Nothing Special   »   [go: up one dir, main page]

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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (293)

Search Parameters:
Keywords = angiopoietin-1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 11548 KiB  
Review
Molecular and Cellular Mechanisms Involved in the Pathophysiology of Retinal Vascular Disease—Interplay Between Inflammation and Oxidative Stress
by Jovana V. Srejovic, Maja D. Muric, Vladimir Lj. Jakovljevic, Ivan M. Srejovic, Suncica B. Sreckovic, Nenad T. Petrovic, Dusan Z. Todorovic, Sergey B. Bolevich and Tatjana S. Sarenac Vulovic
Int. J. Mol. Sci. 2024, 25(21), 11850; https://doi.org/10.3390/ijms252111850 - 4 Nov 2024
Viewed by 914
Abstract
Retinal vascular diseases encompass several retinal disorders, including diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, and retinal vascular occlusion; these disorders are classified as similar groups of disorders due to impaired retinal vascularization. The aim of this review is to address the [...] Read more.
Retinal vascular diseases encompass several retinal disorders, including diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, and retinal vascular occlusion; these disorders are classified as similar groups of disorders due to impaired retinal vascularization. The aim of this review is to address the main signaling pathways involved in the pathogenesis of retinal vascular diseases and to identify crucial molecules and the importance of their interactions. Vascular endothelial growth factor (VEGF) is recognized as a crucial and central molecule in abnormal neovascularization and a key phenomenon in retinal vascular occlusion; thus, anti-VEGF therapy is now the most successful form of treatment for these disorders. Interaction between angiopoietin 2 and the Tie2 receptor results in aberrant Tie2 signaling, resulting in loss of pericytes, neovascularization, and inflammation. Notch signaling and hypoxia-inducible factors in ischemic conditions induce pathological neovascularization and disruption of the blood–retina barrier. An increase in the pro-inflammatory cytokines—TNF-α, IL-1β, and IL-6—and activation of microglia create a persistent inflammatory milieu that promotes breakage of the blood–retinal barrier and neovascularization. Toll-like receptor signaling and nuclear factor-kappa B are important factors in the dysregulation of the immune response in retinal vascular diseases. Increased production of reactive oxygen species and oxidative damage follow inflammation and together create a vicious cycle because each factor amplifies the other. Understanding the complex interplay among various signaling pathways, signaling cascades, and molecules enables the development of new and more successful therapeutic options. Full article
Show Figures

Figure 1

Figure 1
<p>Representation of the physiological structure of the retina and retinal vascularization.</p>
Full article ">Figure 2
<p>Pathological hallmarks of non-proliferative (NPDR) and proliferative (PDR) stages of DR, including aneurysms, hemorrhages, hard exudates, cotton wool spots in NPDR, and abnormal neovascularization and macular edema, combined with previous for PDR.</p>
Full article ">Figure 3
<p>Retinopathy of prematurity (ROP)—different phases of ROP including impaired retinal vascular development in utero induced by low VEGF values, and pathological changes followed by increased VEGF production such as abnormal neovascularization, excessive fibrous tissue growth, and retinal detachment. Anti-VEGF therapy, applied at an early stage as prevention, or when the disease appears, can lead to resolution.</p>
Full article ">Figure 4
<p>Age-related macular degeneration—the main pathological hallmark of the dry form of AMD is drusen formation, while patients in the wet form of the disease have exudation and hemorrhages as crucial characteristics.</p>
Full article ">Figure 5
<p>Retinal vascular occlusions—the main etiological factors involved in the pathogenesis of retinal vascular occlusions and clinical presentation of retinal artery and retinal vein occlusion.</p>
Full article ">Figure 6
<p>Carotid artery stenosis and ocular implications. Various risk factors contribute to the development of carotid artery stenosis with the consequent development of different aspects of ocular ischemic syndrome and chronic ocular ischemic disease.</p>
Full article ">Figure 7
<p>Molecular and cellular signaling in the pathogenesis of ischemic retinal diseases—the key role of VEGF. Abbreviations: Ang2, angiopoietin 2; EPO, erythropoietin; HIF-1α, hypoxia-inducible factor-1α; PDGF, platelet-derived growth factor; AGEs, advanced glycation end products; IGF-1, insulin-like growth factor 1; VEGF, vascular endothelial growth factor; VEGFR2, vascular endothelial growth factor receptor 2; Jag1,2, Jagged 1 and 2; Dll1,3,4, Delta-like ligands 1, 3 and 4; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-kappa B; PKB, protein kinase B; NOS, nitric oxide synthase; BRB, blood–retina barrier.</p>
Full article ">Figure 8
<p>Inflammatory pathways in retinal vascular disorders—the interplay between different interleukins, chemokines, microglia, and development of neovascularization and vascular retinal diseases. Abbreviations: IL-1β, interleukin 1β; IL-6, interleukin 6; IL-10, interleukin 10; TNF-α, tumor necrosis factor α; MCP-1, monocyte chemoattractant protein 1; CCL2, CC motif chemokine ligand 2; CCL5, CC motif chemokine ligand 5; CXCL10, CXC motif chemokine ligand 10; DAMPs, damage-associated molecular pattern molecules; TLR4, toll like receptor 4; NF-κB, nuclear factor-kappa B; BRB, blood–retina barrier.</p>
Full article ">Figure 9
<p>Oxidative stress and damage of retinal cells. Abbreviations: Nox/Duox, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase/dual oxidase; NOS, nitric oxide synthase; eNOS, endothelial nitric oxide synthase; nNOS, neuronal nitric oxide synthase; iNOS, inducible nitric oxide synthase; <sup>•</sup>HO, hydroxyl radical; <sup>•</sup>O<sub>2</sub><sup>2</sup>, peroxide ion radical; <sup>•</sup>O<sub>2</sub>, superoxide anion radical; <sup>•</sup>ONOO, peroxynitrite; <sup>•</sup>NO, nitric oxide; ROS, reactive oxygen species; RNS, reactive nitrogen species.</p>
Full article ">Figure 10
<p>The relation of reactive species and oxidative stress in the development of diabetic retinopathy and age-related macular degeneration. Abbreviations: AGE/RAGE, advanced glycation end products/receptors; GSH, reduced glutathione; IL-1β, interleukin 1β; iNOS, inducible nitric oxide synthase; MDA, malondialdehyde; NF-κB, nuclear factor-kappa B; NO, nitric oxide; Nox, NADPH oxidase system; PKC, protein kinase C; TNF-α, tumor necrosis factor α.</p>
Full article ">
12 pages, 10244 KiB  
Article
Angiopoietin 1 Attenuates Dysregulated Angiogenesis in the Gastrocnemius of DMD Mice
by Andrew McClennan and Lisa Hoffman
Int. J. Mol. Sci. 2024, 25(21), 11824; https://doi.org/10.3390/ijms252111824 - 4 Nov 2024
Viewed by 712
Abstract
Duchenne muscular dystrophy (DMD) is a degenerative neuromuscular disease caused by a lack of functional dystrophin. Ang 1 paracrine signalling maintains the endothelial barrier of blood vessels, preventing plasma leakage. Chronic inflammation, a consequence of DMD, causes endothelial barrier dysfunction in skeletal muscle. [...] Read more.
Duchenne muscular dystrophy (DMD) is a degenerative neuromuscular disease caused by a lack of functional dystrophin. Ang 1 paracrine signalling maintains the endothelial barrier of blood vessels, preventing plasma leakage. Chronic inflammation, a consequence of DMD, causes endothelial barrier dysfunction in skeletal muscle. We aim to elucidate changes in the DMD mouse’s gastrocnemius microvascular niche following local administration of Ang 1. Gastrocnemii were collected from eight-week-old mdx/utrn+/− and healthy mice. Additional DMD cohort received an intramuscular injection of Ang 1 to gastrocnemius and contralateral control. Gastrocnemii were collected for analysis after two weeks. Using immunohistochemistry and real-time quantitative reverse transcription, we demonstrated an abundance of endothelial cells in DMD mouse’s gastrocnemius, but morphology and gene expression were altered. Myofiber perimeters were shorter in DMD mice. Following Ang 1 treatment, fewer endothelial cells were present, and microvessels were more circular. Vegfr1, Vegfr2, and Vegfa expression in Ang 1-treated gastronemii increased, while myofiber size distribution was consistent with vehicle-only gastrocnemii. These results suggest robust angiogenesis in DMD mice, but essential genes were underexpressed—furthermore, exogenous Ang 1 attenuated angiogenesis. Consequentially, gene expression increased. The impact must be investigated further, as Ang 1 therapy may be pivotal in restoring the skeletal muscle microvascular niche. Full article
Show Figures

