the Phenotype and Metabolism of Peritoneal Macrophages in Mucin-2 Knockout Mice and Partial Restoration of Their Functions In Vitro After L-Fucose Treatment
by Elena L. Arzhanova, Yulia Makusheva, Elena G. Pershina, Snezhanna S. Medvedeva and Ekaterina A. Litvinova
Int. J. Mol. Sci. 2025, 26(1), 13; https://doi.org/10.3390/ijms26010013 - 24 Dec 2024
Abstract
In the development of inflammatory bowel disease (IBD), peritoneal macrophages contribute to the resident intestinal macrophage pool. Previous studies have demonstrated that oral administration of L-fucose exerts an immunomodulatory effect and repolarizes the peritoneal macrophages in vivo in mice. In this study, we [...] Read more.
In the development of inflammatory bowel disease (IBD), peritoneal macrophages contribute to the resident intestinal macrophage pool. Previous studies have demonstrated that oral administration of L-fucose exerts an immunomodulatory effect and repolarizes the peritoneal macrophages in vivo in mice. In this study, we analyzed the phenotype and metabolic profile of the peritoneal macrophages from Muc2−/− mice, as well as the effect of L-fucose on the metabolic and morphological characteristics of these macrophages in vitro. The investigation utilized flow cytometry, quantitative PCR (qPCR), measurement of the intracellular ATP and Ca2+ concentrations, an analysis of mitochondrial respiration and membrane potential, and transmission electron microscopy (TEM) for ultrastructural evaluations. The Muc2−/− mice exhibited lower intracellular ATP and Ca2+ levels in their peritoneal macrophages, a higher percentage of stellate macrophages, and an increased oxygen consumption rate (OCR), combined with a higher percentage of mitochondria displaying an abnormal ultrastructure. Additionally, there was a five-fold increase in condensed mitochondria compared to their level in C57BL/6 mice. The number of CD209+ peritoneal macrophages was reduced three-fold, while the number of M1-like cells increased two-fold in the Muc2−/− mice. L-fucose treatment enhanced ATP production and reduced the expression of the Parp1, Mt-Nd2, and Mt-Nd6 genes, which may suggest a reduction in pro-inflammatory factor production and a shift in the differentiation of peritoneal macrophages towards the M2 phenotype. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Comparison of marker expression, shapes, OCR, and ECAR of peritoneal macrophages derived from C57BL/6 and <span class="html-italic">Muc2<sup>−/−</sup></span> mice. (<b>A</b>). Percentage of CD209<sup>+</sup> macrophages (M2-like type) from two mouse strains. (<b>B</b>). Percentage of CD80<sup>+</sup> macrophages (M1-like type) from two mouse strains. (<b>C</b>). Percentage of CD206<sup>+</sup> macrophages (M2-like type) from two mouse strains. (<b>D</b>). Percentage of CD86<sup>+</sup> macrophages (M1-like type) from two mouse strains. (<b>E</b>). Percentage of different shapes of macrophages from two mouse strains. (<b>F</b>). The OCRs were measured following treatment with oligomycin, FCCP, and the antimycin A/rotenone complex; (<b>G</b>). Seahorse assay scheme showing the types of respiration. (<b>H</b>). Means of different types of respiration: basal, ATP-linked, proton leak, maximal respiration, and reserve capacity; (<b>I</b>). The ECAR was measured following the treatment with oligomycin, FCCP, and the antimycin A/rotenone complex. (<b>J</b>). Means of basal glycolysis and glycolytic capacity. “C57BL/6” vs. “<span class="html-italic">Muc2<sup>−/−</sup></span>”: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 according to the PERMANOVA test.</p>
Full article ">Figure 2
<p>Ultrastructures of macrophages from the lamina propria of colons derived from C57BL/6 and <span class="html-italic">Muc2<sup>−/−</sup></span> mice. (<b>A</b>). Morphology of the macrophages from the two mouse strains. <span class="html-italic">Muc2<sup>−/−</sup></span> mice macrophages have a lot of altered mitochondria. (<b>B</b>). Mitochondria with “empties” (white arrows show the “empties”) (<b>C</b>). Different structural defects and functional types of ultrastructures in the mitochondria in the lamina propria of the colons of the <span class="html-italic">Muc2<sup>−/−</sup></span> mice, white arrows show the defects (<b>D</b>). The number of mitochondria in 1 μm<sup>2</sup> of the cytoplasm. (<b>E</b>). Percentages of mitochondria with normal and altered ultrastructures in <span class="html-italic">Muc2<sup>−/−</sup></span> (dark blue) and C57BL/6 (light blue) mice (<b>F</b>). Percentages of mitochondria with empty spaces and the number of cristae per mitochondria; “C57BL/6” vs. “<span class="html-italic">Muc2<sup>−/−</sup></span>”; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001; <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Ultrastructure and mitochondrial membrane potential of peritoneal macrophages derived from C57BL/6 and <span class="html-italic">Muc2<sup>−/−</sup></span> mice. (<b>A</b>). Morphology of the peritoneal macrophages from the two mouse strains. white arrows show dilated cristae in condensed mitochondria. (<b>B</b>). Morphology of the mitochondria in the peritoneal macrophages from the two mouse strains. (<b>C</b>). Size of peritoneal macrophages and the mitochondria in them in the two mouse strains. (<b>D</b>). Percentages of mitochondria with normal and altered ultrastructures in the <span class="html-italic">Muc2<sup>−/−</sup></span> (dark blue) and C57BL/6 (light blue) mice. (<b>E</b>). A FACS graph with the percentage of peritoneal macrophages with a different mitochondrial membrane potential (ΔѰm) analyzed using JC-1 staining. (<b>F</b>). Percentages of peritoneal macrophages with a high and low mitochondrial membrane potential (ΔѰm). “C57BL/6” vs. “<span class="html-italic">Muc2<sup>−/−</sup></span>”; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>Addition of fucose to the cell culture medium affected the production of cytokines, ATP and Ca<sup>2+</sup> levels, and surface and intracellular expression of CD38 of peritoneal macrophages derived from C57BL/6 and <span class="html-italic">Muc2<sup>−/−</sup></span> mice. (<b>A</b>). Inflammatory cytokine levels in cell culture medium of peritoneal macrophages from the two mouse strains with and without the addition of 0.1% L-fucose; the median, min, and max for each cytokine is presented in pg/mg protein. (<b>B</b>). ATP levels in the peritoneal macrophages of two mouse strains incubated with and without 0.1% L-fucose. (<b>C</b>). Ca<sup>2+</sup> levels in the peritoneal macrophages of the two mouse strains incubated with and without 0.1% L-fucose. (<b>D</b>). Percentage of peritoneal macrophages with surface CD38 expression from the two mouse strains incubated with and without 0.1% L-fucose. (<b>E</b>). Percentage of peritoneal macrophages with intracellular CD38 expression from the two mouse strains incubated with and without 0.1% L-fucose. “C57BL/6” vs. “<span class="html-italic">Muc2<sup>−/−</sup></span>“ and “with 0.1% L-fucose” vs. “without 0.1% L-fucose”: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. Two-way PERMANOVA test.</p>
Full article ">Figure 5
<p>Effect of fucose on the expression of the <span class="html-italic">Parp1</span>, <span class="html-italic">Parg</span>, <span class="html-italic">Il1b</span>, <span class="html-italic">Ltc4</span>, <span class="html-italic">Mt-nd2</span>, and <span class="html-italic">Mt-nd6</span> genes and on Tlr2 and Tlr4 expression on the surface of the peritoneal macrophages. (<b>A</b>). Expression of the <span class="html-italic">Parp1</span> gene in peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>B</b>). Expression of the <span class="html-italic">Parg</span> gene in peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>C</b>). Expression of the <span class="html-italic">Il1b</span> gene in peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>D</b>). Expression of the <span class="html-italic">Ltc4</span> gene in peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>E</b>). Expression of the <span class="html-italic">Mt-nd2</span> gene in peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>F</b>). Expression of the <span class="html-italic">Mt-nd6</span> gene in peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>G</b>). Percentage of Tlr2-positive peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. (<b>H</b>). Percentage of Tlr4-positive peritoneal macrophages from the two mouse strains incubated with and without 0.1% L-fucose. “C57BL/6” vs. “<span class="html-italic">Muc2<sup>−/−</sup></span>” and “with 0.1% L-fucose” vs. “without 0.1% L-fucose”: * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01. Two-way PERMANOVA test.</p>
Full article ">
21 pages, 3589 KiB  
Article
In Vitro Toxicity of a DEHP and Cadmium Mixture on Sheep Cumulus–Oocyte Complexes
by Antonella Mastrorocco, Letizia Temerario, Valeria Vurchio, Susanna Cotecchia, Nicola Antonio Martino and Maria Elena Dell’Aquila
Int. J. Mol. Sci. 2025, 26(1), 5; https://doi.org/10.3390/ijms26010005 - 24 Dec 2024
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) and Cadmium (Cd) affect female reproduction. To date, toxicological research has focused on the effects of individual contaminants, whereas living beings are exposed to mixtures. This study analyzed the effects of a DEHP/Cd mixture on nuclear and cytoplasmic maturation of [...] Read more.
