[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,618)

Search Parameters:
Keywords = endothelial dysfunction

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1641 KiB  
Article
Expression of Myeloperoxidase in Patient-Derived Endothelial Colony-Forming Cells—Associations with Coronary Artery Disease and Mitochondrial Function
by Weiqian Eugene Lee, Elijah Genetzakis, Giannie Barsha, Joshua Vescovi, Carmen Mifsud, Stephen T. Vernon, Tung Viet Nguyen, Michael P. Gray, Stuart M. Grieve and Gemma A. Figtree
Biomolecules 2024, 14(10), 1308; https://doi.org/10.3390/biom14101308 (registering DOI) - 16 Oct 2024
Abstract
Background and Aims: Myeloperoxidase (MPO) plays a critical role in the innate immune response and has been suggested to be a surrogate marker of oxidative stress and inflammation, with elevated levels implicated in cardiovascular diseases, such as atherosclerosis and heart failure, as well [...] Read more.
Background and Aims: Myeloperoxidase (MPO) plays a critical role in the innate immune response and has been suggested to be a surrogate marker of oxidative stress and inflammation, with elevated levels implicated in cardiovascular diseases, such as atherosclerosis and heart failure, as well as in conditions like rheumatoid arthritis and cancer. While MPO is well-known in leukocytes, its expression and function in human endothelial cells remain unclear. This study investigates MPO expression in patient-derived endothelial colony-forming cells (ECFCs) and its potential association with CAD and mitochondrial function. Methods: ECFCs were cultured from the peripheral blood of 93 BioHEART-CT patients. MPO expression and associated functions were examined using qRT-PCR, immunochemistry, flow cytometry, and MPO activity assays. CAD presence was defined using CT coronary angiography (CACS > 0). Results: We report MPO presence in patient-derived ECFCs for the first time. MPO protein expression occurred in 70.7% of samples (n = 41) which had nuclear co-localisation, an atypical observation given its conventional localisation in the granules of neutrophils and monocytes. This suggests potential alternative roles for MPO in nuclear processes. MPO mRNA expression was detected in 66.23% of samples (n = 77). CAD patients had a lower proportion of MPO-positive ECFCs compared to non-CAD controls (57.45% vs. 80%, p = 0.04), a difference that persisted in the statin-naïve sub-cohort (53.85% vs. 84.62%, p = 0.02). Non-CAD patients with MPO expression showed upregulated mitochondrial-antioxidant genes (AIFM2, TXNRD1, CAT, PRDX3, PRDX6). In contrast, CAD patients with MPO gene expression had heightened mROS production and mitochondrial mass and decreased mitochondrial function compared to that of CAD patients without MPO gene expression. Conclusions: MPO is present in the nucleus of ECFCs. In non-CAD ECFCs, MPO expression is linked to upregulated mitochondrial-antioxidant genes, whereas in CAD ECFCs, it is associated with greater mitochondrial dysfunction. Full article
Show Figures

Figure 1

Figure 1
<p>MPO protein expression in patient-derived ECFCs. (<b>A</b>) Representative Western blot of MPO protein expression in corresponding patient-derived ECFCs using anti-MPO antibody and anti-β-actin for loading control. (<b>B</b>) MPO protein is co-localised to the nuclei of patient-derived ECFCs. Representative immunocytochemistry images of MPO protein expression co-localised to the nucleus of patient-derived ECFCs, showing nuclei (blue) and MPO granules (red) within the cell (20× magnification). The scale bar represents 100 μM. (<b>C</b>) Representative Western blot of subcellular expression of MPO in patient-derived ECFCs at the soluble nuclear and chromatin-bound nuclear subfractions. At least three biological replicates were used. Original images can be found in <a href="#app1-biomolecules-14-01308" class="html-app">Supplementary Materials</a> file.</p>
Full article ">Figure 2
<p>CAD patients were less likely to express MPO gene, as identified by qRT-PCR. Stacked bar plots showing the association between proportion in MPO gene expression and the presence of CAD in (<b>A</b>) all patients (<span class="html-italic">n</span> = 77; No CAD = 30, CAD = 47), (<b>B</b>) male patients (<span class="html-italic">n</span> = 40; No CAD = 14, CAD = 26) and (<b>C</b>) female patients (<span class="html-italic">n</span> = 36; No CAD = 16, CAD = 20). (<b>D</b>) Statin-naïve patients (<span class="html-italic">n</span> = 54; No CAD = 27, CAD = 27), (<b>E</b>) statin-naïve male patients (<span class="html-italic">n</span> = 27; No CAD = 12, CAD = 15) and (<b>F</b>) statin-naïve female patients (<span class="html-italic">n</span> = 26; No CAD = 14, CAD = 12). Statistical association was analysed using Pearson’s chi-square test (categorial variables). Categorical measurements are shown as percentages. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2 Cont.
<p>CAD patients were less likely to express MPO gene, as identified by qRT-PCR. Stacked bar plots showing the association between proportion in MPO gene expression and the presence of CAD in (<b>A</b>) all patients (<span class="html-italic">n</span> = 77; No CAD = 30, CAD = 47), (<b>B</b>) male patients (<span class="html-italic">n</span> = 40; No CAD = 14, CAD = 26) and (<b>C</b>) female patients (<span class="html-italic">n</span> = 36; No CAD = 16, CAD = 20). (<b>D</b>) Statin-naïve patients (<span class="html-italic">n</span> = 54; No CAD = 27, CAD = 27), (<b>E</b>) statin-naïve male patients (<span class="html-italic">n</span> = 27; No CAD = 12, CAD = 15) and (<b>F</b>) statin-naïve female patients (<span class="html-italic">n</span> = 26; No CAD = 14, CAD = 12). Statistical association was analysed using Pearson’s chi-square test (categorial variables). Categorical measurements are shown as percentages. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>MPO gene expression is associated with dysregulated mitochondrial function and dynamics. (<b>A</b>–<b>C</b>) CAD ECFCs with MPO gene expression at baseline had increased (<b>A</b>) mROS production and (<b>B</b>) mitochondrial mass and decreased (<b>C</b>) mitochondrial function. N = 8; no MPO gene expression: N = 3, MPO gene expression: N = 5. Data are represented as mean ± S.E.M. Welch’s <span class="html-italic">t</span>-test was performed.</p>
Full article ">Figure 4
<p>Antioxidant genes were upregulated with MPO gene expression in non-CAD patients. Relative fold gene expression was evaluated in ECFCs without CAD at baseline in (<b>A</b>) <span class="html-italic">AIFM2</span>, (<b>B</b>) <span class="html-italic">TXNRD1</span>, (<b>C</b>) <span class="html-italic">CAT</span>, (<b>D</b>) <span class="html-italic">PRDX3</span> and (<b>E</b>) <span class="html-italic">PRDX6</span>. Each sample was performed in triplicate. N = 30; no MPO gene expression: N = 6, MPO gene expression: N = 24. Data are represented as mean ± S.E.M. Student’s <span class="html-italic">t</span>-test was performed.</p>
Full article ">
14 pages, 5396 KiB  
Article
Impaired Endothelium-Dependent Vasodilation and Increased Levels of Soluble Fms-like Tyrosine Kinase-1 Induced by Reduced Uterine Perfusion Pressure in Pregnant Rats: Evidence of Protective Effects with Sodium Nitrite Treatment in Preeclampsia
by Maria Luiza Santos Da Silva, Sáskia Estela Biasotti Gomes, Laisla Zanetoni Martins, Serginara David Rodrigues, Cristal de Jesus Toghi and Carlos Alan Dias-Junior
Int. J. Mol. Sci. 2024, 25(20), 11051; https://doi.org/10.3390/ijms252011051 (registering DOI) - 15 Oct 2024
Viewed by 202
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy and is associated with increases in soluble fms-like tyrosine kinase-1 (sFlt-1) and reductions in nitric oxide (NO) levels. Placental ischemia and hypoxia are hypothesized as initial pathophysiological events of PE. Nitrite (NO metabolite) may be [...] Read more.
Preeclampsia (PE) is a hypertensive disorder of pregnancy and is associated with increases in soluble fms-like tyrosine kinase-1 (sFlt-1) and reductions in nitric oxide (NO) levels. Placental ischemia and hypoxia are hypothesized as initial pathophysiological events of PE. Nitrite (NO metabolite) may be recycled back to NO in ischemic and hypoxic tissues. Therefore, this study examined the sodium nitrite effects in an experimental model of PE. Pregnant rats received saline (Preg group) or sodium nitrite (Preg + Na-Nitrite group). Pregnant rats submitted to the placental ischemia received saline (RUPP group) or sodium nitrite (RUPP + Na-Nitrite group). Blood pressure, placental and fetal weights, and the number of pups were recorded. Plasma levels of NO metabolites and sFlt-1 were also determined. Vascular and endothelial functions were also measured. Blood pressure, placental and fetal weights, the number of pups, NO metabolites, sFlt-1 levels, vascular contraction, and endothelium-dependent vasodilation in the RUPP + Na-Nitrite rats were brought to levels comparable to those in Preg rats. In conclusion, sodium nitrite may counteract the reductions in NO and increases in sFlt-1 levels induced by the placental ischemia model of PE, thus suggesting that increased blood pressure and vascular and endothelial dysfunctions may be attenuated by sodium nitrite-derived NO. Full article
(This article belongs to the Special Issue Molecular Pathogenesis and Treatment of Pregnancy Complications)
Show Figures

