[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (959)

Search Parameters:
Keywords = sirtuin1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1645 KiB  
Article
Cross-Sectional Analysis of Hypoxia-Regulated miRNA-181a, miRNA-199a, HIF-1α, and SIRT1 in the Development of Type 2 Diabetes in Patients with Obstructive Sleep Apnea—Preliminary Study
by Filip Franciszek Karuga, Piotr Kaczmarski, Marcin Sochal, Bartosz Szmyd, Piotr Białasiewicz and Agata Gabryelska
J. Clin. Med. 2024, 13(24), 7644; https://doi.org/10.3390/jcm13247644 (registering DOI) - 15 Dec 2024
Abstract
Introduction: Obstructive sleep apnea (OSA) is recognized as an independent risk factor for diabetes mellitus type 2 (T2DM) development, which is twice as common in patients with OSA compared to non-OSA patients. Objectives: This study aimed to investigate changes in oxygen metabolism [...] Read more.
Introduction: Obstructive sleep apnea (OSA) is recognized as an independent risk factor for diabetes mellitus type 2 (T2DM) development, which is twice as common in patients with OSA compared to non-OSA patients. Objectives: This study aimed to investigate changes in oxygen metabolism and their role in T2DM development among OSA patients through epigenetic processes via miRNA-181a, miRNA-199a, and enzymatic processes via SIRT1 and HIF-1α. Methods: Based on polysomnography, apnea–hypopnea index and the presence of T2DM patients were divided into three groups: control group (n = 17), OSA group (n = 11), OSA&T2DM (n = 20) group. Total RNA was extracted from the buffy coat. Moreover, HOMA-IR (Homeostatic Model Assessment for Insulin Resistance) was counted. Results: Morning miRNA-181a expression was significantly higher in the OSA&T2DM group than in the control group: 67.618 vs. 32.685 (p = 0.036). Evening miRNA-199a expression was significantly higher in the OSA group than in the control group: 5.043 vs. 2.081 (p = 0.042), while its morning expression was significantly higher in the OSA&T2DM group when compared to the control: 4.065 vs. 1.605 (p = 0.036). MiRNA-181a evening expression revealed a negative correlation with the SIRT1 evening and morning expressions (R = −0.367, p = 0.010 and R = −0.405, p = 0.004, respectively). Moreover, morning miRNA-181a was positively correlated with HOMA-IR (R = 0.321, p = 0.034). MiRNA-199a evening expression presented a moderate positive correlation with the SIRT1 morning expressions (R = 0.48, p < 0.001) and HOMA-IR (R = 0.35, p = 0.02). Conclusions: Patients suffering from OSA and T2DM had an increased expression of miRNA-181a. Moreover, a negative correlation between miRNA-181a and SIRT1 expression was observed, while a correlation between miRNA-181a and insulin resistance was positive. This phenomenon might suggest a possible epigenetic pathway for an increased incidence of T2DM in OSA patients however further research is needed. Full article
20 pages, 7923 KiB  
Article
Repurposing Dapagliflozin for Mitigation of the Kidney Injury Triggered by Cadmium in Rats: Role of Autophagy, Apoptosis, and the SIRT1/Nrf2/HO-1 Pathway
by Hany H. Arab, Musaad M. Althobaiti, Abdulaziz S. Alharthi, Emad O. Almalki, Saif S. Alsoubie, Jawad M. Qattan, Saeed A. Almalki, Ahmed M. Ashour and Ahmed H. Eid
Pharmaceuticals 2024, 17(12), 1690; https://doi.org/10.3390/ph17121690 (registering DOI) - 15 Dec 2024
Viewed by 129
Abstract
Background/Objectives: The antioxidant/antiapoptotic features of dapagliflozin (DPG) have mediated its beneficial actions against several experimental models. However, no studies have been conducted to determine whether DPG mitigates the renal injury triggered by cadmium (Cd). Herein, DPG was studied for its potential to attenuate [...] Read more.
Background/Objectives: The antioxidant/antiapoptotic features of dapagliflozin (DPG) have mediated its beneficial actions against several experimental models. However, no studies have been conducted to determine whether DPG mitigates the renal injury triggered by cadmium (Cd). Herein, DPG was studied for its potential to attenuate kidney damage in Cd-intoxicated rats, as well as to unravel the mechanisms involving oxidative events, autophagy, and apoptosis. Methods: Histopathological analysis, immunohistochemical staining, and ELISA were conducted on kidney tissue samples. Results: Cd administration (5 mg/kg/day; p.o.) prompted significant renal damage, as evidenced by histopathological changes, elevated kidney injury molecule-1 (KIM-1) expression, and increased serum creatinine and urea. Interestingly, DPG (1 mg/kg/day; p.o.) significantly mitigated these harmful effects without affecting renal Cd metal accumulation. Mechanistically, DPG curbed Cd-induced renal pro-oxidant response and stimulated the antioxidant sirtuin 1 (SIRT1)/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase 1 (HO-1) axis. Moreover, DPG restored autophagy by decreasing sequestosome-1/protein 62 (SQSTM-1/p62) accumulation and stimulating the AMP-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) pathway. In tandem, DPG suppressed Cd-induced apoptosis by lowering renal Bcl-2 associated-x protein (Bax) and cytochrome C (Cyt C) levels and caspase 3 activity. Conclusions: These findings indicate that DPG attenuates Cd-induced nephrotoxicity by enhancing the SIRT1/Nrf2/HO-1 antioxidant pathway, promoting AMPK/mTOR-directed autophagy, and inhibiting apoptotic cell death. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Illustration of dapagliflozin’s chemical structure.</p>
Full article ">Figure 2
<p>Dapagliflozin improved Cd-triggered kidney dysfunction in rats. These effects were proven by reduced serum creatinine (<b>A</b>) and blood urea nitrogen (BUN) (<b>B</b>), alongside the dampened renal expression of KIM-1 (<b>C</b>), compared to the Cd group. Moreover, serum glucose levels (<b>D</b>) and renal Cd metal content (<b>E</b>) were examined in all experimental groups (<b>D</b>). For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 3
<p>Dapagliflozin improved Cd-induced renal histomorphological aberrations in rats. (<b>A</b>) The renal cortex from the control as well as the DPG-treated rats manifested typical histological features, including an intact proximal and distal convoluted tubule epithelium (black arrow) and typical glomerulus (black asterisk; scale bar: 20 µm). On the contrary, the Cd-treated group showed severe atrophy of the glomerular tufts together with an increased Bowman’s space (red asterisk). Moreover, there was evident vacuolation (red arrow), necrosis of the tubular epithelium (blue arrow), and hemorrhage (green arrow; scale bar: 20 µm). DPG and Cd co-administration afforded moderate mitigation of the histological aberrations by improving the size/shape of renal corpuscles and decreasing vacuolization, necrosis, and hemorrhages. (<b>B</b>) According to the EGTI’s scoring of renal histomorphological damage, renal tissue demonstrated reduced damage scores in response to DPG co-administration with Cd, implying an improved kidney histopathology. For <span class="html-italic">n</span> = 6, the bars represent the median with the interquartile range. Statistical significance was described by * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 4
<p>Dapagliflozin improved the Cd-triggered renal oxidative milieu in rats. These effects were proven by the reduced levels of renal lipid peroxides (<b>A</b>), NOX-1 (<b>B</b>), and 8-OHdG (<b>C</b>), alongside an augmented content of renal GSH (<b>D</b>), compared to the Cd group. For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 5
<p>Dapagliflozin counteracted Cd-triggered inhibition in the SIRT1/Nrf2/HO-1 axis of rats. These effects were proven by the augmented levels of renal SIRT1 (<b>A</b>), Nrf2 (<b>B</b>), and HO-1 (<b>C</b>) compared to the Cd group. For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 6
<p>Dapagliflozin counteracted Cd-triggered renal pro-apoptotic events in rats. These effects were proven by the reduced activity of renal caspase 3 (<b>A</b>), alongside the levels of Cyt C (<b>B</b>), and Bax (<b>C</b>), compared to the Cd group. Moreover, DPG augmented renal Bcl-2 protein expression (<b>D</b>). For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 7
<p>Dapagliflozin counteracted the Cd-triggered renal autophagy blockade in rats. This effect was proven by the lowered levels of SQSTM-1/p62. (<b>A</b>) Representative immunohistochemical images of renal SQSTM-1/p62 staining (scale bar: 20 µm). (<b>B</b>) SQSTM-1/p62 protein quantification displaying area percentage (calculated using six non-overlapping microscopic fields). For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 8
<p>Dapagliflozin reversed Cd-triggered renal Beclin 1 downregulation in rats. (<b>A</b>) Representative immunohistochemical images of renal Beclin 1 staining (scale bar: 20 µm). (<b>B</b>) Beclin 1 protein quantification displaying area percentage (calculated using six non-overlapping microscopic fields). For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by * <span class="html-italic">p</span> &lt; 0.05 or ** <span class="html-italic">p</span> &lt; 0.01. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 9
<p>Dapagliflozin counteracted the Cd-triggered inhibition of the renal AMPK/mTOR pathway. These effects were proven by the augmented p-AMPK(Ser487)/total AMPK levels (<b>A</b>), together with the diminished levels of p-mTOR(Ser2448)/total mTOR (<b>B</b>). For <span class="html-italic">n</span> = 6, the graph bars represent the mean ± standard deviation. Statistical significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Cd, cadmium chloride; DPG, dapagliflozin.</p>
Full article ">Figure 10
<p>An illustration of dapagliflozin’s beneficial actions in improving Cd-induced renal outcomes in rats. In reference to the present findings, dapagliflozin attenuated renal pathological and functional disturbances in vivo by (<b>A</b>) antioxidant actions and stimulation of the renal SIRT1/Nrf2/HO-1 pathway; (<b>B</b>) rescue of renal autophagy dysfunction with AMPK/mTOR stimulation; and (<b>C</b>) inhibition of renal pro-apoptotic events, thereby promoting cellular survival. In this figure, there are two types of arrows: solid arrows representing activation and blunt arrows representing inhibition.</p>
Full article ">
11 pages, 788 KiB  
Article
The Influence of Nordic Walking Training on the Serum Levels of Sirtuins, FOXO3a, and Vitamin D Metabolites in Patients with Multiple Myeloma
by Olga Czerwińska-Ledwig, Małgorzata Żychowska, Artur Jurczyszyn, Joanna Kryst, Adrianna Dzidek, Roxana Zuziak, Anna Jurczyszyn and Anna Piotrowska
Curr. Oncol. 2024, 31(12), 7960-7970; https://doi.org/10.3390/curroncol31120587 (registering DOI) - 14 Dec 2024
Viewed by 157
Abstract
Background: Multiple myeloma, a malignancy of plasma cells, often involves the disruption of vitamin D metabolism. Vitamin D, acting through its receptor (VDR), affects transcription factors like FOXO and sirtuins, which regulate cellular processes. The impact of physical activity on these markers in [...] Read more.
Background: Multiple myeloma, a malignancy of plasma cells, often involves the disruption of vitamin D metabolism. Vitamin D, acting through its receptor (VDR), affects transcription factors like FOXO and sirtuins, which regulate cellular processes. The impact of physical activity on these markers in multiple myeloma patients is unclear. Therefore, the objective of this study was to evaluate the effects of a 6-week training program on these parameters. Material and methods: The study was completed by 30 patients, including 16 in the Nordic walking training group (TG) and 14 in the control group (non-exercising, CG). All participants underwent a thorough medical interview before starting the project. Venous blood samples were collected from all participants four times—at baseline, after 3 weeks, after 6 weeks, and after 9 weeks (follow-up). The serum concentrations of sirtuin 1, sirtuin 3, Foxo3a, vitamin D receptor (VDR), 25(OH)D3, and 1,25(OH)2D were determined. Body composition, physical fitness, and physical activity level were assessed at baseline and after 6 weeks. Results: No statistically significant changes were observed in the serum levels of sirtuins, the FOXO3a protein, and 1,25(OH)2D. A statistically significant difference was observed in the levels of VDR for both time and group factors, but this was not confirmed in the post hoc test. Vitamin 25(OH)D3 level increased significantly in the study group with time. Conclusions: The applied 6-week Nordic walking training cycle positively affected the level of vitamin 25(OH)D3 but did not influence the rest of the biochemical parameters studied. The obtained results also indicate that the applied intervention is safe for patients and does not interfere with body composition. Full article
(This article belongs to the Special Issue Diet and Physical Activity Management during Cancer)
Show Figures