Di-(2-ethylhexyl) phthalate (DEHP) and Cadmium (Cd) affect female reproduction. To date, toxicological research has focused on the effects of individual contaminants, whereas living beings are exposed to mixtures. This study analyzed the effects of a DEHP/Cd mixture on nuclear and cytoplasmic maturation of sheep cumulus–oocyte complexes (COCs) compared with single compounds. COCs recovered from slaughterhouses-derived sheep ovaries were in vitro exposed to 0.5 μM DEHP, 0.1 μM Cd, or DEHP/Cd mixture at the same concentrations during 24 h of in vitro maturation (IVM). After IVM, oocyte nuclear chromatin configuration was evaluated, and bioenergetic/oxidative parameters were assessed on expanded cumulus cells (CCs) and matured oocytes (chi-square test and one-way ANOVA; p < 0.05). Under examined conditions, oocyte nuclear maturation was never impaired. However, COC bioenergetics was affected with stronger effects for the mixture than single compounds. Indeed, the percentages of matured oocytes with healthy mitochondrial distribution patterns were reduced (p < 0.001 and p < 0.05 for mixture and single compounds, respectively). Oocyte mitochondrial membrane potential, intracellular ROS levels, and mitochondria/ROS co-localization were reduced, with the same significance level, in all contaminated conditions. CCs displayed increased ROS levels only upon mixture exposure (p < 0.001). In conclusion, in vitro exposure to the DEHP/Cd mixture affected COC quality in the sheep to a greater extent than separate compounds. Full article
Show Figures

Figure 1

Figure 1
<p>Dose-dependence curve of the in vitro effects of DEHP on mitochondrial membrane potential (ΔΨm), intracellular ROS levels, and mitochondrial/ROS co-localization in single metaphase II stage oocytes expressed as MitoTracker Orange CMTM Ros (<b>A</b>) and DCF (<b>B</b>) fluorescence intensities and overlap coefficients of MitoTracker Orange CMTM Ros and DCF fluorescent labeling (<b>C</b>). Values are means ± standard deviations and are expressed as percentages of vehicle CTRL. Numbers of analyzed oocytes per group are indicated on the bottom of each histogram. One-way ANOVA test followed by Tukey’s post hoc test, comparisons among groups; * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 for DEHP-exposed vs. vehicle CTRL.</p>
Full article ">Figure 2
<p>Effects of DEHP/Cd mixture and individual compounds on mitochondrial membrane potential (ΔΨm), intracellular ROS levels, and mitochondrial/ROS co-localization in single metaphase II stage oocytes expressed as MitoTracker Orange CMTM Ros (<b>A</b>) and DCF (<b>B</b>) fluorescence intensities and overlap coefficients of MitoTracker Orange CMTM Ros and DCF fluorescent labeling (<b>C</b>). Values are means ± standard deviations of examined oocytes and are presented as percentages of vehicle CTRL. Numbers of analyzed oocytes per experimental condition are indicated at the bottom of each bar. One-way ANOVA test followed by Tukey’s post hoc test, different superscripts indicate statistically significant differences: comparisons among groups; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 for exposed vs. vehicle CTRL.</p>
Full article ">Figure 3
<p>Mitochondrial distribution pattern and ROS localization in sheep matured oocytes exposed to DEHP/Cd mixture, DEHP and Cd. For each oocyte, corresponding UV light (blue pictures in (<b>A1</b>),(<b>B1</b>),(<b>C1</b>),(<b>D1</b>)) and confocal laser scanning images showing mitochondrial distribution pattern (red pictures in (<b>A2</b>),(<b>B2</b>),(<b>C2</b>),(<b>D2</b>)), intracellular ROS localization (green pictures in (<b>A3</b>),(<b>B3</b>),(<b>C3</b>),(<b>D3</b>)) and mitochondria/ROS merge (orange pictures in (<b>A4</b>),(<b>B4</b>),(<b>C4</b>),(<b>D4</b>)) are shown. Oocytes are representative of heterogeneous (perinuclear/subplasmalemmal; (<b>A</b>) and homogeneous (<b>B</b>–<b>D</b>) mitochondrial distribution patterns, respectively. White scale bar represents 40 µm.</p>
Full article ">Figure 4
<p>Effects of DEHP/Cd mixture on mitochondrial membrane potential (ΔΨm), intracellular ROS levels, and mitochondrial/ROS co-localization in CCs from sheep COCs expressed as MitoTracker Orange CMTM Ros (panel (<b>A</b>)) and DCF (panel (<b>B</b>)) fluorescence intensities and overlap coefficient of mitochondria/ROS co-localization (panel (<b>C</b>)). Values are means ± standard deviations of examined fields and are expressed as percentages of vehicle CTRL. Around 200 CCs per experimental condition were analyzed by LSCM. One-way ANOVA test followed by Tukey’s post hoc test: comparisons among groups; different superscripts indicate statistically significant differences: *** <span class="html-italic">p</span> &lt; 0.001 for DEHP/Cd mixture-exposed vs. vehicle CTRL, 0.5 µM DEHP, and 0.1 µM Cd.</p>
Full article ">Figure 5
<p>Photomicrographs of representative CCs from COCs of juvenile ewes matured in vitro in presence/absence of DEHP and Cd. Lanes show representative CC fields taken by laser scanning confocal microscopy. In each field, all CCs included in the yellow defined boundary underwent quantification analysis of mitochondrial membrane potential, ROS levels, and mt/ROS co-localization, whose results are presented in <a href="#ijms-26-00005-f004" class="html-fig">Figure 4</a>. In columns 1 and 2, cells stained with MitoTracker Orange and DCF are shown, respectively, whereas column 3 shows the mitochondrial/ROS merge. Increased intracellular ROS levels (expressed as DCF fluorescent intensity) can be seen in the CCs exposed to DEHP/Cd mixture (<b>B2</b>) compared with the controls (<b>A2</b>). White scale bars represent 40 µm.</p>
Full article ">
31 pages, 41731 KiB  
Article
Hepatitis C Virus NS5A Activates Mitophagy Through Cargo Receptor and Phagophore Formation
by Yuan-Chao Hsiao, Chih-Wei Chang, Chau-Ting Yeh and Po-Yuan Ke
Pathogens 2024, 13(12), 1139; https://doi.org/10.3390/pathogens13121139 - 23 Dec 2024
Abstract
Chronic HCV infection is a risk factor for end-stage liver disease, leading to a major burden on public health. Mitophagy is a specific form of selective autophagy that eliminates mitochondria to maintain mitochondrial integrity. HCV NS5A is a multifunctional protein that regulates the [...] Read more.
Chronic HCV infection is a risk factor for end-stage liver disease, leading to a major burden on public health. Mitophagy is a specific form of selective autophagy that eliminates mitochondria to maintain mitochondrial integrity. HCV NS5A is a multifunctional protein that regulates the HCV life cycle and may induce host mitophagy. However, the molecular mechanism by which HCV NS5A activates mitophagy remains largely unknown. Here, for the first time, we delineate the dynamic process of HCV NS5A-activated PINK1/Parkin-dependent mitophagy. By performing live-cell imaging and CLEM analyses of HCV NS5A-expressing cells, we demonstrate the degradation of mitochondria within autophagic vacuoles, a process that is dependent on Parkin and ubiquitin translocation onto mitochondria and PINK1 stabilization. In addition, the cargo receptors of mitophagy, NDP52 and OPTN, are recruited to the mitochondria and required for HCV NS5A-induced mitophagy. Moreover, ATG5 and DFCP1, which function in autophagosome closure and phagophore formation, are translocated near mitochondria for HCV NS5A-induced mitophagy. Furthermore, autophagy-initiating proteins, including ATG14 and ULK1, are recruited near the mitochondria for HCV NS5A-triggered mitophagy. Together, these findings demonstrate that HCV NS5A may induce PINK1/Parkin-dependent mitophagy through the recognition of mitochondria by cargo receptors and the nascent formation of phagophores close to mitochondria. Full article
(This article belongs to the Special Issue Pathogenesis of Viral Hepatitis)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>HCV NS5A induces the engulfment of mitochondria within autophagic vacuoles: (<b>A</b>) Huh7 cells were transduced with lentiviruses expressing RFP-LC3 and Mito-GFP, as described in the “Materials and Methods” section, to generate Huh7/RFP-LC3/Mito-GFP cells. Then, Huh7/RFP-LC3/Mito-GFP cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The degree of colocalization between RFP-LC3-labeled autophagic vacuoles and Mito-GFP-expressing mitochondria was quantified. The data are presented as means ± SEMs (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the sequestration of Mito-GFP-labeled mitochondria by RFP-LC3 puncta. (<b>D</b>–<b>G</b>) CLEM analysis of mitochondrial sequestration by autophagic vacuoles in HCV NS5A-expressing cells. (<b>D</b>) Huh7/RFP-LC3/Mito-GFP cells were transduced with lentiviruses expressing HCV NS5A-mTagBFP2 for forty-eight hours and then processed for confocal microscopy. (<b>E</b>) The assembled Z-stacks of the confocal micrographs in (<b>D</b>) were reconstituted into a 3-D image. The white dashed boxes indicate the engulfment of Mito-GFP-expressing mitochondria within RFP-LC3 puncta. (<b>F</b>) The aligned image of the confocal micrograph (IF) and electron micrograph (EM) from the CLEM analysis of cells in (<b>D</b>) is shown. The white dashed boxes in the left panel are enlarged and shown in the magnified images in the right panel. The white arrowheads indicate the sequestration of mitochondria within autophagic vacuoles. (<b>G</b>) The enlarged images show the magnified white dashed boxes in the EM of (<b>F</b>). The white arrows indicate the phagophores wrapped around deformed mitochondria.</p>
Full article ">Figure 2
<p>HCV NS5A induces the translocation of Parkin to mitochondria: (<b>A</b>) Huh7 cells were transduced with lentiviruses harboring RFP-Parkin and Mito-GFP, according to the procedure described in the “Materials and Methods” section, to establish Huh7/RFP-Parkin/Mito-GFP cells. Huh7/RFP-LC3/Mito-GFP cells were transduced with (+) or without (−) pTRIP-HCV NS5A-miRFP670 lentiviruses for forty-eight hours and then analyzed via confocal microscopy. (<b>B</b>) The degree of colocalization between RFP-LC3-Parkin and Mito-GFP-labeled mitochondria was quantified. The data are presented as means ± SEMs (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the translocation of RFP-Parkin to Mito-GFP-expressing mitochondria. (<b>D</b>–<b>F</b>) CLEM analysis of the mitochondrial translocation of RFP-Parkin in HCV NS5A-expressing cells. (<b>D</b>) Huh7/RFP-Parkin/Mito-GFP cells were transduced with lentiviruses expressing HCV NS5A-miRFP670. Forty-eight hours later, the cells were analyzed via confocal microscopy. (<b>E</b>) The Z-stacks of the confocal micrograph shown in (<b>D</b>) were assembled and deconvoluted into a 3-D image. The white dashed boxes indicate the Mito-GFP-expressing mitochondria with RFP-Parkin translocation. (<b>F</b>) The aligned IF and CLEM image of the cells from (<b>D</b>) is presented. The white dashed boxes in the left panel are enlarged and shown in the magnified images in the right panel. The white arrowheads indicate the degradative mitochondria in which Parkin translocates.</p>