Figure 1

Figure 1
<p>Effects of sodium nitrite on systolic blood pressure (SBP) measured on pregnancy day 21 in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 animals per group). Values represent the mean ± SEM. <sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the Preg, RUPP, and RUPP+Na-Nitrite groups; * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the RUPP group.</p>
Full article ">Figure 2
<p>Effects of sodium nitrite on placental weight recorded in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 mothers per group). Values represent the mean ± SEM. <sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the Preg and RUPP groups; * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the RUPP group.</p>
Full article ">Figure 3
<p>Effects of sodium nitrite on fetal weight recorded in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 mothers per group). Values represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg group; <sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the Preg and Preg+Na-Nitrite groups; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the RUPP group.</p>
Full article ">Figure 4
<p>Effects of sodium nitrite on litter size (total number of pups) noted in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 mothers per group). Values represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg group; <sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the Preg and Preg+Na-Nitrite groups; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the RUPP group.</p>
Full article ">Figure 5
<p>Effects of sodium nitrite on plasmatic NO metabolites (nitrite+nitrate levels) in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 animals per group). Values represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg group; <sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the Preg and Preg+Na-Nitrite groups; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the RUPP group.</p>
Full article ">Figure 6
<p>Effects of sodium nitrite on circulating sFlt-1 levels in plasma from the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 animals per group). Values represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg, Preg+Na-Nitrite, and RUPP+Na-Nitrite groups.</p>
Full article ">Figure 7
<p>Effects of sodium nitrite on the vascular reactivity of the thoracic aorta rings in the contraction induced by phenylephrine with (<span class="html-italic">n</span> = 2 rings per animal, (<b>A</b>)) or without (<span class="html-italic">n</span> = 2 rings per animal, (<b>B</b>)) endothelium in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 animals per group). Values represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg, Preg+Na-Nitrite, and RUPP+Na-Nitrite groups; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 for the RUPP and RUPP+Na-Nitrite vs. the Preg and Preg+Na-Nitrite groups.</p>
Full article ">Figure 8
<p>Effects of sodium nitrite on the vascular reactivity of acetylcholine-induced relaxations in the endothelium-intact thoracic aortas (<span class="html-italic">n</span> = 2 rings per animal, (<b>A</b>)) or endothelium-denuded thoracic aortas (<span class="html-italic">n</span> = 2 rings per animal, (<b>B</b>)) or endothelium intact thoracic aortas pre-incubated with L-NAME (<span class="html-italic">n</span> = 2 rings per animal, (<b>C</b>)) in the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 animals per group). Values represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 vs. the Preg, Preg+Na-Nitrite, and RUPP+Na-Nitrite groups; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the Preg group.</p>
Full article ">Figure 9
<p>Effects of sodium nitrite on the vascular reactivity of sodium nitroprusside-induced relaxations in the endothelium-intact thoracic aortas (<span class="html-italic">n</span> = 2 rings per animal, (<b>A</b>)) or endothelium-denuded thoracic aortas (<span class="html-italic">n</span> = 2 rings per animal, (<b>B</b>)) from the Preg, Preg+Na-Nitrite, RUPP, and RUPP+Na-Nitrite groups (<span class="html-italic">n</span> = 8–10 animals per group). Values represent the mean ± SEM.</p>
Full article ">
12 pages, 947 KiB  
Article
Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR), Growth Differentiation Factor-15 (GDF-15), and Soluble C5b-9 (sC5b-9) Levels Are Significantly Associated with Endothelial Injury Indices in CAR-T Cell Recipients
by Eleni Gavriilaki, Christos Demosthenous, Paschalis Evangelidis, Zoi Bousiou, Ioannis Batsis, Anna Vardi, Despina Mallouri, Eudoxia-Evaggelia Koravou, Nikolaos Spyridis, Alkistis Panteliadou, Georgios Karavalakis, Marianna Masmanidou, Tasoula Touloumenidou, Apostolia Papalexandri, Christos Poziopoulos, Evangelia Yannaki, Ioanna Sakellari, Marianna Politou and Ioannis Papassotiriou
Int. J. Mol. Sci. 2024, 25(20), 11028; https://doi.org/10.3390/ijms252011028 - 14 Oct 2024
Viewed by 428
Abstract
Endothelial injury indices, such as Endothelial Activation and Stress Index (EASIX), modified EASIX (m-EASIX), and simplified EASIX (s-EASIX) scores, have been previously associated with chimeric antigen receptor-T (CAR-T) cell immunotherapy complications. Soluble urokinase-type plasminogen activator receptor (suPAR), growth differentiation factor-15 (GDF-15), and soluble [...] Read more.
Endothelial injury indices, such as Endothelial Activation and Stress Index (EASIX), modified EASIX (m-EASIX), and simplified EASIX (s-EASIX) scores, have been previously associated with chimeric antigen receptor-T (CAR-T) cell immunotherapy complications. Soluble urokinase-type plasminogen activator receptor (suPAR), growth differentiation factor-15 (GDF-15), and soluble C5b-9 (sC5b-9) have been described as markers of endothelial injury post-hematopoietic stem cell transplantation. In the current study, we examined whether suPAR, GDF-15, and sC5b-9 levels were associated with endothelial injury indices in adult CAR-T cell recipients. The levels of these markers were measured in patients before CAR-T cell infusion and in healthy individuals with immunoenzymatic methods. We studied 45 CAR-T cell recipients and 20 healthy individuals as the control group. SuPAR, GDF-15, and sC5b-9 levels were significantly higher in the patients’ group compared to the healthy control group (p < 0.001, in all comparisons). SuPAR levels at baseline were associated with the m-EASIX scores calculated at the same time point (p = 0.020), while suPAR and GDF-15 concentrations were correlated with EASIX scores at day 14 post-infusion (p < 0.001 in both comparisons). Moreover, sC5b-9 levels were correlated with the s-EASIX scores at infusion (p = 0.008) and the EASIX scores at day 14 (p = 0.005). In our study, sC5b9, suPAR, and GDF-15 levels were found to reflect endothelial injury in CAR-T cell recipients. Full article
(This article belongs to the Special Issue Novel Insights into Monoclonal Antibodies in Disease)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) sC5b9 levels show a significant correlation with sEASIX0 (<span class="html-italic">p</span> = 0.008) and (<b>B</b>) EASIX14 (<span class="html-italic">p</span> = 0.005) scores. sC5b9 = soluble C5b-9 and s-EASIX0 = simplified Endothelial Activation and Stress Index scores were calculated at the day of CAR-T cell infusion; EASIXDAY14 = Endothelial Activation and Stress Index scores were calculated 14 days post-CAR-T cell infusion.</p>
Full article ">Figure 2
<p>(<b>A</b>) s-EASIX0 score over the median value (1.9, range: 0.74–49.3) was associated with poor OS (<span class="html-italic">p</span> = 0.027); (<b>B</b>) s-EASIX14 score over the median value (3, range: 0.6–74.6) was also associated with poor OS (<span class="html-italic">p</span> = 0.004). Green lines: s-EASIX score below the median; blue lines: s-EASIX score over the median value. S-EASIX0 = simplified Endothelial Activation and Stress Index calculated at the day of the infusion; S-EASIX14 = simplified Endothelial Activation and Stress Index calculated 14 days post-infusion; OS = overall survival.</p>
Full article ">Figure 3
<p>Methodology used in our study. Samples for sC5b-9, suPAR, and GDF-15 measurements were obtained from CAR-T cell recipients before their admission to the cellular therapy unit, and at the same time point, EASIX, mEASIX, and s-EASIX scores were calculated. EASIX and s-EASIX scores were also calculated on the day of CAR-T cell product infusion and 14 days post-infusion. During the post-infusion period, patients were closely monitored for therapy-related toxicities. The minimum follow-up period was 1 month. LDC = lymphodepleting chemotherapy; EASIX = Endothelial Activation and Stress Index; m-EASIX = modified Endothelial Activation and Stress Index; s-EASIX = simplified Endothelial Activation and Stress Index; sC5b-9 = soluble C5b-9; suPAR = soluble urokinase-type plasminogen activator receptor; GDF-15 = growth differentiation factor-15; CRS = cytokine release syndrome; ICANS = immune effector cell-associated neurotoxicity syndrome.</p>
Full article ">
18 pages, 7639 KiB  
Article
Therapeutic Effects of Proanthocyanidins on Diabetic Erectile Dysfunction in Rats
by Xiaoyan Zeng, Lanlan Li and Li Tong
Int. J. Mol. Sci. 2024, 25(20), 11004; https://doi.org/10.3390/ijms252011004 - 13 Oct 2024
Viewed by 368
Abstract
The rising occurrence of erectile dysfunction related to diabetes mellitus (DMED) has led to the creation of new medications. Proanthocyanidins (PROs) is a potential agent for DMED. In this study, the DMED rat model was established using streptozotocin (STZ) and erectile function was [...] Read more.
The rising occurrence of erectile dysfunction related to diabetes mellitus (DMED) has led to the creation of new medications. Proanthocyanidins (PROs) is a potential agent for DMED. In this study, the DMED rat model was established using streptozotocin (STZ) and erectile function was assessed using apomorphine (APO) in rats. Following this, the rats were subjected to oral treatment with PRO. Then, we evaluated the influence of PROs on DMED rats. The findings suggest that PROs significantly enhance erectile function in DMED rats. PROs modulated glucose and lipid metabolism in DMED rats by decreasing blood glucose and lipid levels while increasing liver glycogen and serum insulin levels. Furthermore, PROs enhanced vascular endothelial function in DMED rats by augmenting nitric oxide (NO) levels and reducing the levels of endothelin-1 (ET-1) and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1). Additionally, PROs have been shown to elevate testosterone (T) levels, mitigate pathological testicular damage, and enhance sperm concentration and survival rates. Finally, the core targets were screened using network pharmacology, followed by validation through molecular docking, enzyme-linked immunoassay (ELISA), and real-time PCR methodologies. Our findings imply that PROs may treat DMED by elevating AKT1 levels while concurrently diminishing CASP3 levels, thereby effectively regulating the PI3K-Akt signaling pathway. Overall, these results support using PROs as a potential candidate for the treatment of DMED. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of PROs on body weight, glucose, and lipid metabolism in DMED rats. (<b>A</b>) Changes in body weight of rats after treatment. (<b>B</b>) Changes in blood glucose levels of rats after treatment. (<b>C</b>) Serum insulin levels of rats in each group. (<b>D</b>) Liver glycogen levels of rats in each group. (<b>E</b>) Blood lipid (TC, TG, HDL, LDL) levels of rats in each group. Sixty rats were randomly allocated into six groups: the control group (CON), model group (MOD), PROs high-dose group (PRO-H), PROs medium-dose group (PRO-M), PROs low-dose group (PRO-L), and tadalafil (TAD) group. The data are expressed as the means ± SDs (<span class="html-italic">n</span> = 10). *** <span class="html-italic">p</span> &lt; 0.001 vs. the CON group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the MOD group. TC, total cholesterol; TG, triglyceride; HDL, high-density lipoprotein; LDL, low-density lipoprotein.</p>
Full article ">Figure 2
<p>Erectile function assessment of rats in each group after treatment. ICP, MAP, and ICP/MAP were measured 4 weeks after administration. (<b>A</b>) ICP of rats in each group. (<b>B</b>) MAP of rats in each group. (<b>C</b>) ICP/MAP ratios of rats in each group. The data are expressed as the means ± SDs (<span class="html-italic">n</span> = 10). *** <span class="html-italic">p</span> &lt; 0.001 vs. the CON group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the MOD group. ICP, intracavernosal pressure; MAP, mean arterial pressure.</p>
Full article ">Figure 3
<p>Effects of PROs on sperm quality in DMED rats. The epididymis of rats was collected, and subsequent analyses were conducted to determine sperm concentration and survival rates. (<b>A</b>) Sperm concentration of rats in each group. (<b>B</b>) Sperm survival rates of rats in each group (<span class="html-italic">n</span> = 10). The data are expressed as the means ± SDs. *** <span class="html-italic">p</span> &lt; 0.001 vs. the CON group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the MOD group.</p>
Full article ">Figure 4
<p>Effects of PROs on endothelial function in DMED rats. (<b>A</b>) NO levels of rats in each group. (<b>B</b>) ET-1 levels of rats in each group. (<b>C</b>) LOX-1 levels of rats in each group. The data are expressed as the means ± SDs (<span class="html-italic">n</span> = 10). *** <span class="html-italic">p</span> &lt; 0.001 vs. the CON group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the MOD group. ET-1, of rats in each group; LOX-1, lectin-like oxidized low-density lipoprotein receptor-1.</p>
Full article ">Figure 5
<p>Sex hormone levels of rats in each group after treatment. (<b>A</b>) FSH levels of rats in each group. (<b>B</b>) LH levels of rats in each group. (<b>C</b>) T levels of rats in each group. The data are expressed as the means ± SDs (<span class="html-italic">n</span> = 10). *** <span class="html-italic">p</span> &lt; 0.001 vs. the CON group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. the MOD group. FSH, follicle-stimulating hormone; LH, luteinizing hormone; T, testosterone.</p>
Full article ">Figure 6
<p>Hematoxylin and eosin (HE) staining of rat testis. Rats in the control group exhibited an intact testicular cell structure, tightly organized spermatogonia, and the presence of spermatozoa within the lumen without any apparent pathological alterations. Rats in the model group displayed dislodged spermatogonia, necrosis of spermatocytes, and a significant reduction in the number of spermatozoa, and distinct pathological changes. Conversely, the testicular tissues of the PROs and TAD treatment groups tended normalization. The green arrow indicates spermatocyte necrosis and nuclei consolidated or fragmented. The red arrow indicates eosinophilic tissue fluid exudation. The blue arrow signifies a sparsely arranged varicocele with widened gaps. The black arrow indicates interstitial capillary stasis.</p>
Full article ">Figure 7
<p>Network pharmacology reveals potential targets and pathways for PROs treatment of DMED. (<b>A</b>) Wayne analysis of PROs and DMED intersection targets. (<b>B</b>) PPI network diagram of intersection target of PROs and DMED. The size and coloration of the circles signify the extent of correlation with other targets; specifically, circles that are larger and redder indicate a stronger correlation. (<b>C</b>) GO function enrichment analysis results. (<b>D</b>) KEGG pathway enrichment analysis results.</p>
Full article ">Figure 8
<p>Molecular docking results. (<b>A</b>–<b>E</b>) Schematic diagram of the binding mode and details of the binding pose of PROs to AKT1, ALB, EGFR, ESR1, and CASP3. (<b>F</b>) Molecular docking results heat map of PROs and key targets.</p>
Full article ">Figure 9
<p>Effects of PROs on core target levels and the PI3K-Akt Pathway in DMED rats. (<b>A</b>) Evaluation of AKT1, ALB, CASP3, ESR1, and EGFR expression in the penile corpus cavernosum of rats (<span class="html-italic">n</span> = 10). (<b>B</b>) Relative mRNA expression levels of AKT and CASP3 (<span class="html-italic">n</span> = 5). (<b>C</b>) pAKT levels in the penile corpus cavernosum of rats (<span class="html-italic">n</span> = 10). The data are expressed as the means ± SDs. *** <span class="html-italic">p</span> &lt; 0.001 vs. the CON group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, vs. the MOD group.</p>
Full article ">
43 pages, 4206 KiB  
Review
Advancements in Polymer Biomaterials as Scaffolds for Corneal Endothelium Tissue Engineering
by Kevin Y. Wu, Myriam Belaiche, Ying Wen, Mazen Y. Choulakian and Simon D. Tran
Polymers 2024, 16(20), 2882; https://doi.org/10.3390/polym16202882 (registering DOI) - 12 Oct 2024
Viewed by 600
Abstract
Corneal endothelial dysfunction is a leading cause of vision loss globally, frequently requiring corneal transplantation. However, the limited availability of donor tissues, particularly in developing countries, has spurred on the exploration of tissue engineering strategies, with a focus on polymer biomaterials as scaffolds [...] Read more.
Corneal endothelial dysfunction is a leading cause of vision loss globally, frequently requiring corneal transplantation. However, the limited availability of donor tissues, particularly in developing countries, has spurred on the exploration of tissue engineering strategies, with a focus on polymer biomaterials as scaffolds for corneal endotlhelium regeneration. This review provides a comprehensive overview of the advancements in polymer biomaterials, focusing on their role in supporting the growth, differentiation, and functional maintenance of human corneal endothelial cells (CECs). Key properties of scaffold materials, including optical clarity, biocompatibility, biodegradability, mechanical stability, permeability, and surface wettability, are discussed in detail. The review also explores the latest innovations in micro- and nano-topological morphologies, fabrication techniques such as electrospinning and 3D/4D bioprinting, and the integration of drug delivery systems into scaffolds. Despite significant progress, challenges remain in translating these technologies to clinical applications. Future directions for research are highlighted, including the need for improved biomaterial combinations, a deeper understanding of CEC biology, and the development of scalable manufacturing processes. This review aims to serve as a resource for researchers and clinician–scientists seeking to advance the field of corneal endothelium tissue engineering. Full article
(This article belongs to the Section Biobased and Biodegradable Polymers)
Show Figures