Figure 1

Figure 1
<p>CONSORT patient flow diagram.</p>
Full article ">Figure 2
<p>Serum levels of vitamin 25-OH-D3 in participants of the project. CG—control group; TG—training group.</p>
Full article ">
18 pages, 5796 KiB  
Article
Fermented Gold Kiwifruit Protects Mice Against Non-Alcoholic Fatty Liver Disease in a High-Fat Diet Model
by Jihye Choi, Hwal Choi, Yuseong Jang, Hyeon-Gi Paik, Hyuck-Se Kwon and Jungkee Kwon
Appl. Sci. 2024, 14(24), 11503; https://doi.org/10.3390/app142411503 - 10 Dec 2024
Viewed by 387
Abstract
Gold kiwifruit is known for its high vitamin C content and various benefits. This study investigated the effects and molecular mechanisms of fermented gold kiwifruit (FGK) in a mouse model of high-fat diet (HFD)-induced obesity and hepatic steatosis. FGK powder was prepared using [...] Read more.
Gold kiwifruit is known for its high vitamin C content and various benefits. This study investigated the effects and molecular mechanisms of fermented gold kiwifruit (FGK) in a mouse model of high-fat diet (HFD)-induced obesity and hepatic steatosis. FGK powder was prepared using five strains of lactic acid bacteria: L. paracasei, Lc. lactis, L. acidophilus, L. casei, and L. helveticus. ICR mice were fed an HFD for 8 weeks to induce obesity and hepatic steatosis, and FGK supplementation was evaluated for its therapeutic potential. FGK administration significantly reduced serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol, triglyceride, and glucose compared to the HFD-only group. Histopathological analysis showed that FGK reduced lipid accumulation and hepatic lesions, as confirmed by hematoxylin and eosin (H&E) staining. Furthermore, administration of FGK activated the sirtuin 1(SIRT1)/adenosine monophosphate-activated protein kinase (AMPK) pathway and inhibited expression of the pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α in liver tissue. These findings suggest that FGK could reduce the severity of non-alcoholic fatty liver disease (NAFLD) by inhibiting fat synthesis, promoting fat breakdown, and suppressing inflammation in HFD-induced obese mice. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of fermented gold kiwifruit (FGK) on the fat parameters of ICR mice fed with a high-fat diet (HFD): (<b>A</b>) representative images of the bodies and livers of mice fed a normal diet (ND) or fed the HFD and administered FGK; (<b>B</b>) changes body weight; (<b>C</b>) final body weights; (<b>D</b>) changes in food intake; (<b>E</b>) average daily food intake; and (<b>F</b>) relative organ weights. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–c) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test.</p>
Full article ">Figure 2
<p>Effects of FGK on the fasting blood concentrations of ICR mice fed with a high-fat diet: (<b>A</b>) fasting blood glucose at four and eight weeks; (<b>B</b>) IPGTT; (<b>C</b>) area under the curve (AUC). All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–d) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test.</p>
Full article ">Figure 3
<p>Effects of FGK on serum (<b>A</b>) ALT, (<b>B</b>) AST, (<b>C</b>) glucose, and (<b>D</b>) lipase concentration of ICR mice fed with a high-fat diet. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–c) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test, while NS indicates no significant difference.</p>
Full article ">Figure 4
<p>Effects of FGK on serum lipid profile levels of ICR mice fed with a high-fat diet: (<b>A</b>) total cholesterol (TC); (<b>B</b>) triglyceride (TG); (<b>C</b>) low-density lipoprotein (LDL); (<b>D</b>) high-density lipoprotein (HDL). All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–d) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test.</p>
Full article ">Figure 5
<p>Effects of FGK on the livers of ICR mice fed with a high-fat diet: (<b>A</b>) H&amp;E-stained hepatic tissue sections (×400). Rats induced with HFD exhibited hepatocellular steatosis characterized by intracellular lipid droplets with clear vacuoles indicative of fatty changes (black arrows); (<b>B</b>) steatosis score. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–d) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test.</p>
Full article ">Figure 6
<p>Effects of FGK on the serum cytokines concentration of ICR mice fed with a high-fat diet; (<b>A</b>) Serum TNF-α concentration; (<b>B</b>) Serum IL-1β concentration; (<b>C</b>) Serum IL-6 concentration. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a,b) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test.</p>
Full article ">Figure 7
<p>Effects of FGK on hepatic expression of inflammatory cytokines in ICR mice fed with a high-fat diet. (<b>A</b>) protein expression image; (<b>B</b>) relative optical density as measured using Image J (Java 1.8.0) software. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–c) indicate statistically significant differences between groups at the <span class="html-italic">p</span> &lt; 0.05 level using one-way ANOVA analysis after completing post hoc analysis using Tukey’s test.</p>
Full article ">Figure 8
<p>Effects of FGK on hepatic expression of SIRT1/AMPK pathway proteins in ICR mice fed with a high-fat diet; (<b>A</b>) protein expression image; (<b>B</b>) relative optical density as measured using Image J software. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6), and different letters (a–c) indicate statistically significant differences at the <span class="html-italic">p</span> &lt; 0.05 level after completing post hoc analyses using Tukey’s test.</p>
Full article ">Figure 9
<p>Illustration of SIRT1/AMPK pathway activation by FGK and the inhibitory of FGK on NAFLD in mice. The blue line represents the activation of the SIRT1/AMPK pathway, while the red line indicates the inhibition of inflammation by FGK. Each black arrow signifies the progression to the next step in the pathway. HFD, high-fat diet; SIRT1, sirtuin 1; AMPK, AMP-activated protein kinase; DNL, de novo lipogenesis; TG, triglyceride; LDL, low-density lipoprotein; TNF-α, tumor necrosis factor α; IL-1β, interleukin 1 β; IL-6, interleukin 6.</p>
Full article ">
11 pages, 1169 KiB  
Article
Intraperitoneal Treatment of Cambinol, a Synthetic SIRT1 and SIRT2 Inhibitory Compound, Exacerbates Brucella abortus 544 Burden in the Spleens of Institute of Cancer Research Mice
by Alisha Wehdnesday Bernardo Reyes, Tran Xuan Ngoc Huy, Trang Thi Nguyen, Said Abdi Salad, Ched Nicole Turbela Aguilar, Wongi Min, Hu Jang Lee and Suk Kim
Microorganisms 2024, 12(12), 2533; https://doi.org/10.3390/microorganisms12122533 - 9 Dec 2024
Viewed by 538
Abstract
Our preliminary data using bone marrow-derived macrophages (BMDMs) collected from ICR mice treated with anti-sirtuin (anti-SIRT) 1 antibody showed that Brucella uptake was significantly attenuated. We then further investigated the effect of an inhibitor of SIRT1/2, cambinol, in the progression of Brucella. [...] Read more.
Our preliminary data using bone marrow-derived macrophages (BMDMs) collected from ICR mice treated with anti-sirtuin (anti-SIRT) 1 antibody showed that Brucella uptake was significantly attenuated. We then further investigated the effect of an inhibitor of SIRT1/2, cambinol, in the progression of Brucella. The in vitro results using RAW264.7 cells revealed that cambinol treatment had no effect on adhesion, uptake, intracellular survival and nitric oxide (NO) production during B. abortus infection, nor did it directly affect bacterial growth for up to 72 h. Finally, intraperitoneal treatment of 8-week-old female ICR mice infected with Brucella showed no differences in the total average weights of spleens and livers; however, the treated mice displayed higher Brucella colony-forming units (CFUs) from the spleens. Furthermore, the interleukin (IL)-10 serum level was observed to be lower in treated mice at 7 d post-infection, and none of the cytokines tested showed a change at 14 d post-infection. The overall findings showed that cambinol treatment had no effect on the proliferation of Brucella in RAW264.7 macrophages but exacerbated the splenic proliferation of the bacteria in mice and displayed reduced anti-inflammatory cytokine IL-10 at the first week of infection, suggesting that cambinol as an inhibitory of SIRT1/2 could be beneficial in the context of Brucella dissemination in animal hosts and that exploration of activating SIRTs could be an alternative treatment against Brucella infection. Full article
(This article belongs to the Section Veterinary Microbiology)
Show Figures

