[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

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

Search Results (1,469)

Search Parameters:
Keywords = SIRT

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 19430 KiB  
Article
Thymoquinone-Loaded Chitosan Nanoparticles Combat Testicular Aging and Oxidative Stress Through SIRT1/FOXO3a Activation: An In Vivo and In Vitro Study
by Enas A. Kasem, Gehan Hamza, Nagi M. El-Shafai, Nora F. Ghanem, Shawky Mahmoud, Samy M. Sayed, Mohammed Ali Alshehri, Laila A. Al-Shuraym, Heba I. Ghamry, Magdy E. Mahfouz and Mustafa Shukry
Pharmaceutics 2025, 17(2), 210; https://doi.org/10.3390/pharmaceutics17020210 - 6 Feb 2025
Viewed by 282
Abstract
Background: Aging is a complex biological process characterized by the accumulation of molecular and cellular damage over time, often driven by oxidative stress. This oxidative stress is particularly detrimental to the testes, where it causes degeneration, reduced testosterone levels, and compromised fertility. D-galactose [...] Read more.
Background: Aging is a complex biological process characterized by the accumulation of molecular and cellular damage over time, often driven by oxidative stress. This oxidative stress is particularly detrimental to the testes, where it causes degeneration, reduced testosterone levels, and compromised fertility. D-galactose (D-gal) is commonly used to model aging as it induces oxidative stress, mimicking age-related cellular and molecular damage. Testicular aging is of significant concern due to its implications for reproductive health and hormonal balance. This research examines the protection by thymoquinone (TQ) or thymoquinone-loaded chitosan nanoparticles (NCPs) against D-galactose (D-gal)-induced aging in rat testes, focusing on biochemical, histological, and molecular changes. Aging, which is driven largely by oxidative stress, leads to significant testicular degeneration, reducing fertility. D-gal is widely used to model aging due to its ability to induce oxidative stress and mimic age-related damage. TQ, a bioactive ingredient of Nigella sativa, has earned a reputation for its anti-inflammatory, anti-apoptotic, and antioxidant characteristics, but its therapeutic application is limited by its poor bioavailability. Methods: Thymoquinone was loaded into chitosan nanoparticles (NCPs) to enhance its efficacy, and this was hypothesized to improve its stability and bioavailability. Four groups of male Wistar rats participated in the study: one for the control, one for D-gal, one for D-gal + TQ, and the last one for D-gal + NCP. Results: The results exhibited that D-gal substantially increased oxidative injury, reduced testosterone levels, and caused testicular damage. Treatment with TQ and NCPs significantly reduced oxidative stress, improved antioxidant enzyme levels, and restored testosterone levels, with NCPs showing a stronger protective effect than TQ alone. A histological analysis confirmed that NCPs better preserved testicular structure and function. Additionally, the NCP treatment upregulated the expression of key genes of oxidative stress resistance, mitochondrial function, and reproductive health, including SIRT1, FOXO3a, and TERT. Conclusions: The findings suggest that NCPs offer enhanced protection against aging-related testicular damage compared with TQ alone, which is likely due to the improved bioavailability and stability provided by the nanoparticle delivery system. This research emphasizes the potential of NCPs as a more effective therapeutic strategy for mitigating oxidative stress and age-related reproductive dysfunction. Future research should further explore the mechanisms underlying these protective effects. Full article
Show Figures

Figure 1

Figure 1
<p>Characterization of the fabricated nanomaterials: (<b>a</b>) XRD patterns; (<b>b</b>) zeta potential; (<b>c</b>) dynamic light scattering; (<b>d</b>) UV−Vis spectroscopy; (<b>e</b>) FT−IR spectrum of Cs NPs.</p>
Full article ">Figure 2
<p>SEM micrograph of the fabricated nanomaterials: (<b>a</b>) Cs NPs (1 µm); (<b>b</b>–<b>d</b>) NCPs: 5 µm, 10 µm, and 1 µm; (<b>e</b>) EDX analysis.</p>
Full article ">Figure 3
<p>(<b>A</b>) Inhibition of cellular viability by NCPs (mean ± SE). *: <span class="html-italic">p</span> &lt; 0.05; ***: <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) Dose-response effect of NCPs on cellular viability (IC50). (<b>C</b>) H9C2 cells were incubated for 48 h in the presence or absence of NCPs (0.01, 0.1, 1, 10, and 100 µg/mL) on a cell invasion assay plate.</p>
Full article ">Figure 4
<p>The effects of TQ and NCP administration on body weight and the percentage change in body weight. Data are shown as the mean ± SE. Different subscript letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Effects of D-gal, TQ, and NCPs on SOD, MDA, and CAT levels in the serum and testicular tissues of rats. Panels (<b>A</b>–<b>C</b>) represent the levels of SOD, MDA, and CAT in the testicular tissues, respectively. Panels (<b>D</b>–<b>F</b>) display the corresponding serum levels of SOD, MDA, and CAT. Each bar represents the mean ± SE. * <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, and ns = not significant.</p>
Full article ">Figure 6
<p>Effects of D-gal, TQ, and NCPs on testosterone levels and SDH, LDH, and γ-GTP activity in rats: (<b>A</b>) testicular testosterone; (<b>B</b>) serum testosterone; (<b>C</b>) succinate dehydrogenase (SDH); (<b>D</b>) lactate dehydrogenase (LDH); (<b>E</b>) gamma-glutamyl transpeptidase (γ-GTP). The data are expressed as the mean ± 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; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, and ns = not significant.</p>
Full article ">Figure 7
<p>Effects of D-gal, TQ, and NCPs on testicular 17β-HSD, 3β-HSD, and LDH-C activity in rats: (<b>A</b>) testicular 17β-HSD; (<b>B</b>) testicular 3β-HSD; (<b>C</b>) testicular LDH-C. Data are expressed as the mean ± 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; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, and ns = not significant.</p>
Full article ">Figure 8
<p>Photomicrographs of H&amp;E staining of rat testis sections in different experimental groups and the control. (<b>A</b>) Normal control group showing normal testicular tissue histology with normal arrangement of spermatogonia and Sertoli cells and normal shape of seminiferous tubules (STs) resting on an intact basement membrane (arrow). Normal late-stage sperm (S) in the lumen and normal Leydig cell distribution (*). (<b>B</b>) The D-gal group showed degenerated STs, with numerous degenerated germ cells (curved arrows), absence of late-stage sperm (S), and separation (arrowhead) of basal and adluminal cellular compartments in some STs. (<b>C</b>) The D-gal TQ group shows a nearly normal ST structure with the separation (arrowhead) of basal and adluminal layers in some areas, as well as an intact basement membrane (arrow). (<b>D</b>) The D-gal NCP group shows slight amelioration of most seminiferous tubules (STs), with no noticed histopathological changes, normal late-stage sperm (S) in the lumen with a normal basement membrane (arrow), and Leydig cells (*). Magnification ×: 400.</p>
Full article ">Figure 9
<p>Immunohistochemical staining in testicular sections from the control and experimental groups using P53 immunostaining. (<b>A</b>) Normal control group: P53-immunolabeled cells were rarely present in the testes of control rats with a normal seminiferous tubule (ST) architecture. (<b>B</b>) D-gal group: This was identified by a slight brown staining increase, representing P53-immunolabeled cells as Leydig cells (thick arrows) and spermatogonial cells (thin arrow), suggesting an inflammatory response and a slight increase in apoptosis. (<b>C</b>) D-gal TQ group: P53-immunolabeled cells were moderately increased (arrow), indicating slight testicular section injury. (<b>D</b>) D-gal NCP group: A few cells showed positive expression for P53, indicating amelioration in testicular sections. The brown color indicates immunopositivity for P53 staining; ST: seminiferous tubule. LC: Leydig cells. Magnification: 40×. (<b>E</b>) Percentage area of P53-immunolabeled cells. Data are expressed as the mean ±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, and ns = not significant.</p>
Full article ">Figure 10
<p>Transmission electron microscopy (TEM) of the effects of D-gal, TQ, and NCPs on the seminiferous tubules from the testes of the control group showing (<b>A</b>) normal spermatogonia (G) with a large spherical nucleus (N) and mitochondria (m) resting on a regular basement membrane (arrow) alongside a myoid cell with a flattened nucleus (My). (<b>B</b>) A primary spermatocyte (P) with a rounded nucleus (N) containing heterochromatin clumps (Ch) and normal mitochondria (M). (<b>C</b>) A round spermatid (SP) with an euchromatic nucleus (N), a well-formed acrosomal cap (tailed arrow), an acrosomal granule (AG), and a flattened Golgi body (Go). Mitochondria (M) are peripherally arranged with free ribosomes (R). (<b>D</b>) Transverse sections of the sperm tail show the middle piece (MP) with central microtubules (curved arrows), outer dense fibers (zigzag arrow), a mitochondrial sheath (crossed arrow), and a plasma membrane (arrowhead). In contrast, the end piece (EP) includes a central axoneme within a plasma membrane.</p>
Full article ">Figure 11
<p>Transmission electron microscopy (TEM) of the effects of D-gal, TQ, and NCPs on the seminiferous tubules from the testes from the D-gal group showing (<b>A</b>) spermatogonia (G) with an irregular membrane partially detached from the basement membrane (arrowhead), an oval nucleus (N), and enlarged mitochondria (m) with vacuoles (crossed arrow). A primary spermatocyte (P) with an irregular nucleus (N) and increased cell-to-cell junctions (*). (<b>B</b>) Two primary spermatocytes (P) with distorted shapes, large nuclei (N) with peripheral heterochromatin, and cytoplasm containing mitochondria (m) and vacuoles (crossed arrow). (<b>C</b>) A round spermatid (SP) with an eccentric nucleus (N), abnormally positioned mitochondria (m), widened intercellular spaces and necrotic debris in the intervening spaces (*), and a degenerated acrosomal cap (AC). (<b>D</b>) Shrinkage of the middle (MP) and end pieces (EP) of sperm, with cross-sections showing a well-developed sperm head with a nucleus (N) and vacuolated acrosomal cap (arrow). Vacuoles are present in the cytoplasm.</p>
Full article ">Figure 12
<p>Transmission electron microscopy (TEM) of the effects of D-gal, TQ, and NCPs on seminiferous tubules from the testes of the D-gal TQ group showing (<b>A</b>) spermatogonia (G) with a large, irregular nucleus (N) and cytoplasm containing normally shaped mitochondria (m) but with an abnormal distribution, resting on a thin basement membrane (thick arrow) with numerous lipid droplets (LDs). (<b>B</b>) A primary spermatocyte (P) with an irregularly shaped nucleus (N), small mitochondria (m), and visible vacuoles (crossed arrow). (<b>C</b>) A round spermatid (SP) with a euchromatic nucleus (N), some mitochondria with abnormal shapes scattered throughout the cytoplasm (m), an acrosomal cap (tailed arrow), and an acrosomal granule (AG). (<b>D</b>) Middle (MP) and end pieces (EP) of sperm, each containing a central axoneme (curved arrow), varying sizes of the middle piece with a moderately normal axoneme enclosed by nine thick, dense fibers (zigzag arrow), and mitochondrial sheaths (crossed arrow).</p>
Full article ">Figure 13
<p>Transmission electron microscopy (TEM) of the effects of D-gal, TQ, and NCPs on (<b>A</b>) spermatogonia (G) with a normal nucleus (N). An electron micrograph of several sections of seminiferous tubules taken from the testes of the D-gal NCP group; cytoplasm containing mitochondria (M) with different sizes, cells resting on a regular basement membrane (thick arrow), and multi-vacuoles (V) still appear. (<b>B</b>) The primary spermatocyte (P) has rounded nuclei (N) containing clumps of heterochromatin (Ch) that are scattered all over the nucleoplasm, with the appearance of disorganized mitochondria (m) and few vacuoles (crossed arrow). (<b>C</b>) The spermatid (SP) has a euchromatic nucleus (N) and a well-formed acrosomal cap (tailed arrow). Mitochondria (m) appear near one pole of the nucleus, forming a flattened Golgi body (Go). (<b>D</b>) Most middle pieces (MPs) of sperms in the lumen have a normal central axoneme (curved arrows) with the presence of swallowed middle pieces with mitochondrial sheaths (crossed arrow) and dense fibers (zigzag arrow).</p>
Full article ">Figure 14
<p>Effects of D-gal, TQ, and NCPs on the mRNA expression levels of (<b>A</b>) <span class="html-italic">SIRT1</span>, (<b>B</b>) <span class="html-italic">FOXO3a</span>, (<b>C</b>) <span class="html-italic">IGF-1</span>, (<b>D</b>) <span class="html-italic">PGC1α</span>, (<b>E</b>) <span class="html-italic">PRM1</span>, (<b>F</b>) <span class="html-italic">TERT</span>, (<b>G</b>) SOD2, and (<b>H</b>) CAT in rat testicular tissue. The data represent the mean ± 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; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, and ns = not significant.</p>
Full article ">
31 pages, 3323 KiB  
Article
Plant-Derived B-CGT Hydrogel Accelerates Diabetic Wound Healing Through Multitarget Modulation of Inflammation, Angiogenesis, and Tissue Remodeling
by Fei Ran, Kailang Mu, Lingli Zhou, Leqiang Peng, Gang Liu, Yuchen Liu, Yuxin Pang, Guo Feng, Changmao Guo, Tianjian Wang and Qiumei Luo
Gels 2025, 11(2), 104; https://doi.org/10.3390/gels11020104 - 2 Feb 2025
Viewed by 265
Abstract
Diabetic wound healing presents significant challenges due to impaired angiogenesis, chronic inflammation, and cellular dysfunction. Building on previous research, this study further explores the potential of a plant-derived glucosyloxybenzyl 2-isobutylmalates (B-CGT) hydrogel in promoting diabetic wound healing. Network pharmacology and molecular docking analyses [...] Read more.
Diabetic wound healing presents significant challenges due to impaired angiogenesis, chronic inflammation, and cellular dysfunction. Building on previous research, this study further explores the potential of a plant-derived glucosyloxybenzyl 2-isobutylmalates (B-CGT) hydrogel in promoting diabetic wound healing. Network pharmacology and molecular docking analyses suggest that B-CGT may regulate key mechanisms, such as apoptosis, inflammation, and matrix remodeling, through core targets including SIRT1, CASP8, and MMP8. In vivo studies further demonstrated that B-CGT hydrogel significantly accelerated wound closure in diabetic mice, enhanced angiogenesis, promoted collagen deposition, and achieved immune balance by modulating macrophage polarization, thereby shifting the inflammatory environment toward a repair state. Moreover, B-CGT hydrogel significantly improved the wound microenvironment by upregulating VEGF expression and exerting antioxidant effects. By combining theoretical predictions with experimental validation, this study elucidates the multi-target synergistic regulatory mechanisms of B-CGT hydrogel. These findings provide new research directions for addressing immune imbalance and angiogenesis defects in diabetic wound healing and lay a scientific foundation for the optimization and application of chronic wound treatment strategies. Full article
(This article belongs to the Special Issue Advances in Gels for Wound Treatment)
17 pages, 2163 KiB  
Article
The Chemopreventive Effect of Ginsenoside Compound K Is Regulated by PARP-1 Hyperactivation, Which Is Promoted by p62-Dependent SIRT6 Degradation
by Sang-Hun Kim, Sung-Hwan Ki, Seok-Woo Hyeong and Seon-Hee Oh
Nutrients 2025, 17(3), 539; https://doi.org/10.3390/nu17030539 - 31 Jan 2025
Viewed by 342
Abstract
Background and aims: Ginsenoside compound K (CK), a saponin metabolite of ginseng, exerts anticancer effects; however, its molecular mechanisms of action in lung cancer remain unclear. We investigated the involvement of silent information regulator 6 (SIRT6) and poly (ADP-ribose) polymerase 1 (PARP-1) in [...] Read more.
Background and aims: Ginsenoside compound K (CK), a saponin metabolite of ginseng, exerts anticancer effects; however, its molecular mechanisms of action in lung cancer remain unclear. We investigated the involvement of silent information regulator 6 (SIRT6) and poly (ADP-ribose) polymerase 1 (PARP-1) in the anticancer effects of CK in lung cancer. Methods and Results: CK induced PARP-1 activation-mediated parthanatos via sequestosome-1/p62-mediated SIRT6 degradation and inhibited the proliferation of H460 cells. Although CK reduced procaspase-8 levels, no significant apoptotic cleavage of procaspase-3 or PARP-1 was observed. Furthermore, CK upregulated p27, p21, phospho-p53, and gamma-H2AX levels. CK increased LC3-II levels in a p62-independent manner, but p62 was upregulated by autophagy inhibition, indicating that p62 is involved in CK-induced autophagy. CK-treated cells showed typical features of parthanatos, including PARP-1 hyperactivation, intracellular redistribution of poly ADP-ribose and pro-apoptotic factors, and chromatin fragmentation. SIRT6 was degraded in a CK concentration- and time-dependent manner. SIRT6 protein was upregulated by PARP-1 inhibition, nicotinamide adenine dinucleotide (NAD)+ supplementation, antioxidants, and p62 knockdown, but was decreased by autophagy blockade. PARP-1 activation was negatively correlated with SIRT6 levels, indicating that SIRT6 and PARP-1 activation play complementary roles in CK-induced growth inhibition. Immunofluorescence staining, fractionation studies, and immunoprecipitation were used to confirm the colocalization and interaction between p62 and SIRT6. Conclusions: PARP-1 activation is promoted by p62-mediated SIRT6 degradation, which plays an important role in CK-induced growth inhibition. Therefore, SIRT6 is a potential biomarker for the chemopreventive effect of CK in lung cancer cells, but further studies on SIRT6 are needed for the clinical application of CK. Full article
(This article belongs to the Special Issue Natural Active Substances and Cancer)
Show Figures