Figure 1

Figure 1
<p>Histology of anti-CD31-stained endothelial cells and anti-laminin-stained myofibers in Healthy (<b>a</b>–<b>f</b>), Diseased (<b>g</b>–<b>l</b>), Vehicle-only (<b>m</b>–<b>r</b>), and Ang 1-Treated (<b>s</b>–<b>x</b>) transverse gastrocnemius samples. Scale bar = 50 µm (<b>a</b>,<b>c</b>,<b>e</b>,<b>g</b>,<b>i</b>,<b>k</b>,<b>m</b>,<b>o</b>,<b>q</b>,<b>s</b>,<b>u</b>,<b>w</b>), 10 µm (<b>b</b>,<b>d</b>,<b>f</b>,<b>h</b>,<b>j</b>,<b>l</b>,<b>n</b>,<b>p</b>,<b>r</b>,<b>t</b>,<b>v</b>,<b>x</b>).</p>
Full article ">Figure 2
<p>Microvessel morphometry in Healthy, Diseased, Vehicle-only, and Ang 1-Treated gastrocnemius samples: (<b>a</b>,<b>b</b>) percent area CD31 positively stained tissue, (<b>c</b>,<b>d</b>) capillary-to-fibre perimeter exchange (CFPE) index, (<b>e</b>,<b>f</b>) microvessel area, (<b>g</b>,<b>h</b>) microvessel density, and (<b>i</b>,<b>j</b>) microvessel circularity. Data are mean ± SEM. <span class="html-italic">n</span> = 6–7. * <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 3
<p>Analysis of endothelial cell-related gene expression by Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR). Gene expression was measured in the gastrocnemius of (<b>a</b>) Diseased relative to Healthy and (<b>b</b>) Ang 1-Treated relative to Vehicle-only. Data are mean ± SEM. <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p><span class="html-italic">Ang 1</span>:<span class="html-italic">Ang 2</span> relative fold change ratio of (<b>a</b>) Healthy and Diseased and (<b>b</b>) Vehicle-only and Ang 1-Treated gastrocnemii. Data are mean ± SEM. <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Distribution of myofiber area of (<b>a</b>) Healthy, Diseased <span class="html-italic">p</span> = 0.0642, (<b>b</b>) Vehicle-only, and Ang 1-Treated <span class="html-italic">p</span> = 0.0792 gastrocnemius samples. <span class="html-italic">n</span> = 6.</p>
Full article ">Figure 6
<p>Representative histology of Masson’s Trichrome staining. Blue arrows indicate collagen deposition in (<b>a</b>) Healthy, (<b>b</b>) Diseased, (<b>c</b>) Vehicle-only, and (<b>d</b>) Ang 1-Treated gastrocnemius with scale bar = 150 μm. (<b>e</b>) Healthy, (<b>f</b>) Diseased, (<b>g</b>) Vehicle-only, and (<b>h</b>) Ang 1-Treated gastrocnemius with scale bar = 35 μm.</p>
Full article ">Figure 7
<p>Quantitative analysis of collagen deposition in (<b>a</b>) Healthy and Diseased, and (<b>b</b>) Vehicle-only, and Ang 1-Treated gastrocnemius. <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
10 pages, 757 KiB  
Article
Angiopoietin II in Critically Ill Septic Patients: A Post Hoc Analysis of the DRAK Study
by Veronika Bucher, Helen Graf, Johannes Zander, Uwe Liebchen, Danilo Hackner, Caroline Gräfe, Martin Bender, Michael Zoller and Christina Scharf
Biomedicines 2024, 12(11), 2436; https://doi.org/10.3390/biomedicines12112436 - 23 Oct 2024
Viewed by 548
Abstract
Introduction: Angiopoietin II (Ang-II) plays a pivotal role in the development of microcirculatory dysfunction as it provokes endothelial barrier disruption in patients with sepsis or septic shock. In particular, those with acute kidney injury show high Ang-II concentrations. So far, it is unclear [...] Read more.
Introduction: Angiopoietin II (Ang-II) plays a pivotal role in the development of microcirculatory dysfunction as it provokes endothelial barrier disruption in patients with sepsis or septic shock. In particular, those with acute kidney injury show high Ang-II concentrations. So far, it is unclear which covariates influence Ang-II concentration in the early phase of sepsis, especially if extracorporeal therapies also do. Methods: Ang-II concentrations were measured in 171 patients with sepsis after the first day of antibiotic treatment between 03/2013 and 01/2015. Ang-II was correlated with potential influencing factors (Spearman correlation). A multivariate model was established including the significant correlating parameters. The Mann–Whitney U test and the Kruskal–Wallis test were used to detect significant differences in Ang-II concentration. Results: The median Ang-II concentration was 8015 pg/mL (interquartile range (IQR): 5024–14,185). A total of forty patients were treated with kidney replacement therapy (KRT) and 20 were supported by venovenous extracorporeal membrane oxygenation (vv-ECMO). Sequential organ failure assessment (SOFA) score (r = 0.541), creatinine clearance (r = −0.467), urinary output (r = −0.289), interleukin (IL)-6 (r = 0.529), C-reactive protein (CRP) (r = 0.241), platelet count (r = −0.419), bilirubin (r = 0.565), lactate (r = 0.322), KRT (r = 0.451), and fluid balance (r = 0.373) significantly correlated with Ang-II concentration and were included in the multivariate model. There, creatinine clearance (p < 0.01, b = −26.3, 95% confidence interval (CI) −41.8–−10.8), fluid balance (p = 0.002, b = 0.92, 95% CI 0.33–1.51), and CRP (p = 0.004, b = 127.6, 95% CI 41.6–213.7) were associated with Ang-II concentration. Furthermore, patients with KRT (median: 15,219 pg/mL, IQR: 10,548–20,270) had significantly (p < 0.01) higher Ang-II concentrations than those with vv-ECMO support (median: 6412 pg/mL, IQR: 5246–10,257) or those without extracorporeal therapy (median: 7156 pg/mL, IQR: 4409–12,741). Conclusion: Increased CRP, positive fluid balance, and impaired kidney function were associated with higher Ang-II concentrations in critically ill patients in the early stage of sepsis in this post hoc analysis. In particular, patients with KRT had very high Ang-II concentrations, whereas the use of vv-ECMO was not related to higher Ang-II concentrations. The significance for clinical practice should be clarified by a prospective study with standardized measurements. Full article
(This article belongs to the Special Issue Inflammatory Signaling in Vascular Endothelial Cells)
Show Figures

Figure 1

Figure 1
<p>Ang-II concentrations in three different subgroups (fluid balance, CRP, kidney function). Note: The first three boxplots include patients with a fluid balance &lt; 0 mL/24 h (blue), CRP &lt; 10 mg/dL (orange), and creatinine clearance &gt; 60 mL/min (gray). The second three boxplots include patients with a fluid balance 0–1500 mL/min (blue), CRP 10–20 mg/dL, and creatinine clearance &lt; 60 mL/min (gray). The last three boxplots include patients with a fluid balance &gt; 1500 mL/24 h, CRP &gt; 20 mg/dL (orange), and kidney replacement therapy (gray). The boxes of the boxplots represent the interquartile range (IQR) and the horizontal line represents the median. The whiskers are limited to 1.5 times the IQR. The mean is indicated by the cross.</p>
Full article ">Figure 2
<p>The percentage of patients with Ang-II concentrations &lt; 5000, 5000–10,000, 10,000–20,000, and &gt;20,000 pg/mL in the subgroups with KRT, vv-ECMO, and no extracorporeal therapy.</p>
Full article ">
12 pages, 1012 KiB  
Review
Flavonoid-Mediated Suppression of Tumor Angiogenesis: Roles of Ang-Tie/PI3K/AKT
by Shallu Saini, Hardeep Singh Tuli, Reena V. Saini, Adesh K. Saini, Katrin Sak, Damandeep Kaur, Moyad Shahwan, Ritu Chauhan and Abhishek Chauhan
Pathophysiology 2024, 31(4), 596-607; https://doi.org/10.3390/pathophysiology31040043 - 12 Oct 2024
Viewed by 790
Abstract
Angiogenesis is a process involved in the formation of new blood capillaries from pre-existing ones. It is regulated by several anti-angiogenic molecules involved in tumor growth and metastasis. The endothelial angiopoietin Ang-Tie/PI3K/AKT growth receptor pathway is necessary for healthy vascular development. The activation [...] Read more.
Angiogenesis is a process involved in the formation of new blood capillaries from pre-existing ones. It is regulated by several anti-angiogenic molecules involved in tumor growth and metastasis. The endothelial angiopoietin Ang-Tie/PI3K/AKT growth receptor pathway is necessary for healthy vascular development. The activation of AKT is controlled by a multistep process involving phosphoinositide 3-kinase (PI3K). This article aims to provide an overview of the role and mechanism of the Ang-Tie/PI3K/AKT signaling pathways and the potential of flavonoids as anti-angiogenic drugs. Flavonoids have shown great potential in preventing angiogenesis by targeting signaling pathways and exhibit additional anti-cancer properties. Research studies have revealed that the currently available anti-angiogenic drugs do not meet the safety and efficacy standards for treating tumor growth. Phytocompounds have long been a valuable resource for the development of novel therapeutic drugs. This article explores recent findings explaining the role and mechanism of the Ang-Tie/PI3K/AKT signaling pathways, as well as the interaction of flavonoids with angiogenic signaling pathways as a novel therapeutic approach. Several investigations have shown that synergistic studies of natural phytocompounds have great potential to target these pathways to inhibit tumor growth. Therefore, flavonoid-based medications may offer a more effective synergistic strategy to treat cancer. Full article
Show Figures