Figure 1

Figure 1
<p>Histology of the human cornea. Created with BioRender.com.</p>
Full article ">Figure 2
<p>Penetrating keratoplasty (PK), Descemet’s stripping automated keratoplasty (DSAEK), and Descemet’s membrane endothelial keratoplasty (DMEK). Created with BioRender.com.</p>
Full article ">Figure 3
<p>Corneal endothelium tissue engineering. Created with BioRender.com.</p>
Full article ">Figure 4
<p>Schematic of the ECM structure of Descemet’s membrane [<a href="#B25-polymers-16-02882" class="html-bibr">25</a>]. Reprinted with permission from ref. [<a href="#B25-polymers-16-02882" class="html-bibr">25</a>]. Copyright 2021 Elsevier.</p>
Full article ">Figure 5
<p>Topological features with different shapes and sizes that can be used for surface patterning [<a href="#B113-polymers-16-02882" class="html-bibr">113</a>]. Reprinted with permission from ref. [<a href="#B113-polymers-16-02882" class="html-bibr">113</a>]. Copyright 2016 Elsevier.</p>
Full article ">Figure 6
<p>Enhanced protein adsorption and cell adhesion on micropatterned hydrogels. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 7
<p>Length scale bar illustrating featured resolutions of various biofabrication techniques in comparison with geometric sizes of representative cells and tissues [<a href="#B154-polymers-16-02882" class="html-bibr">154</a>]. Reprinted from ref. [<a href="#B154-polymers-16-02882" class="html-bibr">154</a>], licensed under CC BY 4.0 [<a href="https://creativecommons.org/licenses/by/4.0/" target="_blank">https://creativecommons.org/licenses/by/4.0/</a>]. Accessed on 3 August 2024. “ES” stands for electrospinning and “LEP” stands for low-voltage electrospinning patterning.</p>
Full article ">
14 pages, 1995 KiB  
Article
Cardioprotection by Preconditioning with Intralipid Is Sustained in a Model of Endothelial Dysfunction for Isolated-Perfused Hearts
by Martin Stroethoff, Natalie Schneider, Lea Sung, Jan Wübbolt, André Heinen and Annika Raupach
Int. J. Mol. Sci. 2024, 25(20), 10975; https://doi.org/10.3390/ijms252010975 - 12 Oct 2024
Viewed by 370
Abstract
Endothelial dysfunction (ED) is closely associated with most cardiovascular diseases. Experimental models are needed to analyze the potential impact of ED on cardioprotection in constant pressure Langendorff systems (CPLS). One cardioprotective strategy against ischemia/reperfusion injury (I/RI) is conditioning with the lipid emulsion Intralipid [...] Read more.
Endothelial dysfunction (ED) is closely associated with most cardiovascular diseases. Experimental models are needed to analyze the potential impact of ED on cardioprotection in constant pressure Langendorff systems (CPLS). One cardioprotective strategy against ischemia/reperfusion injury (I/RI) is conditioning with the lipid emulsion Intralipid (IL). Whether ED modulates the cardioprotective effect of IL remains unknown. The aim of the study was to transfer a protocol using a constant flow Langendorff system for the induction of ED into a CPLS, without the loss of smooth muscle cell functionality, and to analyze the cardioprotective effect of IL against I/RI under ED. In isolated hearts of male Wistar rats, ED was induced by 10 min perfusion of a Krebs–Henseleit buffer containing 60 mM KCl (K+), and the vasodilatory response to the vasodilators histamine (endothelial-dependent) and sodium–nitroprusside (SNP, endothelial-independent) was measured. A CPLS was employed to determine cardioprotection of pre- or postconditioning with 1% IL against I/RI. The constant flow perfusion of K+ reduced endothelial response to histamine but not to SNP, indicating reduced vasodilatory functionality of endothelial cells but not smooth muscle cells. Preconditioning with IL reduced infarct size and improved cardiac function while postconditioning with IL had no effect. The induction of ED neither influenced infarct size nor affected the cardioprotective effect by preconditioning with IL. This protocol allows for studies of cardioprotective strategies under ED in CLPS. The protection by preconditioning with IL seems to be mediated independently of a functional endothelium. Full article
Show Figures