Figure 1

Figure 1
<p>Effect of sirtuin inhibitors in macrophages and <span class="html-italic">B. abortus</span>. (<b>A</b>) The uptake of <span class="html-italic">B. abortus</span> in murine bone marrow-derived macrophages neutralized using mouse anti-sirtuin 1 antibody at 0 h post-infection; (<b>B</b>) RAW264.7 cell viability treated with various concentrations of cambinol for 48 h; (<b>C</b>) <span class="html-italic">B. abortus</span> growth treated with three different concentrations of cambinol at 0, 2, 8, 24, 48 and 72 h. Data are expressed as the mean ± standard deviation, with (****) <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Effect of cambinol treatment in macrophages infected with <span class="html-italic">B. abortus</span>. (<b>A</b>) Adhesion assay at 0.5 h post-infection; (<b>B</b>) internalization assay at 0 and 0.5 h post-infection; (<b>C</b>) intracellular killing assay at 2, 24 and 48 h post-incubation; (<b>D</b>) nitric oxide assay at 24 and 48 h post-incubation. Data are expressed as the mean ± standard deviation.</p>
Full article ">Figure 3
<p>Effect of cambinol after intraperitoneal treatment in ICR mice infected with <span class="html-italic">B. abortus</span>. (<b>A</b>) Total average organs collected at 15 d post-infection; (<b>B</b>) bacterial proliferation in the organs. Data are expressed as the mean (n = 5–6) ± standard deviation, with (*) <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Effect of cambinol treatment in the serum cytokine levels in ICR mice infected with <span class="html-italic">B. abortus</span>. (<b>A</b>) The different serum cytokine level at 7 d post-infection; (<b>B</b>) serum cytokine level at 14 d post-infection. Data are expressed as the mean (n = 5–6) ± standard deviation, with (*) <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
26 pages, 3140 KiB  
Article
SIRT2 Inhibition by AGK2 Promotes Perinuclear Cytoskeletal Organisation and Reduces Invasiveness of MDA-MB-231 Triple-Negative Breast Cancer Cells in Confined In Vitro Models
by Emily Jessop, Natalie Young, Beatriz Garcia-Del-Valle, Jack T. Crusher, Boguslaw Obara and Iakowos Karakesisoglou
Cells 2024, 13(23), 2023; https://doi.org/10.3390/cells13232023 - 7 Dec 2024
Viewed by 619
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype characterised by the absence of targetable hormone receptors and increased metastatic rates. As nuclear softening strongly contributes to TNBC’s enhanced metastatic capacity, increasing the nuclear stiffness of TNBC cells may present a [...] Read more.
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype characterised by the absence of targetable hormone receptors and increased metastatic rates. As nuclear softening strongly contributes to TNBC’s enhanced metastatic capacity, increasing the nuclear stiffness of TNBC cells may present a promising therapeutic avenue. Previous evidence has demonstrated the ability of Sirtuin 2 (SIRT2) inhibition to induce cytoskeletal reorganisation, a key factor in regulating nuclear mechanics. Thus, our study aimed to investigate the effect of SIRT2 inhibition on the nuclear mechanics and migratory behaviour of TNBC cells. To achieve this, SIRT2 was pharmacologically inhibited in MDA-MB-231 cells using AGK2, a SIRT2-specific inhibitor. Although SIRT2 inhibition had no effect on LINC complex composition, the AGK2-treated MDA-MB-231 cells displayed more prominent perinuclear organisations of acetylated α-tubulin, vimentin, and F-actin. Additionally, the nuclei of the AGK2-treated MDA-MB-231 cells exhibited greater resistance to collapse under osmotic shock. Scratch-wound assays also revealed that SIRT2 inhibition led to polarity defects in the MDA-MB-231 cells, while in vitro space-restrictive invasion assays highlighted their reduced migratory capacity upon AGK2 treatment. Taken together, our findings suggest that SIRT2 inhibition promotes a perinuclear cytoskeletal organisation in MDA-MB-231 cells, which enhances their nuclear rigidity and impedes their invasion through confined spaces in vitro. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>AGK2-induced SIRT2 inhibition promotes the perinuclear localisation of acetylated α-tubulin and vimentin in MDA-MB-231 cells. (<b>A</b>) Western blot analysis showing SIRT2, acetylated histone 4, acetylated α-tubulin, and vimentin protein expression levels in media-, vehicle-, and AGK2-treated MDA-MB-231 cells. GAPDH expression is used to demonstrate equal protein loading. (<b>B</b>) Immunofluorescence microscopy images of acetylated α-tubulin (AcTub, upper row) and vimentin (lower row) organisation in control (media and vehicle) and AGK2-treated MDA-MB-231 cells. Nuclei are visualised using a DAPI counterstain (blue channel). Insets (showing only the red channel) are higher magnifications of the areas indicated by dashed boxes. Scale bar: 20 μm. (<b>C</b>,<b>D</b>) Quantification of the percentage (%) of control and AGK2-treated cells exhibiting prominent perinuclear acetylated α-tubulin (<b>C</b>) and vimentin (<b>D</b>) rings; &gt;350 cells analysed for each condition. The data are presented as the mean ± SEM (standard error of the mean). Statistical significance was determined using a one-way ANOVA followed by a Dunnett’s post hoc test to compare with the vehicle control. “ns” denotes no significant difference, while *** <span class="html-italic">p</span> ≤ 0.001 and **** <span class="html-italic">p</span> ≤ 0.0001 indicate statistical significance.</p>
Full article ">Figure 2
<p>Pharmacological inhibition of SIRT2 in MDA-MB-231 cells induces the formation of perinuclear F-actin rings. (<b>A</b>) Representative fluorescence microscopy images of filamentous actin (F-actin) organisation in control and AGK2-treated MDA-MB-231 cells stained with TRITC-phalloidin (red channel) and DAPI (blue channel). Insets (showing only the red channel) are higher magnifications of the areas marked by dashed boxes. Scale bar: 10 μm. (<b>B</b>) Quantification of the percentage (%) of cells that displayed a perinuclear F-actin ring; &gt;800 cells analysed per condition. The data are presented as the mean ± SEM, and statistical analysis was determined using a one-way ANOVA and Dunnett’s post hoc test for comparisons with the vehicle control. “ns” indicates no significance while *** <span class="html-italic">p</span> ≤ 0.001 denotes statistical significance.</p>
Full article ">Figure 3
<p>The expression and localisation of Nesprin-1 giant and Nesprin-2 giant are unaffected in AGK2-treated MDA-MB-231 cells. (<b>A</b>) Immunoblot analysis of Nesprin-1 giant and Nesprin-2 giant protein expression in media-, vehicle-, and AGK2-treated MDA-MB-231 cells. GAPDH expression demonstrates equal protein loading. (<b>B</b>,<b>C</b>) Densitometric quantification of Nesprin-1 giant (<b>B</b>) and Nesprin-2 giant (<b>C</b>) protein expression, normalised against the GAPDH loading control and presented as an average fold change relative to the vehicle treatment. The data are presented as the mean ± SEM, n = 3. Statistical analysis was conducted using a one-way ANOVA, and no significant differences (indicated by “ns”) were observed between the media- and AGK2-treated groups, compared to the vehicle-treated control. (<b>D</b>) Immunofluorescence microscopy images of control and AGK2-treated MDA-MB-231 cells stained with N1ABD antibody (red channel), specific to the actin-binding domain of Nesprin-1 (Nes1). Nuclei are visualised using DAPI counterstain (blue channel). Scale bar: 10 μm. (<b>E</b>) Microscopy images of control and AGK2-treated MDA-MB-231 cells immunostained with pAbK1 antibody (red channel), specific to the C-terminus of Nesprin-2 (Nes2). DAPI indicates nuclear staining. Scale bar: 5 μm.</p>
Full article ">Figure 4
<p>SIRT2 inhibition does not impact the expression of SUN1, SUN2, lamin A/C, and lamin B1 proteins in MDA-MB-231 cells. (<b>A</b>) Western blot analysis of SUN1, SUN2, lamin A/C, and Lamin B1 protein expression in media-, vehicle-, and AGK2-treated MDA-MB-231 cells, with GAPDH levels demonstrating equal protein loading. (<b>B</b>–<b>F</b>) Densitometric quantification of SUN1 (<b>B</b>), SUN2 (<b>C</b>), lamin A (<b>D</b>), lamin C (<b>E</b>), and lamin B1 (<b>F</b>) protein expression levels, normalised against the GAPDH loading control and presented as a mean fold change relative to the vehicle treatment. The data are presented as the mean ± SEM, n = 3. Statistical significance was assessed using a one-way ANOVA, comparing each group to the vehicle control. “ns” indicates non-significance.</p>
Full article ">Figure 5
<p>The nuclei of AGK2-treated MDA-MB-231 cells exhibit greater resistance to collapse under osmotic shock compared to controls. (<b>A</b>) A schematic demonstrating the osmotic shock assay as a method to study nuclear mechanics. Subjection to the hypertonic 640 mOsm sucrose solution results in the outward movement of water from the cell. Nuclei that undergo collapse and exhibit nuclear envelope folds suggest an inability to resist the osmotic pressure, indicating nuclear softness. In contrast, nuclei that retain their morphology under osmotic shock imply greater rigidity. (<b>B</b>) Lamin A/C immunofluorescence microscopy images of control and AGK2-treated MDA-MB-231 cells cultured under normal growth conditions (control, upper row) or subjected to osmotic shock (lower row). Lamin A/C stain (red channel) acts as a nuclear envelope marker, enabling the identification of cells exhibiting nuclear envelope folds, which are highlighted by asterisks (*). DAPI stain was used to visualise nuclei (blue channel). Insets (osmotic shock condition, lower row) are higher magnifications of the areas indicated by dashed boxes. Scale bars (lower and upper row): 20 μm. (<b>C</b>) Quantification of the percentage (%) of media-, vehicle-, and AGK2-treated cells exhibiting lamin A/C folds when cultured under control conditions or following osmotic shock; &gt;200 cells analysed for each condition. (<b>D</b>) Quantification of the contour ratio (nuclear circularity) of control and AGK2-treated MDA-MB-231 cells following subjection to osmotic shock; &gt;200 cells analysed per condition. The data in (<b>C</b>,<b>D</b>) are presented as the mean ± SEM. Statistical significance between cells grown under control and osmotic shock conditions for each treatment (<b>C</b>) was determined using a Student’s unpaired t-test, with statistically significant results indicated by underlined labels. Statistical analysis of media-, vehicle-, and AGK2-treated MDA-MB-231 cells subjected to osmotic shock (<b>C</b>,<b>D</b>) was conducted using a one-way ANOVA, followed by a Dunnett’s post hoc test relative to the vehicle treatment. “ns” demonstrates non-significance, while ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001 signify statistical significance.</p>
Full article ">Figure 6
<p>AGK2 treatment impedes the migration and polarisation of MDA-MB-231 cells in a 2D environment. (<b>A</b>) Fluorescence microscopy images of media-, vehicle-, and AGK2-treated MDA-MB-231 cells subjected to the EdU proliferation assay. EdU-positive cells are indicated by the red channel, while Hoechst labelling indicates nuclear staining on the blue channel. Scale bar: 20 μm. (<b>B</b>) Quantification of the percentage (%) of EdU-positive cells for each treatment; &gt;1000 cells analysed per condition. (<b>C</b>) Phase contrast images of the scratch-wound assay at 0 h (upper row) and 24 h (lower row) post-wounding for control and AGK2-treated cells. Scale bar: 100 μm. (<b>D</b>) Quantification of the percentage (%) wound closure at 0, 12, and 24 h of the cell wounding experiment for each treatment, n = 3. (<b>E</b>) A schematic depicting the parameters by which cells were characterised as polarised in the scratch-wound assay. Cells were classified as polarised when the Golgi organelle was situated within the 120° radius (pink-shaded area) facing the wound edge. (<b>F</b>) Quantification of the number of polarised cells for each treatment, displayed as a fold change relative to the vehicle control; &gt;300 cells analysed per condition. The data in (<b>B</b>,<b>D</b>,<b>F</b>) are presented as the mean ± SEM, and statistical significance was assessed using a one-way ANOVA, followed by a Dunnett’s post hoc test comparing the data to the vehicle treatment. “ns” denotes non-significant results, while * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, and *** <span class="html-italic">p</span> ≤ 0.001 indicates statistical significance.</p>
Full article ">Figure 7
<p>SIRT2 inhibition reduces the migration of MDA-MB-231 cells through space-restricted 3D environments. (<b>A</b>) A schematic demonstrating the setup of the 3D space-restrictive invasion assay, where an insert with a porous membrane (indicated by a purple dashed line, pore diameter: 5 μm) acts as a restrictive barrier for cells migrating from an area of serum-free media (light blue shading) to complete media (dark blue shading). (<b>B</b>) Quantification of media-, vehicle-, and AGK2-treated cell migration within the 3D space-restrictive invasion assay, presented as a fold change relative to the vehicle treatment. (<b>C</b>) A schematic showing the setup of the well-established Boyden Chamber assay, in which the restrictive barrier between the serum-free media (light blue shading) and complete media (dark blue shading) consists of a layer of extracellular matrix (ECM, pink shading) as well as a porous membrane (indicated by a pink dashed line, pore diameter: 8 μm). (<b>D</b>) Quantification of media-, vehicle-, and AGK2-treated cell migration within the Boyden Chamber assay, presented as a fold change relative to the vehicle treatment. The data in (<b>B</b>,<b>D</b>) are shown as the mean ± SEM, and statistical significance was determined using a one-way ANOVA followed by a Dunnett’s post hoc test relative to the vehicle treatment. “ns” indicates non-significant results, while * <span class="html-italic">p</span> ≤ 0.05 and ** <span class="html-italic">p</span> ≤ 0.01 indicate statistical significance.</p>
Full article ">Figure 8