Figure 1

Figure 1
<p>The general structure of Flavonoids and their subclasses [<a href="#B9-pathophysiology-31-00043" class="html-bibr">9</a>].</p>
Full article ">Figure 2
<p>Role of Ang1 in the angiogenesis and cell proliferation pathway.</p>
Full article ">Figure 3
<p>Ang-Tie2 Signaling pathway [<a href="#B53-pathophysiology-31-00043" class="html-bibr">53</a>].</p>
Full article ">
10 pages, 1489 KiB  
Article
Role of Plasma Angiopoietin-1 and VEGF Levels as Potential Biomarkers in Chronic Central Serous Chorioretinopathy with Macular Neovascularization
by Michał Chrząszcz, Weronika Pociej-Marciak, Natalia Mackiewicz, Bożena Romanowska-Dixon, Marek Sanak, Sławomir Teper, Maciej Gawęcki and Izabella Karska-Basta
Int. J. Mol. Sci. 2024, 25(19), 10748; https://doi.org/10.3390/ijms251910748 - 6 Oct 2024
Viewed by 611
Abstract
To evaluate the plasma levels of angiopoietin-1 and vascular endothelial growth factor (VEGF) and their association with macular neovascularization (MNV) in patients with chronic central serous chorioretinopathy (cCSC). Correlations between plasma cytokine levels, CSC duration, and mean choroidal thickness (CT) were also investigated. [...] Read more.
To evaluate the plasma levels of angiopoietin-1 and vascular endothelial growth factor (VEGF) and their association with macular neovascularization (MNV) in patients with chronic central serous chorioretinopathy (cCSC). Correlations between plasma cytokine levels, CSC duration, and mean choroidal thickness (CT) were also investigated. Of the 59 patients with cCSC, 10 patients with MNV secondary to cCSC and 10 patients with cCSC without MNV were enrolled in the study. The control group included 15 healthy volunteers matched for age, sex, smoking status, and comorbidities. Chronic CSC was diagnosed based on typical findings on swept-source optical coherence tomography (OCT), fundus fluorescein angiography, and indocyanine green angiography. Additionally, all patients underwent OCT angiography to help detect MNV. Plasma angiopoietin-1 and VEGF levels were assessed using multiplex immunoassay. The plasma angiopoietin-1 levels differed between the 3 groups (p = 0.005). The angiopoietin-1 levels were lower in patients with cCSC with MNV than in controls (p = 0.006). There were no differences in the plasma VEGF levels between all the 3 groups (p = 0.329). The VEGF levels were negatively correlated with mean CT in cCSC patients with MNV (rho = −0.683, p = 0.042) but correlated positively with disease duration in patients with cCSC without MNV (rho = 0.886, p = 0.003). Our study confirms that MNV is a common complication of cCSC and provides new insights into the role of angiopoietin-1 in cCSC and MNV. Reduced angiopoietin-1 levels in cCSC patients, regardless of MNV status, highlight the importance of the Ang–Tie2 pathway in disease pathogenesis and may point to new therapeutic targets and future novel treatments to improve the management of these patients. Full article
Show Figures

Figure 1

Figure 1
<p>Box-and-whisker plot of plasma angiopoetin-1 levels in patients with chronic central serious chorioretinopathy (cCSC) without macular neovascularization (MNV), patients with cCSC with MNV, and controls. Circles indicate cases distant from an interquartile range of 1.5 to 3.</p>
Full article ">Figure 2
<p>Box-and-whisker plot of plasma vascular endothelial growth factor (VEGF) levels in patients with chronic central serious chorioretinopathy (cCSC) with macular neovascularization (MNV), patients with cCSC without MNV, and controls. Circles indicate cases distant from an interquartile range of 1.5 to 3.</p>
Full article ">Figure 3
<p>Correlation between plasma vascular endothelial growth factor (VEGF) levels and mean choroidal thickness in patients with chronic central serious chorioretinopathy (cCSC) with macular neovascularization (MNV).</p>
Full article ">Figure 4
<p>Swept-source optical coherence tomography angiography of macular neovascularization (MNV) secondary to chronic central serous chorioretinopathy: (<b>A</b>)—en-face angiogram at the level of the superficial plexus; (<b>B</b>)—en-face angiogram at the level of the deep plexus; (<b>C</b>)—en-face angiogram at the level of the avascular deep retina with a visible vessel network of MNV; (<b>D</b>)—en-face angiogram at the level of choriocapillaris with a visible vessel network of MNV; (<b>E</b>)—optical coherence tomography showing flat irregular pigment epithelial detachment (FIPED) and subretinal fluid (SRF) B scan; (<b>F</b>)—vessel density map; (<b>G</b>)—color fundus picture.</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 1020
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 ">
10 pages, 3500 KiB  
Article
One Copy Number Variation within the Angiopoietin-1 Gene Is Associated with Leizhou Black Goat Meat Quality
by Qun Wu, Xiaotao Han, Yuelang Zhang, Hu Liu, Hanlin Zhou, Ke Wang and Jiancheng Han
Animals 2024, 14(18), 2682; https://doi.org/10.3390/ani14182682 - 14 Sep 2024
Viewed by 578
Abstract
The ANGPT1 gene plays a crucial role in the regulation of angiogenesis and muscle growth, with previous studies identifying copy number variations (CNVs) within this gene among Leizhou black goats. In this study, we investigated three ANGPT1 CNVs in 417 individuals of LZBG [...] Read more.
The ANGPT1 gene plays a crucial role in the regulation of angiogenesis and muscle growth, with previous studies identifying copy number variations (CNVs) within this gene among Leizhou black goats. In this study, we investigated three ANGPT1 CNVs in 417 individuals of LZBG using quantitative PCR (qPCR), examining the impact of different CNV types on the ANGPT1 gene expression and their associations with growth and meat quality traits. Notably, the ANGPT1 CNV-1 (ARS1_chr14:24950001-24953600) overlaps with protein-coding regions and conserved domains; its gain-of-copies genotype (copies ≥ 3) was significantly correlated with ANGPT1 mRNA expression in muscle tissue (p < 0.01). Furthermore, the gain-of-copies genotype of CNV-1 demonstrated significant correlations with various phenotypic traits, including carcass weight, body weight, shear stress, chest circumference, and cross-sectional area of longissimus dorsi muscle. These findings indicate that the CNV-1 gain-of-copies genotype in the ANGPT1 gene may serve as a valuable marker for selecting Leizhou black goats exhibiting enhanced growth and muscular development characteristics, thereby holding potential applications in targeted breeding programs aimed at improving meat quality. Full article
Show Figures

Figure 1

Figure 1
<p>Biological evolution and conserved domains of the <span class="html-italic">ANGPT1</span> gene. (<b>A</b>) Multiple sequence alignment of the <span class="html-italic">ANGPT1</span> for 10 species. ‘*’ indicates conserved amino acid among different species. (<b>B</b>) Phylogenetic tree analysis for the <span class="html-italic">ANGPT1</span> gene among 10 species. (<b>C</b>) Motif structural analysis for the ANGPT1 protein among 10 species. (<b>D</b>) Protein structure prediction of the ANGPT1 protein.</p>
Full article ">Figure 2
<p>The location of the <span class="html-italic">ANGPT1</span>-CNV1 and the schematic diagram of overlaps between CNV-1-related protein region and conserved domains in the ANGPT1 protein sequence.</p>
Full article ">Figure 3
<p>Comparison analysis of the <span class="html-italic">ANGPT1</span> expression levels and distribution of CNVs. (<b>A</b>) The <span class="html-italic">ANGPT1</span> mRNA expression profile in 10 tissues of the 12 adult-female LZBG. (<b>B</b>) Comparison of the <span class="html-italic">ANGPT1</span> mRNA levels at different times in the longissimus dorsi muscle of the 12 LZBG. (<b>C</b>–<b>E</b>), Distribution of CNVs different types and comparison of the <span class="html-italic">ANGPT1</span> expression levels among different CNVs and different genotypes in longissimus dorsi muscle and gluteofemoral triceps in the LZBG. Loss, Normal, and Gain were defined as copy number &lt;2, =2, or ≥3, respectively. Different letters represent significant differences (a, b, c : <span class="html-italic">p</span> &lt; 0.05; A, B: <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
22 pages, 7071 KiB  
Perspective
Angiopoietin-like Proteins and Lipoprotein Lipase: The Waltz Partners That Govern Triglyceride-Rich Lipoprotein Metabolism? Impact on Atherogenesis, Dietary Interventions, and Emerging Therapies
by Alejandro Gugliucci
J. Clin. Med. 2024, 13(17), 5229; https://doi.org/10.3390/jcm13175229 - 4 Sep 2024
Viewed by 1071
Abstract
Over 50% of patients who take statins are still at risk of developing atherosclerotic cardiovascular disease (ASCVD) and do not achieve their goal LDL-C levels. This residual risk is largely dependent on triglyceride-rich lipoproteins (TRL) and their remnants. In essence, remnant cholesterol-rich chylomicron [...] Read more.
Over 50% of patients who take statins are still at risk of developing atherosclerotic cardiovascular disease (ASCVD) and do not achieve their goal LDL-C levels. This residual risk is largely dependent on triglyceride-rich lipoproteins (TRL) and their remnants. In essence, remnant cholesterol-rich chylomicron (CM) and very-low-density lipoprotein (VLDL) particles play a role in atherogenesis. These remnants increase when lipoprotein lipase (LPL) activity is inhibited. ApoCIII has been thoroughly studied as a chief inhibitor and therapeutic options to curb its effect are available. On top of apoCIII regulation of LPL activity, there is a more precise control of LPL in various tissues, which makes it easier to physiologically divide the TRL burden according to the body’s requirements. In general, oxidative tissues such as skeletal and cardiac muscle preferentially take up lipids during fasting. Conversely, LPL activity in adipocytes increases significantly after feeding, while its activity in oxidative tissues decreases concurrently. This perspective addresses the recent improvements in our understanding of circadian LPL regulations and their therapeutic implications. Three major tissue-specific lipolysis regulators have been identified: ANGPTL3, ANGPTL4, and ANGPTL8. Briefly, during the postprandial phase, liver ANGPTL8 acts on ANGPTL3 (which is released continuously from the liver) to inhibit LPL in the heart and muscle through an endocrine mechanism. On the other hand, when fasting, ANGPTL4, which is released by adipocytes, inhibits lipoprotein lipase in adipose tissue in a paracrine manner. ANGPTL3 inhibitors may play a therapeutic role in the treatment of hypertriglyceridemia. Several approaches are under development. We look forward to future studies to clarify (a) the nature of hormonal and nutritional factors that determine ANGPTL3, 4, and 8 activities, along with what long-term impacts may be expected if their regulation is impaired pharmacologically; (b) the understanding of the quantitative hierarchy and interaction of the regulatory actions of apoCIII, apoAV, and ANGPTL on LPL activity; (c) strategies for the safe and proper treatment of postprandial lipemia; and (d) the effect of fructose restriction on ANGPTL3, ANGPTL4, and ANGPTL8. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