Full article ">Figure 3
<p>HCV NS5A induces mitolysosome formation: (<b>A</b>) Huh7 cells were transduced with pTRIP-Mito-QC lentiviruses, as described in the “Materials and Methods” section, generating Huh7/Mito-QC cells. Huh7/Mito-QC cells were transduced with (+) or without (−) pTRIP-HCV NS5A-miRFP670 lentiviruses. After forty-eight hours, the cells were analyzed via confocal microscopy. (<b>B</b>) The number of RFP<sup>+</sup>/GFP<sup>−</sup> mitolysosomes was quantified with Image J, as described previously [<a href="#B33-pathogens-13-01139" class="html-bibr">33</a>]. The data are presented as means ± SEMs (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the formation of RFP<sup>+</sup>/GFP<sup>−</sup> mitolysosomes. (<b>D</b>–<b>F</b>) CLEM analysis of mitolysosome formation in HCV NS5A-expressing cells. (<b>D</b>) Huh7/Mito-QC cells were transduced with lentiviruses expressing HCV NS5A-miRFP670 for forty-eight hours and then processed for confocal microscopy. (<b>E</b>) The Z-stacks of the confocal micrograph shown in (<b>D</b>) were assembled and deconvoluted into a 3-D image. The white dashed boxes indicate the loci of RFP<sup>+</sup>/GFP<sup>−</sup> mitolysosomes. (<b>F</b>) The aligned IF and CLEM image of the cells from (<b>D</b>) is shown. The white dashed boxes in the left panel are enlarged and shown in the magnified images in the right panel. The white arrowheads indicate the loci of mitolysosomes.</p>
Full article ">Figure 4
<p>HCV NS5A enhances mitophagic flux and induces ubiquitin recruitment to mitochondria: (<b>A</b>) Huh7 cells were transduced with pTRIP-MT-Keima lentiviruses, according to the procedure described in the “Materials and Methods” section, to establish Huh7/MT-Keima cells. Then, Huh7/MT-Keima cells were transduced with (+) or without (−) pTRIP-HCV NS5A-miRFP670 lentiviruses. Forty-eight hours later, the cells were analyzed via confocal microscopy at short (488 nm) and long (561 nm) excitation wavelengths. (<b>B</b>) The percentage of cells containing acidic MT-Keima (excitation at 561 nm) was quantified, as described previously [<a href="#B33-pathogens-13-01139" class="html-bibr">33</a>]. The data are presented as means ± SEMs (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the cells expressing acidic MT-Keima. (<b>D</b>) Huh7 cells were transduced with lentiviruses expressing RFP-LC3, GFP-Ub, or Mito-miRFP670, as described in the “Materials and Methods” section, generating Huh7/RFP-LC3/GFP-Ub/Mito-miRFP670 cells. Huh7/RFP-LC3/GFP-Ub/Mito-miRFP670 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and then analyzed via confocal microscopy. (<b>E</b>) The number of RFP-LC3 puncta containing GFP-Ub on Mito-miRFP670-labeled mitochondria was quantified. The data are presented as means ± SEMs (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). (<b>F</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-Ub to Mito-miRFP670-expressing mitochondria and the subsequent sequestration of RFP-LC3-labeled autophagic vacuoles.</p>
Full article ">Figure 5
<p>HCV NS5A induces ubiquitin Ser65 phosphorylation and PINK1 stabilization in mitochondria: (<b>A</b>,<b>B</b>) (<b>A</b>) Huh7/RFP-LC3/Mito-GFP cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses. Forty-eight hours later, the cells were immunostained with a phospho-ubiquitin (Ub; Ser65) antibody and analyzed via confocal microscopy. (<b>B</b>) Foci of phospho-ubiquitin (Ser65) recruited onto mitophagosomes in which RFP-LC3 puncta sequestered Mito-GFP-labeled mitochondria were quantified. (<b>C</b>,<b>D</b>) (<b>C</b>) Huh7/RFP-Parkin/Mito-GFP cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours. Then, the cells were immunostained with an anti-phospho-ubiquitin (Ser65) antibody and analyzed via confocal microscopy. (<b>D</b>) Foci of phosphor-Ub recruited to RFP-Parkin-translocated Mito-GFP-expressing mitochondria were quantified. (<b>E</b>,<b>F</b>) (<b>E</b>) Huh7/RFP-LC3/Mito-GFP cells were transduced with lentiviruses expressing PINK1-miRFP670, generating Huh7/RFP-LC3/Mito-miRFP670/PINK1-miRFP670 cells. Then, the cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>F</b>) Foci of PINK1-miRFP670 stabilized on mitophagosomes, in which RFP-LC3 puncta sequestered Mito-GFP-expressing mitochondria were quantified. (<b>G</b>,<b>H</b>) (<b>G</b>) Huh7/RFP-Parkin/Mito-GFP cells were transduced with lentiviruses expressing PINK1-miRFP670 to establish Huh7/RFP-LC3/Mito-miRFP670/PINK1-miRFP670 cells. Then, the cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>H</b>) The number of MiRFP670-PINK1 foci recruited onto the RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. The data shown in (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). The magnified field-1 and magnified field-2 in (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) show enlarged images of white dashed boxes 1 and 2 in the top and bottom panels. The white arrowheads indicate colocalized signals.</p>
Full article ">Figure 6
<p>Recruitment of NDP52 and OPTN into HCV NS5A-activated mitophagy: (<b>A,B</b>) (<b>A</b>) Huh7 cells were transduced with lentiviruses expressing RFP-LC3, Mito-miRFP670, or GFP-NDP52, as described in the “Materials and Methods” section, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-NDP52 cells. Then, the cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-NDP52 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>,<b>D</b>) (<b>C</b>) Huh7 cells were transduced with lentiviruses expressing RFP-LC3, Mito-miRFP670, or GFP-OPTN, as described in the “Materials and Methods” section, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-OPTN cells. Then, the cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>D</b>) The number of GFP-OPTN molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>E</b>,<b>F</b>) (<b>E</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with lentiviruses expressing GFP-NDP52, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-NDP52 cells. Then, the cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>F</b>) The number of GFP-NDP52 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>G</b>,<b>H</b>) (<b>G</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with lentiviruses expressing GFP-OPTN to establish Huh7/RFP-Parkin/Mito-miRFP670/GFP-OPTN cells. Then, the cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (H) The number of GFP-OPTN molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. The data shown in (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001). The magnified field-1 and magnified field-2 in (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) show enlarged images of white dashed boxes 1 and 2 in the top and bottom panels. The white arrowheads indicate colocalized signals.</p>
Full article ">Figure 7
<p>Recruitment of ATG5 into close proximity to mitochondria for HCV NS5A-induced mitophagy: (<b>A</b>) Huh7 cells were transduced with lentiviruses expressing RFP-LC3 and mito-miRFP670 to establish Huh7/RFP-LC3/Mito-miRFP670 cells. Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG5, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG5 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG5 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-ATG5 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-ATG5 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7 cells were transduced with lentiviruses expressing RFP-Parkin and Mito-miRFP670 to establish Huh7/RFP-Parkin/Mito-miRFP670 cells. Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG5, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG5 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG5 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-ATG5 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-ATG5 to Mito-miRFP670-labeled mitochondria after translocation by RFP-Parkin. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 7 Cont.
<p>Recruitment of ATG5 into close proximity to mitochondria for HCV NS5A-induced mitophagy: (<b>A</b>) Huh7 cells were transduced with lentiviruses expressing RFP-LC3 and mito-miRFP670 to establish Huh7/RFP-LC3/Mito-miRFP670 cells. Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG5, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG5 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG5 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-ATG5 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-ATG5 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7 cells were transduced with lentiviruses expressing RFP-Parkin and Mito-miRFP670 to establish Huh7/RFP-Parkin/Mito-miRFP670 cells. Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG5, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG5 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG5 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-ATG5 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-ATG5 to Mito-miRFP670-labeled mitochondria after translocation by RFP-Parkin. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 8
<p>Recruitment of DFCP1 into the proximity of mitochondria for HCV NS5A-induced mitophagy: (<b>A</b>) Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-DFCP1, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-DFCP1 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-DFCP1 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-DFCP1 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The frames of selected live images show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-DFCP1 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-DFCP1, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-DFCP1 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-DFCP1 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-DFCP1 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The frames of selected live images show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-DFCP1 to Mito-miRFP670-labeled mitochondria after translocation by RFP-Parkin. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 8 Cont.