<p>HCC38 cells display reduced SIRT2 expression compared to MDA-MB-231 cells and show no change in migratory capacity upon AGK2 treatment. (<b>A</b>) Western blot analysis showing SIRT2, acetylated α-tubulin, and vimentin protein expression levels in MDA-MB-231 cells compared to HCC38 cells. GAPDH expression is used to demonstrate equal protein loading. (<b>B</b>) Densitometric quantification of SIRT2, acetylated α-tubulin, and vimentin protein expression in MDA-MB-231 and HCC38 cells, normalised against the GAPDH loading control and presented as an average fold change relative to the protein expression in MDA-MB-231 cells. The data are presented as the mean ± SEM, n = 3. (<b>C</b>) Immunofluorescence microscopy images of acetylated α-tubulin (AcTub; red channel) organisation in media-, vehicle-, and AGK2-treated HCC38 cells. Nuclei are visualised using a DAPI counterstain (blue channel). Scale bar: 10 μm. (<b>D</b>) Quantification of the percentage (%) of control and AGK2-treated HCC38 cells exhibiting prominent perinuclear acetylated α-tubulin rings; &gt;300 cells analysed per condition. (<b>E</b>) Quantification of media-, vehicle-, and AGK2-treated HCC38 cell migration within the 3D space-restrictive invasion assay (5 μm diameter pores), presented as a fold change relative to the vehicle treatment. Statistical significance in panel (<b>B</b>) was determined by a Student’s unpaired t-test, and in panels (<b>D</b>,<b>E</b>) using a one-way ANOVA, comparing each group to the vehicle control. “ns” indicates non-significance, while * <span class="html-italic">p</span> ≤ 0.05 denotes statistical significance.</p>
Full article ">
27 pages, 3233 KiB  
Review
Sirtuins as Key Regulators in Pancreatic Cancer: Insights into Signaling Mechanisms and Therapeutic Implications
by Surbhi Chouhan, Anil Kumar, Naoshad Muhammad, Darksha Usmani and Tabish H. Khan
Cancers 2024, 16(23), 4095; https://doi.org/10.3390/cancers16234095 - 6 Dec 2024
Viewed by 449
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands as one of the most lethal cancers, marked by rapid progression, pronounced chemoresistance, and a complex network of genetic and epigenetic dysregulation. Within this challenging context, sirtuins, NAD+-dependent deacetylases, have emerged as pivotal modulators of key [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) stands as one of the most lethal cancers, marked by rapid progression, pronounced chemoresistance, and a complex network of genetic and epigenetic dysregulation. Within this challenging context, sirtuins, NAD+-dependent deacetylases, have emerged as pivotal modulators of key cellular processes that drive pancreatic cancer progression. Each sirtuin contributes uniquely to PDAC pathogenesis. SIRT1 influences apoptosis and chemoresistance through hypoxia, enhancing glycolytic metabolism and HIF-1α signaling, which sustain tumor survival against drugs like gemcitabine. SIRT2, conversely, disrupts cancer cell proliferation by inhibiting eIF5A, while SIRT3 exerts tumor-suppressive effects by regulating mitochondrial ROS and glycolysis. SIRT4 inhibits aerobic glycolysis, and its therapeutic upregulation has shown promise in curbing PDAC progression. Furthermore, SIRT5 modulates glutamine and glutathione metabolism, offering an avenue to disrupt PDAC’s metabolic dependencies. SIRT6 and SIRT7, through their roles in angiogenesis, EMT, and metastasis, represent additional targets, with modulators of SIRT6, such as JYQ-42, showing potential to reduce tumor invasiveness. This review aims to provide a comprehensive exploration of the emerging roles of sirtuins, a family of NAD+-dependent enzymes, as critical regulators within the oncogenic landscape of pancreatic cancer. This review meticulously explores the nuanced involvement of sirtuins in pancreatic cancer, elucidating their contributions to tumorigenesis and suppression through mechanisms such as metabolic reprogramming, the maintenance of genomic integrity and epigenetic modulation. Furthermore, it emphasizes the urgent need for the development of targeted therapeutic interventions aimed at precisely modulating sirtuin activity, thereby enhancing therapeutic efficacy and optimizing patient outcomes in the context of pancreatic malignancies. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>SIRT1 and pancreatic cancer. These pathways illustrate SIRT1’s broad impact on various processes critical for pancreatic cancer progression, including EMT, metabolic adaptation, survival under hypoxic conditions and resistance to apoptosis and chemotherapy. Targeting SIRT1 in these pathways holds therapeutic potential for overcoming drug resistance and improving treatment outcomes in pancreatic cancer.</p>
Full article ">Figure 2
<p>Summary of pancreatic cancer pathways influenced by SIRT2. These pathways highlight SIRT2’s multifaceted role in regulating KRAS-driven oncogenic signaling, MYC stability, inflammation and epigenetic remodeling, making it a promising therapeutic target in pancreatic cancer.</p>
Full article ">Figure 3
<p>Overview of signaling pathways impacted by SIRT3 in pancreatic cancer. SIRT3 acts as a tumor suppressor by regulating mitochondrial function, oxidative stress and metabolic reprogramming in pancreatic cancer.</p>
Full article ">Figure 4
<p>Summary of key signaling pathways influenced by SIRT4 in pancreatic cancer. These pathways collectively position SIRT4 as a critical tumor suppressor in pancreatic cancer, influencing metabolic reprogramming, autophagy and mitochondrial function.</p>
Full article ">Figure 5
<p>Overview of the principal signaling pathways affected by SIRT5 in pancreatic cancer. SIRT5 functions as a crucial metabolic gatekeeper in pancreatic cancer, with its modulation presenting potential therapeutic strategies to disrupt the metabolic dependencies of tumor cells.</p>
Full article ">Figure 6
<p>Overview of the major signaling pathways influenced by SIRT6 in pancreatic cancer. SIRT6 regulates critical pathways in pancreatic cancer, including the suppression of glycolysis, the modulation of the NF-κB and HIF1α axes and the stabilization of ATF4 for stress response. It also controls epigenetic regulation via Lin28b, impacting tumor growth and metastasis. Novel SIRT6 inhibitors show promise in enhancing chemotherapy sensitivity and inhibiting angiogenesis.</p>
Full article ">Figure 7
<p>Overview of the key signaling pathways impacted by SIRT7 in pancreatic cancer. SIRT7 regulates pancreatic cancer progression by controlling GLUT3 expression, impacting gemcitabine sensitivity, and inhibiting EMT. O-GlcNAcylation stabilizes SIRT7, allowing it to repress tumor suppressor genes and promote tumor growth. Its low nuclear expression correlates with aggressive tumors, making SIRT7 a key prognostic biomarker and therapeutic target.</p>
Full article ">
14 pages, 9492 KiB  
Article
SIRT1 Activation Suppresses Corneal Endothelial–Mesenchymal Transition via the TGF-β/Smad2/3 Pathway
by Yi Yu, Ruilin Guo, Jie Ling, Chenjia Xu, Minglu Ma, Xiaojuan Dong, Jing Wu and Ting Huang
Curr. Issues Mol. Biol. 2024, 46(12), 13846-13859; https://doi.org/10.3390/cimb46120827 - 6 Dec 2024
Viewed by 410
Abstract
Endothelial–mesenchymal transition (EnMT) is the transversion of endothelial cells to mesenchymal cells under certain physiological or pathological conditions. When EnMT occurs in the corneal endothelium, corneal endothelial cells (CECs) lose their normal function and thus cannot maintain corneal clarity. Studies have shown that [...] Read more.
Endothelial–mesenchymal transition (EnMT) is the transversion of endothelial cells to mesenchymal cells under certain physiological or pathological conditions. When EnMT occurs in the corneal endothelium, corneal endothelial cells (CECs) lose their normal function and thus cannot maintain corneal clarity. Studies have shown that the mechanism of EnMT in CECs involves the transforming growth factor-β (TGF-β) signaling pathway, and one of the important inhibitors of the TGF-β/Smad2/3 pathway is sirtuin-1 (SIRT1). In this study, we used a rat model of corneal endothelium injury and TGF-β1-treated human CECs to induce EnMT, aiming to explore whether SIRT1 activation inhibits corneal EnMT in vivo and in vitro. SIRT1 was activated and suppressed using resveratrol (RSV) and EX527, respectively. The endothelial markers and mesenchymal markers were measured by immunofluorescence and Western blot assays. Co-immunoprecipitation was used to detect the interaction between SIRT1 and Smad2/3. The results showed that after mechanical injury, the group treated with RSV-activated SIRT1 regained corneal transparency and recovered from edema faster than the control group. Moreover, RSV-activated SIRT1 downregulated the expression levels of alpha smooth muscle actin (α-SMA), vimentin, and Snail and upregulated the expression levels of E-cadherin and Na+/K+-ATPase both in vivo and in vitro, but these effects were reversed when SIRT1 was inhibited by EX527. SIRT1 also upregulated the expression levels of TGF-β receptor 1 and phosphorylated Smad2/3. The interaction between SIRT1 and Smad2/3 in vitro was confirmed by co-immunoprecipitation. Overall, our results indicate that SIRT1 activation inhibits corneal EnMT via the TGF-β/Smad2/3 pathway, which may be a potential therapeutic target for corneal endothelium dysfunction. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Effects of SIRT1 activation on corneal clarity and central corneal thickness in injury-induced EnMT rat model. Endothelial layer is indicated in red. (<b>A</b>) Diagram of the establishment of the corneal endothelial injury model. (<b>B</b>) Changes in corneal clarity in rats as observed under a slit lamp microscope on days 1, 3, 5, and 7 after injection with RSV or RSV + EX527. (<b>C</b>) AS-OCT images of central cornea. Scale bar = 200 μm. AS-OCT: anterior segment optical coherence tomography. (<b>D</b>) H&amp;E staining of the cornea. Scale bar = 100 μm. H&amp;E: hematoxylin and eosin. (<b>E</b>) Quantification of the central cornea thickness. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 2
<p>SIRT1 activation inhibited corneal EnMT in rats. (<b>A</b>) Protein level of SIRT1 in cornea. (<b>B</b>) Double immunofluorescence staining results for the expression levels of α-SMA (green) and SIRT1 (red) in corneal endothelium. Scale bar = 20 μm. (<b>C</b>) Western blot assay was performed to detect the expression levels of Na<sup>+</sup>/K<sup>+</sup>-ATPase, Snail, and E-cadherin and of the mesenchymal markers α-SMA and vimentin. GAPDH was used as internal control. * <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">n</span> = 3.</p>
Full article ">Figure 3
<p>SIRT1 activation inhibited the migration of and the occurrence of morphological changes in TGF-β1-treated HCECs in vitro. (<b>A</b>) Morphological changes in HCECs. Scale bar = 100 μm. (<b>B</b>) Cell counting kit-8 assay was used to assess the cytotoxicity of different treatments toward HCECs. (<b>C</b>) Micrographs and statistical graph of HCEC migration in the scratch assay. Red lines indicate the migration edges. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">n</span> = 3. Scale bar = 100 μm. HCEC: Human Corneal Endothelial Cell.</p>
Full article ">Figure 4
<p>SIRT1 activation inhibited TGF-β1-induced EnMT in HCECs. (<b>A</b>) Protein level of SIRT1 in HCECs. (<b>B</b>) Double immunofluorescence staining results demonstrating the expression of α-SMA (green) and SIRT1 (red) in HCECs. Scale bar = 10 μm. (<b>C</b>) The protein levels of Na<sup>+</sup>/K<sup>+</sup>-ATPase, Snail, and VE-cadherin and the mesenchymal markers α-SMA and vimentin were evaluated by Western blot analysis. GAPDH was used as internal control. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">n</span> = 3. HCEC: Human Corneal Endothelial Cell.</p>
Full article ">Figure 5
<p>SIRT1 inhibited the expression of TGF-βR1 and P-Smad2/3 and interacted with Smad2/3 in TGF-β1-treated HCECs. (<b>A</b>) Western blot analysis of the TGF-β/Smad2/3 pathways. (<b>B</b>) Co-immunoprecipitation (CO-IP) results showing the interaction between SIRT1 and Smad2/3. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">n</span> = 3.</p>
Full article ">
18 pages, 2473 KiB  
Article
Quercetin and Mesenchymal Stem Cell Metabolism: A Comparative Analysis of Young and Senescent States
by Alexandra Ivan, Alexandra Teodora Lukinich-Gruia, Iustina-Mirabela Cristea, Maria-Alexandra Pricop, Crenguta Livia Calma, Alina-Georgiana Simina, Călin Adrian Tatu, Atena Galuscan and Virgil Păunescu
Molecules 2024, 29(23), 5755; https://doi.org/10.3390/molecules29235755 - 5 Dec 2024
Viewed by 383
Abstract
Quercetin is a natural flavonoid renowned for its potent antioxidant, anti-inflammatory, anti-diabetic, and antibacterial properties, making it a highly promising candidate for the treatment of various medical conditions. Our current study investigates the influence of quercetin on energy metabolism, fatty acid composition, oxidative [...] Read more.
Quercetin is a natural flavonoid renowned for its potent antioxidant, anti-inflammatory, anti-diabetic, and antibacterial properties, making it a highly promising candidate for the treatment of various medical conditions. Our current study investigates the influence of quercetin on energy metabolism, fatty acid composition, oxidative stress gene expression, and sirtuin expression in early- and late-stage passages of stem cells derived from human exfoliated deciduous teeth (SHEDs). Mitochondrial respiration was analyzed by measuring oxygen consumption following a 24 h quercetin treatment, while fatty acid profiles were examined using gas chromatography–mass spectrometry (GC-MS). Additionally, quantitative PCR (qPCR) was used to assess the expression of oxidative stress genes and sirtuins. In younger SHEDs, quercetin enhances metabolic activity and mitochondrial respiration, although higher doses may decrease mitochondrial activity. Conversely, in older, senescent SHEDs, quercetin supports mitochondrial function at lower concentrations but appears to inhibit respiration at higher doses. These results suggest that quercetin may hold therapeutic potential for maintaining SHED viability and function, especially at lower doses in older cells. Further research is essential to fully elucidate a dose-dependent effect of quercetin and optimize its applications in regenerative medicine. Full article
Show Figures