Figure 1

Figure 1
<p>Difference of endothelial response (ΔER = ER<sub>1</sub> − ER<sub>2</sub>) before (ER<sub>1</sub>) and after (ER<sub>2</sub>) constant flow perfusion (10 min) with Krebs–Henseleit buffer (KHB) alone (white fill) or KHB containing 60 mM KCl (K+, striped green) to 800 nmol histamine (his, orange borders) or 1 µM sodium nitroprusside (SNP, pink borders). As a positive control for ED induction, a bolus of 1 s with 1% triton (grey fill) was used. Data are mean ± SD, <span class="html-italic">n</span> = 4 (SNP, triton), <span class="html-italic">n</span> = 7 (histamine). One-way ANOVA followed by Šidák’s multiple comparison test. *: <span class="html-italic">p</span> &lt; 0.05, ns: not significant (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 2
<p>Infarct sizes of hearts after ischemia/reperfusion with pre- or post-treatment for 10 min with 1% Intralipid (IL) or vehicle (Con). LV: left ventricle. Data are mean ± SD, <span class="html-italic">n</span> = 7. One-way ANOVA, Dunnett’s multiple comparison test, *: <span class="html-italic">p</span> &lt; 0.05, ns: not significant (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 3
<p>Infarct sizes of hearts after ischemia/reperfusion with post-treatment for 20 min with Intralipid (IL) or vehicle (Con). LV: left ventricle. Data are mean ± SD, <span class="html-italic">n</span> = 6. <span class="html-italic">t</span>-test; ns: not significant.</p>
Full article ">Figure 4
<p>Preconditioning with IL under endothelial dysfunction (ED). Hearts were treated for 10 min before ischemia with 1% Intralipid (IL; blue border) or vehicle (Con; black border). ED was induced by 10 min constant flow perfusion of Krebs–Henseleit buffer (KHB) containing 60 mM KCl (K+; green stripes). The other groups received normal KHB under constant flow conditions (white filling). LV: left ventricle. Data are mean ± SD, <span class="html-italic">n</span> = 7. Two-way ANOVA; * <span class="html-italic">p</span> &lt; 0.05 for effect by conditioning; ns = not significant for effect by ED induction and interaction (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 5
<p>Schematic drawing of the utilized Langendorff system. Krebs–Henseleit buffer (KHB) and KHB containing 1% Intralipid (IL) are perfused in a constant pressure mode (80 mmHg) in separate circuits. To induce endothelial dysfunction, KHB containing 60 mM KCl (K+) is perfused in a constant flow mode. To ensure equivalent control conditions for induction of ED, KHB is perfused in a constant flow mode. The individual modules were switched on or off as required by the respective questions during the experimental setups. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 6
<p>Experimental timeline for characterization of endothelial dysfunction. Dashed blue lines mark measuring points for coronary perfusion pressure (CPP). BL1/2: baseline at time point 1/2, P: after perfusion of vasodilator, His: histamine, T: triton, SNP: sodium–nitroprusside, KHB flow: constant flow perfusion with Krebs–Henseleit buffer, K+ flow: constant flow perfusion with KHB containing 60 mM KCl.</p>
Full article ">Figure 7
<p>Experimental protocol: conditioning with Intralipid (IL) using a constant pressure Langendorff system with the ability to switch into a constant flow mode for induction of endothelial dysfunction. (<b>a</b>) Pre- or postconditioning with IL for 10 min. (<b>b</b>) Postconditioning with IL for 20 min. (<b>c</b>) Preconditioning with IL under endothelial dysfunction induced by constant flow perfusion of Krebs–Henseleit buffer (KHB) containing 60 mM KCl (K+ flow) and under control condition with constant flow perfusion of KHB alone (KHB flow).</p>
Full article ">
22 pages, 6409 KiB  
Article
Intracellular Iron Deficiency and Abnormal Metabolism, Not Ferroptosis, Contributes to Homocysteine-Induced Vascular Endothelial Cell Death
by Wenting Shi, Jing Zhang, Wairong Zhao, Meiyan Yue, Jie Ma, Silu Zeng, Jingyi Tang, Yu Wang and Zhongyan Zhou
Biomedicines 2024, 12(10), 2301; https://doi.org/10.3390/biomedicines12102301 - 10 Oct 2024
Viewed by 331
Abstract
Background/Objectives: Homocysteine (Hcy) and iron are factors co-related with the progression of cardiovascular diseases. The vascular endothelium is an important barrier for physiological homeostasis, and its impairment initiates cardiovascular injury. However, the mechanism underlying Hcy-caused vascular endothelial cell injury and the participation of [...] Read more.
Background/Objectives: Homocysteine (Hcy) and iron are factors co-related with the progression of cardiovascular diseases. The vascular endothelium is an important barrier for physiological homeostasis, and its impairment initiates cardiovascular injury. However, the mechanism underlying Hcy-caused vascular endothelial cell injury and the participation of iron are not fully elucidated. This study aims to investigate the Hcy-induced vascular endothelial injury and iron metabolism dysfunction as well as the underlying molecular mechanism. Methods: Human umbilical vein endothelial cells (HUVECs) were employed as the experimental model to examine the Hcy-induced endothelial injury and its underlying mechanism via various biochemical assays. Results: Hcy suppressed the cell viability and proliferation and caused cell death in a concentration-dependent manner. Hcy induced cell cycle arrest, apoptosis, and autophagy as well as impairment of intracellular energy metabolism. Hcy disrupted the intracellular antioxidant system and mitochondrial function by increasing intracellular ROS, MDA and mitochondrial content, and decreasing the SOD activity and mitochondrial membrane potential. Hcy significantly reduced the GSH-Px activity along with the accumulation of intracellular GSH in a concentration-dependent manner. Ferroptosis inhibitors, Ferrostatin-1 (Fer-1), and Deferoxamine (DFO) significantly decreased the Hcy-caused cytotoxicity accompanied by a reduction in dysregulated mitochondria content, but only DFO ameliorated the elevation of intracellular ROS, and neither Fer-1 nor DFO affected the Hcy-caused reduction in intracellular ATP. In addition, Hcy decreased the intracellular concentration of iron, and supplementing Hcy with various concentrations of Fe3+ increased the cell viability and decreased the LDH release in a concentration-dependent manner. Hcy dramatically decreased the mRNA expression level of transferrin receptor while increasing the mRNA expression levels of transferrin, ferritin light chain, ferritin heavy chain, ferroportin, and SLC7A11. Moreover, Hcy suppressed the protein expression of phosphor-Akt, phosphor-mTOR, Beclin-1, LC3A/LC3B, Nrf2, HO-1, phosphor-MEK1/2, phosphor-ERK1/2, and Caspase-3 in concentration- and time-dependent manners. Conclusions: Hcy-induced vascular endothelial injury is likely to be associated with apoptosis and autophagy, but not ferroptosis. The key underlying mechanisms are involved in the disruption of the intracellular antioxidant system and iron metabolism via regulation of PI3K/Akt/mTOR, MAPKs, Nrf2/HO-1, and iron metabolism. Full article
Show Figures