Figure 1
<p><b>Lipoprotein lipase (LPL) is the key enzyme that transforms insoluble, energy-rich triglycerides (TG) into soluble fatty acids (FA); some structural considerations are as follows.</b> (1) Skeletal and cardiac muscle, adipocytes (white and brown), oligodendrocytes, astrocytes, and microglia, among others, produce lipoprotein lipase (2). Anchored in the adjacent endothelium by glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1 (GPIHBP1), it acts on triglyceride-rich lipoproteins (TRL) 3) LPL binds to TRL via (4) hydrophobic residues, its hydrolytic pore closed (5) until the binding occurs. LPL acts on TG (6) when ensuing conformational changes result in (7) opening of the lid of the pore (8) where hydrolysis occurs (9), and in a unidirectional flow, one FA at a time (10) is released, as well as the resulting di- and mono-glycerides (11). The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 2
<p><b>Capillary lipoprotein lipase (LPL) directs the flux and partition of fatty acid supply to tissues: main features.</b> (1) Adipocytes, and cells in oxidative tissues such as skeletal and cardiac muscle, among others, produce lipoprotein lipase. (2) LPL is a complex enzyme that needs heparin sulfate proteoglycans (HSPG) on the cell surface to aid in the molecule’s transcytosis to the microvascular’ luminal surface. (3) Glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1 (GPIHBP1) assists in fixing LPL on the luminal surface of the endothelial cells and supports the configuration of LPL to an active lipolytic enzyme. (4) LPL acts on triglyceride-rich lipoproteins (TRL), namely VLDL (all day) and chylomicrons (postprandial), to hydrolyze triglycerides (TG) (5) into free fatty acids (FA) (6) and glycerol (7). Free fatty acids are employed for oxidation in muscle and myocardial tissue as shown in (8) or storage in adipocytes as shown in (9). (10) About 5 to 30% may remain in the circulation, referred to as “spill over” fatty acids. LPL has a very multifaceted regulation: its chief activators are insulin, apoCII, AIV, and AV, and its key inhibitors are apoCIII and angiopoietin-like proteins (ANGPTL) 3, 4, and 8. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 3
<p><b>Potential waste of energy-rich fatty acids during fasting if LPL were not tissue-specifically regulated. Competition between adipose and oxidative tissues in the fasting state.</b> (1) During fasting, the liver produces VLDL as a source of fatty acids (FA) for most oxidative tissues. (2) VLDL circulates and reaches both adipose and (3) oxidative tissues. (4) Low insulin and high glucagon promote hepatic production of glucose whereas GlutIV is inactive, and (5) most glucose is therefore taken up by the brain and erythrocytes via GlutI and II. As shown in <a href="#jcm-13-05229-f001" class="html-fig">Figure 1</a>, VLDL reaches LPL (6) in oxidative tissues, providing most of the energy as FA. At the same time, VLDL also reaches LPL (7) in adipose, where hormone-sensitive lipase (HSL) fully activated by low insulin catalyzes extensive lipolysis (8), inundating the capillary with intracellular FA that would add to the extracellular FA produced by LPL (9). Given the relative masses of muscle and adipose (rough averages are presented), between 30 and 40% of very energy-demanding productions of TG by the liver, exported as VLDL, would possibly enter a liver-adipose-liver futile FA cycle, which would also add to the acid load of the bloodstream (10). This is prevented by the presence of another level of regulation provided by the axis of ANGPTL3, 4, and 8 that controls the partition of TRL fluxes during fasting and feeding cycles, as depicted in <a href="#jcm-13-05229-f003" class="html-fig">Figure 3</a> and <a href="#jcm-13-05229-f004" class="html-fig">Figure 4</a>. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 4
<p><b>ANGPTL4 acts in an autocrine and paracrine fashion in adipose to divert VLDL to oxidative tissues during fasting.</b> Superimposed on the regulation of LPL activity by apoCII/CIII, there exists a finer control of the enzyme in different tissues that provides for the physiological partition of the TRL load depending on the needs of the body. Basically, during fasting, lipids are preferentially taken up by oxidative tissues such as cardiac and skeletal muscle, and storage at the adipocytes is not favored. Thus, during fasting, the futile cycle depicted in <a href="#jcm-13-05229-f002" class="html-fig">Figure 2</a> is prevented (1) by ANGPTL4, which is produced by adipocytes during fasting, whereas ANGPTL8 is downregulated (2). ANGPTL8 curbs ANGPTL4 inhibition of LPL. The resulting mainly inactive LPL (3) shunts VLDL away from adipose. Additionally, fasting increases the hepatic output of apoAV and CREBH (cAMP response element-binding protein, hepatocyte-specific (4), which inactivates the ANGPTL3/8 complex, a potent inhibitor of oxidative tissue LPL, which is key in the fed state (see <a href="#jcm-13-05229-f004" class="html-fig">Figure 4</a>). The combined effect allows for 5) an unrestricted activity of oxidative tissue LPL (regulated, however, by the apoCII/CIII balance) providing much-needed FA (6) for energy production in glucose-depleted muscle (7). The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 5
<p><b>Hepatic ANGPTL3 and 8 act in an endocrine fashion on muscle to divert VLDL and Chylomicrons to adipose tissue in the fed state.</b> Superimposed on the regulation of LPL activity by apoCII/CIII, there exists a finer control of the enzyme in different tissues that provides for the physiological partition of the TRL load depending on the needs of the body. Upon feeding LPL, activity in adipocytes is much higher, and at the same time, its activity is reduced in oxidative tissues. In a nutshell, ANGPTL3 (secreted all day long) from the liver acts in an endocrine way to inhibit lipoprotein lipase in muscle and heart during the postprandial period. During the fed or postprandial phase, high insulin levels (1) promote the uptake of glucose via GlutIV activation in adipose and muscle (2) as well as lipogenesis in the former (3). Feeding induces high expression of ANGPTL8 (4), which multiplies ANGPTL3 inhibition of LPL several-fold (5). TRL (VLDL and chylomicrons) are then preferentially acted upon on adipose (6). Feeding enhances ANGPTL8 synthesis and reduces ANGPTL4 production in adipose, which then liberates adipose LPL (7). Moreover, plasminogen is attracted to ANGPTL4/8 together with tPA, which turns plasminogen into plasmin. Without influencing the LPL activator APOCII, plasmin then cleaves LPL inhibitors such as ANGPTL3/8, A4, and APOCIII (8). Through this sequence of events, LPL activity is entirely restored locally in adipose, whereas ANGPTL3/8 can precisely block oxidative-tissue LPL (5). One should take note of the competition between intestinal chylomicrons and hepatic VLDL for hydrolysis by LPL, which happens mostly in the capillaries of adipose tissue. Fat is thus preferentially partitioned to adipose tissue for storage following a meal. Note that this regulation is a fine-tuning of the control provided by insulin and the rate of apoCII and apoCIII on TRL. We go into additional detail in this article regarding the potential significance of ANGPTL3 inhibitors as a treatment route for hypertriglyceridemia, given the critical role ANGPTL3 plays in this process and the findings of loss of function studies conducted on humans and animals. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 6
<p><b>Integrated current view of the ANGPTL3,4,8 axis on fine-tuning of LPL activity and fat partition in the fast-fed cycle</b>. (1) The liver produces and secretes relatively steady levels of ANGPTL3. In the fasting state (<b>left</b> column, A), ANGPTL8 is reduced (2), and therefore the levels of the active LPL inhibitor which is a 3/1 ANGPTL3/8 complex are greatly reduced (3). Additionally, fasting increases the hepatic output of apoAV and CREBH (cAMP response element-binding protein, hepatocyte-specific) (4), which inactivates the ANGPTL3/8 complex. Therefore, all inhibition of muscle LPL is lifted, and oxidative tissues receive FA from VLDL (5). In parallel, (6) and (7) fasting greatly increases the production of ANGPTL4 (and lowers ANGPTL8) whereby ANGPTL4 locally blocks adipose LPL (8), preventing VLDL hydrolysis and shunting VLDL to oxidative tissues. Potential futile cycles are prevented. In the fed state (<b>right</b> column, B), hepatic ANGPTL8 is greatly increased (1), and therefore the levels of the active LPL inhibitor, which is a 3/1 ANGPTL3/8 complex, are increased (2), acting as endocrinal mediators on LPL in oxidative tissues to block it (3). Additionally, feeding increases ANGPTL8 expression in adipose while reducing ANGPTL4 (4). The ANGPTL4/8 complex does not inhibit LPL (5) Therefore, all inhibition of adipose LPL is lifted (5), and adipocytes can receive FA from VLDL and chylomicrons for appropriate storage stimulated by high insulin levels (6). The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 7