Figure 1

Figure 1
<p>CK induced morphological changes and cell proliferation inhibition. (<b>A</b>) H460 cell morphological changes after the exposure to increasing CK concentrations for up to 24 h monitored by phase-contrast microscopy. Arrows and red asterisk denote the cytoplasmic vacuoles and floating cells, respectively. (<b>B</b>) Cells were treated as outlined in A, and cell viability was determined using an MTT assay. Data are expressed as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005. (<b>C</b>,<b>D</b>) Cells were exposed for 18 h in ascending order of CK concentration or for up to 24 h with a CK concentration of 35 µg/mL. Lysates were analyzed based on immunoblotting. β-actin was used as a loading control (<span class="html-italic">n</span> &gt; 3).</p>
Full article ">Figure 2
<p>CK induced parthanatos and SIRT6 protein degradation. (<b>A</b>) H460 cells were exposed for 18 h in ascending order of CK concentration or for up to 24 h with a CK concentration of 35 µg/mL. Lysates were analyzed based on immunoblotting. β-actin was used as a loading control (<span class="html-italic">n</span> &gt; 3). (<b>B</b>) H460 cells cultured on coverslips were exposed to CK for 18 h, fixated, and the nuclei were stained with Hoechst 33342. Arrows indicate the parthanatos nuclear features. Images were taken using a fluorescence microscope (×200). (<b>C</b>) Cells were exposed to CK for 6, 12, and 18 h and subsequently performed intracellular fractionation into insoluble (nuclear enriched membrane), cytosolic, and particulate (mitochondrial enriched) fractions. The enrichment of each fraction was assessed via immunoblotting for HDAC1, β-actin, and SOD2 (<span class="html-italic">n</span> &gt; 3). (<b>D</b>) Cells were exposed to CK for 18 h after a pretreatment with 3-AB, ANI, zVAD-fmk, or dimethylsulfoxide (DMSO) control for 2 h. Lysates were analyzed using immunoblotting for the indicated proteins. β-actin was used as the loading control. (<b>E</b>,<b>F</b>) Cells were exposed to increasing NAD+ concentrations (25 and 50 nM) for 18 h after pretreatment and then to CK. Immunoblotting was used to analyze the levels of the indicated proteins. Cells were cultured for 24 h as outlined in E, and cell viability was measured using MTT. Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 3
<p>SIRT6 negatively regulates PARP-1 activation by CK. (<b>A</b>) Immunoblotting was used to evaluate the knockdown efficiency of Sirt6-specific siRNA. NC = negative control. (<b>B</b>) H460 cells infected with NC or Sirt6 siRNA were challenged with CK for 18 h and the levels of the shown proteins were assayed using immunoblotting (<span class="html-italic">n</span> = 3). (<b>C</b>) Morphological changes in cells treated as in B were viewed with phase-contrast microscopy. Arrows represent cytoplasmic vacuoles. (<b>D</b>) After 24 h of transfection with an empty vector or pcDNA3.1-Sirt6, immunoblotting for SIRT6 was performed. (<b>E</b>,<b>F</b>) Cells overexpressing SIRT6 were treated with 35 µg/mL of CK for 18 h, observed for morphological changes, and then harvested and lysed. Immunoblotting was used to analyze the levels of the indicated proteins. β-actin was taken as a loading control. Arrows indicate the cytoplasmic vacuoles. Scale bar = 25 µm.</p>
Full article ">Figure 4
<p>Oxidative stress and autophagy regulate CK-induced PARP-1 activation and SIRT6 protein stability. (<b>A</b>) H460 cells were exposed for 18 h in ascending order of CK concentration or for up to 24 h with a CK concentration of 35 µg/mL. Lysates were analyzed based on immunoblotting. (<b>B</b>) Cells were exposed to CK a maximum of 8 h and were stained with JC-1 dye. Images were captured under a fluorescence microscope. Scale bar = 25 µm. (<b>C</b>) Cells were pretreated with NAC (5 mM) or tocopherol (25 µM) for 2 h and then continuously treated with CK for 18 h. Immunoblotting was used to analyze the levels of the indicated proteins (<span class="html-italic">n</span> ≥ 3). (<b>D</b>) After culturing cells treated as described in C for 24 h, the viability was determined using the MTT assay. Data are expressed as the mean ± SD. # <span class="html-italic">p</span> &lt; 0.001; *** <span class="html-italic">p</span> &lt; 0.0005. (<b>E</b>) Cells were pretreated with BaF1 (100 nM), CQ (50 µM), and DMSO for 2 h and then exposed to CK for 18 h. The lysates were subjected to immunoblotting for the indicated proteins. (<b>F</b>,<b>G</b>) The knockdown efficiency obtained using ATG5-specific siRNA was evaluated using immunoblotting for ATG5. NC = negative control. Cells knocked down with NC or ATG5 siRNA were exposed to CK (35 µg/mL) for 18 h and the levels of each protein were assayed using immunoblotting. β-actin was taken as a loading control (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>The subcellular translocation of SIRT6 depends on p62. (<b>A</b>) The indicated proteins were analyzed in the subcellular fraction samples obtained in <a href="#nutrients-17-00539-f002" class="html-fig">Figure 2</a>C. (<b>B</b>,<b>C</b>) Immunoblotting for p62 was performed to assess the knockdown efficiency of the p62-specific siRNA. NC = negative control. Cells transfused with NC or p62 siRNA were incubated with CK (35 µg/mL) for 18 h. The shown proteins were assayed by immunoblotting. β-actin was taken as a loading control (<span class="html-italic">n</span> = 3). (<b>D</b>) Cells grown on coverslips were transfected with NC or p62 siRNA, exposed to CK for 12 h, fixed, and performed IF immunostaining for both SIRT6 (red) and p62 (green). Nuclei were counterstained with Hoechst 33342 (blue). The yellow arrows indicate p62 in the perinuclear region. Scale bar, 25 µm.</p>
Full article ">Figure 6
<p>SIRT6 can interact with p62 through its ubiquitination. (<b>A</b>) H460 cells were pretreated with LMB (25 nm) or DMSO for 2 h and then continuously treated with CK for 18 h. (<b>B</b>) Cells were harvested and subjected to intracellular fractionation into insoluble (nuclear enriched membrane), cytosolic, and microparticulate fractions. Enrichment of individual fractions was measured using immunoblotting for HDAC1 (nucleus), β-actin (cytosol), SOD2 (mitochondria), LC3-II (autophagosome), and calnexin (endoplasmic reticulum) (<span class="html-italic">n</span> &gt; 3). (<b>C</b>) Cells grown on coverslips were treated with DMSO or LMB (25 nm) for 2 h, continuously exposed to CK for 12 h, fixed, and performed IF staining for SIRT6 (red) and p62 (green). Nuclei were counterstained by Hoechst 33342 (blue). Scale bar, 25 µm. (<b>D</b>,<b>E</b>) Cells were exposed to CK (30 µg/mL) for 12 h, lysates were immunoblotted for SIRT6, and 800 µg of the residual protein was used for IP assays using p62, ubiquitin (Ub), and mouse IgG antibodies, followed by immunoblotting for SIRT6 (<span class="html-italic">n</span> = 2).</p>
Full article ">Figure 7
<p>Schematic diagram of CK-induced PARP-1 activation and its regulatory mechanism.</p>
Full article ">
19 pages, 3898 KiB  
Article
High Rosmarinic Acid Content Melissa officinalis L. Phytocomplex Modulates Microglia Neuroinflammation Induced by High Glucose
by Giacomina Videtta, Chiara Sasia and Nicoletta Galeotti
Antioxidants 2025, 14(2), 161; https://doi.org/10.3390/antiox14020161 - 29 Jan 2025
Viewed by 470
Abstract
Diabetic patients experience hyperglycemia, which can affect multiple organs, including brain function, leading to disabling neurological complications. Hyperglycemia plays a key role in promoting neuroinflammation, the most common complication in diabetic individuals, through the activation of microglia. Attenuating hyperglycemia-related neuroinflammation in microglia may [...] Read more.
Diabetic patients experience hyperglycemia, which can affect multiple organs, including brain function, leading to disabling neurological complications. Hyperglycemia plays a key role in promoting neuroinflammation, the most common complication in diabetic individuals, through the activation of microglia. Attenuating hyperglycemia-related neuroinflammation in microglia may reduce diabetes-associated neurological comorbidities. Natural remedies containing phenolic compounds have shown efficacy in mitigating microglia-mediated neuroinflammation. The aim of this study was to investigate the potential of a Melissa officinalis L. (MO) phytocomplex, obtained from plant cell cultures and enriched in its main polyphenolic constituent, rosmarinic acid (RA), in attenuating hyperglycemia-induced neuroinflammation in microglia. A time-course morphological analysis of BV2 microglial cells exposed to high glucose (HG) levels showed a shift towards a proinflammatory phenotype, peaking after 48 h, which was reversed by pretreatment with MO. Biochemical assays revealed increased expression of the microglial marker CD11b (187%), activation of the NF-κB pathway (179%), expression of iNOS (225%), enhanced phosphorylation of ERK1/2 (180%), and increased expression of the proinflammatory cytokine IL-6 (173%). Pretreatment with MO prevented the aberrant expression of these proinflammatory mediators and restored SIRT1 levels. Exposure of neuronal SH-SY5Y cells to the conditioned medium from HG-exposed microglia significantly reduced cell viability. MO counteracted this effect, exhibiting neuroprotective activity. RA showed efficacy comparable to that of MO. In conclusion, MO and RA attenuated microglia-mediated oxidative imbalance and neuroinflammation under HG exposure by inhibiting the morphological shift toward a proinflammatory phenotype induced by HG and abrogating the subsequent activation of the downstream ERK1/2–NF-κB–iNOS pathway. Full article
(This article belongs to the Special Issue Plant Antioxidants, Inflammation, and Chronic Disease)
Show Figures