Figure 1

Figure 1
<p>The effect of Hcy on endothelial cell morphology, cytotoxicity, and cell viability in HUVECs<b>.</b> (<b>A</b>) The cell morphology of HUVECs treated with indicated concentrations of Hcy for 24 h, n = 3. (<b>B</b>) The cytotoxicity and cell viability of Hcy were examined by LDH release and MTT assays, respectively. Data are presented as folds or percentages of the control group, n = 4. (<b>C</b>) HUVECs were suspended with various concentrations (2, 4, and 8 mM) of Hcy and cultured in an RTCA system for 24 h, and the anti-proliferation effect of Hcy is presented as the Cell Index. The Cell Index of the Hcy-treated group and the control group were also summarized at 24 h, n = 3. (<b>D</b>) HUVECs were cultured and attached for 24 h and then treated with various concentrations (2, 4, and 8 mM) of Hcy for another 24 h. The toxicity effect of Hcy was recorded in real time by RTCA. The Cell Index was normalized to the folds of the value at the time point of adding Hcy. The normalized Cell Index of the Hcy-treated group and control group were also summarized after treatment with Hcy for 24 h, n = 3. (<b>E</b>,<b>F</b>) Presentive images and analysis of live and dead cell staining. The green (calcein-AM) and red (PI) fluorescence demonstrate the live and dead cells, respectively, n = 5. Data are presented as mean ± S.E.M. # <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 vs. the control group.</p>
Full article ">Figure 2
<p>The effects of Hcy on cell cycle, apoptosis, autophagy, and energy metabolism in HUVECs. (<b>A</b>) The cell cycle was analyzed by PI staining, followed by flow cytometry. The cell population percentages of the G0/G1, S, and G2/M phases were summarized in both the control group and the Hcy-treated group, n = 3. (<b>B</b>) The apoptosis cells were detected by annexin V-FITC and PI double-staining using flow cytometry. The percentages of early (LR) and late (UR) apoptotic cells were calculated and summarized, n = 3. (<b>C</b>–<b>G</b>) HUVECs were co-treated with Hcy (8 mM) with various indicated concentrations of Z-VDA-FMK (n = 3), 3-MA (n = 3), Wort (n = 3), LY294002 (n = 3), or Rapa (n = 4) for 24 h. The cytotoxicity was examined by LDH release assay. Results are presented as folds of the control group. (<b>H</b>) The intracellular ATP concentration (µmol/mg protein) was quantified by the commercially available kit, n = 4. Data are presented as mean ± S.E.M. # <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 vs. the control group. * <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 vs. the Hcy-treated group.</p>
Full article ">Figure 3
<p>The effect of Hcy on intracellular ROS and lipid peroxidation in HUVECs. (<b>A</b>,<b>B</b>) The HUVECs were treated with Hcy (8 mM) for 24 h, and then the intracellular ROS were indicated by DHE staining. The fluorescence intensity was calculated using ImageJ software (1.49 V), n = 3. (<b>C</b>) The HUVECs were treated with various indicated concentrations of Hcy for 24 h and the SOD activity was measured using a commercially available kit, n = 3. (<b>D</b>–<b>F</b>) Co-treatment of Hcy (8 mM) with indicated concentrations of DPI (n = 3), NAC (n = 5), VitE (n = 3), and liproxtatin-1 (n = 3) for 24 h, followed by cytotoxicity detection using LDH release kit. (<b>G</b>) The HUVECs were treated with various indicated concentrations of Hcy for 24 h. The intracellular level of MDA was measured using a commercially available kit, n = 8. (<b>H</b>) Co-treatment of Hcy (8 mM) with various indicated concentrations of Liproxtatin-1 for 24 h followed by LDH release assay, n = 3. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <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 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 4
<p>The role of ferroptosis on Hcy-induced vascular endothelial cell toxicity in HUVECs. (<b>A</b>,<b>B</b>) The HUVECs were treated with various concentrations (2, 4, and 8 mM) of Hcy for 24 h. The intracellular GSH-Px activity and GSH were measured by commercially available kits, n = 5. (<b>C</b>,<b>D</b>) The HUVECs were treated with Hcy (8 mM) for 24 h. The mRNA expression of GPX4 (n = 7) and SLC7A11 (n = 5) genes was examined by real-time PCR. (<b>E</b>,<b>F</b>) The HUVECs were co-treated with Hcy (8 mM), with various indicated concentrations of Fer-1 or DFO for 24 h, followed by the LDH release assay, n = 3. (<b>G</b>,<b>H</b>) HUVECs were suspended with Hcy (8 mM), with or without Fer-1 (80 µM) or DFO (80 µM), for 24 h. The cell proliferation was recorded in real time by RTCA, n = 4. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <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 vs. the control group. * <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 vs. the Hcy-treated group.</p>
Full article ">Figure 5
<p>The effect of ferroptosis inhibitors on Hcy-induced intracellular ROS production and mitochondrial dysfunction in HUVECs. (<b>A</b>–<b>C</b>) The HUVECs were treated with Hcy (8 mM), with or without Fer-1 (80 µM) or DFO (80 µM), for 24 h. Then, the intracellular ROS, number of mitochondria, and mitochondria membrane potential were indicated by DHE (<b>A</b>), Mito-tracker (<b>B</b>), and JC-1 (<b>C</b>) staining, respectively, n = 3. (<b>D</b>–<b>F</b>) The fluorescence intensity was calculated using ImageJ software. The results were normalized to folds of the control group. (<b>G</b>) The HUVECs were treated with Hcy (8 mM), with or without Fer-1 (80 µM) or DFO (80 µM), for 24 h. Then, the intracellular concentration of ATP was detected by a commercially available kit, n = 4. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05 and ## <span class="html-italic">p</span> ˂ 0.01 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 6
<p>The role of iron metabolism in Hcy-induced vascular endothelial cell toxicity in HUVECs. (<b>A</b>) HUVECs were treated with Hcy (8 mM) for 24 h. The intracellular level of iron was measured by a commercially available kit according to its manual, n = 3. (<b>B</b>,<b>C</b>) Co-treatment of Hcy (8 mM) with indicated concentrations of Fe<sup>3+</sup> for 24 h, followed by cell viability and cytotoxicity detections using MTT and LDH release assays, respectively, n = 3. (<b>D</b>) The cell morphology was observed by an inverted microscope equipped with 10× and 20× objective lenses. (<b>E</b>) The HUVECs were treated with or without Hcy (8 mM) for 24 h. The mRNA expressions of transferrin receptor (n = 6), transferrin (n = 5), ferritin light chain (n = 5), ferritin heavy chain (n = 6), and ferriportin (n = 6) genes were examined by real-time PCR. Results are presented as folds or percentages of the control group. Data are presented as mean ± S.E.M. # <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 vs. the control group. * <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 vs. the Hcy-treated group.</p>
Full article ">Figure 7
<p>The effect of Hcy on Akt/mTOR autophagy signaling in HUVECs. (<b>A</b>) The HUVECs were treated with various concentrations (2, 4, and 8 mM) of Hcy for 24 h. The representative bands in Western blotting analysis. (<b>B</b>–<b>E</b>) The quantitative protein expressions of phospho-mTOR (n = 3), mTOR (n = 3), phosphor-Akt (n = 4), Akt (n = 4), Beclin-1 (n = 4), LC3A/B (n = 4), and GAPDH (n = 4) were detected by Western blotting analysis. GAPDH served as the internal control. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <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 vs. the control group.</p>
Full article ">Figure 8
<p>The effect of Hcy on MAPKs and Nrf2/HO-1 signaling in HUVECs. (<b>A</b>) The HUVECs were treated with various concentrations (2, 4, and 8 mM) of Hcy for 24 h. The representative bands in Western blotting analysis. (<b>B</b>–<b>F</b>) The quantitative protein expressions of Nrf2 (n = 4), HO-1 (n = 4), phosphor-MEK1/2 (n = 4), MEK1/2 (n = 4), phosphor-ERK1/2 (n = 3), ERK1/2 (n = 3), Caspase-3 (n = 4), and GAPDH (n = 3) were detected by Western blotting analysis. GAPDH served as the internal control. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <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 vs. the control group.</p>
Full article ">Figure 9
<p>Schematic overview of the underlying mechanism of Hcy-induced endothelial death.</p>
Full article ">
21 pages, 1412 KiB  
Review
The Crucial Triad: Endothelial Glycocalyx, Oxidative Stress, and Inflammation in Cardiac Surgery—Exploring the Molecular Connections
by Božena Ćurko-Cofek, Matej Jenko, Gordana Taleska Stupica, Lara Batičić, Antea Krsek, Tanja Batinac, Aleksandra Ljubačev, Marko Zdravković, Danijel Knežević, Maja Šoštarič and Vlatka Sotošek
Int. J. Mol. Sci. 2024, 25(20), 10891; https://doi.org/10.3390/ijms252010891 - 10 Oct 2024
Viewed by 417
Abstract
Since its introduction, the number of heart surgeries has risen continuously. It is a high-risk procedure, usually involving cardiopulmonary bypass, which is associated with an inflammatory reaction that can lead to perioperative and postoperative organ dysfunction. The extent of complications following cardiac surgery [...] Read more.
Since its introduction, the number of heart surgeries has risen continuously. It is a high-risk procedure, usually involving cardiopulmonary bypass, which is associated with an inflammatory reaction that can lead to perioperative and postoperative organ dysfunction. The extent of complications following cardiac surgery has been the focus of interest for several years because of their impact on patient outcomes. Recently, numerous scientific efforts have been made to uncover the complex mechanisms of interaction between inflammation, oxidative stress, and endothelial dysfunction that occur after cardiac surgery. Numerous factors, such as surgical and anesthetic techniques, hypervolemia and hypovolemia, hypothermia, and various drugs used during cardiac surgery trigger the development of systemic inflammatory response and the release of oxidative species. They affect the endothelium, especially endothelial glycocalyx (EG), a thin surface endothelial layer responsible for vascular hemostasis, its permeability and the interaction between leukocytes and endothelium. This review highlights the current knowledge of the molecular mechanisms involved in endothelial dysfunction, particularly in the degradation of EG. In addition, the major inflammatory events and oxidative stress responses that occur in cardiac surgery, their interaction with EG, and the clinical implications of these events have been summarized and discussed in detail. A better understanding of the complex molecular mechanisms underlying cardiac surgery, leading to endothelial dysfunction, is needed to improve patient management during and after surgery and to develop effective strategies to prevent adverse outcomes that complicate recovery. Full article
(This article belongs to the Special Issue Molecular Perspective of Cardiovascular Diseases)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the endothelial glycocalyx (EG) structure under physiological conditions. The EG covers the luminal surface of blood vessels. Some elements (glycoprotein, syndecan, and glypican) are bound to endothelial cells, while others (like heparan sulphate and chondroitin sulphate) have an indirect connection. Some molecules (like orosomucoid and albumins) are “trapped” within the matrix molecules. (ORM—orosomucoid).</p>