Figure 1

Figure 1
<p>β-galactosidase activity was assessed in SHED cells at early (P5) and late passages (P16). Senescent cells stained blue due to β-galactosidase activity, allowing for visual quantification under a Zeiss Axio Observer Z1 microscope, at magnifications of 200× (<b>A</b>,<b>C</b>) and 400× (<b>B</b>,<b>D</b>). Passage 5 (<b>A</b>,<b>B</b>) represents cells at a younger stage, showing a baseline level of senescence, while passage 16 (<b>C</b>,<b>D</b>) reflects an advanced stage with likely higher senescence levels. The increase in blue-stained cells at higher passages reflects a greater accumulation of senescent cells as cell passage advances. The percentage of senescent cells was determined by calculating the ratio of β-gal-positive cells to the total cell count, expressed as a percentage for early-passage (<b>E</b>) and late-passage (<b>F</b>) cells.</p>
Full article ">Figure 2
<p>MTT assay. Effect of quercetin on cell proliferation in younger (P5) and older (P16) SHED cell passages across quercetin dose groups (1 µM, 3 µM, 7 µM, and 10 µM) after 24 h of treatment. Data are presented as mean ± SD relative to cells cultured in control media. No significant differences in cell viability were observed.</p>
Full article ">Figure 3
<p>Oxygen consumption rates of SHED cells following quercetin treatment, measured with the Oroboros O2k System. Oxygen consumption was evaluated in early-passage (P5) and late-passage (P16) SHED cells to determine the effects of quercetin treatment at 1 µM and 10 µM. The impact of quercetin on routine respiration (R), maximum respiration (M), leak respiration (L), and oxidative phosphorylation (OXPHOS) is shown. Statistically significant differences between control and quercetin-treated groups are indicated by asterisks ** <span class="html-italic">p</span> &lt; 0.001; *** <span class="html-italic">p</span> &lt; 0.0001. Representative Oroboros oxygraphs illustrating oxygen consumption and basal respiration, measured using high-resolution respirometry, are available in the <a href="#app1-molecules-29-05755" class="html-app">Supplementary Files (Figure S1)</a>.</p>
Full article ">Figure 4
<p>Effect of quercetin on the fatty acid profile in younger and older SHEDs. Data are presented as mean ± SD from three independent experiments. Statistically significant differences between control and quercetin-treated groups are indicated by asterisks * <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 5
<p>Effect of quercetin (1 µM and 10 µM) on saturated and unsaturated fatty acid levels in younger (P5) and older (P16) SHEDs, as analyzed by GC-MS. Data are presented as mean ± SD from three independent experiments. Statistically significant differences between the older quercetin-treated group and both the younger and older passage control groups are marked with asterisks (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 6
<p>Oxidative stress gene expression levels (PPARγ, ACC, Ahr, SOD, and CYP1A1) in SHEDs were measured by qPCR, with GAPDH used as the reference gene. Data are presented as the mean  ±  SEM from three independent experiments. Statistical significance is indicated as * <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 7
<p>Sirtuins gene expression levels were measured by qPCR, with GAPDH as the reference gene. Data are presented as the mean  ±  SEM from three independent experiments. Statistical significance is indicated as * <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 ">
14 pages, 2366 KiB  
Article
HIV-1 Tat Protein and Cigarette Smoke Mediated ADAM17 Upregulation Can Lead to Impaired Mucociliary Clearance
by Kingshuk Panda, Maria J. Santiago, Md. Sohanur Rahman, Suvankar Ghorai, Stephen M. Black, Irfan Rahman, Hoshang J. Unwalla and Srinivasan Chinnapaiyan
Cells 2024, 13(23), 2009; https://doi.org/10.3390/cells13232009 - 5 Dec 2024
Viewed by 754
Abstract
Human immunodeficiency virus type-1 (HIV-1) associated comorbidities account for the majority of poor health outcomes in people living with HIV (PLWH) in the era of antiretroviral therapy. Lung-related comorbidities such as chronic obstructive pulmonary disease (COPD) and bacterial pneumonia are primarily responsible for [...] Read more.
Human immunodeficiency virus type-1 (HIV-1) associated comorbidities account for the majority of poor health outcomes in people living with HIV (PLWH) in the era of antiretroviral therapy. Lung-related comorbidities such as chronic obstructive pulmonary disease (COPD) and bacterial pneumonia are primarily responsible for increased morbidity and mortality in PLWH, even when compensated for smoking. Smokers and COPD patients demonstrate cilia shortening, attenuated ciliary beat frequency (CBF), dysfunctional ciliated cells along with goblet cell hyperplasia, and mucus hypersecretion. This is exacerbated by the fact that almost 60% of PLWH smoke tobacco, which can exacerbate inflammation and mucociliary clearance (MCC) dysfunction. This study shows that HIV Tat alters the microRNAome in airway epithelial cells and upregulates miR-34a-5p with consequent suppression of its target, Sirtuin 1 (SIRT1). SIRT1 is known to suppress Metalloproteinase 17 (ADAM17), a protease activating Notch signaling. HIV and cigarette smoke (CS) upregulate ADAM17. ADAM17 upregulation followed by SIRT1 suppression can lead to decreased ciliation, mucus hypersecretion, and attenuated MCC, a hallmark of chronic bronchitis in smokers and COPD. It is, therefore, essential to understand the pathophysiological mechanism resulting in acquired Notch dysregulation and its downstream impact on HIV-infected smokers. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>HIV Tat and CS alter the bronchial epithelial microRNAome and suppress SIRT1. (<b>A</b>) Primary NHBE cells were infected with five ng p24 equivalent of HIV BaL (R5-tropic strain). Experiments were terminated after 48 h, and total RNA was analyzed. HIV alters the bronchial epithelial microRNAome and upregulates miR-34a-5p. (<b>B</b>) Primary NHBE ALI cultures were treated with Tat (10 nM) (heat-inactivated Tat as control). HIV Tat upregulates miR-34a-5p, which is known to suppress <span class="html-italic">SIRT1</span>. Even though Tat induces miR-34a-5p by a log2 fold of 8, miR-34a-5p binds to and regulates over 600 different genes with one or more binding sites each (<a href="https://awi.cuhk.edu.cn/~miRTarBase/miRTarBase_2025/php/index.php" target="_blank">https://awi.cuhk.edu.cn/~miRTarBase/miRTarBase_2025/php/index.php</a>) (accessed on 24 January 2024). Hence, the observed magnitude of <span class="html-italic">SIRT1</span> suppression aligns with expectations that an effective increase of miR-34a-5p per target will be much smaller. (<b>C</b>) BEAS-2B airway epithelial cells were transfected with miR-34a-5p mimic (40 nM) using RNAiMAX Lipofectamine reagent. The total RNA was extracted, analyzed by qPCR and the <span class="html-italic">SIRT1</span> suppression was observed. (<b>D</b>) Western blot analysis confirmed the suppression of SIRT1 protein level after transfecting miR-34a-5p mimic. (<b>E</b>) NHBE ALI cultures were treated with HIV Tat (heat inactivated Tat as control). Separately, another set was treated with ATA. HIV Tat suppresses <span class="html-italic">SIRT1</span> mRNA compared to control, and ATA rescues the level of <span class="html-italic">SIRT1</span>. (<b>F</b>): NHBE ALI cultures were exposed to CS (air as control). Separately, another set was treated with ATA. CS suppresses <span class="html-italic">SIRT1</span> mRNA compared to control, and ATA rescues this. n = NHBE ALI cultures from at least three different lungs, n = 3 different experiments using at least BEAS-2B cells, * = significant from control (<span class="html-italic">p</span> &lt; 0.05), S = significant from each other (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>HIV infection and CS suppress <span class="html-italic">SIRT1</span> gene expression through TGF-β signaling. (<b>A</b>) NHBE ALI cultures were exposed to CS and infected with five ng p24 equivalent of HIV BaL (R5- tropic strain). Experiments were terminated after 48 h, and total RNA was analyzed for <span class="html-italic">SIRT1</span> mRNA by qPCR. The combination of HIV and CS exposure suppresses <span class="html-italic">SIRT1</span> expression compared to uninfected. (<b>B</b>) Similar set of experiments were used to analyze for SIRT1 protein level by Western blot. HIV infection alone and combination of HIV and CS exposure suppress SIRT1 protein level. (<b>C</b>) NHBE ALI cultures were treated with recombinant TGF-β (10 ng/mL, vehicle as control). Forty-eight hours post-treatment, total RNA was isolated and analyzed for <span class="html-italic">SIRT1</span> mRNA expression level using qPCR. The <span class="html-italic">SIRT1</span> mRNA level was significantly lower in TGF-β-treated NHBE cells than in the vehicle-treated control. (<b>D</b>) Forty-eight hours post-treatment with TGF-β1, total protein was isolated and analyzed for SIRT1 protein expression level using western blot. The SIRT1 protein level was significantly lower in TGF-β1 treated NHBE cells than in the vehicle control. n = NHBE ALI cultures from at least three different lungs, * = significant from control (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>HIV and CS upregulate <span class="html-italic">ADAM17</span> possibly by <span class="html-italic">SIRT1</span> suppression. (<b>A</b>) To investigate the effect of <span class="html-italic">SIRT1</span> on <span class="html-italic">ADAM17</span> regulation, NHBE cells were transfected with SIRT1-siRNA using RNAiMAX Lipofectamine transfection reagent. Total RNA was isolated and analyzed for gene expression. qPCR analysis confirmed the successful silencing of <span class="html-italic">SIRT1</span>. (<b>B</b>) qPCR analysis confirmed the upregulation of <span class="html-italic">ADAM 17</span> upon <span class="html-italic">SIRT1</span> silencing. (<b>C</b>) NHBE ALI cultures were exposed to CS and infected with five ng p24 equivalent of HIV BaL (R5-tropic strain). Experiments were terminated after 48 h, and total RNA was analyzed for <span class="html-italic">ADAM17</span> mRNA by qPCR. HIV-infected and the combination of HIV plus CS exposure increases the <span class="html-italic">ADAM17</span> mRNA level. (<b>D</b>) Another experimental set was used for ADAM17 protein level by western blot. The upregulation of ADAM17 protein levels were noticed in both HIV and CS-exposed NHBE ALI cultures. n = NHBE ALI cultures from at least 3 different lungs, * = significant from control (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Effect of HIV and CS-mediated MCC dysfunction. (<b>A</b>) NHBE ALI cultures were exposed to CS and infected with five ng p24 equivalent of HIV BaL (R5- tropic strain). Experiments were terminated after 9 days, and total RNA was analyzed for <span class="html-italic">MUC5AC</span> mRNA by qPCR. HIV and CS increase the <span class="html-italic">MUC5AC</span> mRNA level compared to uninfected. (<b>B</b>,<b>C</b>) MUC5AC immunofluorescent staining of NHBE ALI cultures and protocol details described in method section. MUC5AC in green and nuclei stained with DAPI in blue. Upregulation of MUC5AC was observed upon HIV infection and in CS-exposed cells compared to uninfected, scale-10 µM. (<b>D</b>) HIV and CS inhibit ciliogenesis. Primary bronchial epithelial cells were exposed to CS and infected with HIV. On Day 9 post-infection, ciliogenesis was observed in lung-matched uninfected control, and protocol details described in method section. Cilia in green; nuclei stained with DAPI in blue. We observed impaired ciliation in both HIV-infected and CS-exposed NHBE ALI cultures, scale-20 µM. n = NHBE ALI cultures from 3 different lungs at least, * = significant from control (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p><span class="html-italic">SIRT1</span>-mediated <span class="html-italic">ADAM17</span> upregulation activates the cytokine storm. (<b>A</b>–<b>D</b>) NHBE ALI cultures were exposed to CS and infected with five ng p24 equivalent of HIV BaL (R5-tropic strain). Experiments were terminated after 48 h, and the supernatant was analyzed for quantitative measurement of secretory cytokines IL-8/CXCL8 and MCP-1/CCL2, G-CSF and CXCL5. ELISA results demonstrated a significant upregulation of IL-8, CCL2, G-CSF, and CXCL5 in NHBE cells upon infection compared to uninfected controls. (<b>E</b>) Experiments were terminated after 48 h, and total protein was analyzed for proinflammatory cytokine protein levels using RayBio Label-Based (L-Series), Human Antibody Array L-8000 Glass Slide Kit. Heatmap analysis shows the upregulated proinflammatory cytokine protein level, including IL-1α, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, MCP-1, RAGE, S100 A8/A9, TNF-α and TNF-β. n = NHBE ALI cultures from at least three different lungs, * = significant from control (<span class="html-italic">p</span> &lt; 0.05), s = significant between the treatment group.</p>
Full article ">
23 pages, 2142 KiB  
Review
Sirtuins: Emergent Players in Tissue and Organ Regeneration
by Ayla Kyler Núñez, Claudia Marcela Arenas-Gómez and Belfran Alcides Carbonell Medina
Int. J. Transl. Med. 2024, 4(4), 687-709; https://doi.org/10.3390/ijtm4040048 - 2 Dec 2024
Viewed by 443
Abstract
Sirtuins are a family of lysine deacetylases that regulate cellular homeostasis and energy sensing. Regeneration is the process that restores structural and functional homeostasis at the cellular, tissue, organ, and appendage levels. Several cellular processes, such as epithelial–mesenchymal transition (EMT), proliferation, migration, and [...] Read more.
Sirtuins are a family of lysine deacetylases that regulate cellular homeostasis and energy sensing. Regeneration is the process that restores structural and functional homeostasis at the cellular, tissue, organ, and appendage levels. Several cellular processes, such as epithelial–mesenchymal transition (EMT), proliferation, migration, and differentiation, contribute to restoration after an injury. This review highlights the role of sirtuins in tissue, organ, and anatomical structure regeneration, showing how sirtuins modulate signalling pathways by deacetylating targets such as transcription factors. Furthermore, understanding the role of this protein family could help elucidate the molecular and cellular mechanisms underlying tissue regeneration, which may hold significant potential for fields such as regenerative medicine. The review compiles evidence suggesting that sirtuins are emerging factors in the regeneration of various organs (e.g., skin, liver, heart) and tissues (e.g., bone, muscle, cornea, spinal cord). Full article
Show Figures