<p>Recruitment of DFCP1 into the proximity of mitochondria for HCV NS5A-induced mitophagy: (<b>A</b>) Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-DFCP1, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-DFCP1 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-DFCP1 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-DFCP1 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The frames of selected live images show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-DFCP1 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-DFCP1, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-DFCP1 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-DFCP1 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-DFCP1 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The frames of selected live images show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-DFCP1 to Mito-miRFP670-labeled mitochondria after translocation by RFP-Parkin. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 9
<p>Translocation of ATG14 into close proximity to mitochondria for HCV NS5A-induced mitophagy: (<b>A</b>) Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG14, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG14 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG14 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-ATG14 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequester Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-ATG14 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG14, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG14 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG14 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-ATG14 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-ATG14 to Mito-miRFP670-labeled mitochondria after RFP-Parkin translocation. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 9 Cont.
<p>Translocation of ATG14 into close proximity to mitochondria for HCV NS5A-induced mitophagy: (<b>A</b>) Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG14, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG14 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-ATG14 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-ATG14 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequester Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-ATG14 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ATG14, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG14 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-ATG14 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-ATG14 molecules recruited to RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-ATG14 to Mito-miRFP670-labeled mitochondria after RFP-Parkin translocation. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 10
<p>Translocation of ULK1 into the proximity of mitochondria for HCV NS5A-activated mitophagy: (<b>A</b>) Huh7/RFP-LC3/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ULK1, generating Huh7/RFP-LC3/Mito-miRFP670/GFP-ULK1 cells. Then, Huh7/RFP-LC3/Mito-miRFP670/GFP-ULK1 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>B</b>) The number of GFP-ULK1 molecules recruited onto mitophagosomes, in which RFP-LC3 puncta sequestered Mito-miRFP670-expressing mitochondria, was quantified. (<b>C</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>A</b>). The white arrowheads indicate the recruitment of GFP-ULK1 to Mito-miRFP670-labeled mitochondria before sequestration by RFP-LC3 puncta. (<b>D</b>) Huh7/RFP-Parkin/Mito-miRFP670 cells were transduced with retroviruses expressing GFP-ULK1, generating Huh7/RFP-Parkin/Mito-miRFP670/GFP-ULK1 cells. Then, Huh7/RFP-Parkin/Mito-miRFP670/GFP-ULK1 cells were transduced with (+) or without (−) pTRIP-HCV NS5A-mTagBFP2 lentiviruses for forty-eight hours and analyzed via confocal microscopy. (<b>E</b>) The number of GFP-ULK1 molecules recruited to the RFP-Parkin-translocated Mito-miRFP670-labeled mitochondria was quantified. (<b>F</b>) The selected live imaging frames show the magnified area in the white dashed box of the top panel in (<b>D</b>). The white arrowheads indicate the recruitment of GFP-ULK1 to Mito-miRFP670-labeled mitochondria after RFP-Parkin translocation. The data shown in (<b>B</b>,<b>E</b>) represent the mean ± SEM (<span class="html-italic">n</span> = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">
18 pages, 2026 KiB  
Review
Advancements in Cellular Imaging: Expanding Horizons with Innovative Dyes and Techniques
by Payal M. Oak and Akash S. Mali
Colorants 2024, 3(4), 360-377; https://doi.org/10.3390/colorants3040025 - 23 Dec 2024
Abstract
Advancements in cellular imaging have significantly enhanced our understanding of membrane potential and Ca2⁺ dynamics, which are crucial for various cellular processes. Voltage-sensitive dyes (VSDs) are pivotal in this field, enabling non-invasive, high-resolution visualization of electrical activity in cells. This review [...] Read more.
Advancements in cellular imaging have significantly enhanced our understanding of membrane potential and Ca2⁺ dynamics, which are crucial for various cellular processes. Voltage-sensitive dyes (VSDs) are pivotal in this field, enabling non-invasive, high-resolution visualization of electrical activity in cells. This review discusses the various types of VSDs, including electrochromic, Förster Resonance Energy Transfer (FRET)-based, and Photoinduced Electron Transfer (PeT)-based dyes. VSDs are essential tools for studying mitochondrial activity and neuronal function and are frequently used in conjunction with Ca2⁺ indicators to elucidate the complex relationship between membrane potential and Ca2⁺ fluxes. The development of novel dyes with improved photostability and reduced toxicity continues to expand the potential of VSDs in biomedical research. This review underscores the importance of VSDs in advancing our understanding of cellular bioenergetics, signaling, and disease mechanisms. Full article
Show Figures

Figure 1

Figure 1
<p>Electrochromic dyes respond to voltage through a direct interaction between the chromophore and the electric field [<a href="#B23-colorants-03-00025" class="html-bibr">23</a>].</p>
Full article ">Figure 2
<p>FRET-based voltage sensors use lipophilic anions that intercalate into the cellular membrane and distribute between the inner and outer leaflets depending upon the transmembrane potential [<a href="#B23-colorants-03-00025" class="html-bibr">23</a>].</p>
Full article ">Figure 3
<p>Molecular wire PeT VSDs depend upon the voltage-sensitive electron transfer from an electron-rich donor through a membrane-spanning molecular wire to a fluorescent reporter [<a href="#B23-colorants-03-00025" class="html-bibr">23</a>].</p>
Full article ">Figure 4
<p>Chemical structure of ANNINE. Reproduced with permission from [<a href="#B116-colorants-03-00025" class="html-bibr">116</a>], 2003, ACS.</p>
Full article ">Figure 5
<p>Synthesis of the ANNINE dyes. Reproduced with permission from [<a href="#B116-colorants-03-00025" class="html-bibr">116</a>], 2003, ACS.</p>
Full article ">Figure 6
<p>Synthesis of chromene-based VSD [<a href="#B44-colorants-03-00025" class="html-bibr">44</a>].</p>
Full article ">
20 pages, 1414 KiB  
Article
Sperm Functional Status: A Multiparametric Assessment of the Fertilizing Potential of Bovine Sperm
by Timea Sarah Odinius, Mathias Siuda, Matthias Lautner, Claus Leiding, Stefan Neuner, Heinrich Bollwein and Eleni Malama
Vet. Sci. 2024, 11(12), 678; https://doi.org/10.3390/vetsci11120678 - 23 Dec 2024
Abstract
Sperm viability is routinely assessed for the quality control of cryopreserved bovine sperm batches but is not usually conclusive regarding their fertilizing potential. In this study, we investigated the fertility predictive value of bull sperm viability in combination with DNA integrity or the [...] Read more.
Sperm viability is routinely assessed for the quality control of cryopreserved bovine sperm batches but is not usually conclusive regarding their fertilizing potential. In this study, we investigated the fertility predictive value of bull sperm viability in combination with DNA integrity or the functional status of viable sperm. In addition to sperm viability, we flow cytometrically assessed the percentage of sperm with high DNA fragmentation index (%DFI) and the fraction of viable sperm with low intracellular Ca2+ content and functional mitochondria using the Sperm Chromatin Structure Assay and a five-color staining panel in 791 and 733 cryopreserved batches with non-return rate (NRR) records after ≥100 first services, respectively. Using linear mixed-effects models and conditional inference trees, we examined the potential of sperm viability combined with either DNA integrity or the functional status of viable sperm to predict the batch-specific NRR. Batches with a %DFI of ≤6.86% were more likely to have a NRR of >60%, whereas %DFI values of >6.86% were more likely to be associated with a 55–60% or lower NRR. Combining post-thaw viability with the functional status of viable sperm did not reliably predict the NRR of individual batches. Concluding, the incorporation of DNA integrity assessment can considerably improve sperm fertility prognostics. Full article
(This article belongs to the Special Issue Sperm Biotechnology in Animals Reproduction)
Show Figures

Figure 1

Figure 1
<p>The standard deviation (SD, %) of the non-return rate (NRR) of cryopreserved batches 60 to 90 days after artificial insemination (AI), in relation to the minimum number of first AIs recorded for each batch. The mean SD of the NRR computed for different minima of the first services is represented by the horizontal solid red line; the dashed red lines reflect the SD values that lie one standard deviation away from the mean.</p>
Full article ">Figure 2
<p>The proportion of cryopreserved batches (with a minimum of 100 first services) scoring different non-return rates (NRRs) 60 to 90 days after artificial insemination. The NRRs are demonstrated in the form of classes at 5-unit half-open intervals (they do not include their lower limit point).</p>
Full article ">Figure 3
<p>Predicted values of the batch-specific non-return rate (NRR, %) on day 60 to 90 post-insemination in relation to the percentage of sperm with a high DNA fragmentation index (%DFI, %) in the cryopreserved semen batch immediately after thawing (0 h). The plotted NRR values were predicted by fitting a linear mixed-effects model on the sperm quality data of 791 cryopreserved sperm samples produced from 236 sires and conditioned on the fixed effects of sperm viability, %DFI, bull age, season, and weekday of semen collection.</p>
Full article ">Figure 4
<p>Decision tree illustrating the predictability of the non-return rate (NRR) class based on the percentage of sperm with a high DNA fragmentation index (%DFI). Classes of the NRR were built with 5-unit intervals (left-open and right-closed intervals) and included NRR scores between 30% and 80%. In each decision split node (nodes 1 and 2), the <span class="html-italic">p</span> value is presented for the selection of a significant predictor. The %DFI cutoff values are shown along the decision branch. The terminal nodes 3, 4, and 5 are in the form of bar plots showing the likelihood of a batch being assigned in a specific NRR class based on its %DFI score. The number of batches (n) landing in each terminal node is also presented.</p>
Full article ">
35 pages, 8312 KiB  
Article
Comparison of the Effects of UV-C Light in the Form of Flash or Continuous Exposure: A Transcriptomic Analysis on Arabidopsis thaliana L.
by Seyed Mehdi Jazayeri, Jawad Aarrouf, Laurent Urban and Félicie Lopez-Lauri
Int. J. Mol. Sci. 2024, 25(24), 13718; https://doi.org/10.3390/ijms252413718 - 22 Dec 2024
Viewed by 349
Abstract
Ultraviolet C (UV-C) flash treatment represents a promising method for priming plants. This study compared the effects of 1 s (flash) and 60 s (60 s) UV-C exposures on the transcriptome of Arabidopsis thaliana L. plants. A dose of 200 J m−2 [...] Read more.
