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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (7,981)

Search Parameters:
Keywords = remodeling

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 1035 KiB  
Article
CCAAT/Enhancer-Binding Protein β (C/EBPβ) Regulates Calcium Deposition in Smooth Muscle Cells
by Nakwon Choe, Sera Shin, Young-Kook Kim, Hyun Kook and Duk-Hwa Kwon
Int. J. Mol. Sci. 2024, 25(24), 13667; https://doi.org/10.3390/ijms252413667 (registering DOI) - 20 Dec 2024
Abstract
Calcium deposition in vascular smooth muscle cells (VSMCs), a form of ectopic ossification in blood vessels, can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that CCAAT/enhancer-binding protein beta (C/EBPβ) potentiates calcium deposition in VSMCs and [...] Read more.
Calcium deposition in vascular smooth muscle cells (VSMCs), a form of ectopic ossification in blood vessels, can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that CCAAT/enhancer-binding protein beta (C/EBPβ) potentiates calcium deposition in VSMCs and mouse aorta induced by inorganic phosphate (Pi) or vitamin D3. Based on cDNA microarray and RNA sequencing data of Pi-treated rat VSMCs, C/EBPβ was found to be upregulated and thus selected for further evaluation. Quantitative RT-PCR and Western blot analysis confirmed that C/EBPβ was upregulated in Pi-treated A10 cells, a rat VSMC line, as well as vitamin D3-treated mouse aorta. The overexpression of C/EBPβ in A10 cells increased bone runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), and osteopontin (OPN) mRNA in the presence of Pi, as well as potentiating the Pi-induced increase in calcium contents. The Runx2 expression was increased by C/EBPβ through Runx2 P2 promotor. Our results suggest that a Pi-induced increase in C/EBPβ is a critical step in vascular calcification. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
22 pages, 2994 KiB  
Review
Apolipoprotein-L Functions in Membrane Remodeling
by Etienne Pays
Cells 2024, 13(24), 2115; https://doi.org/10.3390/cells13242115 - 20 Dec 2024
Abstract
The mammalian Apolipoprotein-L families (APOLs) contain several isoforms of membrane-interacting proteins, some of which are involved in the control of membrane dynamics (traffic, fission and fusion). Specifically, human APOL1 and APOL3 appear to control membrane remodeling linked to pathogen infection. Through its association [...] Read more.
The mammalian Apolipoprotein-L families (APOLs) contain several isoforms of membrane-interacting proteins, some of which are involved in the control of membrane dynamics (traffic, fission and fusion). Specifically, human APOL1 and APOL3 appear to control membrane remodeling linked to pathogen infection. Through its association with Non-Muscular Myosin-2A (NM2A), APOL1 controls Golgi-derived trafficking of vesicles carrying the lipid scramblase Autophagy-9A (ATG9A). These vesicles deliver APOL3 together with phosphatidylinositol-4-kinase-B (PI4KB) and activated Stimulator of Interferon Genes (STING) to mitochondrion–endoplasmic reticulum (ER) contact sites (MERCSs) for the induction and completion of mitophagy and apoptosis. Through direct interactions with PI4KB and PI4KB activity controllers (Neuronal Calcium Sensor-1, or NCS1, Calneuron-1, or CALN1, and ADP-Ribosylation Factor-1, or ARF1), APOL3 controls PI(4)P synthesis. PI(4)P is required for different processes linked to infection-induced inflammation: (i) STING activation at the Golgi and subsequent lysosomal degradation for inflammation termination; (ii) mitochondrion fission at MERCSs for induction of mitophagy and apoptosis; and (iii) phagolysosome formation for antigen processing. In addition, APOL3 governs mitophagosome fusion with endolysosomes for mitophagy completion, and the APOL3-like murine APOL7C is involved in phagosome permeabilization linked to antigen cross-presentation in dendritic cells. Similarly, APOL3 can induce the fusion of intracellular bacterial membranes, and a role in membrane fusion can also be proposed for endothelial APOLd1 and adipocyte mAPOL6, which promote angiogenesis and adipogenesis, respectively, under inflammatory conditions. Thus, different APOL isoforms play distinct roles in membrane remodeling associated with inflammation. Full article