Full article ">Figure 2
<p>Schematic representation of EG exposed to shear stress. Note the detachment of heparan and chondroitin sulphate. Various proteases (primary matrix metalloproteinases (MMP), heparanase, and hyaluronidase) are released by activated leukocytes or induced by mechanical stress. Proteases cleave the core proteins of proteoglycans and glycoproteins in the endothelial glycocalyx, leading to their degradation. (ORM—orosomucoid).</p>
Full article ">Figure 3
<p>Inflammatory response in heart damage. This Figure represents the autophagy process in response to cardiac injury, such as ischemia, sepsis, or ischemia–reperfusion injury. Following cardiac injury, an inflammatory response is triggered, which activates the autophagy pathway. Damaged cells, including apoptotic cells, viruses, bacteria, damage-associated molecular patterns (DAMPs), and damaged mitochondria, are encapsulated in a double-membrane structure called an autophagosome. LC3-II is a marker protein involved in the formation of the autophagosome. The autophagosome then fuses with a lysosome, forming an autolysosome. The lysosomal enzymes degrade the encapsulated cell debris within the autolysosome, leading to its breakdown and recycling, thereby aiding cellular recovery and homeostasis.</p>
Full article ">
19 pages, 1785 KiB  
Review
The Cardiometabolic Risk in Women with Polycystic Ovarian Syndrome (PCOS): From Pathophysiology to Diagnosis and Treatment
by Sotirios Pililis, Stamatios Lampsas, Aikaterini Kountouri, Loukia Pliouta, Emmanouil Korakas, Sarantis Livadas, John Thymis, Melpomeni Peppa, Sophia Kalantaridou, Evangelos Oikonomou, Ignatios Ikonomidis and Vaia Lambadiari
Medicina 2024, 60(10), 1656; https://doi.org/10.3390/medicina60101656 - 10 Oct 2024
Viewed by 435
Abstract
Polycystic Ovarian Syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, with significant variations in presentation characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology. Beyond reproductive health, it may also pose crucial long-term cardiometabolic risks, especially for women with [...] Read more.
Polycystic Ovarian Syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, with significant variations in presentation characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology. Beyond reproductive health, it may also pose crucial long-term cardiometabolic risks, especially for women with specific types of PCOS, contributing to early subclinical cardiovascular atherosclerotic alterations such as endothelial dysfunction, increased arterial stiffness, and coronary artery calcium levels, respectively. Moreover, the precise relationship between clinical cardiovascular disease (CVD) and PCOS remains debated, with studies demonstrating an elevated risk while others report no significant association. This review investigates the pathophysiology of PCOS, focusing on insulin resistance and its link to subclinical and clinical cardiovascular disease. Diagnostic challenges and novel management strategies, including lifestyle interventions, medications like metformin and glucagon-like peptide-1 receptor agonists (GLP-1RAs), hormonal contraceptives, and bariatric surgery, are further discussed. Recognizing the cardiometabolic risks associated with PCOS, a comprehensive approach and early intervention should address both the reproductive and cardiometabolic dimensions of the syndrome. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Diagnostic criteria for polycystic ovarian syndrome (PCOS). The key criteria used for diagnosing PCOS, including hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology, based on the Rotterdam criteria and other relevant clinical guidelines. Parts of the figure were drawn using pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (<a href="https://creativecommons.org/licenses/by/3.0/" target="_blank">https://creativecommons.org/licenses/by/3.0/</a> (accessed on 26 August 2024)).</p>
Full article ">Figure 2
<p>Polycystic ovarian syndrome (PCOS) phenotypes. PCOS is characterized by different variations of clinical features such as hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology, as classified by the Rotterdam criteria.</p>
Full article ">Figure 3
<p>Cardiometabolic risk in patients with polycystic ovarian syndrome (PCOS). The key cardiometabolic risks associated with PCOS, including insulin resistance, hyperandrogenism, obesity, dyslipidemia, hypertension, increased risk of type 2 diabetes mellitus (DM), chronic low-grade inflammation, metabolic syndrome, subclinical cardiovascular disease (CVD), and clinical CVD. ↑: higher.</p>
Full article ">Figure 4
<p>Subclinical cardiovascular disease (CVD) outcomes in patients with polycystic ovarian syndrome (PCOS). Subclinical CVD outcomes in women with PCOS, including impaired endothelial function, increased arterial stiffness, elevated coronary artery calcium levels, an increased carotid intima–media thickness, and the presence of carotid plaques.</p>
Full article ">Figure 5
<p>Cardiometabolic management in patients with polycystic ovarian syndrome (PCOS). Primary individualized interventions focusing on dietary changes are administered as the [Mediterranean diet (Med Diet), ketogenic diet (Keto Diet) or low-glycemic index (Low G.I.) diet] and secondary non-dietary interventions, depending on the body mass index. The non-dietary interventions are distinguished between obese and non-obese individuals: (i) non-obese—metformin and/or myo-inositol and/or oral contraceptives; and (ii) obese—glucagon-like peptide-1 receptor agonists (GLP-1RAs) and/or bariatric surgery. ±: combined treatment or separate treatment, individually per patient medical history.</p>
Full article ">
21 pages, 306 KiB  
Article
Analysis of ICAM-1 rs3093030, VCAM-1 rs3783605, and E-Selectin rs1805193 Polymorphisms in African Women Living with HIV and Preeclampsia
by Samukelisiwe Sibiya, Zinhle Pretty Mlambo, Mbuso Herald Mthembu, Nompumelelo P. Mkhwanazi and Thajasvarie Naicker
Int. J. Mol. Sci. 2024, 25(19), 10860; https://doi.org/10.3390/ijms251910860 - 9 Oct 2024
Viewed by 616
Abstract
Intercellular adhesion molecule-1 (ICAM-1), vascular adhesion molecule-1 (VCAM-1), and E-selectin are cell adhesion molecules that play a significant role in inflammation and are implicated in the pathophysiology of preeclampsia development and HIV infection. More specifically, the immune expression of ICAM-1, VCAM-1, and E-selectin [...] Read more.
Intercellular adhesion molecule-1 (ICAM-1), vascular adhesion molecule-1 (VCAM-1), and E-selectin are cell adhesion molecules that play a significant role in inflammation and are implicated in the pathophysiology of preeclampsia development and HIV infection. More specifically, the immune expression of ICAM-1, VCAM-1, and E-selectin within cyto- and syncytiotrophoblast cells are dysregulated in preeclampsia, indicating their role in defective placentation. This study investigates the associations of ICAM-1, VCAM-1, and E-selectin gene variants (rs3093030, rs3783605, and rs1805193, respectively) with preeclampsia comorbid with HIV infection in women of African ancestry. It also examines the susceptibility to preeclampsia development and the effect of highly active antiretroviral therapy (HAART). A total of 405 women were enrolled in this study. Out of these women, 204 were preeclamptic and 201 were normotensive. Clinical characteristics were maternal age, weight, blood pressure (systolic and diastolic), and gestational age. Whole blood was collected, DNA was extracted, and genotyping of the ICAM-1 (rs3093030 C>T), VCAM-1(rs3783605 A>G), and E-selectin (rs1805193 A>C) gene polymorphisms was performed. Comparisons were made using the Chi-squared test. Our results demonstrated that preeclamptic women exhibited a higher frequency of analyzed variants, in contrast to those with the duality of preeclampsia and HIV infection. Additionally, the C allele of the ICAM-1 (rs3093030 C>T) and G allele of the VCAM-1 (rs3783605 A>G) genes were found to have a greater role in the co-morbidity and may be considered as a risk factor for preeclampsia development in women of African ancestry. In contrast, the SNP of rs1805193 of the E-selectin gene indicated that A>C was only significantly associated with HIV infection and not with preeclampsia. These findings highlight a strong association of the rs3093030 SNP of the ICAM-1 gene and of the VCAM-1 rs3783605 gene with the development of preeclampsia, indicating their role in the defective trophoblast invasion of preeclampsia. Sub-group analysis further reveals an association of the AA genotype with late-onset preeclampsia, a less severe form of disease indicating differing genetic predispositions between early and late-onset forms. Full article
19 pages, 2473 KiB  
Systematic Review
The Role of Curcumin in Modulating Vascular Function and Structure during Menopause: A Systematic Review
by Amanina Athirah Mad Azli, Norizam Salamt, Amilia Aminuddin, Nur Aishah Che Roos, Mohd Helmy Mokhtar, Jaya Kumar, Adila A. Hamid and Azizah Ugusman
Biomedicines 2024, 12(10), 2281; https://doi.org/10.3390/biomedicines12102281 - 8 Oct 2024
Viewed by 452
Abstract
The risk of developing cardiovascular disease (CVD) escalates in women during menopause, which is associated with increased vascular endothelial dysfunction, arterial stiffness, and vascular remodeling. Meanwhile, curcumin has been demonstrated to enhance vascular function and structure in various studies. Therefore, this study systematically [...] Read more.
The risk of developing cardiovascular disease (CVD) escalates in women during menopause, which is associated with increased vascular endothelial dysfunction, arterial stiffness, and vascular remodeling. Meanwhile, curcumin has been demonstrated to enhance vascular function and structure in various studies. Therefore, this study systematically reviewed the recent literature regarding the potential role of curcumin in modulating vascular function and structure during menopause. The Ovid MEDLINE, PubMed, Scopus, and Web of Science electronic databases were searched to identify relevant articles. Clinical and preclinical studies involving menopausal women and postmenopausal animal models with outcomes related to vascular function or structure were included. After thorough screening, seven articles were selected for data extraction, comprising three animal studies and four clinical trials. The findings from this review suggested that curcumin has beneficial effects on vascular function and structure during menopause by addressing endothelial function, arterial compliance, hemodynamic parameters, and the formation of atherosclerotic lesions. Therefore, curcumin has the potential to be utilized as a supplement to enhance vascular health in menopausal women. However, larger-scale clinical trials employing gold-standard techniques to evaluate vascular health in menopausal women are necessary to validate the preliminary results obtained from small-scale randomized clinical trials involving curcumin supplementation (INPLASY, INPLASY202430043). Full article
(This article belongs to the Special Issue Compounds from Natural Products as Sources for Drug Discovery)
Show Figures