Ultraviolet C (UV-C) flash treatment represents a promising method for priming plants. This study compared the effects of 1 s (flash) and 60 s (60 s) UV-C exposures on the transcriptome of Arabidopsis thaliana L. plants. A dose of 200 J m−2 delivered in one second was observed to effectively stimulate plant defenses without causing any adverse effects on plant health. A total of 3054 and 1865 differentially expressed genes (DEGs) were identified in the flash and 60 s treatments, respectively, in comparison to the control plants. Of these, 1131 were common to both treatments. The flash treatment affected a greater number of transcription factors (415 genes) than the 60 s treatment (254 genes), indicating more pronounced alterations in gene expression. The flash treatment resulted in a significant overexpression of heat shock proteins (HSPs), heat shock factors (HSFs), and their associated genes, which impacted oxidative stress, proteostasis, genome stability, cell survival, and thermotolerance. The majority of mitochondrial genes were found to be upregulated, while photosynthetic genes exhibited a downregulation. These expression patterns coordinate electron transport and crosstalk between the nucleus, chloroplasts, and mitochondria, eliciting an adaptive protective response to UV-C flash. Additionally, the flash treatment resulted in alterations to several genes involved in cell cycle regulation, division, and DNA replication. These included ATP BMMs, BRCA2 s, IQDs, kinesin complex, MCM complex, CYCs, and CDKs, which ultimately led to cell cycle arrest as a temporary preparation for subsequent conditions. The present study demonstrates that a 1 s exposure to UV-C induces distinctive plant responses through coordinated gene expression. The findings suggest that the flash treatment is an innovative method that triggers a unique cellular response, prioritizing repair mechanisms and potentially enhancing plant immunity, resilience, and priming. It can be used as a plant resistance inducer and stimulator. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Figure 1

Figure 1
<p>Analyses of differentially expressed genes (DEGs). (<b>A</b>) Volcano diagrams of DEGs identified in leaves after 4 h of UV-C flash (<b>left</b>) and UV-C 60 s treatments (<b>right</b>) vs. control. Spots above the threshold line (fold change cutoff ≥ 1 or fold change cutoff <span class="underline">&lt;</span> −1, FDR <span class="underline">&lt;</span> 0.05) indicate that differences are significant. Downregulated genes are displayed in green, while upregulated genes are displayed in red. Genes in grey are not DEGs. (<b>B</b>) Venn diagram representing the numbers of non-overlapped and overlapped DEGs in the four categories. ‘Up_1s flash vs. Control’ and ‘Down_1s flash vs. Control’ refer to upregulated and downregulated DEGs detected in 1 s UV-C (Flash) treated leaves compared to the control, respectively. ‘Up_60s vs. Control’ and ‘Down_60s vs. Control’ refer to upregulated and downregulated DEGs detected in 60 s UV-C treated leaves compared to control.</p>
Full article ">Figure 2
<p>The comparisons of DEGs of the flash treatment and 60 s treatment based on log2FC of more than 3 or less than −3 and FDR ≤ 0.05. The number of specific upregulated DEGs with a fold change more than 10 for the flash treatment is 205, while there are 65 for the 60 s treatment. On the other hand, the specific downregulated DEGs are 21 and 39 for the flash and 60 s treatment, respectively. This suggests a higher gene activation rate by the flash treatment.</p>
Full article ">Figure 3
<p>The cellular component (CC) of GO enrichment analysis. The up- and downregulated DEGs are employed as separate entities to undertake GO-based enrichment analysis. (<b>A</b>) The GO enrichment of upregulated DEGs of the flash treatment. (<b>B</b>) The GO enrichment of upregulated DEGs of the 60 s treatment. (<b>C</b>) The GO enrichment of downregulated DEGs of the flash treatment. (<b>D</b>) The GO enrichment of downregulated DEGs of the 60 s treatment. In the case of the flash treatment, the affected cellular components are related to the chloroplast, mitochondrion, cell wall, nucleus, extracellular matrix, and their related compartmentalization. The targeted cellular parts are the vesicle and vacuole, as well as endoplasmic reticulum. It is noteworthy that the GO terms related to the MCM complex, kinesin, and microtubule are specific to the flash treatment. This suggests that UV-C is able to penetrate cell structures effectively within a 1 s timeframe.</p>
Full article ">Figure 4
<p>The biological process (BP) of GO enrichment analysis. The up- and downregulated DEGs were subjected to a GO-based enrichment analysis. (<b>A</b>) The GO enrichment of upregulated DEGs of the flash treatment. (<b>B</b>) The GO enrichment of upregulated DEGs of the 60 s treatment. (<b>C</b>). The GO enrichment of downregulated DEGs of the flash treatment. (<b>D</b>) The GO enrichment of downregulated DEGs of the 60 s treatment. The BP GO terms associated with the flash treatment pertain to thermotolerance, cell division, and transport. With regard to the 60 s treatment, the GO terms are associated with metabolite biosynthesis, responses to stresses, and detoxification.</p>
Full article ">Figure 5
<p>The molecular function (MF) of GO enrichment analysis. The up- and downregulated DEGs are employed as separate entities for the purpose of conducting a GO-based enrichment analysis. (<b>A</b>) The GO enrichment of upregulated DEGs of the flash treatment. (<b>B</b>) The GO enrichment of upregulated DEGs of the 60 s treatment. (<b>C</b>) The GO enrichment of downregulated DEGs of the flash treatment. (<b>D</b>) The GO enrichment of downregulated DEGs of the 60 s treatment. The GO terms of MF for the flash treatment are associated with DNA, RNA, protein, and chlorophyll. In the case of the 60 s treatments, the GO terms are assigned to enzymatic activities that are involved in metabolism of a variety of metabolites.</p>
Full article ">Figure 6
<p>The metabolism overview pathway of MapMan. <b>Above</b>: the metabolism overview pathway of the flash treatment. <b>Below</b>: the metabolism overview pathway of the 60 s treatment. The differential effects of the flash and 60 s treatments on plants are evident from the higher number of differentially expressed genes (DEGs) observed in the former. The majority of sub-pathways within the metabolism overview demonstrate a greater number of DEGs in response to the flash treatment. However, the number of DEGs assigned to the amino acid metabolism section of Phe/Tyr/Trp/misc and the second metabolism of flavonoids and S-misc is greater for the 60 s treatment than for the flash treatment. Furthermore, the shared DEGs are illustrated in both maps for both treatments. The red dots indicate genes that are overexpressed, while the blue dots indicate genes that are underexpressed.</p>
Full article ">Figure 7
<p>An overview of the regulatory pathway represented in MapMan. <b>Above</b>: the regulation overview pathway of the flash treatment. <b>Below</b>: the regulation overview pathway of the 60 s treatment. The differential effects of the flash and 60 s treatments on plants are evident from the higher number of DEGs observed in the former. All subcategories within the regulation overview exhibit a greater number of DEGs in response to the flash treatment. This discrepancy indicates a greater impact on the plant regulatory system affected with the flash treatment, suggesting that this treatment can stimulate downstream genes involved in transcription, regulation, and signaling, as well as protein modification and degradation. However, the flash treatment yielded a greater number of receptor kinases, with 65 DEGs, of which 51 were downregulated and 14 were upregulated. In contrast, the 60 s treatment resulted in the upregulation of 27 receptor kinase genes, while the 13 downregulated genes were associated with the 60 s treatment. Moreover, the genes that were differentially expressed in both treatments are illustrated in both maps. The red dots indicate genes that are overexpressed, while the blue dots indicate genes that are underexpressed.</p>
Full article ">Figure 8
<p>The transcription factors (TFs), transcription regulators (TRs), and chromatin regulators (CRs) for the flash (blue) and 60 s (green) treatments. The categorization was performed using the PlantTFcat tool. The number of regulatory and transcription system genes is 415 and 254, respectively, for the flash treatment and 60 s treatment. The greater number of DEGs within these three categories in the flash treatment group indicates that this treatment affects the transcriptional regulatory system in a more effective manner than the 60 s treatment. With respect to TFs and TRs, the flash treatment appears to exert a more pronounced influence on transcriptional regulation. With regard to CRs, the flash treatment prompts epigenetic occurrences and subsequently initiates priming.</p>
Full article ">Figure 9
<p>The MapMan photosynthesis pathway. The figure illustrates the differential expression of 82 and 20 genes in plants treated with the flash (<b>above</b>) and 60 s (<b>below</b>) treatments, respectively. The number of genes altered by the flash treatment is four times greater than that of the 60 s treatment. This indicates that the photosynthetic apparatus is more profoundly impacted by the flash treatment than by the 60 s treatment. In the case of the flash treatment, 31 and 16 DEGs are associated with PSII and PSI, respectively, while seven and three genes are associated with PSII and PSI, respectively. The genes of photorespiration comprise 12 and four in the flash and 60 s treatments, respectively. The genes that are overexpressed are indicated in red, while those that are underexpressed are indicated in blue.</p>
Full article ">Figure 9 Cont.