<p><b>Interaction of LPL with TRL and HDL, active forms.</b> We represent chylomicrons here; a similar process occurs in VLDL. (1) Intestinal chylomicrons, after passage through lymph (2), reach tissue capillaries which express LPL anchored by GPIHBP1 on the luminal surface of the endothelial cells and which supports the configuration of LPL to an active lipolytic enzyme (3). Important interactions with HDL occur all along the hydrolytic process: exchange activators such as apoCII and apoAV and inhibitors such as apoCIII, cholesterol, and phospholipids, and the action of CETP are key (4). Note that, in parallel, some LPL molecules can travel directly on the TRL particle, where it keeps its activity as it achieves optimal interface (5) and the particles are reduced in size by loss of TG to tissues and surface PL, etc. to HDL (6). Remnants are acted upon by hepatic lipase (HL) and endothelial lipase (EL) (7) to be finally taken up by several hepatic receptors including the LDL-receptor (LDL-R). Uptake is facilitated by the presence of LPL in the particles (8). LRP-1: LDL-like receptor 1; SRB1: scavenger receptor B1. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 8
<p><b>The potential role of drugs targeting the ANGPTL3,4,8 axis in the treatment of TRL dyslipidemia.</b> In the past few years, elegant research on animals and loss of function studies in humans has earmarked both apoCIII and ANGPTL3 as pharmacological targets. Several trials are ongoing. ANGPTL3: Three approaches have been developed for the inhibition of ANGPTL3, ASO, siRNA, and mab. (1) Vupanorsen is an N-AcGal liver-targeted ASO of great effect but discontinued in late 2022. (2) Zodasiran is an ARO-ANG3, a N-AcGal liver-targeted siRNA and the latest addition to the armamentarium with two ongoing trials and (3) Evinacumab is a promising monoclonal antibody that provides up to 83% reduction in triglycerides and reduces LDL cholesterol. Elipse and two trials were completed, and two ongoing trials are taking place. Gene editing is feasible, and CRISPR has been conducted in animals but has obvious ethical issues (4). ANGPTL8: Given its pivotal role as a potential gatekeeper, intense research is being conducted at the preclinical level, so far (5). ANGPTL4 mAb is effective in TG reduction, though preclinical phase, obesity, and lymphadenopathy are concerns (6). The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">Figure 9
<p><b>Future directions for ANGPTL3, 4, and 8 research.</b> The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.</p>
Full article ">
11 pages, 630 KiB  
Systematic Review
Real-World Efficacy of Intravitreal Faricimab for Diabetic Macular Edema: A Systematic Review
by Safiullah Nasimi, Nasratullah Nasimi, Jakob Grauslund, Anna Stage Vergmann and Yousif Subhi
J. Pers. Med. 2024, 14(9), 913; https://doi.org/10.3390/jpm14090913 - 28 Aug 2024
Viewed by 877
Abstract
Background: Diabetic macular edema (DME) is a prevalent exudative maculopathy, and anti-vascular endothelial growth factor (anti-VEGF) therapy is the first-line choice for treatment. Faricimab, a novel anti-VEGF and anti-angiopoietin-2 bispecific agent, has recently been approved for the treatment of DME. In this study, [...] Read more.
Background: Diabetic macular edema (DME) is a prevalent exudative maculopathy, and anti-vascular endothelial growth factor (anti-VEGF) therapy is the first-line choice for treatment. Faricimab, a novel anti-VEGF and anti-angiopoietin-2 bispecific agent, has recently been approved for the treatment of DME. In this study, we systematically reviewed the real-world evidence of the efficacy of faricimab for the treatment of DME. Methods: We searched 11 databases for eligible studies. Study selection and data extraction were made independently by two authors in duplicate. Eligible studies were reviewed qualitatively. Results: We identified 10 eligible studies that summarized data from a total of 6054 eyes with a mean follow-up of between 55 days and 12 months. Five studies reported outcomes in a population of both treatment-naïve and previously treated eyes, and five studies reported outcomes exclusively in relation to eyes that were previously treated. Faricimab improved the best-corrected visual acuity and macular thickness. The extension of the treatment interval was possible in 61–81% of treatment-naïve eyes and 36–78% of previously treated eyes. Conclusions: Faricimab for DME yields clinical outcomes similar to those known from previous anti-VEGF treatments but with extended treatment intervals, thus lowering the burden of therapy for patients. Long-term real-world studies are warranted. Full article
(This article belongs to the Special Issue Personalized Diagnosis and Therapies in Retinal Diseases)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram of the study selection process. Number (n) of studies in each category is highlighted throughout the study selection process.</p>
Full article ">
16 pages, 4020 KiB  
Article
SPRi Biosensor for Simultaneous Determination of HIF-1α, Angiopoietin-2, and Interleukin-1β in Blood Plasma
by Zuzanna Zielinska, Lukasz Oldak, Tomasz Guszcz, Adam Hermanowicz and Ewa Gorodkiewicz
Sensors 2024, 24(17), 5481; https://doi.org/10.3390/s24175481 - 24 Aug 2024
Viewed by 688
Abstract
A new analytical method, based on SPRi biosensors, has been developed for the simultaneous determination of the pro-angiogenic factors HIF-1α, angiopoietin-2 (ANG-2), and interleukin-1β (IL-1β) in biological fluids. These proteins take part in the process of angiogenesis, i.e., the creation of new blood [...] Read more.
A new analytical method, based on SPRi biosensors, has been developed for the simultaneous determination of the pro-angiogenic factors HIF-1α, angiopoietin-2 (ANG-2), and interleukin-1β (IL-1β) in biological fluids. These proteins take part in the process of angiogenesis, i.e., the creation of new blood vessels, which is a key stage of cancer development and metastasis. A separate validation process was carried out for each individual compound, indicating that the method can also be used to study one selected protein. Low values of the limit of detection (LOD) and quantification (LOQ) indicate that the developed method enables the determination of very low concentrations, in the order of pg/mL. The LOD values obtained for HIF-1α, ANG-2, and IL-1β were 0.09, 0.01, and 0.01 pg/mL, respectively. The LOQ values were 0.27, 0.039, and 0.02 pg/mL, and the response ranges of the biosensor were 5.00–100.00, 1.00–20.00, and 1.00–15.00 pg/mL. Moreover, determining the appropriate validation parameters confirmed that the design offers high precision, accuracy, and sensitivity. To prove the usefulness of the biosensor in practice, determinations were made in plasma samples from a control group and from a study group consisting of patients with diagnosed bladder cancer. The preliminary results obtained indicate that this biosensor can be used for broader analyses of bladder cancer. Each of the potential biomarkers, HIF-1α, ANG-2, and IL-1β, produced higher concentrations in the study group than in the control group. These are preliminary studies that serve to develop hypotheses, and their confirmation requires the analysis of a larger number of samples. However, the constructed biosensor is characterized by its ease and speed of measurement, and the method does not require special preparation of samples. SPRi biosensors can be used as a sensitive and highly selective method for determining potential blood biomarkers, which in the future may become part of the routine diagnosis of cancers. Full article
(This article belongs to the Section Biosensors)
Show Figures