Figure 1

Figure 1
<p><sup>1</sup>H NMR profile of MO. The regions of δ 4.94−4.78, δ 3.34−3.30, 0.20−0.20 were excluded from the analysis because of the residual signals of solvents and standard. 1 = rosmarinic acid, 2 = sucrose, 3 = α-glucose, 4 = β-glucose, 5 = unknown sugar, 6 = citrate, 7 = acetate, 8 = alanine, 9 = ethanol, 10 = valine.</p>
Full article ">Figure 2
<p>UPLC-DAD profile of the MO extract. The main peak at a retention time of 8.1 min corresponds to rosmarinic acid.</p>
Full article ">Figure 3
<p>Time-dependent reduction in BV2 cell number by high glucose (HG) exposure. (<b>A</b>) BV2 cell number in cultures exposed to normal glucose (NG; 5.5 mM), high glucose (HG; 25 mM), or high mannitol (HM; 25 mM) concentrations at different time points (2, 8, 24, 48, and 72 h). Scatter plots of data obtained after 24 h (<b>B</b>) or 48 (<b>C</b>) hours of treatment. * <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. Representative images of HG-exposed cells after 24 h (<b>D</b>) and 48 h (<b>E</b>). Scale bar: 50 µm.</p>
Full article ">Figure 4
<p>Morphological analysis of BV2 cells exposed to high glucose (HG). (<b>A</b>) Soma cell surface area of BV2 cells exposed to normal glucose (NG; 5.5 mM), high glucose (HG; 25 mM), or high mannitol (HM; 25 mM) concentrations at different time points (2, 8, 24, 48, and 72 h). * <span class="html-italic">p</span> &lt; 0.05 vs. NG; ° <span class="html-italic">p</span> &lt; 0.05 versus HG. Scatter plots of data obtained after 24 h (<b>B</b>) or 48 (<b>C</b>) hours of treatment. **** <span class="html-italic">p</span> &lt; 0.0001. (<b>D</b>) BV2 cell length after exposure to NG, HG, and HM at different time points (2, 8, 24, 48, and 72 h). * <span class="html-italic">p</span> &lt; 0.05 vs. NG; ° <span class="html-italic">p</span> &lt; 0.05 versus HG. Scatter plots of data obtained after 24 h (<b>E</b>) or 48 (<b>F</b>) hours of treatment. **** <span class="html-italic">p</span> &lt; 0.0001. (<b>G</b>) Increase in the percentage of cells in the proinflammatory state after stimulation with HG at different time points (2, 8, 24, 48, and 72 h) in comparison with NG and HM. * <span class="html-italic">p</span> &lt; 0.05 vs. NG; ° <span class="html-italic">p</span> &lt; 0.05 versus HG. Scatter plots of data obtained after 24 h (<b>H</b>) or 48 (<b>I</b>) hours of treatment. **** <span class="html-italic">p</span> &lt; 0.0001. Representative images of HG-exposed cells after 24 h (<b>J</b>) and 48 h (<b>K</b>). Scale bar: 50 µm.</p>
Full article ">Figure 5
<p>Effect of MO on cell viability. (<b>A</b>) Lack of alteration of cell viability by MO (0.1–100 µg/mL) in unstimulated BV2 cells. (<b>B</b>) MO (10 µg/mL) and RA (0.4 µg/mL) attenuation of HG-induced reduction of cell viability. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01*** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Attenuation by MO and RA of HG–induced proinflammatory morphological phenotype in BV2 cells. MO (10 µg/mL) and RA (0.4 µg/mL) attenuation of HG–induced cell number reduction (quantitative analysis (<b>A</b>); representative images (<b>B</b>)), cell length (<b>C</b>), percentage of cells in the proinflammatory state (<b>D</b>), and cell surface area (<b>E</b>). (<b>F</b>) Redistribution of HG–exposed BV2 cell subpopulations by cell surface area after 48 h of stimulation in the presence or absence of MO and RA. Cells were classified as either small (&lt;200 μm<sup>2</sup>), mid-sized (200–400 μm<sup>2</sup>), or large (&gt;400 μm<sup>2</sup>). (<b>G</b>) Effect of MO and CH on HG–induced variation of the percentage of distribution of cells by size. * <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. (<b>H</b>) Representative images of HG-exposed cells. Scale bar: 50 µm.</p>
Full article ">Figure 7
<p>MO and RA inhibition of microglia activation and neuroinflammation. (<b>A</b>) Increase in CD11b expression by HG exposure and attenuation by MO (10 µg/mL) ND RA 0.4 µg/mL). Inhibition of HG-induced p65 overphosphorylation (<b>B</b>), induction of iNOS expression (<b>C</b>), IL-6 expression (<b>D</b>), and ERK1/2 (<b>E</b>) increased phosphorylation by MO and RA. * <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. Representative blots for western blotting analysis of CD11b, IL-6, and pERK1/2 are reported.</p>
Full article ">Figure 8
<p>Neuroprotective effect of MO and RA on SH-SY5Y. (<b>A</b>) Schematic representation of experimental protocol and (<b>B</b>) reduction of cell viability in SH-SY5Y cells exposed to conditioned medium from HG-stimulated BV2 cells compared to NG- and HM-stimulated. CTRL cells were not exposed to the BV2 medium. (<b>C</b>) Schematic representation of experimental protocol and (<b>D</b>) protection by MO and RA treatment from neurotoxicity in SH-SY5Y cells induced by conditioned medium from HG-exposed BV2 cells. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>Antioxidant activity of MO and RA. (<b>A</b>) Anti-radical scavenger activity of rosmarinic acid (RA) (1–100 μg/mL) in the DPPH test. Ascorbic acid was used as a reference drug. (<b>B</b>) Reduction of the overexpression of SIRT1 by MO of HG-exposed BV2 cells. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
39 pages, 2961 KiB  
Review
Do Lifestyle Interventions Mitigate the Oxidative Damage and Inflammation Induced by Obesity in the Testis?
by Ruben J. Moreira, Pedro F. Oliveira, Maria Angélica Spadella, Rita Ferreira and Marco G. Alves
Antioxidants 2025, 14(2), 150; https://doi.org/10.3390/antiox14020150 - 27 Jan 2025
Viewed by 628
Abstract
Obesity results from a disproportionate accumulation of fat and has become a global health concern. The increase in adipose tissue is responsible for several systemic and testicular changes including hormone levels (leptin, adiponectin, testosterone, estrogen), inflammatory cytokines (increase in TNF-α and IL-6 and [...] Read more.
Obesity results from a disproportionate accumulation of fat and has become a global health concern. The increase in adipose tissue is responsible for several systemic and testicular changes including hormone levels (leptin, adiponectin, testosterone, estrogen), inflammatory cytokines (increase in TNF-α and IL-6 and decrease in IL-10), and redox state (increase in reactive oxygen species and reduction in antioxidant enzymes). This results in poor sperm quality and compromised fertility in men with obesity. Lifestyle modifications, particularly diet transition to caloric restriction and physical exercise, are reported to reverse these negative effects. Nevertheless, precise mechanisms mediating these benefits, including how they modulate testicular oxidative stress, inflammation, and metabolism, remain to be fully elucidated. The main pathway described by which these lifestyle interventions reverse obesity-induced oxidative damage is the Nrf2-SIRT1 axis, which modulates the overexpression of antioxidant defenses. Of note, some of the detrimental effects of obesity on the testis are inherited by the descendants of individuals with obesity, and while caloric restriction reverses some of these effects, no significant work has been carried out regarding physical exercise. This review discusses the consequences of obesity-induced testicular oxidative stress on adult and pediatric populations, emphasizing the therapeutic potential of lifestyle to mitigate these detrimental effects. Full article
(This article belongs to the Special Issue Oxidative Stress in Obesity—3rd Edition)
Show Figures

Figure 1

Figure 1
<p>Mechanisms linking high-fat diet to adipose tissue expansion and systemic effects of inflammation and oxidative stress. A high-fat diet leads to hyperplasia and hypertrophy of adipose tissue. This expansion contributes to hyperleptinemia and leptin resistance. Enlarged adipose tissue experiences hypoxia, which activates hypoxia-inducible factor 1-alpha (HIF-1α) and increases plasminogen activator inhibitor-1 (PAI-1). Additionally, unfolded adiponectin triggers the unfolded protein response (UPR), especially the PERK (protein kinase R-like ER kinase) and IRE1 (inositol-requiring enzyme 1) pathways, responsible for the degradation of misfolded adiponectin and consequent hypoadiponectinemia and insulin resistance. Macrophages infiltrate the adipose tissue Adipose tissue also synthesizes monocyte chemoattractant protein-1 (MCP-1), which disturbs insulin signaling by disrupting Akt phosphorylation. MCP-1 contributes to inflammation by secreting inflammatory cytokines such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin-6 (IL-6), Interleukin-1β (IL-1β), and nitric oxide (NO). Inflammatory and oxidative stress markers, including NADPH oxidase (NOX) activity, exacerbate systemic inflammation and oxidative stress. Together, these pathways contribute to dysfunctions associated with metabolic syndrome. ↑—upregulation.</p>
Full article ">Figure 2
<p>High-fat-diet-induced testicular dysfunction and its transgenerational effects. A high-fat diet induces adipose tissue hypertrophy and aromatase activity, increasing estradiol levels. Nitric oxide (NO) released by macrophages and adipose tissue contributes to nitrosative stress, impairing the hypothalamus–pituitary axis and disrupting LH signaling. Other inflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin-6 (IL-6), Interleukin-1β (IL-1β), exacerbate inflammation, further impairing Leydig cell function. Leydig cells exhibit reduced steroidogenic activity due to the activation of the JNK/ERK/MAPK pathway that culminates in the increased Nuclear Factor Kappa B (NF-κB) and DAX-1, which inhibits Nur77 and steroidogenic factor 1 (SF1), transcription factors involved in the expression of steroidogenic enzymes (steroidogenic acute regulatory protein—StAR; Cytochromes P450 CYP11a and CYP17a; and β-Hydroxysteroid dehydrogenases—β-HSD, resulting in decreased testosterone production. The seminiferous tubules show increased apoptosis, disrupted Sertoli cell activity, and compromised blood–testis barrier integrity, contributing to poor sperm quality. Elevated oxidative stress, driven by reduced nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant defenses and increased NADPH oxidase (NOX) activity, exacerbates lipid peroxidation and testicular damage. Transgenerational effects observed in the F1 and F2 generations include reduced testicular weight, decreased sperm parameters, and impaired antioxidant capacity, accompanied by metabolic, lipidic, and transcriptomic inheritance. Red arrows represent the negative effects induced (regular arrow—effect induced; inhibition arrow—process inhibition). ↓—downregulation. ↑—upregulation. Red box—negative effects. Blue box—transgenerational effects.</p>
Full article ">Figure 3
<p>Effects of dietary transition from a high-fat diet to caloric restriction on male reproductive health and transgenerational outcomes. Transitioning from a high-fat diet to caloric restriction leads to improvements in serum markers, including increased testosterone and adiponectin levels, and reduced insulin and leptin levels. In the testes, enhanced expression of key genes—fat mass and obesity-associated protein (FTO), melanocortin 4 receptor (MC4R), sirtuin 1 (SIRT1), glucosamine-6-phosphate deaminase 2 (GNPDA2), transmembrane protein 18 (TMEM18)—promotes antioxidant enzyme activity (via Nrf2), sperm viability, and insulin sensitization. However, caloric restriction is associated with an increase in abnormal sperm head morphology. Transgenerational benefits observed in the F1 and F2 generations include restored testicular weight, improved sperm quality, increased testicular antioxidant capacity, and enhanced metabolic, lipidic, and transcriptomic profiles, suggesting intergenerational inheritance of positive dietary effects. ↓—decrease. ↑—increase. Green box—positive effects. Red box—negative effects. Blue box—transgenerational effects.</p>
Full article ">Figure 4
<p>Effects of physical exercise on male reproductive health and potential transgenerational effects. Moderate-intensity physical exercise decreases inflammatory cytokines (Tumor Necrosis Factor-alpha—TNF-α; Interleukin-6—IL-6; Interleukin-1β—IL-1β; nitric oxide—NO; and monocyte chemoattractant protein-1—MCP-1), insulin, and leptin levels, while increasing testosterone levels, improving sperm motility, count, and normal morphology, and resulting in insufficient apoptosis. The positive effects of moderate exercise loads are associated with decrease in microRNA mir34a, an increase in sirtuin 1 (SIRT1) and nuclear factor erythroid 2-related factor 2 (Nrf2), which is associated with higher testicular antioxidant capacity. High-intensity physical exercise shows no changes in inflammatory cytokines, testosterone production, or sperm motility, count, and normal morphology, but increases Sertoli cell activity. Transgenerational effects of physical exercise on damaged testicles remain unknown. ↓—downregulation. ↑—upregulation. Green box—positive effects. Red box—negative effects. Blue box—transgenerational effects.</p>
Full article ">
21 pages, 4529 KiB  
Article
NAD+ Suppresses EV-D68 Infection by Enhancing Anti-Viral Effect of SIRT1
by Yue Wang, Haiyu Li, Xia Huang, Yan Huang, Mingqi Lv, Hong Tang, Xinyue Han, Juntong Liu, Yan Liang, Guangchao Zang, Nan Lu and Guangyuan Zhang
Viruses 2025, 17(2), 175; https://doi.org/10.3390/v17020175 - 26 Jan 2025
Viewed by 359
Abstract
Enterovirus 68 (EV-D68) is a non-enveloped virus with a positive-sense single-stranded RNA genome that causes respiratory diseases and acute flaccid myelitis, posing significant threats to human health. However, an effective vaccine remains undeveloped. SIRT1, a nicotinamide adenine dinucleotide (NAD+)-dependent enzyme, plays a key [...] Read more.
Enterovirus 68 (EV-D68) is a non-enveloped virus with a positive-sense single-stranded RNA genome that causes respiratory diseases and acute flaccid myelitis, posing significant threats to human health. However, an effective vaccine remains undeveloped. SIRT1, a nicotinamide adenine dinucleotide (NAD+)-dependent enzyme, plays a key role in cellular metabolism, but its interaction with NAD+ during viral infections is not well understood. In this study, through a metabolomics analysis, we demonstrate that EV-D68 infection influences cellular metabolism. Additionally, we show that NAD+ inhibits EV-D68 infection both in vivo and in vitro. EV-D68 reduces cellular NAD+ levels by regulating the expression of enzymes involved in NAD+ consumption and synthesis. Moreover, the infection increases the expression of sirtuin 1 (SIRT1), which inhibits EV-D68 replication in turn. Mechanistically, SIRT1 suppresses EV-D68 5′UTR-mediated translation, and the antiviral effect of SIRT1 on EV-D68 replication is enhanced by NAD+. Collectively, our findings highlight the critical role of NAD+ metabolism in EV-D68 infection and reveal the antiviral potential of SIRT1, providing valuable insights for the development of antiviral strategies. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