Figure 1

Figure 1
<p>Sirtuins regulate cell migration, proliferation, and differentiation. SIRT1 negatively (red grid) or positively (green grid) modulates cell migration. Deacetylation by SIRT1 activates or inhibits the functions of proteins, while SIRT1 inhibits SMAD and YY1 and activates beclin 1 through deacetylation. SIRT1 modulates cell proliferation through the JAK/STAT and WNT pathways, and the deacetylation of STAT3 permits its phosphorylation and (subsequent) activation. Similarly, the deacetylation of p21 permits its ubiquitination and (subsequent) degradation. SIRT1 deacetylates and activates FOXO3a, and FOXO3a binds to the FOXO response element (FRE) in the <span class="html-italic">runx2</span> (an osteogenesis factor) promoter and enhances its expression. SIRT1 also indirectly participates in osteogenesis through the inhibition of osteogenesis inhibitors such as NF-κB and p65. In general, sirtuins promote osteogenesis and chondrogenesis and impair adipogenesis, except SIRT4, which increases adipogenesis. Abbreviations: ACSL1, acyl CoA synthetase long-chain family member 1; ACTA2, actin alpha 2; ALP, alkaline phosphatase; BSP, bone sialoprotein; C/EBP α, CCAAT/enhancer-binding protein α; CDK, cyclin-dependent kinase; COL, collagen; ECM, extracellular matrix; EMT, epithelial–mesenchymal transition; FABP4, fatty acid-binding protein 4; FAS, fatty acid synthase; FN, fibronectin; FRE, FOXO response element; FOXO3a, forkhead box O3a; IWP2, inhibitor of Wnt production 2; KLF5, Krüppel-like factor 5; MMP, matrix metalloproteinase; NF-κB, nuclear factor kappa-light-chain enhancer of activated B cells; OC, osteocalcin; OPN, osteopontin; OSX, osterix; PPARγ2, peroxisome proliferator-activated receptor gamma 2; PRDM16, positive regulatory domain zinc finger region protein 16; RUNX2, runt-related transcription factor 2; SMAD4, small mothers against decapentaplegic 4; STAT, signal transducer and activator of transcription; WNT, wingless-related integration; YY1, Yin Yang 1; α-SMA, alpha smooth muscle actin.</p>
Full article ">Figure 2
<p>Sirtuins participate in tissue regeneration. Inhibition of sirtuins reduces proliferation, retarding wound closure in the corneal epithelium. Chemical activation of SIRT1 improves autophagy and reduces negative events (inflammation and apoptosis) in spinal cord regeneration. Additionally, in muscle regeneration, experimental modulation of PGC1-α has no effects on muscle regeneration. Abbreviations: ALP, alkaline phosphatase; CCND, cyclin D; CDK, cyclin-dependent kinase; COL, collagen; IL, interleukin; LC-3B, light chain 3 beta; MYF5, myogenic factor 5; MyoD1, myoblast determination protein 1; OC, osteocalcin; OPN, osteopontin; OSX, osterix; PGC1-α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha; RUNX2, runt-related transcription factor 2; TNF-α, tumour necrosis factor alpha.</p>
Full article ">Figure 3
<p>Sirtuins participate in organ regeneration. Sirtuins improve skin regeneration by increasing some cellular processes (angiogenesis, autophagy, etc.) and reducing other ones (fibrosis, inflammation, etc.). In heart regeneration, a direct target of a sirtuin is known: SIRT7 deacetylates and activates GATA4 (a transcription factor that induces the gene expression of <span class="html-italic">Vegf</span>). Additionally, SIRT3 has no effect on the bone parameters in mouse digit regeneration. Abbreviations: ATP, adenosine triphosphate; CCND, cyclin D; CLPP, caseinolytic protease proteolytic subunit; COL, collagen; EGF, epidermal growth factor; FGF, fibroblast growth factor; GATA4, guanine, adenine, thymine, adenine 4; HSP, heat shock protein; IGF, insulin-like growth factor; IκB, inhibitory kappa B; LONP1, Lon protease 1; NF-κB, nuclear factor kappa-light-chain enhancer of activated B cells; NOS, nitric oxide synthase; ROS, reactive oxygen species; TGF-β, transforming growth factor beta; TNF-α, tumour necrosis factor alpha; UPR, unfolded protein response; VEGF, vascular endothelial growth factor.</p>
Full article ">
27 pages, 1188 KiB  
Review
Sirtuin Proteins and Memory: A Promising Target in Alzheimer’s Disease Therapy?
by Francesca Fernandez, Lyn R. Griffiths, Heidi G. Sutherland, Michael H. Cole, J. Helen Fitton, Pia Winberg, Daniel Schweitzer, Lloyd N. Hopkins and Barbara J. Meyer
Nutrients 2024, 16(23), 4088; https://doi.org/10.3390/nu16234088 - 27 Nov 2024
Viewed by 524
Abstract
Sirtuins (SIRTs), nicotine adenine dinucleotide (+)-dependent histone deacetylases, have emerged as critical regulators in many signalling pathways involved in a wide range of biological processes. Currently, seven mammalian SIRTs have been characterized and are found across a number of cellular compartments. There has [...] Read more.
Sirtuins (SIRTs), nicotine adenine dinucleotide (+)-dependent histone deacetylases, have emerged as critical regulators in many signalling pathways involved in a wide range of biological processes. Currently, seven mammalian SIRTs have been characterized and are found across a number of cellular compartments. There has been considerable interest in the role of SIRTs in the brain due to their role in a plethora of metabolic- and age-related diseases, including their involvement in learning and memory function in physiological and pathophysiological conditions. Although cognitive function declines over the course of healthy ageing, neurological disorders including Alzheimer’s disease (AD) can be associated with progressive cognitive impairments. This review aimed to report and integrate recent advances in the understanding of the role of SIRTs in cognitive function and dysfunction in the context of AD. We have also reviewed the use of selective and/or natural SIRT activators as potential therapeutic agents and/or adjuvants for AD. Full article
Show Figures