Figure 1

Figure 1
<p>HIF-1α protein structure [<a href="#B9-sensors-24-05481" class="html-bibr">9</a>].</p>
Full article ">Figure 2
<p>ANG-2 protein structure [<a href="#B18-sensors-24-05481" class="html-bibr">18</a>].</p>
Full article ">Figure 3
<p>IL-1β protein structure [<a href="#B33-sensors-24-05481" class="html-bibr">33</a>].</p>
Full article ">Figure 4
<p>Schematic diagram of the SPRi apparatus with device components. The LED laser emits a beam of light that passes through a system of polarizers and lenses and then hits a prism with a biosensor, a plate with a gold layer. During measurement, the light is collected by a CCD camera and sent to a computer. Own elaboration.</p>
Full article ">Figure 5
<p>Construction of the biosensor for the determination of HIF-1α, ANG-2, and IL-1β (A—BK7 glass layer; B—titanium layer; C—gold layer; D—polymer foil; E—thiol–mercaptoundecanoic acid 11-MUA; F<sub>1</sub>—IL-1β-specific antibody; F<sub>2</sub>—ANG-2-specific antibody; F<sub>3</sub>—HIF-1α-specific antibody; G<sub>1</sub>—IL-1β protein; G<sub>2</sub>—ANG-2 protein; G<sub>3</sub>—HIF-1α protein). Each generated receptor layer (thiol–antibody) is separate for a given protein. In this way, three measurement spots for detecting each protein were created on one chip, which can be determined simultaneously in one measurement cycle. Own elaboration.</p>
Full article ">Figure 6
<p>The course of model SPR curves. For each of the distinguished layers present on the biosensor surface, data were collected at angles from 34 to 37 degrees, in 0.1-degree increments. Each of the curves for a given layer is shifted towards larger angle values relative to the curve of the preceding layer.</p>
Full article ">Figure 7
<p>HIF-1α, ANG-2, and IL-1β calibration curves. Ligand/antibody concentration for HIF-1α = 50.00 ng/mL, ligand/antibody concentration for ANG-2 = 5.00 μg/mL, and ligand/antibody concentration for IL-1β = 100.00 ng/mL. Calibration curves were prepared using standard solutions prepared in PBS buffer.</p>
Full article ">Figure A1
<p>Steps for immobilization of the antibody layer (illustration by the authors). To immobilize the antibody on the gold surface with a thiol layer, a mixture of EDC and NHS is applied in a 1:1 volume ratio. The carboxyl group of the thiol is converted into an ester group by EDC, while NHS modifies the ester into a short-lived NHS-ester. In the next step, the applied antibody forms a covalent amide bond with the NHS-ester. To prevent non-specific adsorption, ethanolamine is applied, which deactivates active NHS-esters and also prevents the attachment of other components to thiol molecules that have not bound to the antibody.</p>
Full article ">Figure A2
<p>Antibody saturation curve for HIF-1α.</p>
Full article ">Figure A3
<p>Antibody saturation curve for ANG-2.</p>
Full article ">Figure A4
<p>Antibody saturation curve for IL-1β.</p>
Full article ">
15 pages, 2235 KiB  
Article
The Impact of Liver Steatosis on Interleukin and Growth Factors Kinetics during Chronic Hepatitis C Treatment
by Leona Radmanic Matotek, Snjezana Zidovec-Lepej, Nikolina Salek, Adriana Vince and Neven Papic
J. Clin. Med. 2024, 13(16), 4849; https://doi.org/10.3390/jcm13164849 - 16 Aug 2024
Viewed by 749
Abstract
Background/Objectives: Various biological response modifiers play important roles in the immunopathogenesis of chronic hepatitis C (CHC). While serum levels of cytokines and growth factors change with the disease severity and treatment responses, the impact of concomitant liver steatosis on systemic inflammatory response is [...] Read more.
Background/Objectives: Various biological response modifiers play important roles in the immunopathogenesis of chronic hepatitis C (CHC). While serum levels of cytokines and growth factors change with the disease severity and treatment responses, the impact of concomitant liver steatosis on systemic inflammatory response is largely unknown. The aim of this study was to analyze the characteristics and kinetics of serum profiles of interleukins and growth factors in CHC patients with steatotic liver disease (SLD). Methods: Serum concentrations of 12 cytokines (IL-5, IL-13, IL-2, IL-6, IL-9, IL-10, IFN-γ, TNF-α, IL-17A, IL-17F, IL-4 and IL-22) and 6 growth factors (Angiopoietin-2, EGF, EPO, HGF, SCF, VEGF) were analyzed in 56 CHC patients at four time points (baseline, week 4, week 8 and SVR12) with bead-based flow cytometry assay. Results: At baseline, patients with SLD had significantly lower IL-9, IL-10, IL-13 and IL-22 and higher serum concentrations of EGF, VEGF and ANG. In a subgroup of patients with advanced liver fibrosis, SLD was linked with lower serum concentrations of IL-4, IL-5, IL-9, IL-10, IL-13 and IL-22 and higher concentrations of HGH and VEGF. Distinct cytokine kinetics during DAA treatment was observed, and SLD was identified as the main source of variation for IL-5, IL-9, IL-10, IL-13, IL-17A, IL-22, EGF, VEGF and ANG. Patients with SLD at SVR12 had significantly higher VEGF and HGF serum concentrations. Conclusions: SLD is associated with distinct cytokine and growth factor profiles and kinetics during CHC treatment, which might be associated with disease severity and the capacity for liver regeneration and contribute to fibrosis persistence. Full article
(This article belongs to the Special Issue The Latest Advances and Challenges in Viral Hepatitis)
Show Figures

Figure 1

Figure 1
<p>Baseline cytokine and growth factor serum concentrations according to presence of steatosis, stratified by fibrosis stage. Shown are medians with IQR, and difference significance is calculated by the Mann–Whitney test. *: <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. Abbreviations: interleukine, IL; Epidermal growth factor, EGF; Hepatocyte growth factor, HGF; Vascular endothelial growth factor, VEGF.</p>
Full article ">Figure 2
<p>Spearman’s correlation correlogram. The strength of the correlation between two variables is represented by the color at the intersection of those variables. Colors range from dark blue (strong negative correlation; r = −1.0) to red (strong positive correlation; r = 1.0).</p>
Full article ">Figure 3
<p>Comparison of cytokine and growth factor serum concentrations at four selected time points (before treatment, at weeks (wks) 4 and 8 of treatment and at time of sustained virological response, SVR), stratified by the presence of liver steatosis. Medians with IQRs are shown. Repeated measures two-way ANOVA with Tukey’s multiple comparisons test was used to calculate the source of the variations. *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01; ***: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Comparison of growth factor and cytokine concentrations at four selected time points (before treatment, at weeks (wks) 4 and 8 of treatment and at time of sustained virological response. SVR), stratified by the presence of liver steatosis and liver fibrosis (F0–2 vs. F3,4). Medians with IQRs are shown. Repeated measures three-way ANOVA with Tukey’s multiple comparisons test was used to calculate the source of variations. <span class="html-italic">p</span>-values are shown; <span class="html-italic">p</span> &lt; 0.05 is considered significant.</p>
Full article ">Figure 5
<p>Comparison of growth factor concentrations before treatment and at SVR12 stratified by the presence of liver steatosis at SVR12. A Wilcoxon rank sum test was performed to analyze the differences in time within the groups, and the Mann–Whitney test analyzed the differences between the groups. *: <span class="html-italic">p</span> &lt; 0.05; ****: <span class="html-italic">p</span> &lt; 0.0001. Abbreviations: Epidermal growth factor, EGF; Hepatocyte growth factor, HGF; Vascular endothelial growth factor, VEGF; sustained virological response, SVR.</p>
Full article ">
28 pages, 1193 KiB  
Review
Effects of Angiogenic Factors on the Epithelial-to-Mesenchymal Transition and Their Impact on the Onset and Progression of Oral Squamous Cell Carcinoma: An Overview
by Silvia Pomella, Ombretta Melaiu, Maria Dri, Mirko Martelli, Marco Gargari and Giovanni Barillari
Cells 2024, 13(15), 1294; https://doi.org/10.3390/cells13151294 - 31 Jul 2024
Viewed by 1191
Abstract
High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development [...] Read more.
High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development of new blood and lymphatic vessels that nourish the growing OPMDs or OSCCs and provide the latter with metastatic routes. Notably, VEGF, FGF-2, and ANG-2 are also linked to the epithelial-to-mesenchymal transition (EMT), a trans-differentiation process that respectively promotes or exasperates the invasiveness of normal and neoplastic oral epithelial cells. Here, we have summarized published work regarding the impact that the interplay among VEGF, FGF-2, ANG-2, vessel generation, and EMT has on oral carcinogenesis. Results from the reviewed studies indicate that VEGF, FGF-2, and ANG-2 spark either protein kinase B (AKT) or mitogen-activated protein kinases (MAPK), two signaling pathways that can promote both EMT and new vessels’ formation in OPMDs and OSCCs. Since EMT and vessel generation are key to the onset and progression of OSCC, as well as to its radio- and chemo-resistance, these data encourage including AKT or MAPK inhibitors and/or antiangiogenic drugs in the treatment of this malignancy. Full article
Show Figures