(This article belongs to the Special Issue Evolution, Structure, and Functions of Apolipoproteins L)
Show Figures

Figure 1

Figure 1
<p>Structural features of APOL1 and APOL3. The colored cylinders represent different α-helices, some of which are numbered, according to Ultsch et al. [<a href="#B5-cells-13-02115" class="html-bibr">5</a>]. HC1, HC2 = hydrophobic clusters 1, 2; LZ1, LZ2 = leucine zippers 1, 2; CRAC-1, CRAC-2 = cholesterol recognition amino acid consensuses 1, 2 (represented by red stars); TM = potential transmembrane hairpin helix; MAD = membrane-addressing domain. At acidic pH, the APOL1 TM hairpin can form weak anion pores, but pH neutralization confers high cation conductance. HC2 amino acids involved in pore pH-gating are highlighted in yellow. The boxes illustrate the folding of the N- and C-terminal APOL1 domains. In the isolated N-terminal domain, helix 5 can adopt two positions, preventing (bound) or not preventing (open) helix 4 accessibility [<a href="#B5-cells-13-02115" class="html-bibr">5</a>]. APOL1 SID represents the Smallest Interacting Domain between N- and C-terminal regions. This interaction, driven by LZ1-LZ2 pairing, is affected either by acidic conditions, as in trypanosome endosomes, or by LZ2 mutations, as in the natural G1 or G2 variants. In APOL3, LZ2 interacts with helix 5.</p>
Full article ">Figure 2
<p>APOL2 sequence comparison with APOL1 and APOL3. Hydrophobic residues characterizing HC2 and LZ2 are highlighted in violet and pink, respectively. APOL1 CRAC-2 residues are boxed. Key APOL2 HC2 and LZ2 differences from APOL1 are in orange and red, respectively. The boxed sequence alignments show antisense pairing between helix 5 and LZ2, based on hydrophobic heptad repeats (highlighted in green).</p>
Full article ">Figure 3
<p>WT or C-terminal variant APOL1 interactions and activities. The same symbols and colors as in <a href="#cells-13-02115-f001" class="html-fig">Figure 1</a>. In the last scheme, hypothetical cation driving to the membrane pore at neutral pH [<a href="#B14-cells-13-02115" class="html-bibr">14</a>] is symbolized by a dotted red arrow.</p>
Full article ">Figure 4
<p>APOL3 interactions and activities. The same symbols, colors and numbers as in <a href="#cells-13-02115-f001" class="html-fig">Figure 1</a>. NCS1 and CALN1 are alternative APOL3 binders activating or inhibiting PI4KB, depending on calcium concentration. ARF1 binds to APOL3, and inflammation-mediated ARF1 activation promotes its binding to PI4KB, possibly dissociating APOL3-PI4KB interaction. VAMP8 interacts with both helices 4–5 and MAD, promoting membrane fusion.</p>
Full article ">Figure 5
<p>Intracellular traffic of proteins involved in infection-induced changes in membrane dynamics. Detection of pathogen DNA triggers the synthesis of cyclic GMP-AMP (cGAMP), which binds to STING and disrupts STING-cholesterol interactions, allowing STING binding to PI(4)P for translocation to the Golgi. In the Golgi, STING undergoes oligomerization, which induces IFN-I inflammatory signaling. IFN-I activates ARF1, leading to STING, PI4KB and APOL3 dissociation from the Golgi in ATG9A vesicles trafficking to MERCSs, promoting membrane fission and fusion events linked to auto/mitophagy and apoptosis. This pathway allows inflammation termination due to STING autophagic degradation. Through association with NM2A and PHB2, APOL1 could direct ATG9A vesicles to MERCSs, where mitophagy is initiated. Red stars represent cholesterol interactions. The double-arrowed black dotted line represents the involvement of endolysosomes in both mitochondrion fission and autophagosome formation by ATG9A vesicles.</p>
Full article ">Figure 6
<p>Sequence alignment between human APOL3 (above) and mouse APOL7c (below), using Clustal Omega (<a href="https://www.ebi.ac.uk/Tools/msa/clustalo/" target="_blank">https://www.ebi.ac.uk/Tools/msa/clustalo/</a> (accessed on 4 November 2024)). Insertion of clustered acidic residues, highlighted in red, characterizes the murine APOL7 family.</p>
Full article ">Figure 7