Figure 1
<p>EV-D68 infection influences cellular metabolism. RD cells cultured in 10 cm dishes infected with EV-D68 Fermon at an MOI of 1 and non-infected as control. After 18 h infection, cells were harvested for LC-MS analysis (n = 3 samples per group). (<b>a</b>) Principal component analysis of metabolomics data in the mock and EV-D68-infected group. (<b>b</b>) Volcano map of differentially expressed metabolites in EV-D68 compared with that in the mock group. (<b>c</b>) VIP analysis of the top 30 differentially expressed metabolites between the mock and EV-D68 group. The color of each rectangle in the right panel represents the relative level of the metabolites. The metabolites with VIP &gt; 1 and <span class="html-italic">p</span> &lt; 0.05 were determined as significantly different metabolites. The full name of metabolites were shown in in <a href="#app1-viruses-17-00175" class="html-app">Supplementary Tables S1–S4</a>. (<b>d</b>) Pie charts showing the HMDB classifications of the 93 differential metabolites. (<b>e</b>) KEGG pathway enrichment analysis of the significantly altered metabolites between the mock and EV-D68 group.</p>
Full article ">Figure 2
<p>NAD+ suppresses EV-D68 proliferation in vitro. (<b>a</b>,<b>b</b>) RD cells were incubated with different concentrations of flavin adenine dinucleotide (FAD), L-Cystathionine (L-Cth), 8-Hydroxyguanosine (8-OHG), L-Acetylcarnitine (ALCAR), or nicotinamide adenine dinucleotide (NAD+) as indicated for 48 h. Then the cell viability was assessed by Cell Counting Kit-8 (CCK-8) assay. RD cells were treated with different concentrations of exogenous FAD (0, 2.5, 5, and 10 µM), 8-OHG (0, 5, 10, and 20 nM), L-Cth (0, 0.1, 0.5, and 1 mM), ALCAR (0, 0.5, 1, and 2 mM), and NAD+ (0, 0.5, 1, and 2 mM) 6 h prior to infection with EV-D68 Fermon at an MOI of 1 for 18 h. Viral VP1 protein levels were assessed by Western blot. (<b>c</b>) As with the cell line and experiment mentioned above, RD cells were treated with different concentrations of exogenous NAD+ (0, 0.5, 1, and 2 mM) 6 h prior to infection with EV-D68 Fermon at an MOI of 1, and after 24 h the supernatant of the cells was collected. Viral titers were determined using TCID50. Data in all quantitative panels are normalized based on β-actin presented as the mean ± SD of n = 3 replicates. Error bars indicate SD (n = 3). “−” represents absence, and “+” represents presence. *** <span class="html-italic">p</span> &lt; 0.001; ns, not significant.</p>
Full article ">Figure 3
<p>NAD+ suppresses EV-D68 proliferation in vivo. (<b>a</b>) Schematic representation of the experimental design for NAD+ treatment in neonatal mice infected with EV-D68(ATCC VR-1824). Mice were intracranially inoculated with EV-D68 (1 × 10<sup>5</sup> PFU/mouse) and treated with continuous NAD+ therapy until the seventh day. Viral loads in major organs and tissues were evaluated using RT-qPCR on the 3rd day, and survival rates were monitored until the 14th day. (<b>b</b>) Representative images showing symptoms of healthy mice in the mock group and the mice with limb paralysis in the EV-D68(ATCC VR-1824) infection group on 7 dpi. (<b>c</b>) Bar graph showing the percentage of mice with paralysis between the EV-D68-infected and NAD+ treatment groups. (<b>d</b>,<b>e</b>) Levels of EV-D68 viral nucleic acids and VP1 protein in major organs and muscle tissues were quantified using Western blot and RT-qPCR on the third day. (<b>f</b>) Histopathological changes in tissues from mock, EV-D68-infected, and EV-D68-infected with NAD+ treatment groups (n = 3) were analyzed. Pathological images of spine muscles and hind limb muscles, scale bar = 100 μm. Yellow arrow: Vacuolar degeneration of muscle cells. Brown arrow: Fibrosis of interstitial fibrous connective tissue. Red arrow: Infiltration of lymphocytes and granulocytes. Black arrow: Necrosis of chondrocytes. Pathological images of the spinal cord, scale bar = 50 μm. Blue arrow: Atrophied neurons. Yellow arrow: Proliferation of gliosis. Green arrow: Neuronal necrosis. Orange arrow: Vascular congestion. (<b>g</b>) Day of paralysis onset in mice after injection with EV-D68(ATCC VR-1824). (<b>h</b>) Survival rate statistics for the NAD+ treatment (n = 23) and saline control groups (n = 22) following EV-D68 infection were compared. Data in all quantitative panels are normalized based on β-actin or GAPDH presented as the mean ± SD of n = 3 replicates. Error bars indicate SD (n = 3). “−” represents absence, and “+” represents presence. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; ns, not significant.</p>
Full article ">Figure 4
<p>Viral infection regulated the expression of enzymes in the NAD+ salvage pathway, leading to a reduction in NAD+ level. (<b>a</b>) RD cells were cultured in 6-well plates and infected with EV-D68 Fermon at an MOI of 0 or 1 for 24 h. WST—detection of NAD+ in cells by WST-8 reaction colorimetry. (<b>b</b>–<b>e</b>) RD and A549 cells were infected with EV-D68 Fermon at an MOI of 1. Then, 18 h later, the expression changes of NAD+ synthesis genes (Nmnat1 to 3, Nampt, and Nmrk2) and NAD+ consuming genes (Sirt1 to 7, Parp1 to 6, Parp9, 10, 12, 14, and CD38) were measured using RT-qPCR. (<b>f</b>) A549 cells were infected with EV-D68 Fermon at an MOI of 1. Then, 18 h later, the protein levels of VP1, SIRT1, SIRT2, NAMPT, NMNAT2, and CD38, compared to the control β-actin, were determined using Western blot. Data in all quantitative panels are normalized based on β-actin or GAPDH presented as the mean ± SD of n = 3 replicates. Error bars indicate SD (n = 3). “−” represents absence, and “+” represents presence. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>NAD+-consuming enzyme SIRT1 inhibits EV-D68 replication. (<b>a</b>) A549 cells were infected with EV-D68 Fermon at a multiplicity of infection (MOI) of 0, 0.5, and 1. Samples were collected after 24 h. Another group were infected with EV-D68 Fermon at an MOI of 1; samples were collected at 0, 8, and 16 h post-infection (hpi). Protein levels of SIRT1 and VP1 were detected using Western blot. (<b>b</b>) RD cells infected with EV-D68 Fermon at an MOI of 1 for 24 h. Cytoplasm extracts (CEs) and nuclear extracts (NEs) were prepared. SIRT1, β-actin, and lamin A expression levels were detected by Western blot analyses using the corresponding antibodies. (<b>c</b>) RD cells were infected with EV-D68 Fermon at an MOI of 1, and SIRT1 localization was assessed by immunofluorescence at 24 hpi. The SIRT1 was stained with the preliminary antibody rabbit anti-SIRT1 and secondary florescent affiliate antibody goat anti-rabbit AlexaFluor488. The nuclei were counterstained with DAPI. Scale bar represents 50 μm. (<b>d</b>–<b>g</b>) 293T cells were transfected with pBudCE4.1-Sirt1 or pBudCE4.1-mtSirt1 at 2, 4 μg, with pBudCE4.1 as the control. After 48 h, cells were infected with EV-D68 Fermon at an MOI of 1 for 24 h. Protein levels of SIRT1 and viral VP1 were quantified by Western blot. The intensity of Western blot band signals was quantified behind by Image J 1.54g (<b>d</b>,<b>f</b>). And viral titers were measured using TCID50 (<b>e</b>,<b>g</b>). (<b>h</b>,<b>i</b>) 293T cells were transfected with si-Sirt1-1, si-Sirt1-2, and si-SIRT1-3 (30 nM), with si-NC as the control, for 48 h. After that, cells were infected with EV-D68 Fermon at an MOI of 1 for 24 h. Protein levels of SIRT1 and viral VP1 were quantified by Western blot. The intensity of Western blot band signals was quantified behind by Image J (<b>h</b>). And viral titers were measured using TCID50 (<b>i</b>). (<b>j</b>,<b>k</b>) RD cells were treated with Resveratrol and Sirtinol in different doses (0, 20, 40 µM). After 8 h, cells were infected with EV-D68 Fermon for 24 h at an MOI of 1. Protein levels of viral VP1 were quantified by Western blot. The intensity of Western blot band signals was quantified behind by Image J. Data in all quantitative panels are normalized based on β-actin or GAPDH presented as the mean ± SD of n = 3 replicates. Error bars indicate SD (n = 3). “−” represents absence, and “+” represents presence. * <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; ns, not significant.</p>
Full article ">Figure 6
<p>NAD+ augments the antiviral effect of SIRT1. (<b>a</b>,<b>b</b>) 293T cells were transfected with si-Sirt1-1 and si-NC for 30 nM; 48 h later, cells were treated with NAD+ for 8 h and then infected with EV-D68 Fermon at an MOI of 1 for 24 h. Protein levels of SIRT1 and viral VP1 were detected by Western blot. The intensity of Western blot band signals was quantified behind by Image J (<b>a</b>). And viral titers were measured using TCID50 (<b>b</b>). (<b>c</b>,<b>d</b>) 293T cells were transfected with si-Sirt1-3 and si-NC for 30 nM. Then, 48 h later, followed the same experiment mentioned before (<b>a</b>,<b>b</b>). (<b>e</b>,<b>f</b>) 293T cells were transfected with pBudCE4.1-Sirt1 and pBudCE4.1 for 2 μg. Then, followed the same experiment mentioned before (<b>a</b>,<b>b</b>). (<b>g</b>) RD cells were treated with NAD+ for 8 h, and SIRT1 localization was assessed by immunofluorescence. The SIRT1 was stained with the preliminary antibody rabbit anti-SIRT1 and secondary florescent affiliate antibody goat anti-rabbit AlexaFluor488. Thr nuclei were counterstained with DAPI. Scale bar represents 10 μm. (<b>h</b>) Schematic presentation of the luciferase reporter plasmid, containing a pol I promoter, the 5′ UTR region of EV-D68, and the Renilla luciferase gene (Rluc) infused with the first 15 bps of EV-D68 ORF, and a Firefly luciferase reporter (Fluc) gene driven by TK promoter. (<b>i</b>) Cells were co-transfected with p5Ferm and either pBudCE4.1-SIRT1 or the control vector. After 24 h, cells were treated with NAD+ for 8 h. Cells were lysed and their Rluc and Fluc activities measured. Determining the ratio of Rluc activity to Fluc activity yielded the relative IRES activity. Data in all quantitative panels are normalized based on β-actin or GAPDH presented as the mean ± SD of n = 3 replicates. Error bars indicate SD (n = 3). “−” represents absence, and “+” represents presence. * <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; ns, not significant.</p>
Full article ">Figure 7
<p>Schematic diagram illustrating the regulatory mechanism of NAD+ and SIRT1 during EV-D68 infection. EV-D68 infection decreases the level of cellular NAD+ (Black arrow: decrease), which promotes the expression of SIRT1. SIRT1 has an inhibitory effect on EV-D68, and it is strengthened by NAD+.</p>
Full article ">
15 pages, 5367 KiB  
Article
Sirtuin-1 Regulates Mitochondrial Calcium Uptake Through Mitochondrial Calcium Uptake 1 (MICU1)
by Xinyi Zhang, Shuhu Liu, Yanshan Su, Ling Zhang, Ting Guo and Xuemin Wang
Life 2025, 15(2), 174; https://doi.org/10.3390/life15020174 - 25 Jan 2025
Viewed by 464
Abstract
Mitochondria play a central role in cell biological processes, functioning not only as producers of ATP but also as regulators of Ca2+ signaling. Mitochondrial calcium uptake occurs primarily through the mitochondrial calcium uniporter channel (mtCU), with the mitochondrial calcium uptake subunits 1, [...] Read more.
Mitochondria play a central role in cell biological processes, functioning not only as producers of ATP but also as regulators of Ca2+ signaling. Mitochondrial calcium uptake occurs primarily through the mitochondrial calcium uniporter channel (mtCU), with the mitochondrial calcium uptake subunits 1, 2, and 3 (MICU1, MICU2, and MICU3) serving as the main regulatory components. Dysregulated mitochondrial calcium uptake is a hallmark of cellular degeneration. Sirtuin 1 (SIRT1), a key regulator of cellular metabolism, plays a critical role in aging and various neurodegenerative conditions. By blocking SIRT1 using EX527 or shSIRT1, we observed mitochondrial structural fragmentation as well as intensified and prolonged mitochondrial calcium overload. Our study revealed a direct interaction between SIRT1 and MICU1. Notably, SIRT1 inhibition resulted in reduced MICU1 expression, hence led to mitochondrial calcium overload, illustrating the unconventional role of SIRT1 in governing mitochondrial function. Full article
(This article belongs to the Section Physiology and Pathology)
Show Figures