Figure 1

Figure 1
<p>Overview of SIRTs in the context of learning and memory in AD. Briefly, learning and memory process are supported by synaptic plasticity activation and dendrites formation and branching in neurons. SIRT1 and SIRT3 stimulate these processes directly and indirectly via regulation of cellular oxidation, inflammation and degradation of NFTs via ubiquitination. SIRT1 is also involved in the indirect stimulation of the non-amyloidogenic pathway, leading to a decrease in the formation of Aβ in AD. Via its inhibition of AMPA Rc internalisation, SIRT2 promotes synaptic plasticity and memory. SIRT1 participates in the regulation of <span class="html-italic">BACE1</span> and <span class="html-italic">ADAM10</span> genes, both critical genes in the context of AD. SIRT1, SIRT3 and SIRT6 also modulate the transcription of apoptotic and survival genes in the neurons. Abbreviations: ADAM 10: A Disintegrin and Metalloprotease 10, AMPK: 5′adenosine-monophosphate-activated protein kinase; AMPA Rc: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, APP: Amyloid precursor protein, sAPPβ: secreted amino-terminals APPβ fragment, BACE 1: β-secretase enzyme, CamK: Ca 2+/calmodulin-dependent protein kinase, cdck5cyclin dependent kinase, Aβ: amyloid-β peptides, AICD: APP Intracellular Cytoplasmic/<span class="html-italic">C</span>-terminal Domains, Fynk: Fyn kinase, GSK3β: Glycogen synthase kinase 3 beta, IL-1: Interleukin 1, IL-6: Interleukin 6, mAch and nACh: Metabotropic and nicotinic Acetyl-choline receptors, NF-kB: nuclear factor kappa-light-chain-enhancer of activated B cells, NFTs: Neurofibrillary tangles, NMDA Rc: methyl-D-aspartate receptors receptor, TNF-α: Tumor Necrosis factor alpha, ROS: reactive oxygen species, PSD95: post-synaptic density protein 95.</p>
Full article ">Figure 2
<p>Overview of the modulation of SIRTs by natural compounds (resveratrol, curcumin, and seaweeds) and exercise. Dotted arrows represent an activation of SIRTs reported in vitro.</p>
Full article ">
11 pages, 920 KiB  
Article
Role of Nicotinamide in the Pathogenesis of Actinic Keratosis: Implications for NAD+/SIRT1 Pathway
by Riccardo Belardi, Francesca Pacifici, Terenzio Cosio, Sara Lambiase, Ruslana Gaeta Shumak, Fabio Artosi, Antonia Rivieccio, Danilo Cavalloro, Elena Dellambra, Luca Bianchi, David Della-Morte and Elena Campione
Biomolecules 2024, 14(12), 1512; https://doi.org/10.3390/biom14121512 - 27 Nov 2024
Viewed by 590
Abstract
Actinic keratosis (AK) is a precursor to invasive squamous cell carcinoma, making early diagnosis and treatment essential to prevent progression. Among available therapeutic options, nicotinamide (NAM) has shown potential in reducing AK progression. NAM is a precursor of nicotinamide adenine dinucleotide (NAD+ [...] Read more.
Actinic keratosis (AK) is a precursor to invasive squamous cell carcinoma, making early diagnosis and treatment essential to prevent progression. Among available therapeutic options, nicotinamide (NAM) has shown potential in reducing AK progression. NAM is a precursor of nicotinamide adenine dinucleotide (NAD+), which activates sirtuin (SIRT)1, a protein with anti-cancer properties. Although the role of SIRT1 in AK is still debated, no data currently exist on the systemic modulation of this protein in AK. Therefore, this study aims to evaluate whether NAM, by increasing serum NAD+ levels, may promote SIRT1 activation in peripheral blood mononuclear cells (PBMCs) in AK patients. Thirty patients were enrolled and treated with NAM for 24 months. Hematological, biochemical, and skin condition assessments were conducted, alongside the measurement of SIRT1 and NAD+ levels. A decrease in basophils, monocytes, total cholesterol, and blood glucose levels was observed in the study group, along with a reduction in AK lesions. Notably, NAM treatment significantly enhanced serum NAD+ levels, and nuclear SIRT1 activity in PBMCs. In conclusion, NAM administration significantly reduced AK progression in a NAD+/SIRT1-dependent manner, supporting its role as a chemopreventive agent in AK management. Full article
(This article belongs to the Special Issue NAD Metabolism in Physiology and Pathology)
Show Figures

Figure 1

Figure 1
<p>Evaluation of Actinic Keratosis (AK) lesion number in patients at T0 and T24, along with a representative patient’s image (** <span class="html-italic">p</span> &lt; 0.005).</p>
Full article ">Figure 2
<p>Sera levels of Nicotinamide Adenine Dinucleotide (NAD): (<b>a</b>) sera levels of Total NAD (NADt) in patients at T0 and T24; (<b>b</b>) sera levels of NAD + Hydrogen (NADH) in patients at T0 and T24; (<b>c</b>) sera levels of NAD Plus (NAD<sup>+</sup>) in patients at T0 and T24. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. pmol, picomoles.</p>
Full article ">Figure 3
<p>(<b>a</b>) Sera levels of NAD<sup>+</sup>-dependent deacetylases of the Sirtuin 1 (SIRT1) in patients at T0 and T24. (<b>b</b>) Nuclear activity of SIRT1 in peripheral blood mononuclear cells (PBMCs:) comparison of nuclear activity, monitored over 1 h, of Sirtuin1 (SIRT1), extracted from PBMCs, in patients at T0 and T24. (**** <span class="html-italic">p</span> &lt; 0.0001). FI: fluorescence intensity.</p>
Full article ">
15 pages, 1672 KiB  
Article
Sirtuin-5 Is Recruited to Hepatic Peroxisomes in Mice Fed Dodecanedioic Acid but Has Little Impact on the Peroxisomal Succinylome
by Yuxun Zhang, Bob B. Zhang, Sivakama S. Bharathi, Joanna Bons, Jacob P. Rose, Samah Shah, Steven F. Dobrowolski, Sunder Sims-Lucas, Birgit Schilling and Eric S. Goetzman
Biomolecules 2024, 14(12), 1508; https://doi.org/10.3390/biom14121508 - 26 Nov 2024
Viewed by 433
Abstract
Lysine succinylation, and its reversal by sirtuin-5 (SIRT5), is known to modulate mitochondrial fatty acid β-oxidation (FAO). We recently showed that feeding mice dodecanedioic acid, a 12-carbon dicarboxylic acid (DC12) that can be chain-shortened four rounds to succinyl-CoA, drives high-level protein [...] Read more.
Lysine succinylation, and its reversal by sirtuin-5 (SIRT5), is known to modulate mitochondrial fatty acid β-oxidation (FAO). We recently showed that feeding mice dodecanedioic acid, a 12-carbon dicarboxylic acid (DC12) that can be chain-shortened four rounds to succinyl-CoA, drives high-level protein hypersuccinylation in the peroxisome, particularly on peroxisomal FAO enzymes. However, the ability of SIRT5 to reverse DC12-induced peroxisomal succinylation, or to regulate peroxisomal FAO in this context, remained unexplored. Here, we showed that feeding DC12 strongly recruits SIRT5 into hepatic peroxisomes. Knocking out SIRT5 impaired peroxisomal FAO as evidenced by reduced 14C-DC12 flux in liver homogenates and elevated levels of partially shortened DC12 catabolites in urine. Further, mass spectrometry revealed a trend toward less peroxisomal protein succinylation in SIRT5 knockout liver. This is consistent with a reduced flux of DC12 through the peroxisomal FAO pathway, thereby reducing the production of the succinyl-CoA that chemically reacts with lysine residues to produce protein succinylation. Mass spectrometry comparisons of site-level succinylation in wildtype and SIRT5 knockout liver did not reveal any clear pattern of SIRT5 target sites in the peroxisome after DC12 feeding. However, SIRT5 co-immunoprecipitated with 15 peroxisomal proteins, including the key peroxisomal FAO enzymes acyl-CoA oxidase-1 and enoyl-CoA/3-hydroxyacyl-CoA dehydrogenase (EHHADH). In vitro, recombinant SIRT5 partially desuccinylated chemically modified recombinants ACOX1a, ACOX1b, and EHHADH. Desuccinylation by SIRT5 had no effect on enzyme activity for ACOX1a and EHHADH. For ACOX1b, SIRT5-mediated desuccinylation decreased activity by ~15%. Possible interpretations of these data are discussed. Full article
(This article belongs to the Special Issue Research on Fatty Acid Oxidation and Fatty Acid Oxidation Disorders)
Show Figures