<p>The two APOL1-like domains of APOLd1. Positively charged residues of helix 5 are highlighted in red, and the two helices of the putative transmembrane domain are highlighted in blue, with acidic residues in green. Hydrophobic residues characterizing the HC2 and LZ2 helices are highlighted in violet and pink, respectively. The amino acids involved in pH gating of the APOL1 pore are highlighted in yellow. The APOL1 residues defining CRAC-2 are boxed, and the loop sequences between the two helices of the double-stranded HC2-LZ2 helix hairpin are in bold. The boxed sequence alignment shows antisense pairing between APOLd1 helix 5 and LZ2, based on hydrophobic heptad repeats (highlighted in green).</p>
Full article ">
28 pages, 3739 KiB  
Review
The Interplay of Molecular Factors and Morphology in Human Placental Development and Implantation
by Ioana Vornic, Victor Buciu, Cristian George Furau, Flavia Zara, Dorin Novacescu, Alina Cristina Barb, Alin Adrian Cumpanas, Silviu Constantin Latcu, Ioan Sas, Denis Serban, Talida Georgiana Cut and Cristina Stefania Dumitru
Biomedicines 2024, 12(12), 2908; https://doi.org/10.3390/biomedicines12122908 - 20 Dec 2024
Abstract
The placenta is a vital organ that supports fetal development by mediating nutrient and gas exchange, regulating immune tolerance, and maintaining hormonal balance. Its formation and function are tightly linked to the processes of embryo implantation and the establishment of a robust placental-uterine [...] Read more.
The placenta is a vital organ that supports fetal development by mediating nutrient and gas exchange, regulating immune tolerance, and maintaining hormonal balance. Its formation and function are tightly linked to the processes of embryo implantation and the establishment of a robust placental-uterine interface. Recent advances in molecular biology and histopathology have shed light on the key regulatory factors governing these processes, including trophoblast invasion, spiral artery remodeling, and the development of chorionic villi. This review integrates morphological and molecular perspectives on placental development, emphasizing the roles of cytokines, growth factors, and signaling pathways, such as VEGF and Notch signaling, in orchestrating implantation and placental formation. The intricate interplay between molecular regulation and morphological adaptations highlights the placenta’s critical role as a dynamic interface in pregnancy. This review synthesizes current findings to offer clinicians and researchers a comprehensive understanding of the placenta’s role in implantation, emphasizing its importance in maternal-fetal medicine. By integrating these insights, the review lays the groundwork for advancing diagnostic and therapeutic approaches that can enhance pregnancy outcomes and address related complications effectively. Full article
(This article belongs to the Special Issue Role of Factors in Embryo Implantation and Placental Development)
Show Figures

Figure 1

Figure 1
<p>Development and Maturation of the Villous Tree.</p>
Full article ">Figure 2
<p>Histological section of the decidua at 20× magnification, showing large, polygonal decidual cells (indicated by red arrows) with abundant cytoplasm rich in glycogen and lipids.</p>
Full article ">Figure 3
<p>Histological section of the placenta at 20× magnification, illustrating the the structure of chorionic tertiary villi within the intervillous space. The image highlights various components essential for placental function, including the mesenchymal core (MC), cytotrophoblast cells (C), syncytiotrophoblast cells (S), decidual cells (D), and intervillous space (IV).</p>
Full article ">Figure 4
<p>Illustration of the developing placenta and early uteroplacental circulation. The left side shows an embryo within the amniotic cavity surrounded by the trophoblast and early placental structures. The right side zooms into the placental interface, highlighting the chorionic villi’s branching structure within the intervillous space, where maternal blood circulates to facilitate nutrient and gas exchange.</p>
Full article ">Figure 5