Figure 1

Figure 1
<p>Expression of SIRT1 in mitochondria of HeLa cells and rat cortical neurons. (<b>a</b>) Western blot analysis of SIRT1 expression in mitochondria, cytoplasm, and whole-cell lysates of HeLa cells. (<b>b</b>) Confocal microscopy determining the SIRT1 colocalization with mitochondria in HeLa cells. Red, MitoTracker; Green, SIRT1. Magnification is 60×. Scale bars, 5 μm. (<b>c</b>) The levels of colocalization between SIRT1 and mitochondria expressed as MCC for a fraction of SIRT1 pixels that are shared with mitochondria (black bar) or vice versa (red bar) in HeLa cells. (<b>d</b>) Confocal microscopy determining the SIRT1 colocalization with mitochondria in SD rat cortical neurons. Red, MitoTracker; Green, SIRT1; Blue, Dapi. Magnification is 60×. Scale bars, 5 μm. (<b>e</b>) The levels of colocalization between SIRT1 and mitochondria expressed as MCC for a fraction of SIRT1 pixels that are shared with mitochondria (black bar) or vice versa (red bar) in SD rat cortical neurons. (<b>f</b>) Representative images of confocal microscopy determining 10 μM of EX527 to mitochondrial morphology in HeLa cells. Red, MitoTracker. Magnification is 60×. Scale bars, 5 μm. (<b>g</b>,<b>h</b>) The footprint of mitochondria and mean branch length of HeLa cells treated with DMSO or EX527 for 1 h. Data presented as mean ± standard deviation (SD). ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001. The number of cells analyzed/the number of independent experiments is indicated in the bars.</p>
Full article ">Figure 2
<p>EX527 affected mitochondrial calcium uptake in HeLa cells. (<b>a</b>) Representative confocal microscopy images acquired at 40 s, 76 s, and 160 s depict mitochondrial calcium levels in HeLa cells following treatment with either DMSO or 10 μM EX527 for 1 h. Continuous recording was performed, with one frame captured every 4 s. At the 60 s time point, 100 μM histamine was added. Rhod-2 AM; Magnification is 40×. Scale bars, 20 μm. (<b>b</b>) The mitochondrial calcium level in HeLa cells treated with DMSO or EX527 for 1 h was determined by Rhod-2 AM staining. Overall, 100 μM histamine was used to induce cellular calcium release. (<b>c</b>) The fold change in mitochondrial calcium level in HeLa cells treated with DMSO or EX527 for 1 h, normalized to baseline. (<b>d</b>) Average Rhod-2 AM fluorescence intensity before 100 μM histamine treatment. (<b>e</b>) Maximum fold change in Rhod-2/AM fluorescence intensity after 100 μM histamine treatment. For each group, 30 cells from three independent experiments were analyzed. The data are presented as mean values ± SD. ns, not significant; * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>shSIRT1 affected mitochondrial calcium uptake in HeLa cells. (<b>a</b>) Representative confocal images (40 s, 76 s, and 160 s) of mitochondrial calcium in HeLa cells co-transfected with shSIRT1 or shSIRT2 interference plasmids and the CMV-mito-R-GECO1 mitochondrial calcium-targeting fluorescence plasmid, followed by treatment with 100 μM histamine to induce cellular calcium release. Continuous recording was performed, with one frame captured every 4 s. At the 60 s time point, 100 μM histamine was added. Red, mito-Red; Magnification is 40×. Scale bars, 50 μm. (<b>b</b>) Mitochondrial calcium fluorescence intensity changes in HeLa cells determined by mito-Red intensity. Overall, 100 μM histamine was used to induce cellular calcium release. (<b>c</b>) The fold change in mitochondrial calcium level in HeLa cells normalized to baseline. (<b>d</b>) Average mitochondrial calcium fluorescence intensity before treatment with 100 μM histamine. (<b>e</b>) Maximum fold change in mitochondrial calcium fluorescence intensity after treatment with 100 μM histamine. (<b>f</b>) The fold change in mitochondrial calcium fluorescence intensity at 300 s. For each group, 30 cells from three independent experiments were analyzed. The data are presented as mean values ± SD. ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>MICU1 and SIRT1 interacted with each other. (<b>a</b>–<b>d</b>) SIRT1, MCU, and MICU1 protein expression levels after HeLa cells transfection of shSIRT1 (<span class="html-italic">n</span> = 3 biological replicates; three technical replicates were achieved). (<b>e</b>) GST assay was performed to determine the interaction of SIRT1 and MICU1. Purified GST and GST-SIRT1 proteins were incubated with cell lysate (Input) containing MICU1-HA proteins, followed by examination of the proteins bound to GST and GST-SIRT1 via Western blot analysis. (<b>f</b>) Co-IP was determined in SIRT1 immunoprecipitates by Western blot analysis. SIRT1-flag + pcmv-HA, pcmv-tag4a + MICU1-HA, SIRT1-flag + MICU1-HA, and SIRT1-flag + MICU1-HA constructs were expressed in HeLa cells, and anti-FLAG, anti-HA, and anti-SIRT1 antibodies were employed for immunoprecipitation. (<b>g</b>) SIRT1 and MICU1 exhibited colocalization in HeLa cells. Red, HA; Green, SIRT1. Magnification is 60×. Scale bars, 5 μm. (<b>h</b>) The levels of colocalization between SIRT1 and MICU1-HA expressed as MCC for a fraction of SIRT1 pixels that are shared with HA (black bar) or vice versa (red bar) in HeLa cells. (<b>i</b>) Lysates from rat neurons cultured for 7 days were collected. SIRT1 antibody was utilized for immunoprecipitation, and anti-SIRT1 and anti-MICU1 antibodies were used for Western blot analysis. (<b>j</b>) MICU1-HA was transfected into primary cultured rat neurons, and antibodies targeting HA and SIRT1 were applied as immunofluorescent co-labels. Red, SIRT1; Green, HA. Magnification is 60×. Scale bars, 50 μm. (<b>k</b>) The levels of colocalization between SIRT1 and mitochondria expressed as MCC for a fraction of SIRT1 pixels that are shared with HA (black bar) or vice versa (red bar) in SD rat cortical neurons. The data are presented as mean values ± SD. ns, not significant; * <span class="html-italic">p</span> &lt; 0.05. Statistical analysis was carried out using the Mann–Whitney U Test (<b>b</b>–<b>d</b>). The number of cells analyzed/the number of independent experiments is indicated above the bars.</p>
Full article ">Figure 5
<p>SIRT1 regulated mitochondrial calcium uptake through MICU1. (<b>a</b>) HeLa cells were co-transfected with MICU1-HA or pCMV-C-HA, along with the mitochondrial calcium-targeting fluorescent plasmid CMV-mito-R-GECO1. Representative confocal images (40 s, 120 s, and 300 s) of mitochondrial calcium in HeLa cells treated with DMSO or 10 μM EX527 for 1 h. Overall, 100 μM Histamine was used to induce cellular calcium release. Red, mito-Red; Magnification is 40×. Scale bars, 50 μm. Continuous recording was performed, with one frame captured every 4 s. At the 60 s time point, 100 μM histamine was added. (<b>b</b>) The fold change in mitochondrial calcium levels in HeLa cells, normalized to baseline values. (<b>c</b>) Average mitochondrial calcium fluorescence intensity before treatment with 100 μM histamine. (<b>d</b>) The fold change in the peak mitochondrial calcium fluorescence intensity following treatment with 100 μM histamine. For each group, 30 cells from three independent experiments were analyzed (two-way ANOVA and Tukey’s post hoc test). The data are presented as mean values ± SD. ns, not significant; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
23 pages, 1066 KiB  
Review
The Potential of Polyphenols in Modulating the Cellular Senescence Process: Implications and Mechanism of Action
by Larissa Della Vedova, Giovanna Baron, Paolo Morazzoni, Giancarlo Aldini and Francesca Gado
Pharmaceuticals 2025, 18(2), 138; https://doi.org/10.3390/ph18020138 - 22 Jan 2025
Viewed by 580
Abstract
Background: Cellular senescence is a biological process with a dual role in organismal health. While transient senescence supports tissue repair and acts as a tumor-suppressive mechanism, the chronic accumulation of senescent cells contributes to aging and the progression of age-related diseases. Senotherapeutics, [...] Read more.
Background: Cellular senescence is a biological process with a dual role in organismal health. While transient senescence supports tissue repair and acts as a tumor-suppressive mechanism, the chronic accumulation of senescent cells contributes to aging and the progression of age-related diseases. Senotherapeutics, including senolytics, which selectively eliminate senescent cells, and senomorphics, which modulate the senescence-associated secretory phenotype (SASP), have emerged as promising strategies for managing age-related pathologies. Among these, polyphenols, a diverse group of plant-derived bioactive compounds, have gained attention for their potential to modulate cellular senescence. Methods: This review synthesizes evidence from in vitro, in vivo, and clinical studies on the senolytic and senomorphic activities of bioactive polyphenols, including resveratrol, kaempferol, apigenin, and fisetin. The analysis focuses on their molecular mechanisms of action and their impact on fundamental aging-related pathways. Results: Polyphenols exhibit therapeutic versatility by activating SIRT1, inhibiting NF-κB, and modulating autophagy. These compounds demonstrate a dual role, promoting the survival of healthy cells while inducing apoptosis in senescent cells. Preclinical evidence indicates their capacity to reduce SASP-associated inflammation, restore tissue homeostasis, and attenuate cellular senescence in various models of aging. Conclusions: Polyphenols represent a promising class of senotherapeutics for mitigating age-related diseases and promoting healthy lifespan extension. Further research should focus on clinical validation and the long-term effects of these compounds, paving the way for their development as therapeutic agents in geriatric medicine. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic representation of senescence cell (SC) formation.</p>
Full article ">Figure 2
<p>Schematic representation of the main features of senescence cells (SCs).</p>
Full article ">
21 pages, 4995 KiB  
Article
Ontogeny of Fetal Cardiometabolic Pathways: The Potential Role of Cortisol and Thyroid Hormones in Driving the Transition from Preterm to Near-Term Heart Development in Sheep
by Reza Amanollahi, Stacey L. Holman, Melanie R. Bertossa, Ashley S. Meakin, Kent L. Thornburg, I. Caroline McMillen, Michael D. Wiese, Mitchell C. Lock and Janna L. Morrison
J. Cardiovasc. Dev. Dis. 2025, 12(2), 36; https://doi.org/10.3390/jcdd12020036 - 21 Jan 2025
Viewed by 537
Abstract
Understanding hormonal and molecular changes during the transition from preterm to near-term gestation is essential for investigating how pregnancy complications impact fetal heart development and contribute to long-term cardiovascular risks for offspring. This study examines these cardiac changes in fetal sheep, focusing on [...] Read more.
Understanding hormonal and molecular changes during the transition from preterm to near-term gestation is essential for investigating how pregnancy complications impact fetal heart development and contribute to long-term cardiovascular risks for offspring. This study examines these cardiac changes in fetal sheep, focusing on the changes between 116 days (preterm) and 140 days (near term) of gestation (dG, term = 150) using Western blotting, LC-MS/MS, and histological techniques. We observed a strong correlation between cortisol and T3 (Triiodothyronine) in heart tissue in near-term fetuses, highlighting the role of glucocorticoid signalling in fetal heart maturation. Protein expression patterns in the heart revealed a decrease in multiple glucocorticoid receptor isoforms (GRα-A, GR-P, GR-A, GRα-D2, and GRα-D3), alongside a decrease in IGF-1R (a marker of cardiac proliferative capacity) and p-FOXO1(Thr24) but an increase in PCNA (a marker of DNA replication), indicating a shift towards cardiomyocyte maturation from preterm to near term. The increased expression of proteins regulating mitochondrial biogenesis and OXPHOS complex 4 reflects the known transition from glycolysis to oxidative phosphorylation, essential for meeting the energy demands of the postnatal heart. We also found altered glucose transporter expression, with increased pIRS-1(ser789) and GLUT-4 but decreased GLUT-1 expression, suggesting improved insulin responsiveness as the heart approaches term. Notably, the reduced protein abundance of SIRT-1 and SERCA2, along with increased phosphorylation of cardiac Troponin I(Ser23/24), indicates adaptations for more energy-efficient contraction in the near-term heart. In conclusion, these findings show the complex interplay of hormonal, metabolic, and growth changes that regulate fetal heart development, providing new insights into heart development that are crucial for understanding pathological conditions at birth and throughout life. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Hormone concentrations of fetal cardiac tissue. The fetal cardiac concentration of cortisol (<b>A</b>) and cortisone (<b>B</b>) were not different between preterm and near-term fetuses. The cortisol: cortisone ratio (<b>C</b>) was higher, while 11-deoxycortisol (<b>D</b>) and corticosterone (<b>E</b>) were lower with no change in progesterone (<b>F</b>) in the near-term compared to preterm fetuses. T<sub>4</sub> (<b>G</b>) was lower with no change in T<sub>3</sub> (<b>H</b>) concentrations in the near-term compared to preterm fetuses. In the near-term fetuses only, there were positive linear relationships of T<sub>3</sub> with cortisol (<b>I</b>). Males (M) = circles, females (F) = triangles. preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; hormone = 2M, 5F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; hormone = 3M, 4F). One sample per animal was analysed via LC-MS/MS. Data were excluded due to a technical error in hormone concentration. Up to one outlier was excluded per group using the Grubbs method (Alpha = 0.05), when applicable. Data are expressed as mean ± SD and were analysed using either an unpaired <span class="html-italic">t</span>-test or simple linear regression. Data for progesterone, T<sub>3</sub>, and T<sub>4</sub> failed the normality test and were consequently analysed using the Mann–Whitney test. (*) indicates a statistically significant difference between the groups. <span class="html-italic">p</span> &lt; 0.05 was considered significant. AU: arbitrary unit.</p>
Full article ">Figure 2
<p>Abundance of glucocorticoid receptor isoforms in the fetal heart. The cardiac protein abundance of glucocorticoid receptors (GR) including GRα-A (<b>A</b>), GR-P (<b>B</b>), GR-A (<b>C</b>), GRα-D2 (<b>D</b>), and GRα-D3 (<b>E</b>) was lower in the near-term compared to preterm fetuses. In the near-term fetuses only, there was a positive linear relationship between cortisol and GRα-D2 (<b>F</b>). Males (M) = circles, females (F) = triangles. Preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; protein = 3M, 5F; hormone = 2M, 5F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; protein = 3M, 4F; hormone = 3M, 4F). One sample per animal was run per Western blot and LC-MS/MS. Data were excluded due to a technical error in hormone concentration. Up to one outlier was excluded per group using the Grubbs method (Alpha = 0.05) when applicable. Data were expressed as mean ± SD and were analysed using either an unpaired <span class="html-italic">t</span>-test or simple linear regression. (*) indicates a statistically significant difference between the groups. <span class="html-italic">p</span> &lt; 0.05 was considered significant. AU: arbitrary unit. (X) indicates data excluded from analysis (due to a defect on the band/s).</p>
Full article ">Figure 3
<p>Molecular markers of fetal cardiac growth. The cardiac protein expression of IGF-1R (<b>A</b>) and p-FOXO1:FOXO1 ratio (<b>B</b>) was lower, while PCNA (<b>C</b>) was higher in the near term compared to preterm fetuses. There was no difference in the p-mTOR:mTOR ratio (<b>D</b>), p-Akt:Akt ratio (<b>E</b>), and p-P70 S6K:P70 S6K ratio (<b>F</b>) between the groups. Males (M) = circles, females (F) = triangles. Preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; protein = 3M, 5F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; protein = 3M, 4F). One sample per animal was run per Western blot. Up to one outlier was excluded per group using the Grubbs method (Alpha = 0.05), when applicable. Data are expressed as mean ± SD and were analysed either using an unpaired <span class="html-italic">t</span>-test or simple linear regression. (*) indicates a statistically significant difference between the groups. <span class="html-italic">p</span> &lt; 0.05 was considered significant. AU: arbitrary unit. (X) indicates data excluded from analysis (due to a defect on the band/s).</p>
Full article ">Figure 4
<p>Molecular markers of fetal cardiac OXPHOS and mitochondrial content. The cardiac protein abundance of complex 4 (<b>D</b>) was higher in the near-term compared to preterm fetuses, while there was no difference in complex 1 (<b>A</b>), 2 (<b>B</b>), 3 (<b>C</b>), and 5 (<b>E</b>). The MT-COXI: SDHA ratio (<b>F</b>), a marker of mitochondrial content) was higher in the near-term compared to preterm fetuses. CS activity (<b>G</b>) did not differ between the groups, while CS activity: mitochondrial content ratio (<b>H</b>) was lower in the near-term compared to preterm fetuses. Males (M) = circles, females (F) = triangles. Preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; protein/CS activity = 3M, 5F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; protein/CS activity = 3M, 4F). One sample per animal was run per Western blot and CS activity. Up to one outlier was excluded per group using the Grubbs method (Alpha = 0.05), when applicable. Data are expressed as mean ± SD and were analysed using either an unpaired <span class="html-italic">t</span>-test or simple linear regression. (*) indicates a statistically significant difference between the groups. <span class="html-italic">p</span> &lt; 0.05 was considered significant. AU: arbitrary unit. (X) indicates data excluded from analysis (due to a defect on the band/s).</p>
Full article ">Figure 5
<p>Molecular markers of fetal cardiac glucose metabolism. The ratio of p-IRS-1:IRS-1 ratio (<b>A</b>) and GLUT-4 (<b>C</b>) were higher, while the ratio of p-AS160:AS160 (<b>B</b>) was not different, and GLUT-1 (<b>D</b>) was lower in the near-term compared to preterm fetuses. The abundance of PDK-4 protein (<b>E</b>), and activity of LDH (<b>F</b>) were not different in preterm and near-term fetuses. In the preterm fetuses only, there were positive linear relationships between GRα-D2 and GLUT-1 (<b>G</b>), as well as GRα-D3 and GLUT-1 (<b>H</b>). Males (M) = circles, females (F) = triangles. Preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; protein/LDH activity = 3M, 5F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; protein/LDH activity = 3M, 4F). One sample per animal was run per Western blot and LDH activity. Up to one outlier was excluded per group using the Grubbs method (Alpha = 0.05), when applicable. Data are expressed as mean ± SD and were analysed using either an unpaired <span class="html-italic">t</span>-test or simple linear regression. Data for p-AS160:AS160 ratio failed the normality test and were consequently analysed using the Mann–Whitney test. (*) indicates a statistically significant difference between the groups. <span class="html-italic">p</span> &lt; 0.05 was considered significant. AU: arbitrary unit. (X) indicates data excluded from analysis (due to a defect on the band/s).</p>
Full article ">Figure 6
<p>Molecular markers of fetal cardiac contractility. The expression of SIRT-1 (<b>A</b>) and SERCA2 (<b>B</b>) in cardiac tissue was lower, while there was no difference in the ratio of p-PLN:PLN (<b>C</b>) in the near-term compared to preterm fetuses. The ratio of p-TroponinI:TroponinI (<b>D</b>) was higher, while NOX-2 (<b>E</b>) was lower in near-term compared to preterm fetuses. Males (M) = circles, females (F) = triangles. Preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; protein = 3M, 5F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; protein = 3M, 4F). One sample per animal was run per Western blot. Up to one outlier was excluded per group using the Grubbs method (Alpha = 0.05), when applicable. Data are expressed as mean ± SD and were analysed using either an unpaired <span class="html-italic">t</span>-test or simple linear regression. (*) indicates a statistically significant difference between the groups. <span class="html-italic">p</span> &lt; 0.05 was considered significant. AU: arbitrary unit.</p>
Full article ">Figure 7
<p>Fetal cardiac glycogen, collagen, and Ki67 staining: 20× magnification representative micrograph of glycogen staining using PAS (black arrow indicates glycogen stained in magenta) in preterm (<b>A</b>) and near term (<b>B</b>). 20× magnification representative micrograph of collagen staining using Masson’s trichrome (black arrow indicates collagen stained in blue) in preterm (<b>D</b>) and near term (<b>E</b>). 40× magnification representative micrograph of Ki67 staining using IHC (black arrow) in preterm (<b>G</b>) and near term (<b>H</b>). The fetal cardiac glycogen (<b>C</b>), collagen (<b>F</b>), and Ki67 (<b>I</b>) staining were not different between preterm and near-term fetuses. Males (M) = circles, females (F) = triangles. Preterm, left ventricle (LV) tissue from fetuses at 116 days of gestation (dG) (open symbols; histology/IHC = 3M, 2F). Near-term, LV tissue from fetuses at 140 dG (closed symbols; histology/IHC = 2M, 3F). One sample per animal was run per histology and IHC. A smaller subset of animals was included in this analysis due to missing fixed tissue samples. Scale bars = 100 μm. Data are expressed as mean ± SD and were analysed using an unpaired <span class="html-italic">t</span>-test. <span class="html-italic">p</span> &lt; 0.05 was considered significant.</p>
Full article ">
28 pages, 766 KiB  
Review
Unveiling the Utilization of Grape and Winery By-Products in Cosmetics with Health Promoting Properties
by Olga I. Tsiapali, Efthymia Ayfantopoulou, Athanasia Tzourouni, Anna Ofrydopoulou, Sophia Letsiou and Alexandros Tsoupras
Appl. Sci. 2025, 15(3), 1007; https://doi.org/10.3390/app15031007 - 21 Jan 2025
Viewed by 560
Abstract
Winemaking by-products, such as grape pomace and grape seed oil, provide sustainable and eco-friendly resources for cosmetics and are rich in bioactive compounds like phenolic bioactives, proteins, and lipids (i.e., unsaturated fatty acids, bioactive polar lipids, and carotenoids). These compounds, extracted using advanced [...] Read more.
Winemaking by-products, such as grape pomace and grape seed oil, provide sustainable and eco-friendly resources for cosmetics and are rich in bioactive compounds like phenolic bioactives, proteins, and lipids (i.e., unsaturated fatty acids, bioactive polar lipids, and carotenoids). These compounds, extracted using advanced techniques such as ultrasound, microwave, and enzyme-assisted methods, exhibit antioxidant, antimicrobial, anti-aging, and anti-inflammatory properties. In vitro and in vivo studies on keratinocytes and fibroblasts demonstrate their efficacy in enhancing skin hydration, elasticity, and UV protection while reducing oxidative stress and inflammation through pathways like SIRT1 and HSP47. Encapsulation techniques further improve their stability and bioavailability. The aim of this review is to investigate in detail the advanced techniques for the extraction of bioactive compounds from winemaking by-products and to evaluate their effectiveness in the isolation of phenolic compounds, proteins, and lipids. At the same time, it focuses on the application of the extracted compounds in the cosmetics industry, highlighting their contribution to products with antioxidant, anti-aging, antimicrobial, and anti-inflammatory properties. Finally, special emphasis is given to encapsulation techniques to improve their stability and bioavailability, with the aim of developing innovative and sustainable cosmetic products. Full article
(This article belongs to the Special Issue Bioactive-Based Cosmeceuticals)
Show Figures