Figure 1

Figure 1
<p>The chemical structure of curcumin.</p>
Full article ">Figure 2
<p>Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 flow diagram for systematic reviews.</p>
Full article ">Figure 3
<p>Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) risk of bias summary: the reviewers’ assessments of the risk of bias for each item in every animal study included in the review, namely Abd. Aziz et al. 2012 [<a href="#B26-biomedicines-12-02281" class="html-bibr">26</a>], Jusoh et al. 2013 [<a href="#B27-biomedicines-12-02281" class="html-bibr">27</a>] and Morrone et al. 2016 [<a href="#B25-biomedicines-12-02281" class="html-bibr">25</a>].</p>
Full article ">Figure 4
<p>The Cochrane risk of bias tool for every randomized controlled trial included in the review, namely Akazawa et al. 2012 [<a href="#B28-biomedicines-12-02281" class="html-bibr">28</a>], Akazawa et al. 2013 [<a href="#B29-biomedicines-12-02281" class="html-bibr">29</a>], Sugawara et al. 2012 [<a href="#B30-biomedicines-12-02281" class="html-bibr">30</a>] and Farshbaf et al. 2022 [<a href="#B20-biomedicines-12-02281" class="html-bibr">20</a>].</p>
Full article ">Figure 5
<p>Meta-analysis of the effects of curcumin versus placebo on brachial blood pressure based on the findings of Akazawa et al. 2012 [<a href="#B28-biomedicines-12-02281" class="html-bibr">28</a>], Akazawa et al. 2013 [<a href="#B29-biomedicines-12-02281" class="html-bibr">29</a>] and Sugawara et al. 2012 [<a href="#B30-biomedicines-12-02281" class="html-bibr">30</a>]. The green square with horizontal line indicates individual study effect size (mean difference) together with its 95% confidence interval (CI), whilst the black diamond indicates the summary effect size together with its 95% CI.</p>
Full article ">
11 pages, 812 KiB  
Review
Age-Related Changes in Insulin Resistance and Muscle Mass: Clinical Implications in Obese Older Adults
by Ali A. Rizvi and Manfredi Rizzo
Medicina 2024, 60(10), 1648; https://doi.org/10.3390/medicina60101648 - 8 Oct 2024
Viewed by 346
Abstract
The older segment of the global population is increasing at a rapid pace. Advancements in public health and modern medicine lengthened life expectancy and reduced the burden of disease in communities worldwide. Concurrent with this demographic change is the rise in overweight people [...] Read more.
The older segment of the global population is increasing at a rapid pace. Advancements in public health and modern medicine lengthened life expectancy and reduced the burden of disease in communities worldwide. Concurrent with this demographic change is the rise in overweight people and obesity, which is evident in all age groups. There is also an aging-related reduction in muscle mass and function, or sarcopenia, that is exacerbated by sedentary lifestyle and poor nutrition. The coexistence of muscle loss and elevated body mass index, termed “sarcopenic obesity”, has particularly deleterious consequences in older individuals. Worsening insulin resistance and a proinflammatory state operate at the pathophysiologic level and lead to adverse health outcomes such as a proclivity to cardiovascular disease, type 2 diabetes, and even cognitive dysfunction. Although the concept of sarcopenic obesity as a disease construct is being increasingly recognized, a clearer understanding is warranted in order to define its components and health impact. Research is needed at the molecular-cellular level to tie together derangements in insulin action, cytokines, myokines, and endothelial dysfunction with clinical outcomes. Lifestyle modifications as well as targeted nonpharmacologic approaches, such as supplements and antioxidants, appear to have a promising role in reducing the chronic burden of this emerging disorder. Breakthroughs in drug therapies that retard or even reverse the underlying dynamics of sarcopenia and obesity in older persons are being actively explored. Full article
(This article belongs to the Section Endocrinology)
Show Figures

Figure 1

Figure 1
<p>The relationships of insulin resistance, glucose effectiveness, and first-phase insulin secretion in men and women with age (from: Huang et al., 2023, available at <a href="https://www.mdpi.com/2075-4418/13/13/2158" target="_blank">https://www.mdpi.com/2075-4418/13/13/2158</a>, accessed on 1 August 2024).</p>
Full article ">Figure 2
<p>Sarcopenia: EWGSOP2 algorithm for case-finding, making a diagnosis and quantifying severity in practice. The steps of the pathway are represented as find-assess-confirm-severity or F-A-C-S. (from: Cruz-Jentoft et al., 2019, available at: <a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6322506/" target="_blank">https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6322506/</a>, accessed on 1 August 2024).</p>
Full article ">Figure 3
<p>Diagrammatic depiction of myokines exerting paracrine and autocrine effects because of contraction of skeletal muscles. Release of stimulatory cytokines and the inhibitory mediator are shown by green and red arrows, respectively, while black arrows indicate downstream effects. GDF-15 = growth differentiation factor-15, IL-6 = interleukin-6, and FGF-21 = fibroblast growth factor-21. (From: Feraco et al., 2021, available at <a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8430804/" target="_blank">https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8430804/</a>, accessed on 1 August 2024).</p>
Full article ">Figure 4
<p>HOMA-IR Index in the control (nonobese–nonsarcopenic), obese–nonsarcopenic, nonobese–sarcopenic, and obese–sarcopenic study groups: mean HOMA-IR values were 2.35, 4.38, 2.60, and 2.22, respectively, (from Rizvi et al., 2024, available at: <a href="https://doi.org/10.2337/db24-158-OR" target="_blank">https://doi.org/10.2337/db24-158-OR</a>).</p>
Full article ">
20 pages, 4075 KiB  
Review
Treatment-Related Reversible Cerebral Vasoconstriction Syndrome
by Giulia Avola and Alessandro Pezzini
J. Clin. Med. 2024, 13(19), 5930; https://doi.org/10.3390/jcm13195930 - 4 Oct 2024
Viewed by 504
Abstract
Reversible cerebral vasoconstriction syndrome (RCVS) is a rare but significant cause of intracranial arteriopathy and stroke in young adults. The syndrome encompasses a spectrum of disorders radiologically characterized by reversible narrowing and dilation of intracranial arteries, often triggered by vasoactive drugs or the [...] Read more.
Reversible cerebral vasoconstriction syndrome (RCVS) is a rare but significant cause of intracranial arteriopathy and stroke in young adults. The syndrome encompasses a spectrum of disorders radiologically characterized by reversible narrowing and dilation of intracranial arteries, often triggered by vasoactive drugs or the postpartum period. The hallmark clinical feature of RCVS is thunderclap headache with or without other neurological signs. Though endothelial dysfunction and sympathetic hyperactivation are hypothesized to be key mechanisms, the exact pathogenesis of RCVS is still unclear. RCVS’s diagnosis could be challenging, since vasospasm proceeds centripetally, initially involving distal small pial and cortical arteries, and angiographic studies, especially brain magnetic resonance angiography (MRA) and computed tomography angiography (CTA), may miss it in the early phase of the disease, while early signs such as vascular hyperintensities may be visible on T2/FLAIR sequences before vasospasm onset. Catheter angiography is the gold standard and it could be used to assess vasospasm reversibility post-intra-arterial vasodilator administration. Treatment is mainly symptomatic, and nimodipine is the most commonly administered therapy, given orally or intra-arterially in severe cases. Since many aspects of RCVS remain partially known, further research is needed to better understand the complex pathophysiology of this unique clinical condition and to optimize specific management strategies. Full article
Show Figures