Figure 1

Figure 1
<p>Molecular mechanisms leading to EMT in oral mucosa. Arrows symbolize directions of connections. Abbreviations: AKT, protein kinase B; E-cadherin, epithelial-cadherin; EGF, epidermal growth factor; EMT, epithelial-to-mesenchymal transition; GLUT, glucose transporter protein; HIF, hypoxia-inducible factor; IC, inflammatory cytokines; MAPK, mitogen-activated protein kinase; MMPs, matrix metalloproteinases; N-cadherin, neuronal-cadherin; NF-κB, nuclear factor kappa B; OPMDs, oral potentially malignant disorders; OSCC, oral squamous cell carcinoma; SNAI, zinc finger snail homolog; TGF, transforming growth factor; TWIST, basic helix–loop–helix twist homolog; VEGF, vascular endothelial growth factor; ZEB, zinc finger E-box-binding homeobox. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>FGF-2 promotes EMT in oral mucosa. Arrows symbolize directions of connections. Abbreviations: AKT, protein kinase B; CSCs, cancer stem cells; EMT, epithelial-to-mesenchymal transition; FGF, fibroblast growth factor; GLUT, glucose transporter protein; HIF, hypoxia-inducible factor; MAPK, mitogen-activated protein kinase; MMPs, matrix metalloproteinases; OSCC, oral squamous cell carcinoma; SNAI, zinc finger snail homolog; TWIST, basic helix–loop–helix twist homolog; VEGF, vascular endothelial growth factor; ZEB, zinc finger E-box-binding homeobox. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
16 pages, 4917 KiB  
Article
Eucalyptol Ameliorates Retinal Microvascular Defects through Modulating ER Stress and Angiopoietin–Tie Signaling in Diabetic Eyes
by Dong Yeon Kim, Sin-Hye Park, Zaee Yoon, Jimin Kim, Min-Kyung Kang and Young-Hee Kang
Int. J. Mol. Sci. 2024, 25(14), 7826; https://doi.org/10.3390/ijms25147826 - 17 Jul 2024
Cited by 1 | Viewed by 941
Abstract
Loss of the inner blood–retinal barrier (BRB) integrity is a main feature of ocular diseases such as diabetic macular edema. However, there is a lack of clarity on how inner BRB function is modulated within the diabetic retina. The current study examined whether [...] Read more.
Loss of the inner blood–retinal barrier (BRB) integrity is a main feature of ocular diseases such as diabetic macular edema. However, there is a lack of clarity on how inner BRB function is modulated within the diabetic retina. The current study examined whether eucalyptol inhibited inner BRB destruction and aberrant retinal angiogenesis in 33 mM glucose-exposed human retinal microvascular endothelial (RVE) cells and db/db mice. This study further examined the molecular mechanisms underlying endothelial dysfunction including retinal endoplasmic reticulum (ER) stress and angiopoietin (Ang)/Tie axis in conjunction with vascular endothelial growth factor (VEGF). Eucalyptol is a naturally occurring monoterpenoid and an achiral aromatic component of many plants including eucalyptus leaves. Nontoxic eucalyptol reduced the production of amyloid-β (Aβ) protein in glucose-loaded RVE cells and in diabetic mice. This natural compound blocked apoptosis of Aβ-exposed RVE cells in diabetic mouse eyes by targeting ER stress via the inhibition of PERK-eIF2α-ATF4-CHOP signaling. Eucalyptol promoted activation of the Ang-1/Tie-2 pathway and dual inhibition of Ang-2/VEGF in Aβ-exposed RVE cells and in diabetic eyes. Supply of eucalyptol reversed the induction of junction proteins in glucose/Aβ-exposed RVE cells within the retina and reduced permeability. In addition, oral administration of eucalyptol reduced vascular leaks in diabetic retinal vessels. Taken together, these findings clearly show that eucalyptol inhibits glucose-induced Aβ-mediated ER stress and manipulates Ang signaling in diabetic retinal vessels, which ultimately blocks abnormal angiogenesis and loss of inner BRB integrity. Therefore, eucalyptol provides new treatment strategies for diabetes-associated RVE defects through modulating diverse therapeutic targets including ER stress, Ang-1/Tie-2 signaling, and Ang-2/VEGF. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chemical structure of eucalyptol (<b>A</b>), eucalyptol cytotoxicity (<b>B</b>), eucalyptol viability in high glucose-exposed human retinal microvascular endothelial (RVE) cells (<b>C</b>) and amyloid-β (Aβ)-exposed RVE cells (<b>D</b>), temporal responses of Aβ to glucose (<b>E</b>), and the inhibitory effect of eucalyptol on Aβ induction (<b>F</b>). Human RVE cells were cultured in 33 mM glucose media or with Aβ in the absence and presence of 1–20 μM eucalyptol for up to 5 days. Cells were also incubated with 5.5 mM glucose plus 27.5 mM mannitol as osmotic controls. Cell viability was measured by MTT assay (<b>B</b>–<b>D</b>). Bar graphs for viability (mean ± SEM, n = 5) were expressed as percent cell survival, compared to glucose control. Whole-cell lysates were subject to SDS-PAGE and Western blot with a specific antibody against Aβ (<b>E</b>,<b>F</b>). β-Actin protein was used as a cellular internal control of RVE cells. Bar graphs (mean ± SEM, n = 3) in the bottom panels represent densitometric results of upper blot bands. The db/db mice were orally supplemented with 10 mg/kg eucalyptol daily for 8 weeks. Immunohistochemical staining was performed to visualize the induction of Aβ in diabetic retinal tissues (<b>G</b>). Green fluorescein isothiocyanate (FITC)-conjugated secondary antibody was used for visualizing Aβ, being counterstained with 4′,6-diamidino-2-phenylindole for the blue nuclear staining. Each microphotograph (mean ± SEM, n = 3) was obtained by using an Axiomager microscope system. Scale bar = 25 μm. All the respective values do not share a common letter difference at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Effects of eucalyptol on induction of bcl-2, bax and cleaved caspase-12 (<b>A</b>), and apoptotic DNA fragmentation (<b>B</b>). Human retinal endothelial cells were treated with 1–20 μM eucalyptol for 3 days in culture media containing 5 μM amyloid-β (Aβ). Cells were also incubated in 33 mM glucose media. Whole-cell lysates were subject to SDS-PAGE and Western blot with a specific antibody against bcl-2, bax, or cleaved caspase-12 (<b>A</b>). β-Actin protein was used as a cellular internal control of RVE cells. The bar graphs (mean ± SEM, n = 3) represent quantitative results of blots in the left panel obtained from a densitometer. DNA fragmentation was measured with a TUNEL assay and nuclear counterstaining was done with blue-emitting fluorescent 4′,6-diamidino-2-phenylindole (<b>B</b>). Representative microphotographs were obtained by fluorescent microscopy with a fluorescein green filter. Fluorescence intensity was quantified by using an Axiomager microscope system. Scale bar = 100 μm. All the respective values not sharing a letter are different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Reciprocal changes in retinal tissue levels of bcl-2 and bax in eucalyptol-supplemented db/db mice. The db/db mice were orally supplemented with 10 mg/kg eucalyptol daily for 8 weeks. Immunohistochemical staining of bcl-2 and bax was performed to visualize the induction of these proteins in diabetic retinas by using green fluorescein isothiocyanate (FITC)-conjugated secondary bcl-2 antibody and red Cy3-conjugated secondary bax antibody. Nuclear staining was conducted using blue 4′,6-diamidino-2-phenylindole (DAPI). Representative microphotographs were obtained using fluorescent microscopy with fluorescein green or red filters. Scale bar = 25 μm. The relative staining intensity was measured, and the respective values are expressed as mean ± SEM (n = 3 in each group). Values in bar graphs not sharing a letter indicate significant different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Western blot data showing inhibition of protein kinase R-like ER kinase (PERK) and eukaryotic initiation factor 2 alpha (eIF2α) activation (<b>A</b>), activating transcription factor-4 (ATF4) induction (<b>B</b>), and C/EBPα-homologous protein (CHOP) expression (<b>C</b>) by eucalyptol. Human retinal endothelial (RVE) cells were treated with 1–20 μM eucalyptol for 3 days in culture media containing 5 μM amyloid-β (Aβ). Cells were also incubated in 33 mM glucose media. Whole-cell lysates were subject to SDS-PAGE and Western blot with a specific antibody against PERK, phospho-PERK, elF2α, phospho-eIF2α, ATF4, or CHOP (<b>A</b>–<b>C</b>). β-Actin protein was used as a cellular internal control of RVE cells. The bar graphs (mean ± SEM, n = 3) represent quantitative results of blots in the upper panels obtained from a densitometer. Respective values not sharing a letter are different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Inhibition of induction of phospho-protein kinase R-like ER kinase (PERK), phospho-eukaryotic initiation factor 2 alpha (eIF2α), activating transcription factor-4 (ATF4) and C/EBPα-homologous protein (CHOP) in eyes by eucalyptol. The db/db mice were orally supplemented with 10 mg/kg eucalyptol daily for 8 weeks. Whole eye tissue extracts were subject to SDS-PAGE and Western blot with a specific antibody against phospho-PERK, phospho-eIF2α, ATF4, or CHOP (<b>A</b>–<b>C</b>). β-Actin protein was used as an internal control of eye tissue extracts. The bar graphs (mean ± SEM, n = 3) represent quantitative results of blots in the upper panels obtained from a densitometer. Respective values not sharing a letter are different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Effects of eucalyptol on expression of VEGF and induction of angiopoietin (Ang)-1, Ang-2, and Tie-2. Human retinal endothelial (RVE) cells were treated with 1–20 μM eucalyptol for 3 days in culture media containing 5 μM amyloid-β (Aβ). Cells were also incubated in 33 mM glucose media. Whole-cell lysates were subject to SDS-PAGE and Western blot with a specific antibody against VEGF, Ang-1, Ang-2, or Tie-2 (<b>A</b>–<b>C</b>). β-Actin protein was used as a cellular internal control of RVE cells. The bar graphs (mean ± SEM, n = 3) represent quantitative results of blots in the upper panels obtained from a densitometer. Respective values not sharing a letter are different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Western blot data showing eye tissue levels of VEGF, angiopoietin (Ang)-1, Ang-2, and Tie-2 in eucalyptol-supplemented db/db mice. The db/db mice were orally supplemented with 10 mg/kg eucalyptol daily for 8 weeks. Whole eye tissue extracts were subject to SDS-PAGE and Western blot with a specific antibody against VEGF, Ang-1, Ang-2, and Tie-2 (<b>A</b>–<b>C</b>). β-Actin protein was used as an internal control of eye tissue extracts. The bar graphs (mean ± SEM, n = 3) represent quantitative results of blots in the upper panels obtained from a densitometer. Respective values not sharing a letter are different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 8
<p>Effects of eucalyptol on induction of VE-cadherin and occludin-1 (<b>A</b>,<b>B</b>), and vascular permeability (<b>C</b>). Human retinal endothelial (RVE) cells were treated with 1–20 μM eucalyptol for 3 days in culture media containing 5 μM amyloid-β (Aβ). The db/db mice were orally supplemented with 10 mg/kg eucalyptol daily for 8 weeks. Whole-cell lysates and eye tissue extracts were subject to SDS-PAGE and Western blot with a specific antibody against VE-cadherin and occludin-1 (<b>A</b>,<b>B</b>). β-Actin protein was used as an internal control of RVE cells or eye tissue extracts. The bar graphs (mean ± SEM, n = 3) represent quantitative results of blots in the upper panels obtained from a densitometer. For the measurement of RVE injury, the albumin permeability assay was performed with fluorescein isothiocyanate (FITC)-labeled BSA (<b>C</b>). Respective values not sharing a letter are different at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>Inhibition of microvascular endothelial leakage by eucalyptol. The db/db mice were orally supplemented with 10 mg/kg eucalyptol daily for 8 weeks. The images below are magnified images within a square of each of the images above. Retinas were dissected, flat-mounted, and observed by confocal microscopy. Leakage of the green-colored fluorescent fluorescein isothiocyanate (FITC)-conjugated dextran appeared in diabetic retinal vessels (red arrows). Scale bar = 100 μm.</p>
Full article ">
11 pages, 1262 KiB  
Article
Analysis of Expression of the ANG1, CaSR and FAK Proteins in Uterine Fibroids
by Anna Markowska, Mateusz de Mezer, Paweł Kurzawa, Wiesława Bednarek, Anna Gryboś, Monika Krzyżaniak, Janina Markowska, Marian Gryboś and Jakub Żurawski
Int. J. Mol. Sci. 2024, 25(13), 7164; https://doi.org/10.3390/ijms25137164 - 28 Jun 2024
Viewed by 1014
Abstract
Understanding the molecular factors involved in the development of uterine myomas may result in the use of pharmacological drugs instead of aggressive surgical treatment. ANG1, CaSR, and FAK were examined in myoma and peripheral tissue samples taken from women after myoma surgery and [...] Read more.
Understanding the molecular factors involved in the development of uterine myomas may result in the use of pharmacological drugs instead of aggressive surgical treatment. ANG1, CaSR, and FAK were examined in myoma and peripheral tissue samples taken from women after myoma surgery and in normal uterine muscle tissue samples taken from the control group. Tests were performed using tissue microarray immunohistochemistry. No statistically significant differences in ANG1 expression between the tissue of the myoma, the periphery, and the normal uterine muscle tissue of the control group were recorded. The CaSR value was reduced in the myoma and peripheral tissue and normal in the group of women without myomas. FAK expression was also lower in the myoma and periphery compared to the healthy uterine myometrium. Calcium supplementation could have an effect on stopping the growth of myomas. Full article
Show Figures