Figure 1

Figure 1
<p>Illustration of different classes of phenolic bioactives of grape and winery by-products [<a href="#B46-applsci-15-01007" class="html-bibr">46</a>].</p>
Full article ">
18 pages, 5599 KiB  
Article
The Essential Role of Monte Carlo Simulations for Lung Dosimetry in Liver Radioembolization—Part B: 166Ho Microspheres
by Edoardo d’Andrea, Andrea Politano, Bartolomeo Cassano, Nico Lanconelli, Marta Cremonesi, Vincenzo Patera and Massimiliano Pacilio
Appl. Sci. 2025, 15(2), 958; https://doi.org/10.3390/app15020958 - 19 Jan 2025
Viewed by 714
Abstract
This study compares dosimetric approaches for lung dosimetry in 166 radioembolization (Ho-TARE) with direct Monte Carlo (MC) simulations on a voxelized anthropomorphic phantom derived from a real patient’s CT scan, preserving the patient’s lung density distribution. Lung dosimetry was assessed for five lung [...] Read more.
This study compares dosimetric approaches for lung dosimetry in 166 radioembolization (Ho-TARE) with direct Monte Carlo (MC) simulations on a voxelized anthropomorphic phantom derived from a real patient’s CT scan, preserving the patient’s lung density distribution. Lung dosimetry was assessed for five lung shunt (LS) scenarios with conventional methods: the mono-compartmental organ-level approach (MIRD), voxel S-value convolution for soft tissue (kST, ICRU soft tissue with 1.04 g/cm3) and lung tissue (kLT, ICRU lung tissue with 0.296 g/cm3), local density rescaling (kSTL and kLTL, respectively, for soft tissue and lung tissue), or global rescaling for a lung mean density of 0.221 g/cm3 (kLT221). Significant underestimations in the mean absorbed dose (AD) were observed, with relative differences with respect to the reference (MC) of −64% for MIRD, −93% for kST, −56% for kSTL, −76% for kLT, −68% for kLT221, and −60% for kLTL. Given the high heterogeneity of lung tissue, standard dosimetric approaches cannot accurately estimate the AD. Additionally, MC results for 166Ho showed notable spatial absorbed dose inhomogeneity, highlighting the need for tailored lung dosimetry in Ho-TARE accounting for the patient-specific lung density distribution. MC-based dosimetry thus proves to be essential for safe and effective radioembolization treatment planning in the presence of LS. Full article
Show Figures