<p>The MapMan photosynthesis pathway. The figure illustrates the differential expression of 82 and 20 genes in plants treated with the flash (<b>above</b>) and 60 s (<b>below</b>) treatments, respectively. The number of genes altered by the flash treatment is four times greater than that of the 60 s treatment. This indicates that the photosynthetic apparatus is more profoundly impacted by the flash treatment than by the 60 s treatment. In the case of the flash treatment, 31 and 16 DEGs are associated with PSII and PSI, respectively, while seven and three genes are associated with PSII and PSI, respectively. The genes of photorespiration comprise 12 and four in the flash and 60 s treatments, respectively. The genes that are overexpressed are indicated in red, while those that are underexpressed are indicated in blue.</p>
Full article ">Figure 10
<p>The MapMan mitochondrial electron transport pathway. The data indicate that there are 52 and 23 DEGs in plants treated with the flash (<b>above</b>) and 60 s (<b>below</b>) treatments, respectively. Similarly, the mitochondrial apparatus is more profoundly activated by the flash than by the 60 s treatment. Twelve genes associated with metabolite transporters exhibit differential overexpression in the flash treatment, with seven being exclusive to this treatment. For the 60 s treatment, six metabolite transporter genes are found. Eight genes associated with cytochrome c are overexpressed by the flash treatment, while no gene is altered by the 60 s treatment. For complex I of the electron transport, the flash treatment alters 11 genes, with 10 specific ones. The genes that are overexpressed are indicated in red, while those that are underexpressed are indicated in blue.</p>
Full article ">Figure 10 Cont.
<p>The MapMan mitochondrial electron transport pathway. The data indicate that there are 52 and 23 DEGs in plants treated with the flash (<b>above</b>) and 60 s (<b>below</b>) treatments, respectively. Similarly, the mitochondrial apparatus is more profoundly activated by the flash than by the 60 s treatment. Twelve genes associated with metabolite transporters exhibit differential overexpression in the flash treatment, with seven being exclusive to this treatment. For the 60 s treatment, six metabolite transporter genes are found. Eight genes associated with cytochrome c are overexpressed by the flash treatment, while no gene is altered by the 60 s treatment. For complex I of the electron transport, the flash treatment alters 11 genes, with 10 specific ones. The genes that are overexpressed are indicated in red, while those that are underexpressed are indicated in blue.</p>
Full article ">Figure 11
<p>Hypothetical vision of plant responses to the flash treatment. 1. UV-C light flashes. 2. Perception: Plants receive UV-C light via receptors, proteins, and molecules in the membrane, cell wall, and extracellular and intercellular zones. 3. Signaling initiation: interaction of receptors, cell wall, and membrane proteins begins signaling. 4. Cell wall modification: enzymes and proteins modify cell wall integrity, producing signaling molecules from compounds like pectin and cellulose. 5. ROS production: chloroplasts and mitochondria produce and scavenge ROS, which are also used as signaling molecules. Electron transport flow and energy remodeling occur. 6. Transcription regulation: nucleus, nucleolus, and other organelles coordinate interactions between genes and proteins, notably in mitochondria and chloroplasts. Expression of ATCG and ATMG is regulated by cell genes. 7. Protein modifications: overexpression of HSPs, HSFs, and related genes indicates protein modifications involved in signaling and transcriptional regulation. 8. PCD: PCD genes are expressed, collaborating with ROS, cell cycle, DNA repair, and signaling activities. 9. Defense activation: Differential gene expression activates defense, immunity, and priming systems. Genes in blue are overexpressed, red are underexpressed, and pink show differential expression patterns (both over- and underexpression).</p>
Full article ">
13 pages, 4995 KiB  
Article
BODIPY-Based Ratiometric Fluorescent Probe for Sensing Peroxynitrite in Inflammatory Cells and Tissues
by Qian Wu, Ziwei Hu, Guoyang Zhang, Yulong Jin and Zhuo Wang
Biosensors 2024, 14(12), 638; https://doi.org/10.3390/bios14120638 - 22 Dec 2024
Viewed by 352
Abstract
Peroxynitrite (ONOO) plays an important role in many physiological and pathological processes. Excessive ONOO in cells leads to oxidative stress and inflammation. However, precise monitoring of ONOO levels in specific organelles (e.g., mitochondria) is still lacking and urgently needed. [...] Read more.
Peroxynitrite (ONOO) plays an important role in many physiological and pathological processes. Excessive ONOO in cells leads to oxidative stress and inflammation. However, precise monitoring of ONOO levels in specific organelles (e.g., mitochondria) is still lacking and urgently needed. Herein, we rationally designed a mitochondria-targeted ratiometric fluorescent probe, MOBDP-I, for imaging of ONOO in the mitochondria of inflammatory cells and model mice. This probe, MOBDP-I, was synthesized by conjugating a BODIPY fluorophore to a mitochondria-targeting moiety–indole-salt group by a carbon–carbon double bond (C=C). In the presence of ONOO, the C=C bond between the BODIPY backbone and the indole-salt group was oxidized and broken, leading to an 18-fold enhancement of fluorescence at 510 nm, along with a significant fluorescence decrease at 596 nm. The ratiometric response property bestowed the probe with advantages in the precise quantification of ONOO in cells, thus allowing estimation of the extent of inflammation in living cells and mouse models of rheumatoid arthritis, peritonitis, and brain inflammation. MOBDP-I could act as an effective molecular tool to study the relationship between ONOO and the occurrence and development of inflammatory diseases. Full article
(This article belongs to the Special Issue State-of-the-Art Biosensors in China (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) UV-vis absorption and fluorescence spectra of MOBDP-I (10 μM) in PBS (10 mM, pH = 7.4, 0.1% DMSO). (<b>b</b>) Normalized absorption spectra of MOBDP-I (10 μM) in various solvents, including DCM, THF, EA, Me<sub>2</sub>CO, EtOH, MeCN, DMSO, PBS. (<b>c</b>) Absorption spectra of MOBDP-I (10 μM) with and without ONOO<sup>−</sup> (10 μM) in PBS (pH = 7.4). Inset: − (without ONOO<sup>−</sup>), + (with ONOO<sup>−</sup>). (<b>d</b>) The response time of MOBDP-I (10 μM) to ONOO<sup>−</sup> (10 μM) in PBS (pH 7.4), λ<sub>ex</sub> = 460 nm.</p>
Full article ">Figure 2
<p>(<b>a</b>) Fluorescence spectra of MOBDP-I (10 μM) in response to ONOO<sup>−</sup> (0~10 μΜ) in PBS (pH = 7.4), λ<sub>ex</sub> = 460 nm. (<b>b</b>) Linear relationship between the fluorescence intensity ratio (I<sub>510</sub>/I<sub>596</sub>) and ONOO<sup>−</sup> concentration from 0 to 10 μM. (<b>c</b>) Fluorescence spectra of MOBDP-I (10 μM) in the presence of 10 μM of ONOO<sup>−</sup> or 50 μM of other relevant analytes. (<b>d</b>) The fluorescence response intensity (I<sub>510</sub>/I<sub>596</sub>) of MOBDP-I to ONOO<sup>−</sup> (10 μM) and other relevant analytes (50 μM). Analytes: <sup>1</sup>O<sub>2</sub>, ClO<sup>−</sup>, H<sub>2</sub>O<sub>2</sub>, ROO<sup>−</sup>, ·OH, NO<sub>3</sub><sup>−</sup>, NO<sub>2</sub><sup>−</sup>, H<sub>2</sub>S, SO<sub>3</sub><sup>2−</sup>, HSO<sub>3</sub><sup>−</sup>, GSH, Cys, K<sup>+</sup>, Na<sup>+</sup>, Ca<sup>2+</sup>, Mg<sup>2+</sup>, Zn<sup>2+</sup>, Cu<sup>2+</sup>, Ni<sup>2+</sup>, Co<sup>2+</sup>. Samples were measured in PBS (pH = 7.4) at room temperature with λ<sub>ex</sub> = 460 nm. The error bars represent ± standard deviation (SD) (n = 3).</p>
Full article ">Figure 3
<p>Co-localization imaging in HeLa cells with probe MOBDP-I (10 μM) and Rhodamine 123 (5 μM). Green channel: λ<sub>ex</sub> = 488, λ<sub>em</sub> = 510–540 nm. Red channel: λ<sub>ex</sub> = 543, λ<sub>em</sub> = 580–650 nm, scale bar: 75 μm.</p>
Full article ">Figure 4
<p>(<b>a</b>) Fluorescence imaging of exogenous ONOO<sup>−</sup> (0, 2, 4, 6, 8, and 10 μM) in HeLa cells stained with MOBDP-I (10 μM). scale bar: 50 μm. (<b>b</b>) Relative fluorescence intensities corresponding to panel (<b>a</b>) in both the green and red channels. (<b>c</b>) Fluorescence intensity ratio corresponding to panel (<b>a</b>). Green channel: λ<sub>ex</sub> = 488 nm, λ<sub>em</sub> = 510–540 nm. Red channel: λ<sub>ex</sub> = 543 nm, λ<sub>em</sub> = 580–650 nm. The error bars represent ± standard deviation (SD) (n = 3).</p>
Full article ">Figure 5
<p>Fluorescence imaging of endogenous ONOO<sup>−</sup> in HeLa cells. (<b>a</b>) The cells were treated with LPS (1 μg/mL) for different times and then incubated with MOBDP-I (10 μM). Green channel: λ<sub>ex</sub> = 488 nm, λ<sub>em</sub> = 510–540 nm. Red channel: λ<sub>ex</sub> = 543 nm, λ<sub>em</sub> = 580–650 nm, scale bar: 50 μm. (<b>b</b>) Fluorescence intensity ratio corresponding to panel (<b>a</b>). (<b>c</b>) The cells were treated with different concentrations of LPS for 6 h and then incubated with MOBDP-I (10 μM). Green channel: λ<sub>ex</sub> = 488 nm, λ<sub>em</sub> = 510–540 nm. Red channel: λ<sub>ex</sub> = 543 nm, λ<sub>em</sub> = 580–650 nm, scale bar: 10 μm; (<b>d</b>) Fluorescence intensity ratio corresponding to panel (<b>c</b>). The error bars represent ± standard deviation (SD) (n = 3).</p>