Figure 1

Figure 1
<p>The proposed model of the pathophysiology of RCVS. The development of RCVS is sequential, which may require both predisposition and precipitating factors to initiate and perpetuate a vicious cycle of pathogenic mechanisms (showed in the red box) that result in the clinical and radiological manifestations of RCVS (as indicated by the gradient arrow on the left of the figure). Dysregulation of cerebral vascular tone and disruption of the blood–brain barrier (BBB) are supposed to be crucial in the pathophysiology of RCVS. Both of them are a consequence of endothelial disfunction, sympathetic overactivity and oxidative stress, mediated by mechanical and biochemical stimuli. When the autoregulation and BBB disruption worsen and the endogenous protective mechanisms fail, headache, vasoconstriction and complications may ensue. In particular, hemorrhagic complications (cSAH and ICH) or PRES may be attributed to the breakdown of the BBB, while ischemic stroke is related to hypoperfusion caused by vasoconstriction of major cerebral arteries. White matter hyperintensity lesions could be attributed to either increased BBB permeability or partial ischemia due to cerebral hypoperfusion. The thunderclap headache could be attributed to the dilatation of distal arterioles or meningeal arteries that activate the trigeminovascular nociceptive fibers. RCVS: reversible cerebral vasoconstriction syndrome. PRES: posterior reversible encephalopathy syndrome. ICH: intracerebral hemorrhage. cSAH: convexity subarachnoid hemorrhage. WMHs: white matter hyperintensities. TCH: thunderclap headache.</p>
Full article ">Figure 2
<p>Hyperintense vessels on FLAIR images in two patients with RCVS (arrows).</p>
Full article ">Figure 3
<p>A 58-year-old female admitted to the Emergency Department due to thunderclap headache, diagnosed with RCVS. (<b>a</b>) The initial Angio CT showed bilateral V4 vasoconstriction (arrow) and narrowing posterior cerebral arteries; (<b>b</b>) three months later, the follow-up Angio CT showed complete resolution of vertebral artery vasoconstriction (arrows); (<b>c</b>) the follow-up imaging also showed complete recanalization of posterior cerebral arteries. RCVS: reversible cerebral vasoconstriction syndrome.</p>
Full article ">Figure 4
<p>A 37-year-old female with cocaine-induced RCVS, admitted to the Emergency Department due to headache and bilateral hypovision. Perfusion CT showed (<b>a</b>,<b>b</b>) an increased TMAX and reduced CBF in the left occipital lobe; (<b>c</b>,<b>d</b>) an increased TMAX and a reduced CBF in the left cerebellar hemisphere. (<b>e</b>) CT showed a cerebral infarct on the left cerebellar hemisphere corresponding to the area indicated on perfusion CT. RCVS: reversible cerebral vasoconstriction syndrome. TMAX: time-to-maximum. CBF: cerebral blood flow.</p>
Full article ">Figure 5
<p>An overview of the potential complications of RCVS in four different patients. (<b>a</b>) Posterior reversible encephalopathy syndrome appearing as hyperintense lesions on fluid-attenuated inversion recovery imaging (FLAIR); (<b>b</b>) ischemic strokes (arrow) highlighted by diffusion-weighted imaging (DWI); (<b>c</b>) convexity subarachnoid hemorrhage shown as the hypointensity, demonstrated by gradient echo imaging that indicates the presence of blood within the cortical sulci; (<b>d</b>) temporo-parietal and frontal lobar parenchymal hemorrhages. RCVS: reversible cerebral vasoconstriction syndrome. FLAIR: fluid-attenuated inversion recovery. DWI: diffusion-weighted imaging.</p>
Full article ">
16 pages, 545 KiB  
Review
Exploring the Impact of Extracorporeal Membrane Oxygenation on the Endothelium: A Systematic Review
by Yakun Li, Carolien Volleman, Dionne P. C. Dubelaar, Alexander P. J. Vlaar and Charissa E. van den Brom
Int. J. Mol. Sci. 2024, 25(19), 10680; https://doi.org/10.3390/ijms251910680 - 3 Oct 2024
Viewed by 543
Abstract
Extracorporeal membrane oxygenation (ECMO) is a life-saving intervention for patients with circulatory and/or pulmonary failure; however, the rate of complications remains high. ECMO induces systemic inflammation, which may activate and damage the endothelium, thereby causing edema and organ dysfunction. Advancing our understanding in [...] Read more.
Extracorporeal membrane oxygenation (ECMO) is a life-saving intervention for patients with circulatory and/or pulmonary failure; however, the rate of complications remains high. ECMO induces systemic inflammation, which may activate and damage the endothelium, thereby causing edema and organ dysfunction. Advancing our understanding in this area is crucial for improving patient outcomes during ECMO. The goal of this review is to summarize the current evidence of the effects of ECMO on endothelial activation and damage in both animals and patients. PubMed and Embase databases were systematically searched for both clinical and animal studies including ECMO support. The outcome parameters were markers of endothelial activation and damage or (in)direct measurements of endothelial permeability, fluid leakage and edema. In total, 26 studies (patient n = 16, animal n = 10) fulfilled all eligibility criteria, and used VA-ECMO (n = 13) or VV-ECMO (n = 6), or remained undefined (n = 7). The most frequently studied endothelial activation markers were adhesion molecules (ICAM-1) and selectins (E- and P-selectin). The levels of endothelial activation markers were comparable to or higher than in healthy controls. Compared to pre-ECMO or non-ECMO, the majority of studies showed stable or decreased levels. Angiopoietin-2, von Willebrand Factor and extracellular vesicles were the most widely studied circulating markers of endothelial damage. More than half of the included studies showed increased levels when compared to normal ranges, and pre-ECMO or non-ECMO values. In healthy animals, ECMO itself leads to vascular leakage and edema. The effect of ECMO support in critically ill animals showed contradicting results. ECMO support (further) induces endothelial damage, but endothelial activation does not, in the critically ill. Further research is necessary to conclude on the effect of the underlying comorbidity and type of ECMO support applied on endothelial dysfunction. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>PRISMA diagram representing the flowchart of study selection. PRISMA, Preferred Reporting Items for Systematic Reviews.</p>
Full article ">
13 pages, 9196 KiB  
Article
Sera from Rheumatoid Arthritis Patients Induce Oxidative Stress and Pro-Angiogenic and Profibrotic Phenotypes in Human Endothelial Cells
by Roberta Giordo, Anna Maria Posadino, Paola Maccioccu, Giampiero Capobianco, Angelo Zinellu, Gian Luca Erre and Gianfranco Pintus
J. Clin. Med. 2024, 13(19), 5913; https://doi.org/10.3390/jcm13195913 - 3 Oct 2024
Viewed by 405
Abstract
Background: Rheumatoid arthritis (RA) is a long-term autoimmune condition marked by persistent inflammation of the joints and various systemic complications, including endothelial dysfunction, atherosclerosis, and pulmonary fibrosis. Oxidative stress is a key contributor to the pathogenesis of RA, potentially exacerbating vascular damage and [...] Read more.
Background: Rheumatoid arthritis (RA) is a long-term autoimmune condition marked by persistent inflammation of the joints and various systemic complications, including endothelial dysfunction, atherosclerosis, and pulmonary fibrosis. Oxidative stress is a key contributor to the pathogenesis of RA, potentially exacerbating vascular damage and promoting pro-angiogenic and profibrotic processes. Objective: This study aims to investigate the effects of sera from RA patients on human umbilical vein endothelial cells (HUVECs), focusing on the induction of oxidative stress, endothelial cell proliferation, migration, and collagen type I synthesis. Methods: Twenty-eight serum samples were collected from RA patients and healthy donors (HDs). HUVECs were exposed to these sera, and intracellular reactive oxygen species (ROS) levels were fluorescently detected using H2DCF-DA. Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell migration was evaluated through a scratch wound assay, and collagen type I synthesis was measured using a lentiviral vector expressing the green fluorescent protein (GFP) under the control of the human COL1A1 gene promoter. Results: Exposure to RA sera resulted in a significant increase in intracellular ROS levels in HUVECs compared to HD sera, indicating an elevated state of oxidative stress. RA sera also promoted endothelial cell proliferation and migration, suggesting a pro-angiogenic stimulus. Additionally, RA sera significantly increased collagen type I synthesis in HUVECs, implicating a potential role in profibrotic processes associated with RA. Conclusion: The results of this study emphasize the importance of circulating factors in RA sera in promoting oxidative stress, endothelial dysfunction, and pro-angiogenic and profibrotic phenotypes in endothelial cells. These processes may contribute to the vascular and fibrotic complications observed in RA, highlighting the necessity for additional research into focused therapeutic approaches to alleviate these effects. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of RA sera on intracellular ros levels in human umbilical vein endothelial cells (HUVECs). To investigate the effect of sera from rheumatoid arthritis (RA) patients on intracellular reactive oxygen species (ROS) levels, sub-confluent HUVECs were pre-loaded with 10 μM of H2-DCFDA, a fluorescent probe for ROS detection. The cells were then grown in a basal medium supplemented with 5% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) sera from either RA patients or healthy donors (HDs). The ROS levels inside the cells were measured kinetically over a 4 h period, with the data from the first hour used for comparative analysis. Fluorescence intensity data, indicative of intracellular ROS levels, were normalized to protein content and expressed as relative fluorescence units (RFUs). The results are presented as mean values, with standard deviations indicated by horizontal lines. A Welch’s correction-modified unpaired <span class="html-italic">t</span>-test was employed to analyze the statistical differences among the studied groups, considering <span class="html-italic">p</span>-values lower than 0.05 statistically significant.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Impact of RA sera on the proliferation of human umbilical vein endothelial cells (HUVECs). Sub-confluent HUVECs were grown for 24 h (<b>A</b>) and 48 h (<b>B</b>) in a basal medium supplemented with 5% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) sera from either rheumatoid arthritis (RA) patients or healthy donors (HDs). After the respective incubation periods, cell viability was assessed according to the protocol described in the Materials and Methods section. The absorbance (ABS) results are presented as mean values, with standard deviations indicated by horizontal lines. A Welch’s correction-modified unpaired <span class="html-italic">t</span>-test was employed to analyze the statistical differences among the studied groups, considering <span class="html-italic">p</span>-values lower than 0.05 statistically significant.</p>
Full article ">Figure 3
<p>(<b>A</b>,<b>B</b>) Effect of RA sera on the migration of human umbilical vein endothelial cells (HUVECs). Sub-confluent HUVECs were grown for 24 and 48 h in a basal medium supplemented with 5% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) sera from rheumatoid arthritis (RA) patients or healthy donors (HDs). Wound-healing assays were conducted at 0, 24, and 48 h to assess cell migration in both RA-treated HUVECs and HD-treated HUVECs, which served as controls. (<b>A</b>) Representative phase-contrast microscopy images illustrate the area covered by cells at 0, 24, and 48 h following wounding. Original magnification: 10×. After 24 h, HUVECs treated with RA sera migrated faster than those treated with HD sera, nearly closing the wound gap after 48 h, as indicated by the manually drawn overlays. (<b>B</b>) The percentage of wound closure was quantitatively assessed by measuring the wound edges with ImageJ software, version 1.53t. The results are presented as mean values, with standard deviations indicated by horizontal lines. Statistical differences between the groups were assessed using a two-way analysis of variance, followed by Tukey’s post hoc test for multiple comparisons, considering <span class="html-italic">p</span>-values &lt; 0.05 statistically significant.</p>
Full article ">Figure 4
<p>Effects of RA sera on collagen promoter activity in HUVECs. Sub-confluent HUVEC/pCol1GFP-pEFα-FP602 cells were cultured in a standard medium supplemented with 5% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) sera from rheumatoid arthritis (RA) patients and healthy donors (HDs). The activation of the collagen type I (COL1A1) promoter was monitored kinetically over a 4 h period, with the first hour’s values used for comparative analysis. Data were expressed as relative fluorescence units (RFUs) and corrected for transduction efficiency using the ratio of pCOL1A1-LV-tGFP (green fluorescence) to EF-LV-FP602 (red fluorescence). The results are presented as mean values, with standard deviations indicated by horizontal lines. A Welch’s correction-modified unpaired <span class="html-italic">t</span>-test was employed to analyze the statistical differences among the studied groups, considering <span class="html-italic">p</span>-values lower than 0.05 statistically significant.</p>
Full article ">Figure 5
<p>Correlation matrix illustrating the relationships between the clinical parameters of patients and the in vitro experimental data, as well as the correlations among the various in vitro experimental outcomes. The arrow highlights the significant correlation between ROS levels and cell migration at 48 h.</p>
Full article ">
Back to TopTop