Figure 1

Figure 1
<p>VSV kernels (available as Supporting Material of this paper) represented as a plot of the AD per unit decay to the target voxel (y-axis) versus the source–target voxel distance (x-axis) for the soft tissue (<b>a</b>) and lung tissue (<b>b</b>) for <sup>166</sup>Ho on a square voxel of 2.21 mm side.</p>
Full article ">Figure 2
<p>Correlation plot of the mean absorbed dose in the lungs (<math display="inline"><semantics> <mover> <mrow> <mi>A</mi> <mi>D</mi> </mrow> <mo>¯</mo> </mover> </semantics></math>) per GBq of administered activity, obtained from MC simulations with the reference phantom (x-axis), compared with those obtained using the methods listed in <a href="#applsci-15-00958-t002" class="html-table">Table 2</a> (y-axis). Each point in the data series represents an increasing LS value (10%, 20%, 30%, and 40%), with a line representing the linear interpolation of each dataset, provided as a qualitative visual guide only.</p>
Full article ">Figure 3
<p>Example slices in the coronal view of the AD spatial distributions (left) for <math display="inline"><semantics> <msub> <mi>kLT</mi> <mi>L</mi> </msub> </semantics></math> (<b>a</b>), <math display="inline"><semantics> <msub> <mi>kST</mi> <mi>L</mi> </msub> </semantics></math> (<b>b</b>), and MC (<b>c</b>), along with their respective color scales, are shown, whereas the plot (right) reports the corresponding DVH (<b>d</b>). All dosimetric approaches show significant heterogeneity in the AD spatial distribution. The values of the AD maps and the DVH are given in Gy per GBq of administered activity.</p>
Full article ">Figure 4
<p>Example slices in the coronal view of the ADr maps (left) of <sup>166</sup>Ho (<b>a</b>) and <sup>90</sup>Y (<b>b</b>), along with the corresponding DrVHs (<b>c</b>), are shown for LS = 10%. The data demonstrate a different degree of inhomogeneity between the two radionuclides, due to the distinct physical characteristics of their decay spectra.</p>
Full article ">Figure 5
<p>Cumulative DVHs of <math display="inline"><semantics> <mrow> <mi>E</mi> <mi>Q</mi> <msub> <mi>D</mi> <mn>2</mn> </msub> </mrow> </semantics></math> for <sup>90</sup>Y (blue band) and <sup>166</sup>Ho (yellow band) for the 0.5–1 h range of <math display="inline"><semantics> <msub> <mi mathvariant="normal">T</mi> <mi>μ</mi> </msub> </semantics></math> along with the volumetric constraints (red dots) listed in <a href="#applsci-15-00958-t004" class="html-table">Table 4</a>.</p>
Full article ">Figure 6
<p><math display="inline"><semantics> <mover> <mrow> <mi>A</mi> <mi>D</mi> </mrow> <mo>¯</mo> </mover> </semantics></math> NTCP model for RP incidence in partial lung irradiation treatments from EBRT as reported in QUANTEC [<a href="#B42-applsci-15-00958" class="html-bibr">42</a>]. The black solid line is the logistic model according to Equation (<a href="#FD6-applsci-15-00958" class="html-disp-formula">6</a>) with parameters <math display="inline"><semantics> <mrow> <msub> <mi>b</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>−</mo> <mn>3.87</mn> </mrow> </semantics></math> and <math display="inline"><semantics> <mrow> <msub> <mi>b</mi> <mn>1</mn> </msub> <mo>=</mo> <mn>0.126</mn> <mspace width="4.pt"/> <msup> <mi>Gy</mi> <mrow> <mo>−</mo> <mn>1</mn> </mrow> </msup> </mrow> </semantics></math>, the blue dots refer to <sup>90</sup>Y cases, and the yellow ones to <sup>166</sup>Ho cases, each for the LS = 10% case and for the labeled <math display="inline"><semantics> <msub> <mi mathvariant="normal">T</mi> <mi>μ</mi> </msub> </semantics></math>.</p>
Full article ">
27 pages, 1493 KiB  
Review
Autophagy and Mitophagy in Diabetic Kidney Disease—A Literature Review
by Alina Mihaela Stanigut, Liliana Tuta, Camelia Pana, Luana Alexandrescu, Adrian Suceveanu, Nicoleta-Mirela Blebea and Ileana Adela Vacaroiu
Int. J. Mol. Sci. 2025, 26(2), 806; https://doi.org/10.3390/ijms26020806 - 18 Jan 2025
Viewed by 588
Abstract
Autophagy and mitophagy are critical cellular processes that maintain homeostasis by removing damaged organelles and promoting cellular survival under stress conditions. In the context of diabetic kidney disease, these mechanisms play essential roles in mitigating cellular damage. This review provides an in-depth analysis [...] Read more.
Autophagy and mitophagy are critical cellular processes that maintain homeostasis by removing damaged organelles and promoting cellular survival under stress conditions. In the context of diabetic kidney disease, these mechanisms play essential roles in mitigating cellular damage. This review provides an in-depth analysis of the recent literature on the relationship between autophagy, mitophagy, and diabetic kidney disease, highlighting the current state of knowledge, existing research gaps, and potential areas for future investigations. Diabetic nephropathy (DN) is traditionally defined as a specific form of kidney disease caused by long-standing diabetes, characterized by the classic histological lesions in the kidney, including mesangial expansion, glomerular basement membrane thickening, nodular glomerulosclerosis (Kimmelstiel–Wilson nodules), and podocyte injury. Clinical markers for DN are albuminuria and the gradual decline in glomerular filtration rate (GFR). Diabetic kidney disease (DKD) is a broader and more inclusive term, for all forms of chronic kidney disease (CKD) in individuals with diabetes, regardless of the underlying pathology. This includes patients who may have diabetes-associated kidney damage without the typical histological findings of diabetic nephropathy. It also accounts for patients with other coexisting kidney diseases (e.g., hypertensive nephrosclerosis, ischemic nephropathy, tubulointerstitial nephropathies), even in the absence of albuminuria, such as a reduction in GFR. Full article
(This article belongs to the Special Issue Molecular Mechanism of Diabetic Kidney Disease (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Molecular mechanism of autophagy: cytosolic components are included in autophagosomes. The autophagosome fuses with the lysosome becomes autophagolysosome and then degrade the cytosolic components.</p>
Full article ">Figure 2
<p>Molecular mechanisms of mitophagy: there are three well-described pathways of mitophagy: PINK1-PARK2 pathway, BNIP3/NIX receptor pathway, and FUNDC1 receptor pathway.</p>
Full article ">Figure 3
<p>Autophagy and mitophagy in Diabetic Nephropathy and potential therapeutic targets. Autophagy prevents tubular hypertrophy, endoplasmic reticulum (ER) stress, accumulation of AGEs (advanced glycation end products), and loss of podocytes. Mitophagy inhibits mitochondria specific oxidative stress (mROS—mitochondria-derived reactive oxygen species), attenuates NLRP3 inflammasome-mediated tubular injury (NLR family pyrin domain containing 3), inflammation and fibrosis, AS-IV, astragaloside IV; CoQ10, coenzyme Q10; MitoQ, mitoquinone; mTORC1, mechanistic target of rapamycin (mTOR) kinase complex 1; SGLT2, Naþ-glucose cotransporter-2; SIRT1, sirtuin; TFEB, transcription factor EB; ULK1, uncoordinated-51-like protein kinase 1.</p>
Full article ">Figure 4
<p>The potential mechanisms of mitophagy in diabetic nephropathy. Impaired mitophagy leads to an accumulation of damaged mitochondria which plays an important role in the pathogenesis of diabetic nephropathy.</p>
Full article ">
18 pages, 4065 KiB  
Article
Effects of Astragaloside IV and Formononetin on Oxidative Stress and Mitochondrial Biogenesis in Hepatocytes
by Quoc-Anh Tran, Grant Van Tran, Sanel Velic, Hou-Mai Xiong, Jaspreet Kaur, Zuhurr Moosavi, Phuong Nguyen, Nhi Duong, Vy Tran Luu, Gurjot Singh, Tram Bui, Melanie Rose and Linh Ho
Int. J. Mol. Sci. 2025, 26(2), 774; https://doi.org/10.3390/ijms26020774 - 17 Jan 2025
Viewed by 573
Abstract
Over-accumulation of reactive oxygen species (ROS) causes hepatocyte dysfunction and apoptosis that might lead to the progression of liver damage. Sirtuin-3 (SIRT3), the main NAD+-dependent deacetylase located in mitochondria, has a critical role in regulation of mitochondrial function and ROS production as well [...] Read more.
Over-accumulation of reactive oxygen species (ROS) causes hepatocyte dysfunction and apoptosis that might lead to the progression of liver damage. Sirtuin-3 (SIRT3), the main NAD+-dependent deacetylase located in mitochondria, has a critical role in regulation of mitochondrial function and ROS production as well as in the mitochondrial antioxidant mechanism. This study explores the roles of astragaloside IV (AST-IV) and formononetin (FMR) in connection with SIRT3 for potential antioxidative effects. It was shown that the condition of combined pre- and post-treatment with AST-IV or FMR at all concentrations statistically increased and rescued cell proliferation. ROS levels were not affected by pre-or post-treatment individually with AST-IV or pre-treatment with FMR; however, post-treatment with FMR resulted in significant increases in ROS in all groups. Significant decreases in ROS levels were seen when pre- and post-treatment with AST-IV were combined at 5 and 10 μM, or FMR at 5 and 20 μM. In the condition of combined pre- and post-treatment with 10 μM AST-IV, there was a significant increase in SOD activity, and the transcriptional levels of Sod2, Cat, and GPX1 in all treatment groups, which is indicative of reactive oxygen species detoxification. Furthermore, AST-IV and FMR activated PGC-1α and AMPK as well as SIRT3 expression in AML12 hepatocytes exposed to t-BHP-induced oxidative stress, especially at high concentrations of FMR. This study presents a novel mechanism whereby AST-IV and FMR yield an antioxidant effect through induction of SIRT3 protein expression and activation of an antioxidant mechanism as well as mitochondrial biogenesis and mitochondrial content and potential. The findings suggest these agents can be used as SIRT3 modulators in treating oxidative-injury hepatocytes. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of (<b>A</b>) astragaloside IV (a cycloartane-type triterpene glycoside); and (<b>B</b>) formononetin (O-methylated isoflavone).</p>
Full article ">Figure 2
<p>SIRT3 expression (2 isoforms) levels in AML12 cells at different concentrations of AST-IV and FMR. (<b>A</b>) Expression of SIRT3 and (<b>B</b>) quantification of SIRT3 expression intensity by WB in cells treated with AST-IV at concentrations of 2.5, 5, and 10 µM compared to controls (untreated cells and cells treated with <span class="html-italic">t</span>-BHP). (<b>C</b>) Expression of SIRT3 and (<b>D</b>) quantification of SIRT3 expression intensity by WB in cells treated with FMR at concentrations of 5, 10, and 20 µM compared to controls (untreated cells and cells treated with <span class="html-italic">t</span>-BHP). Tubulin was used as a loading control. Data were assessed from at least three independent experiments.</p>
Full article ">Figure 3
<p>Rescue effect on cell proliferation of AML12 hepatocytes in the condition of pre-treatment with AST-IV or FMR. Rescue effect of AST-IV or FMR on cell proliferation of AML12 hepatocytes in the condition of pre-treatment with AST-IV or FMR for 12 h, then oxidative stress induced by <span class="html-italic">t</span>-BHP for another 12 h. (<b>A</b>) Effects on cell proliferation of AST-IV at concentrations of 2.5, 5, and 10 µM compared to <span class="html-italic">t</span>-BHP-treated cells. (<b>B</b>) Effects on cell proliferation of FMR at concentrations of 5, 10, and 20 µM compared to <span class="html-italic">t</span>-BHP-treated cells. Data were collected from at least three independent experiments. NS: not significant; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Rescue effect on cell proliferation of AML12 hepatocytes in the condition of post-treatment with AST-IV or FMR. Rescue effect of AST-IV or FMR on cell proliferation of AML12 hepatocytes in the condition of oxidative stress induced by <span class="html-italic">t</span>-BHP, then post-treatment with AST-IV or FMR for 12 h. (<b>A</b>) Effects on cell proliferation of AST-IV at concentrations of 2.5, 5, and 10 µM compared to <span class="html-italic">t</span>-BHP-treated cells. (<b>B</b>) Effects on cell proliferation of FMR at concentrations of 5, 10, and 20 µM compared to <span class="html-italic">t</span>-BHP-treated cells. Data were collected from at least three independent experiments. NS: not significant; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Rescue effect on cell proliferation of AML12 hepatocytes in the condition of combined pre- and post-treatment with AST-IV or FMR. Rescue effect of AST-IV or FMR on cell proliferation of AML12 hepatocytes in the condition of combined pre- and post-treatment with AST-IV or FMR for 12 h, then oxidative stress induced by <span class="html-italic">t</span>-BHP for another 12 h. (<b>A</b>) Effects on cell proliferation of AST-IV at concentrations of 2.5, 5, and 10 µM compared to <span class="html-italic">t</span>-BHP-treated cells. (<b>B</b>) Effects on cell proliferation of FMR at concentrations of 5, 10, and 20 µM compared to <span class="html-italic">t</span>-BHP-treated cells. Data were collected from at least three independent experiments. NS: not significant, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>ROS measurement from cells treated with AST-IV or FMR in AML12 hepatocytes in the condition of pre- or post-, or combined pre- and post-treatment with AST-IV or FMR. (<b>A</b>) Effects on ROS levels of pre-treatment with AST-IV at concentrations of 2.5, 5, and 10 µM, and FMR at concentrations of 5, 10, and 20 µM, compared to <span class="html-italic">t</span>-BHP-treated cells. (<b>B</b>) Effects on ROS levels of post-treatment with AST-IV at concentrations of 2.5, 5, and 10 µM, and FMR at concentrations of 5, 10, and 20 µM, compared to <span class="html-italic">t</span>-BHP-treated cells. (<b>C</b>) Effects on ROS levels of combined pre- and post-treatment with AST-IV at concentrations of 2.5, 5, and 10 µM, and FMR at concentrations of 5, 10, and 20 µM, compared to <span class="html-italic">t</span>-BHP-treated cells. Data were collected from at least three independent experiments. NS: not significant, * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>SOD activity and glutathione level measurement from cells treated with AST-IV or FMR in AML12 hepatocytes in the condition of combined pre- and post-treatment with AST-IV or FMR. (<b>A</b>) SOD activity in AML12 hepatocytes pre- and post-treatment with various concentrations of AST-IV or FMR; (<b>B</b>) glutathione (GSH) level measured in AML12 hepatocytes pre- and post-treatment with various concentrations of AST-IV or FMR. Data were collected from at least three independent experiments.</p>
Full article ">Figure 8
<p>Gene expression of antioxidant genes (<b>A</b>) SOD2, (<b>B</b>) CAT, and (<b>C</b>) GPX1 in AML12 hepatocytes in the condition of combined pre- and post-treatment with AST-IV or FMR. Data were collected from at least three independent experiments and analyzed using Graphpad Prism software version 9.1.1 (223), and statistical analyses using ordinary one-way ANOVA followed by post hoc Dunnett’s multiple comparisons. ns: not significant; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>AST-IV and FMR increased mitochondrial biogenesis for their antioxidant effect against oxidative stress induced by t-BHP. (<b>A</b>–<b>C</b>) Expression and intensity quantification of AMPK and PGC-1α by WB in pre- and post-treatment condition in AML12 cells with AST-IV at concentrations of 2.5, 5, and 10 µM compared to controls (t-BHP-treated cells). (<b>D</b>–<b>F</b>) Expression and intensity quantification of AMPK and PGC-1α by WB in pre- and post-treatment condition in AML12 cells with FMR at concentrations of 5, 10, and 20 µM compared to controls (t-BHP-treated cells). Tubulin was used as a loading control. Data were assessed from at least three independent experiments. Statistical analyses using ordinary one-way ANOVA followed by post hoc Dunnett’s multiple comparisons. NS: not significant; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>AST-IV and FRM enhance mitochondrial membrane potential and mitochondrial content. AML12 hepatocytes were treated in the condition of combined pre- and post-treatment with AST-IV and FMR, then mitochondrial potential and VDAC were measured. (<b>A</b>) Mitochondrial membrane potential of AST-IV- or FMR-treated samples at various concentrations, analyzed using Graphpad Prism Software version 9.1.1 (223), and statistical analyses using ordinary one-way ANOVA followed by post hoc Dunnett’s multiple comparisons. NS: not significant; * <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>,<b>C</b>) Expression of VDAC and SIRT3 (2 isoforms) and VDAC expression quantitation relative to Actin by WB in pre- and post-treatment condition with AST-IV at concentrations of 2.5, 5, and 10 µM compared to controls (untreated cells and <span class="html-italic">t</span>-BHP-treated cells). (<b>D</b>,<b>E</b>) Expression of VDAC and SIRT3 and VDAC expression quantitation relative to Actin by WB in combined pre- and post-treatment condition with FMR at concentrations of 5, 10, and 20 µM compared to controls (untreated cells and <span class="html-italic">t</span>-BHP-treated cells) in AML12. Data were assessed from at least three independent experiments.</p>
Full article ">
23 pages, 314 KiB  
Review
Molecular Mechanisms and Therapeutic Potential of Mulberry Fruit Extract in High-Fat Diet-Induced Male Reproductive Dysfunction: A Comprehensive Review
by Kannika Adthapanyawanich, Kanyakorn Aitsarangkun Na Ayutthaya, Siriporn Kreungnium, Peter J. Mark, Hiroki Nakata, Wai Chen, Kroekkiat Chinda, Patcharada Amatyakul and Yutthapong Tongpob
Nutrients 2025, 17(2), 273; https://doi.org/10.3390/nu17020273 - 13 Jan 2025
Viewed by 946
Abstract
High-fat diet (HFD)-induced obesity represents a significant challenge to male reproductive health, affecting approximately 13% of the global adult population. This comprehensive review synthesizes current evidence regarding mulberry (Morus alba L.) fruit extract’s therapeutic potential for HFD-induced male reproductive dysfunction. Through comprehensive [...] Read more.
High-fat diet (HFD)-induced obesity represents a significant challenge to male reproductive health, affecting approximately 13% of the global adult population. This comprehensive review synthesizes current evidence regarding mulberry (Morus alba L.) fruit extract’s therapeutic potential for HFD-induced male reproductive dysfunction. Through comprehensive analysis of the peer-reviewed literature from multiple databases (PubMed, Web of Science, Scopus, and Google Scholar; 2005–2024), we evaluated mulberry extract’s effects on testicular morphology, spermatogenesis, sperm parameters, and the underlying molecular mechanisms. Mechanistic studies reveal that standardized mulberry extract mediates protective effects through multiple pathways: enhanced antioxidant enzyme activities (SOD: +45%, Catalase: +38%, GPx: +35%), reduced inflammatory markers (TNF-α: −64%, IL-6: −58%), and modulated NF-κB signaling (−42.3%). These effects are facilitated by mulberry’s rich phytochemical profile, particularly anthocyanins (2.92–5.35 mg/g dry weight) and polyphenols (4.23–6.38 mg/g). The extract demonstrates particular efficacy in preserving seminiferous tubule integrity and maintaining blood-testis barrier function, with treated groups maintaining up to 85% of normal tubular architecture compared to HFD controls. Key molecular mechanisms include AMPK/SIRT1 pathway activation (2.3-fold increase), enhanced mitochondrial function (67% increase in mtDNA copy number), and epigenetic regulation of metabolic pathways. Temporal analysis indicates optimal therapeutic effects after 28 days of treatment, with initial improvements observable within 14 days. While current evidence is promising, limitations include predominant reliance on rodent models and lack of standardized extraction protocols. Future research priorities include well-designed human clinical trials, standardization of preparation methods, and investigation of potential synergistic effects with other therapeutic agents. This comprehensive review indicates that mulberry extract is a promising therapeutic candidate for obesity-related male infertility, warranting further clinical investigation. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">
16 pages, 5570 KiB  
Article
Bone Adaptations to a Whole Body Vibration Protocol in Murine Models of Different Ages: A Preliminary Study on Structural Changes and Biomarker Evaluation
by Ida Cariati, Roberto Bonanni, Cristian Romagnoli, Lucio Caprioli, Giovanna D’Arcangelo, Virginia Tancredi and Giuseppe Annino
J. Funct. Morphol. Kinesiol. 2025, 10(1), 26; https://doi.org/10.3390/jfmk10010026 - 10 Jan 2025
Viewed by 571
Abstract
Background/Objectives: Whole body vibration (WBV) is a valuable tool to mitigate physiological adaptations related to age and inactivity. Although significant benefits have been found at the musculoskeletal level, including increased bone mass and reduced muscle atrophy, the underlying biological mechanisms remain largely [...] Read more.
Background/Objectives: Whole body vibration (WBV) is a valuable tool to mitigate physiological adaptations related to age and inactivity. Although significant benefits have been found at the musculoskeletal level, including increased bone mass and reduced muscle atrophy, the underlying biological mechanisms remain largely unknown. Therefore, our study aimed to evaluate the effects of vibratory training on bone tissue in murine models of different age groups by investigating the structural and distribution changes in some crucial biomarkers involved in musculoskeletal homeostasis. Methods: Specifically, 4-, 12-, and 24-month-old mice were trained with a WBV protocol characterized by three series of 2 min and 30 s, interspersed with a recovery period of the same duration, on a 3-weekly frequency for 3 months. At the end of the training, histological and morphometric analyses were conducted, in association with immunohistochemical analysis to investigate changes in the distribution of fibronectin type III domain-containing protein 5 (FNDC5), NADPH oxidase 4 (NOX4), and sirtuin 1 (SIRT1). Results: Our preliminary results showed that WBV improves musculoskeletal health by preserving bone architecture and promoting up-regulation of FNDC5 and SIRT1 and down-regulation of NOX4. Conclusions: Our study confirms vibratory training as a viable alternative to counter musculoskeletal decline in elderly and/or sedentary subjects. Further investigations should be conducted to deepen knowledge in this field and explore the role of other molecular mediators in physiological adaptations to vibration. Full article
(This article belongs to the Special Issue Role of Exercises in Musculoskeletal Disorders—7th Edition)
Show Figures