Full article ">Figure 6
<p>(<b>a</b>) Fluorescence imaging of MOBDP-I in a mouse model of rheumatoid arthritis. Control group: the left hind limb of mice, in situ injection of PBS (50 μL, 10 mM, pH = 7.0); experimental group: the right hind limb of mice, in situ injection of λ-carr (50 μL, 5 mg/mL) or λ-carr (50 μL, 5 mg/mL) + NAC (50 μL, 30 mg/kg). (<b>b</b>) Relative fluorescence intensities corresponding to panel (<b>a</b>); (<b>c</b>) Fluorescence imaging of MOBDP-I in a mouse model of peritonitis. Control group: intraperitoneal injection of PBS (50 μL, 10 mM, pH = 7.0); experimental group: intraperitoneal injection of LPS (50 μL, 2 mg/mL) or LPS (50 μL, 2 mg/mL) + NAC (50 μL, 30 mg/kg). (<b>d</b>) Relative fluorescence intensities corresponding to panel (<b>c</b>). Green channel, λ<sub>ex</sub> = 460 nm, λ<sub>em</sub> = 510–550 nm. Red channel, λ<sub>ex</sub> = 570 nm, λ<sub>em</sub> = 650–700 nm.</p>
Full article ">Figure 7
<p>(<b>a</b>) Fluorescence imaging of MOBDP-I in mouse model of brain inflammation. Control group (left): tail vein injection of PBS (50 μL, 10 mM, pH = 7.0), and then MOBDP-I (20 μM, 100 μL in 1:9 DMSO/PBS <span class="html-italic">v</span>/<span class="html-italic">v</span>); experimental group (right): tail vein injection of LPS (100 μL, 2 mg/mL, 8 h), and then MOBDP-I (20 μM, 100 μL in 1:9 DMSO/PBS <span class="html-italic">v</span>/<span class="html-italic">v</span>). (<b>b</b>) Relative fluorescence intensities corresponding to the LPS-treated group in panel (<b>a</b>). Red channel, λ<sub>ex</sub> = 570 nm, λ<sub>em</sub> = 650–700 nm. (<b>c</b>) Fluorescence imaging of the dissected mouse brains. (<b>d</b>) Relative fluorescence intensities corresponding to panel (<b>c</b>). Green channel, λ<sub>ex</sub> = 460 nm, λ<sub>em</sub> =510–550 nm. Red channel, λ<sub>ex</sub> = 570 nm, λ<sub>em</sub> = 650–700 nm. The error bars represent ± standard deviation (SD) (n = 3).</p>
Full article ">Scheme 1
<p>The response mechanism of MOBDP-I to ONOO<sup>−</sup> and the fluorescence imaging of MOBDP-I in various inflammatory mouse models.</p>
Full article ">
12 pages, 3953 KiB  
Article
Sexual Dimorphism of Ethanol-Induced Mitochondrial Dynamics in Purkinje Cells
by Rehana Khatoon, Jordan Fick, Abosede Elesinnla, Jaylyn Waddell and Tibor Kristian
Int. J. Mol. Sci. 2024, 25(24), 13714; https://doi.org/10.3390/ijms252413714 - 22 Dec 2024
Viewed by 305
Abstract
The cerebellum, a key target of ethanol’s toxic effects, is associated with ataxia following alcohol consumption. However, the impact of ethanol on Purkinje cell (PC) mitochondria remains unclear. To investigate how ethanol administration affects mitochondrial dynamics in cerebellar Purkinje cells, we employed a [...] Read more.
The cerebellum, a key target of ethanol’s toxic effects, is associated with ataxia following alcohol consumption. However, the impact of ethanol on Purkinje cell (PC) mitochondria remains unclear. To investigate how ethanol administration affects mitochondrial dynamics in cerebellar Purkinje cells, we employed a transgenic mouse model expressing mitochondria-targeted yellow fluorescent protein in Purkinje cells (PC-mito-eYFP). Both male and female PC-mito-eYFP mice received an intraperitoneal injection of ethanol or vehicle. One hour after ethanol administration, the animals were perfusion fixed or their cerebellum tissue or isolated mitochondria were collected. Cerebellum sections were analyzed using confocal microscopy to assess changes in mitochondrial length distribution. In vivo superoxide levels were measured using dihydroethidium (DHE), and mitochondrial NAD levels were determined by high-performance liquid chromatography (HPLC). Our findings revealed a sex-dependent response to ethanol administration in mitochondrial size distribution. While male Purkinje cell mitochondria exhibited no significant changes in size, female mitochondria became more fragmented after one hour of ethanol administration. This coincided with elevated phosphorylation of the fission protein Drp1 and increased superoxide production, as measured by DHE fluorescence intensity. Similarly, mitochondrial NAD levels were significantly reduced in female mice, but no changes were observed in males. Our results demonstrate that ethanol induced mitochondrial fragmentation through increased free radical levels, due to reduced NAD and increased p-Drp1, in PC cells of the female cerebellum. Full article
(This article belongs to the Special Issue New Insights into Mitochondria in Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>Mitochondrial network in Purkinje neurons. Transgenic mouse model that expresses mitochondria-targeted eYFP, driven by Camk2α promotor, was used to visualize mitochondria in Purkinje neurons. Panels (<b>A</b>,<b>B</b>) represent epifluorescent images of mitochondria within the Purkinje cells’ (PCs) soma and dendritic processes. Panel (<b>C</b>) shows staining of PCs with calbindin antibody (red). Scale bar represents 10 µm.</p>
Full article ">Figure 2
<p>Purkinje cell mitochondria. Image generated from z-stack confocal images. Control, vehicle-treated male mouse (<b>left</b> panel), and following ethanol administration (<b>right</b> panel). Triangle indicates axonal mitochondria distributed along the axonal axes. Scale bar represents 10 µm.</p>
Full article ">Figure 3
<p>Ethanol metabolism leads to increased fragmentation of female PC mitochondria. (<b>A</b>) Images of mitochondrial network within dendritic tree of control (left panel (<b>A</b>)) and ethanol-treated male mouse (right panel (<b>A</b>)). (<b>B</b>) Quantification of mitochondrial size distribution in control PC and 1 h after ethanol administration using Volocity 6.3.1 software. There were no significant changes in relative number of mitochondria in the populations of 0.2–1 μm, 1–5 μm, 5–15 μm, and 15–40 μm length groups after ethanol administration. One-way ANOVA for multiple groups (n = 16 images/group). (<b>C</b>) Representative images of PC mitochondria from control and ethanol-treated female mouse cerebella. (<b>D</b>) Quantification of the changes in relative length distribution 1 h following alcohol administration. The relative distribution of individual mitochondria populations shows that ethanol administration increased the number of short mitochondria (0.2–1 μm and 1–5 μm) when compared to control. Conversely, in the longer mitochondria population (5–15 μm and 15–40 μm), the relative number decreased. ** <span class="html-italic">p</span> &lt; 0.01; * <span class="html-italic">p</span> &lt; 0.05 One-way ANOVA for multiple groups (n = 16 images/group). Scale bar represents 10 µm.</p>
Full article ">Figure 4
<p>Ethanol administration increases the p-Drp1(Ser6161) levels in female cerebellum. (<b>A</b>) p-Drp1(Ser616)-to-Drp1 ratio in male cerebellum following ethanol treatment. There was no significant effect on Drp1 phosphorylation levels induced by ethanol metabolism. (<b>B</b>) In female cerebellum, the p-Drp1 levels were increased at 30 min and 1 h after the ethanol administration. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, n = 4.</p>
Full article ">Figure 5
<p>Ethanol administration increases superoxide levels in female cerebellum. Left panel shows ethidium fluorescence in control (<b>A</b>) and after ethanol (<b>B</b>) in male cerebellum. Right panel shows images of ethidium fluorescent taken from female cerebellum sections ((<b>D</b>) control vehicle, (<b>E</b>) ethanol). (<b>C</b>,<b>F</b>) Quantification of fluorescent intensity normalized to area. There is a significant increase in florescent intensity in female cerebellum following ethanol administration. **** <span class="html-italic">p</span> &lt; 0.001 when compared to vehicle, <span class="html-italic">t</span>-test, n = 12.</p>
Full article ">Figure 6
<p>Effect of ethanol metabolism on cerebellar mitochondria NAD levels. (<b>A</b>) Ethanol administration did not alter the mitochondria NAD levels isolated from male cerebellum. (<b>B</b>) The NAD pools in female mitochondria were significantly reduced one hour following ethanol administration. * <span class="html-italic">p</span> &lt; 0.05, n = 4–6.</p>
Full article ">
15 pages, 1125 KiB  
Review
Alpha-Synuclein Effects on Mitochondrial Quality Control in Parkinson’s Disease
by Lydia Shen and Ulf Dettmer
Biomolecules 2024, 14(12), 1649; https://doi.org/10.3390/biom14121649 - 22 Dec 2024
Viewed by 256
Abstract
The maintenance of healthy mitochondria is essential for neuronal survival and relies upon mitochondrial quality control pathways involved in mitochondrial biogenesis, mitochondrial dynamics, and mitochondrial autophagy (mitophagy). Mitochondrial dysfunction is critically implicated in Parkinson’s disease (PD), a brain disorder characterized by the progressive [...] Read more.