Figure 1

Figure 1
<p>SIRT5 is recruited to liver peroxisomes by feeding mice DC12. (<b>a</b>) Liver peroxisomes were isolated from wildtype mice on standard diet under basal conditions (Fed) or after a 16 h fast (Fast). Immunoblotting shows the presence of SIRT5 in peroxisomes. Heat-shock protein-60 (HSP60) was used as a mitochondrial marker and acyl-CoA oxidase-1 (ACOX1) as a peroxisomal marker to demonstrate purity. (<b>b</b>) Liver heavy mitochondria (HM), light mitochondria (LM) and peroxisomes (P) were separated from wildtype and SIRT5KO mice on standard diet and blotted with anti-SIRT5. All lanes contain 16 µg of protein. Ponceau staining was used as loading control. (<b>c</b>) Wildtype mice were fed either standard laboratory diet (Control) or 10% <span class="html-italic">w</span>/<span class="html-italic">w</span> DC12 for five weeks. Peroxisomes and mitochondria were purified from <span class="html-italic">n</span> = 4 livers and blotted for SIRT5, catalase (CAT) as a peroxisomal marker, and HSP60. Densitometry was used to quantify the bands and generate bar graphs, which show means and standard deviations. ns, not significant; **** <span class="html-italic">p</span> &lt; 0.0001 by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 2
<p>SIRT5 knockout mice have impaired DC12 catabolism. (<b>a</b>,<b>b</b>) Liver homogenates from wildtype (WT) and SIRT5KO mice (T5KO) fed DC12 at 10% <span class="html-italic">w</span>/<span class="html-italic">w</span> for five weeks were used to measure peroxisomal fatty acid oxidation (FAO) using 14C-C24 and 14C-DC12 as substrates. Results are normalized to total protein. (<b>c</b>) Mass spectrometry proteomics was used to quantify the expression of peroxisomal FAO proteins in liver after five weeks of DC12 feeding (N = 5). Heatmap represents fold-changes. Abcd3, ATP-binding cassette sub-family D member 3; Acox1, acyl-CoA oxidase-1; Ehhadh, enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase; Hsd17b4, peroxisomal multifunctional enzyme type 2; Thika, 3-ketoacyl-CoA thiolase A, peroxisomal; Thikb, 3-ketoacyl-CoA thiolase B, peroxisomal. (<b>d</b>) Overnight urine samples (n = 3) in mice maintained on DC12 diet were used for mass spectrometry to quantify DC12 and partially shortened products of DC12 FAO. Data were normalized to urine creatinine. (<b>e</b>,<b>f</b>) SIRT5KO mice were fed standard diet (Ctrl) or DC12 for five weeks. EchoMRI was used to quantify total fat mass and lean mass (n = 4). All bar graphs represent means and standard deviations. ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Knocking out SIRT5 has minimal effects on the peroxisomal succinylome. (<b>a</b>) Liver peroxisomes from wildtype and SIRT5 knockout mice (T5KO) on either standard diet (Ctl) or after feeding DC12 for 7 days were used for anti-succinyllysine immunoblotting. PMP70 was used as a peroxisomal marker (and loading control), and Tim23 was used as a mitochondrial marker to demonstrate purity. (<b>b</b>–<b>e</b>) <span class="html-italic">n</span> = 5 wildtype (WT) and SIRT5KO (T5KO) mice were maintained on standard diet or DC12 diet for five weeks. Mass spectrometry was used to quantify site-level changes in lysine succinylation in the peroxisome. A total of 253 unique succinylated peptides representing 45 peroxisomal proteins were quantified. Succinylated peptides were normalized to protein abundance. Each dot in panels (<b>b</b>–<b>d</b>) represents a succinylated peptide. In panel (<b>b</b>), the abundance of each peptide in WT mice on standard diet (Ctrl) was set = 1 (Log<sub>2</sub> fold change = 0, red line). The other three groups are shown as log<sub>2</sub> ratios to WT control. Panels c and d are volcano plots showing statistical significance for T5KO/WT succinylated peroxisomal peptide ratios on standard diet and DC12 diet, respectively. Gold dots represent putative SIRT5 target peptides that increased significantly upon SIRT5 ablation, blue dots are peptides for which succinylation significantly decreased upon SIRT5 ablation, gray dots had no significant change. (<b>e</b>) All putative SIRT5 target peptides, defined as peptides with significantly increased succinylation in the absence of SIRT5, sorted by subcellular localization.</p>
Full article ">Figure 4
<p>SIRT5 desuccinylates recombinant ACOX1 and EHHADH. (<b>a</b>) Recombinant human ACOX1a, ACOX1b, and EHHADH proteins were succinylated in vitro with 0.1 mM succinyl-CoA, and either mock-treated or treated with recombinant human SIRT5 and NAD+. The succinylation signal was detected by anti-succinylysine immunoblotting. The experiment was repeated with similar results. (<b>b</b>) Cartoon illustrating the EHHADH activity assay. Recombinant ACOX1 proteins were used to generate enoy-CoA substrates for EHHADH in a coupled assay. EHHADH activity was detected as the reduction of NAD+ to NADH. (<b>c</b>,<b>d</b>) Succinylated EHHADH protein was incubated with either inactive mutant SIRT5 (H158Y) or wildtype SIRT5. The resulting proteins were (<b>c</b>) immunoblotted for succinylation levels and (<b>d</b>) used for the activity assay illustrated in panel B. (<b>e</b>,<b>f</b>) Recombinant human ACOX1a and ACOX1b were succinylated and then incubated with either inactive mutant SIRT5 (H158Y) or wildtype SIRT5. The resulting proteins were (<b>e</b>) immunoblotted for succinylation levels and (<b>f</b>) used for a fluorometric ACOX1 activity assay that detects the formation of H<sub>2</sub>O<sub>2</sub>. (<b>g</b>) ACOX1b, which showed reduced activity after treatment with SIRT5 (panel <b>f</b>), was succinylated with increasing concentrations of succinyl-CoA and tested for activity with two monocarboxylic substrates, C12-CoA and C16-CoA, in triplicate. Data are plotted as means and standard deviations. * <span class="html-italic">p</span> &lt; 0.05 by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">
21 pages, 6640 KiB  
Article
Combining Network Pharmacology and Transcriptomic Strategies to Explore the Pharmacological Mechanism of Total Ginsenoside Ginseng Root and Its Impact on Antidepressant Effects
by Weijia Chen, Pengli Guo, Lili Su, Xiangjuan Guo, Meiling Shi, Jianan Geng, Ying Zong, Yan Zhao, Rui Du and Zhongmei He
Int. J. Mol. Sci. 2024, 25(23), 12606; https://doi.org/10.3390/ijms252312606 - 24 Nov 2024
Viewed by 500
Abstract
Depression is one of the most common neurological diseases, which imposes a substantial social and economic burden on modern society. The purpose of this study was to explore the mechanism of total ginsenoside ginseng root (TGGR) in the treatment of depression through a [...] Read more.
Depression is one of the most common neurological diseases, which imposes a substantial social and economic burden on modern society. The purpose of this study was to explore the mechanism of total ginsenoside ginseng root (TGGR) in the treatment of depression through a comprehensive strategy combining network pharmacology, transcriptomics, and in vivo experimental validation. The Traditional Chinese Medicine Systematic Pharmacology (TCMSP) database and literature were used to collect the main components and targets of TGGR. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were applied to explore the underlying mechanisms. In addition, the chronic unpredictable mild stress (CUMS)-induced C57BL/6 mouse model was used to evaluate the antidepressant activity of TGGR. The results showed that TGGR improved depression-like behavior in mice and increased the decrease in serum 5-hydroxytryptamine (5-HT) and brain-derived neurotrophic factor (BDNF) levels caused by CUMS. Combined network pharmacology and transcriptomic analysis showed that the AMP-activated kinase (AMPK) signaling pathway mainly enriched the core target. Immunohistochemistry, Western blotting, and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used to confirm whether TGGR exerts antidepressant effects by regulating this pathway. The results showed that TGGR has a regulatory impact on related proteins in the AMPK pathway, and the regulatory effect of TGGR on proteins was inhibited after the administration of related pathway inhibitors. In summary, total ginsenosides may regulate the AMPK signaling pathway and activate the sirtuin 1 (SIRT1) peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) pathway to have therapeutic effects on depression. Full article
(This article belongs to the Special Issue Pathophysiology and Pharmacology in Psychiatry)
Show Figures

Figure 1

Figure 1
<p>Potential targets of TGGR antidepressants and enrichment analyses. (<b>A</b>) Ginsenoside and depression target Venn plots. (<b>B</b>) Construction of the intersection target PPI network map of ginsenoside. (<b>C</b>,<b>D</b>) Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses.</p>
Full article ">Figure 2
<p>(<b>A</b>) Effects of TGGR on body weight of mice. (<b>B</b>) The effect of TGGR on sucrose preference in mice. (<b>C</b>,<b>D</b>) Effects of TGGR administration on resting time of FST and TST mice. (<b>E</b>) Heat map of MWM mouse activity. (<b>F</b>) The residence time of mice in the MWM target quadrant. (<b>G</b>) The levels of serum 5-HT and MDA, determined by ELISA. (<b>H</b>) Apoptosis of hippocampal neurons in each group (Nissl staining).The second picture for each group is a larger picture inside the red dashed box of the first picture. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 were significantly different from the control group, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from model group.</p>
Full article ">Figure 3
<p>(<b>A</b>) DEGs crossover results in the control group, CUMS group, and TGGR group. (<b>B</b>,<b>C</b>) DEG volcanic distribution map. Blue dots indicate downregulated mRNA; red dots indicate upregulated mRNA. (<b>D</b>) Heat map analysis of low- and high-expression DEG. (<b>E</b>) GO enrichment analysis was performed on DEGs’ cellular components, biological processes, and molecular functions. The first 10 items are shown in the figure. (<b>F</b>) The top 10 pathways of DEGs after performing KEGG enrichment.</p>
Full article ">Figure 4
<p>The key targets and pathways of TGGR in the prevention and treatment of depression and the preliminary validation of this target and pathway of action, analyzed with a combination of network pharmacology and transcriptomics. (<b>A</b>) Cross-targets of TGGR and depression in network pharmacology and cross-targets of transcriptomic differential genes. (<b>B</b>) Cross-target GO enrichment analysis. (<b>C</b>) The top 10 KEGG enrichment pathways for cross-targets. (<b>D</b>,<b>E</b>) Expression of SIRT1 and PGC-1α in mouse hippocampus (immunohistochemical staining). (<b>F</b>) Western blot analysis of AMPK, SIRT1, and PGC-1α in the hippocampus of mice in each group. Protein expression was normalized to β-actin for quantitative analysis, and its value expressed as an average (<b>G</b>). Normalization of the data to β-actin. Values are expressed as average ± SD. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from model group.</p>
Full article ">Figure 5
<p>(<b>A</b>) Changes in body weight of mice. (<b>B</b>) Changes in sucrose preference of mice. (<b>C</b>,<b>D</b>) Effects of TGGR administration on resting time of TST and FST mice. (<b>E</b>) Heat maps of MWM mouse activity. (<b>F</b>) Residence time of mice in the MWM target quadrant. (<b>G</b>,<b>H</b>) ELISA-detected serum 5-HT and MDA levels. (<b>I</b>) Apoptosis of hippocampal neurons in each group (Nissl staining). ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 was significantly different from model group, △ <span class="html-italic">p</span> &lt; 0.05 was significantly different from TGGR pair, ns &gt; 0.05 was not significantly different.</p>
Full article ">Figure 6
<p>(<b>A</b>) Ultrastructure of hippocampal neurons and mitochondria of mice in each group. (<b>B</b>) ATP content in mouse hippocampus. (<b>C</b>) ROS levels in mouse brain tissue. (<b>D</b>) Expression of SIRT1 and PGC-1α in mouse hippocampus (immunohistochemical staining). (<b>E</b>) Western blot analysis of SIRT1 and PGC-1α in the hippocampus of mice in each group. (<b>F</b>) Protein expression normalized to β-actin for quantitative analysis, and its value expressed as an average. (<b>G</b>) SIRT1 and PGC-1α mRNA levels detected by RT-qPCR. Data are normalized to β-actin. Values are expressed as average ± SD. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 was substantially different from model group, and <sup>△</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>△△</sup> <span class="html-italic">p</span> &lt; 0.01 were substantially different from TGGR group.</p>
Full article ">
Back to TopTop