Figure 1

Figure 1
<p>Effects of a whole body vibration (WBV) protocol on weight in murine models of different age groups. (<b>a</b>,<b>b</b>) The most significant weight changes were measured in 4-month-old young mice, with a significant increase in mean weight at the 6 weeks of training (IN phase) and at the end of training (POST) (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>c</b>,<b>d</b>) WBV promoted a significant increase in body weight in 12-month-old adult mice in the IN (** <span class="html-italic">p</span> &lt; 0.01) and POST (**** <span class="html-italic">p</span> &lt; 0.0001) phases compared with sedentary animals. (<b>e</b>,<b>f</b>) Significant weight changes were induced by vibratory training in 24-month-old mice in the IN (**** <span class="html-italic">p</span> &lt; 0.0001) and POST (**** <span class="html-italic">p</span> &lt; 0.0001) phases.</p>
Full article ">Figure 2
<p>Hematoxylin and eosin (H&amp;E) sections of lumbar segment of the spine from all experimental groups and measurement of significant bone morphometric parameters. (<b>a</b>–<b>e</b>) The 4-month-old young mice in the WBV group showed the highest values of bone volume (BV/TV) and trabecular thickness (Tb.Th), in association with reduced space between bone trabeculae (Tb.S), compared with sedentary animals (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>f</b>–<b>j</b>) In 12-month-old adult mice, WBV significantly increased BV/TV (** <span class="html-italic">p</span> &lt; 0.01) and Tb.Th (**** <span class="html-italic">p</span> &lt; 0.0001), as well as significantly reduced Tb.S (<span class="html-italic">p</span> &lt; 0.0001). (<b>k</b>–<b>o</b>) WBV also improved bone architecture in 24-month-old mice, promoting a significant increase in BV/TV (*** <span class="html-italic">p</span> &lt; 0.001) and Tb.Th (**** <span class="html-italic">p</span> &lt; 0.0001) and a significant reduction in Tb.S (**** <span class="html-italic">p</span> &lt; 0.0001). For 20× images, the scale bar represents 100 μm. For each condition, the experiment was conducted in triplicate (n = 15 from N = 5 experiments).</p>
Full article ">Figure 3
<p>Evaluation of fibronectin type III domain-containing protein 5 (FNDC5) immunolocalization in bone tissue of young, adult, and aged mice by immunohistochemistry analysis. (<b>a</b>–<b>c</b>) The highest levels of FNDC5 (arrows) were measured in young, trained mice, with a significant increase in FNDC5 distribution compared with the CTRL group (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>d</b>–<b>f</b>) A significant increase in FNDC5 distribution (arrow) was observed in 12-month-old adult mice exposed to WBV compared with sedentary animals (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>g</b>–<b>i</b>) FNDC5 levels (arrow) were significantly increased in the bone tissue of 24-month-old mice after WBV exposure compared with the CTRL group (**** <span class="html-italic">p</span> &lt; 0.0001). For 20× images, the scale bar represents 100 μm. For each condition, the experiment was conducted in triplicate (n = 15 from N = 5 experiments).</p>
Full article ">Figure 4
<p>Evaluation of NADPH oxidase 4 (NOX4) immunolocalization in bone tissue of young, adult, and aged mice by immunohistochemistry analysis. (<b>a</b>–<b>c</b>) A significant reduction in NOX4 distribution (arrow) was observed in 4-month-old young mice exposed to WBV compared with sedentary animals (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>d</b>–<b>f</b>) NOX4 levels (arrow) were significantly reduced in the bone tissue of 12-month-old adult mice after WBV exposure compared with the CTRL group (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>g</b>–<b>i</b>) The highest NOX4 levels (arrows) were measured in 24-month-old sedentary mice, while protein distribution was significantly reduced in the WBV group of the same age group (**** <span class="html-italic">p</span> &lt; 0.0001). For 20× images, the scale bar represents 100 μm. For each condition, the experiment was conducted in triplicate (n = 15 from N = 5 experiments).</p>
Full article ">Figure 5
<p>Evaluation of sirtuin 1 (SIRT1) immunolocalization in bone tissue of young, adult, and aged mice by immunohistochemistry analysis. (<b>a</b>–<b>c</b>) Bone tissue of young, trained mice showed the highest levels of SIRT1 (arrows), with a significant increase in its distribution compared with the CTRL group (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>d</b>–<b>f</b>) A significant increase in SIRT1 distribution (arrow) was observed in 12-month-old adult mice after WBV training compared with sedentary animals (**** <span class="html-italic">p</span> &lt; 0.0001). (<b>g</b>–<b>i</b>) The distribution of SIRT1 (arrow) was significantly increased in the bone tissue of 24-month-old trained mice compared with the CTRL group (*** <span class="html-italic">p</span> &lt; 0.001). For 20× images, the scale bar represents 100 μm. For each condition, the experiment was conducted in triplicate (n = 15 from N = 5 experiments).</p>
Full article ">
Back to TopTop