Figure 1

Figure 1
<p>Immunohistochemical expression of the analyzed protein in the uterus of the same patients: ANG1 (<b>A</b>), CaSR (<b>B</b>), and FAK (<b>C</b>). Magnification for all pictures is 400×.</p>
Full article ">Figure 2
<p>The boxplot for ANG1 (<b>A</b>), CaSR (<b>B</b>), and FAK (<b>C</b>) shows the comparison of expression levels determined by measurement of the intensity of stain resulting from the attachment of the appropriate antibody between the tumor center, the tumor periphery, and the control group.</p>
Full article ">
11 pages, 1456 KiB  
Article
Differential Impact of Tumor Endothelial Angiopoietin-2 and Podoplanin in Lymphatic Endothelial Cells on HCC Outcomes with Tyrosine Kinase Inhibitor Treatment According to Sex
by Simone Lasagni, Rosina Maria Critelli, Fabiola Milosa, Dario Saltini, Filippo Schepis, Adriana Romanzi, Francesco Dituri, Grazia Serino, Lorenza Di Marco, Alessandra Pivetti, Filippo Scianò, Gianluigi Giannelli and Erica Villa
Biomedicines 2024, 12(7), 1424; https://doi.org/10.3390/biomedicines12071424 - 26 Jun 2024
Viewed by 1254
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. Curative treatments are available to a minority of patients, as HCC is often diagnosed at an advanced stage. For patients with unresectable and multifocal HCC, tyrosine kinase inhibitor drugs (TKIs) are [...] Read more.
Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. Curative treatments are available to a minority of patients, as HCC is often diagnosed at an advanced stage. For patients with unresectable and multifocal HCC, tyrosine kinase inhibitor drugs (TKIs) are the only potential treatment option. Despite extensive research, predictors of response to these therapies remain elusive. This study aimed to analyze the biological and histopathological characteristics of HCC patients treated with TKIs, focusing on angiogenesis and lymphangiogenesis. Immunohistochemistry quantified the expression of angiopoietin-2 (Ang2), lymphatic endothelial cells (LEC) podoplanin, and C-type Lectin Domain Family 2 (CLEC-2), key factors in neoangiogenesis and lymphangiogenesis. An increased expression of endothelial Ang2 and LEC podoplanin predicted a lower risk of metastasis. Female patients had significantly longer overall survival and survival on TKIs, associated with higher tumor expression of endothelial Ang2 and LEC podoplanin. Moreover, LEC podoplanin expression and a longer time on TKIs were independently correlated with improved survival on TKI therapy at Cox regression analysis. These findings suggest that endothelial Ang2 and LEC podoplanin could be potential biomarkers for predicting treatment outcomes and guiding therapeutic strategies in HCC patients treated with TKIs. Full article
(This article belongs to the Special Issue Recent Advances of Receptor Tyrosine Kinases in Solid Tumors)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier analysis of overall survival from time of HCC diagnosis to death (<b>A</b>) and survival time from starting TKIs to death (<b>B</b>). For both survival analyses, females showed a significantly longer survival. Most importantly, females showed a significantly higher survival on TKIs.</p>
Full article ">Figure 2
<p>Comparison of Ang2, podoplanin, and CLEC-2 staining in female and male patients treated with TKIs (50 µm scale bar). (<b>A</b>) 63× image of Ang2 staining in tumor and endothelial tissue (red arrows for endothelia) of a female patient. (<b>B</b>) 63× image of Ang2 staining in tumor and endothelial tissue (red arrows for endothelia) of a male patient. (<b>C</b>) 63× image of podoplanin staining at the level of the lymphatic endothelium in a female patient. (<b>D</b>) 63× image of podoplanin staining at the level of the lymphatic endothelium in a male patient. (<b>E</b>) 63× image of CLEC-2 staining at the level of tumor tissue in a female patient. (<b>F</b>) 63× image of CLEC-2 staining at the level of tumor tissue in a male patient.</p>
Full article ">Figure 3
<p>Receiver operating characteristic (ROC) curve analysis to evaluate the diagnostic power of the LEC podoplanin (<b>A</b>) and of endothelial Ang2 (<b>B</b>) to predict extra-hepatic spread. The area under the ROC curves (AUCs) was analyzed using the Hanley and McNeil method [<a href="#B21-biomedicines-12-01424" class="html-bibr">21</a>]. The AUC values were 0.839 (95% CI 0.679 to 0.999; <span class="html-italic">p</span> &lt; 0.0001) and 0.821 (95% CI 0.652 to 0.989; <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
Back to TopTop