The maintenance of healthy mitochondria is essential for neuronal survival and relies upon mitochondrial quality control pathways involved in mitochondrial biogenesis, mitochondrial dynamics, and mitochondrial autophagy (mitophagy). Mitochondrial dysfunction is critically implicated in Parkinson’s disease (PD), a brain disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Consequently, impaired mitochondrial quality control may play a key role in PD pathology. This is affirmed by work indicating that genes such as PRKN and PINK1, which participate in multiple mitochondrial processes, harbor PD-associated mutations. Furthermore, mitochondrial complex-I-inhibiting toxins like MPTP and rotenone are known to cause Parkinson-like symptoms. At the heart of PD is alpha-synuclein (αS), a small synaptic protein that misfolds and aggregates to form the disease’s hallmark Lewy bodies. The specific mechanisms through which aggregated αS exerts its neurotoxicity are still unknown; however, given the vital role of both αS and mitochondria to PD, an understanding of how αS influences mitochondrial maintenance may be essential to elucidating PD pathogenesis and discovering future therapeutic targets. Here, the current knowledge of the relationship between αS and mitochondrial quality control pathways in PD is reviewed, highlighting recent findings regarding αS effects on mitochondrial biogenesis, dynamics, and autophagy. Full article
(This article belongs to the Section Biomacromolecules: Proteins)
Show Figures

Figure 1

Figure 1
<p>Potential pathological interactions of αS with mitochondrial quality control pathways in PD. (<b>a</b>) αS and mitochondrial biogenesis. (<b>i</b>) αS may act as a transcriptional modulator of PGC-1α under oxidative stress, binding to its promoter sequence to repress PGC-1α function. (<b>ii</b>) An excess of exogenous αS oligomers and fibrils may interfere with Parkin’s degradation of PARIS, thus increasing the PARIS-mediated transcriptional repression of PGC-1α. (<b>b</b>) αS and mitochondrial dynamics. (<b>i</b>) Pathogenic αS may increase the cleavage of OPA1 in mitochondrial fusion. (<b>ii</b>) αS-induced alterations to mitochondrial fission may be independent of or dependent upon DRP1: αS may interact directly with mitochondrial membranes or may increase the translocation of DRP1 to mitochondria. (<b>c</b>) αS and mitochondrial autophagy. (<b>i</b>) The overexpression of αS may stabilize Miro proteins, which are required for the formation of mitochondrial-derived vesicles (MDVs). (<b>ii</b>) αS may downregulate Parkin expression and activity as described above, having negative impacts on MDV trafficking. (<b>iii</b>) During autophagosome formation in mitophagy, αS may aberrantly stabilize Miro at the OMM as previously described, causing delays in mitophagy initiation. αS may also impact autophagosome formation by causing a reduction in Parkin levels, affecting the ubiquitination of mitochondrial proteins. (<b>iv</b>) By binding to spectrin, αS may excessively stabilize the actin cytoskeleton, resulting in the mislocalization of key proteins involved in autophagosome trafficking. This mislocalization may also have global effects, disrupting other forms of cellular trafficking. (<b>v</b>) Overexpressed αS may decrease SNAP29 activity, affecting the SNARE complex that mediates autophagosome–lysosome fusion during the last step of mitophagy. Figure created with BioRender. Partially adapted from Thorne and Tumbarello [<a href="#B32-biomolecules-14-01649" class="html-bibr">32</a>].</p>
Full article ">
14 pages, 1632 KiB  
Article
Cannabis (THC) Aggravates the Deleterious Effects of Alcohol (EtOH) on Skeletal Muscles’ Mitochondrial Respiration: Modulation by Age and Metabolic Phenotypes
by Anne-Laure Charles, Margherita Giannini, Alain Meyer, Anne Charloux, Samy Talha, Thomas Vogel, Jean-Sébastien Raul, Valérie Wolff and Bernard Geny
Biology 2024, 13(12), 1080; https://doi.org/10.3390/biology13121080 - 21 Dec 2024
Viewed by 426
Abstract
The anti-inflammatory and analgesic properties of cannabis might be useful to treat muscle diseases, including those linked or not to alcohol. Nevertheless, delta 9 tetrahydrocannabinol (THC) and ethanol (EtOH), often used concomitantly, can have deleterious effects on cardiac mitochondria. We therefore determined whether [...] Read more.
The anti-inflammatory and analgesic properties of cannabis might be useful to treat muscle diseases, including those linked or not to alcohol. Nevertheless, delta 9 tetrahydrocannabinol (THC) and ethanol (EtOH), often used concomitantly, can have deleterious effects on cardiac mitochondria. We therefore determined whether EtOH, alone and associated with THC, impairs skeletal muscle mitochondrial respiration. Further, we investigated potential modulation by metabolic phenotype and age by analyzing predominantly glycolytic gastrocnemius and oxidative soleus muscles in young and middle-aged rats (12 and 49 weeks). Considering the gastrocnemius, EtOH impaired mitochondrial respiration in a similar manner in young- and middle-aged muscles (−34.97 ± 2.97% vs. −37.50 ± 6.03% at 2.1 × 10−5 M; p < 0.05). Interestingly, concomitant THC aggravated EtOH-related mitochondrial impairment in young gastrocnemius (−49.92 ± 1.69%, vs. −34.97 ± 2.97 p < 0.05). Concerning the soleus, EtOH alone mainly decreased young muscle mitochondrial respiration (−42.39 ± 2.42% vs. −17.09 ± 7.61% at 2.1 × 10−5 M, p < 0.001, at 12 and 49 weeks). The soleus was less impaired at 12 weeks by THC and EtOH association than the gastrocnemius (−49.92 ±1.69 vs. −27.22 ± 8.96% in gastrocnemius and soleus, respectively, p < 0.05). In conclusion, EtOH, alone and associated with THC, significantly impairs skeletal muscle mitochondrial respiration and THC aggravates EtOH-induced effects on young glycolytic muscle. Age and metabolic phenotypes modulate these deleterious effects, with the glycolytic muscles of young rats being more prone to impairments than oxidative muscles. Full article
(This article belongs to the Special Issue Musculoskeletal Biology: Impact of Ageing and Disease)
Show Figures

Figure 1

Figure 1
<p>Baseline gastrocnemius and soleus muscles’ mitochondrial respiration before THC or EtOH addition at 12 weeks (<b>a</b>) or 49 weeks (<b>b</b>). Mitochondrial respiration before THC or EtOH addition, corresponding to the 100% values shown in other figures. * <span class="html-italic">p</span> &lt; 0.05. n = 4 (14–15 runs) for 12 weeks; n = 3 (n = 15–17 runs) for 49 weeks.</p>
Full article ">Figure 2
<p>Dose–response effect of EtOH alone and of EtOH with THC on gastrocnemius mitochondrial respiration in 12-week-old and 49-week-old rats. (<b>a</b>) Effect of EtOH alone. (<b>b</b>) Effect of concomitant EtOH and THC. (<b>c</b>) Effect of THC and EtOH contributions to mitochondrial respiration in the gastrocnemius at 12 weeks or 49 weeks. The injection numbers correspond to the concentrations of EtOH (from 0.1 × 10<sup>−5</sup> to 2.1 × 10<sup>−5</sup> M) or THC/EtOH (from 1 × 10<sup>−5</sup>, to 0.2 × 10<sup>−3</sup> M). Values are means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 vs. baseline. Comparisons between groups <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> &lt; <span class="html-italic">p</span> &lt; 0.01. EtOH: ethanol. THC: tetrahydrocannabinoid. n = 4 for 12 weeks; n = 3 for 49 weeks.</p>
Full article ">Figure 3
<p>Dose–response effect of EtOH alone and a combination of EtOH and THC on soleus mitochondrial respiration in 12-week-old and 49-week-old rats. (<b>a</b>) Effect of EtOH alone. (<b>b</b>) Effect of concomitant EtOH and THC. (<b>c</b>) Effect of THC and EtOH contributions to mitochondrial respiration in the soleus at 12 weeks or 49 weeks. The injection numbers correspond to the concentrations of EtOH (from 0.1 × 10<sup>−5</sup> to 2.1 × 10<sup>−5</sup> M) or THC/EtOH (from 1 × 10<sup>−5</sup>, to 0.2 × 10<sup>−3</sup> M). Values are means ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline of each group. EtOH: ethanol. THC: tetrahydrocannabinoid. n = 4 for 12 weeks; n = 3 for 49 weeks.</p>
Full article ">Figure 4
<p>Concomitant THC and EtOH effects are more deleterious on 12-week glycolytic muscle than EtOH alone. These values were obtained for 2.1 × 10<sup>−5</sup> M EtOH, and 0.2 × 10<sup>−3</sup> M THC/EtOH. Values are means ± SEM. * <span class="html-italic">p</span> &lt; 0.05 EtOH: ethanol. THC: tetrahydrocannabinoid. n = 4 for 12 weeks; n = 3 for 49 weeks.</p>
Full article ">Figure 5
<p>Protective effect of the oxidative phenotype of young muscles against EtOH associated with THC-related deleterious effects. Comparison of gastrocnemius and soleus after injection of 0.2 × 10<sup>−3</sup> M THC/EtOH at 12 weeks. Values are means ± SEM. * <span class="html-italic">p</span> &lt; 0.05. n = 4 for 12 weeks; n = 3 for 49 weeks.</p>
Full article ">
Back to TopTop