<p>The left image shows a placenta with a magistral pattern of chorionic plate vessels originating from a marginal insertion of the umbilical cord. In this configuration, blood vessels radiate from the edge of the placenta, distributing blood to the chorionic villi. The right image displays the maternal floor of a term placenta, revealing numerous well-defined cotyledons or lobules. These cotyledons, which are the functional units of the basal plate, contain chorionic villi essential for nutrient and gas exchange between maternal and fetal blood within the intervillous space. The organized arrangement of these cotyledons enhances placental efficiency by maximizing the surface area available for maternal-fetal exchange. Macroscopy images, representative of placental anatomy, obtained during the authors’ clinical practice within the Gynecology ward (private collection).</p>
Full article ">
14 pages, 1710 KiB  
Systematic Review
Histone Deacetylase Inhibitors as a Promising Treatment Against Myocardial Infarction: A Systematic Review
by Eduardo Sanchez-Fernandez, Sol Guerra-Ojeda, Andrea Suarez, Eva Serna and Maria D. Mauricio
J. Clin. Med. 2024, 13(24), 7797; https://doi.org/10.3390/jcm13247797 - 20 Dec 2024
Abstract
Background/Objectives: Acute myocardial infarction (AMI) is a critical medical condition that requires immediate attention to minimise heart damage and improve survival rates. Early identification and prompt treatment are essential to save the patient’s life. Currently, the treatment strategy focuses on restoring blood flow [...] Read more.
Background/Objectives: Acute myocardial infarction (AMI) is a critical medical condition that requires immediate attention to minimise heart damage and improve survival rates. Early identification and prompt treatment are essential to save the patient’s life. Currently, the treatment strategy focuses on restoring blood flow to the myocardium as quickly as possible. However, reperfusion activates several cellular cascades that contribute to organ dysfunction, resulting in the ischaemia/reperfusion (I/R) injury. The search for treatments against AMI and I/R injury is urgent due to the shortage of effective treatments at present. In this regard, histone deacetylase (HDAC) inhibitors emerge as a promising treatment against myocardial infarction. The objective of this systematic review is to analyse the effects of HDAC inhibitors on ventricular function, cardiac remodelling and infarct size, among other parameters, focusing on the signalling pathways that may mediate these cardiovascular effects and protect against AMI. Methods: Original experimental studies examining the effects of HDAC inhibitors on AMI were included in the review using the PubMed and Scopus databases. Non-experimental papers were excluded. The SYRCLE RoB tool was used to assess risk of bias and the results were summarised in a table and presented in sections according to the type of HDAC inhibitor used. Results: A total of 18 studies were included, 10 of them using trichostatin A (TSA) as an HDAC inhibitor and concluding that the treatment improved ventricular function, reduced infarct size, and inhibited myocardial hypertrophy and remodelling after AMI. Other HDAC inhibitors, such as suberoylanilide hydroxamic acid (SAHA), valproic acid (VPA), mocetinostat, givinostat, entinostat, apicidin, and RGFP966, were also analysed, showing antioxidant and anti-inflammatory effects, an improvement in cardiac function and remodelling, and a decrease in apoptosis, among other effects. Conclusions: HDAC inhibitors constitute a significant promise for the treatment of AMI due to their diverse cardioprotective effects. However, high risk of selection, performance, and detection bias in the in vivo studies means that their application in the clinical setting is still a long way off and more research is needed to better understand their benefits and possible side effects. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram.</p>
Full article ">Figure 2
<p>Signalling pathways involved in trichostatin A (TSA)’s effects.</p>
Full article ">Figure 3
<p>Main effects of SAHA, VPA, mocetinostat, givinostat, RGFP966, entinostat, and apicidin.</p>
Full article ">
12 pages, 3368 KiB  
Communication
Flow-Dependent Modulation of Endothelial Ca2+ Dynamics by Small Conductance Ca2+-Activated K+ Channels in Mouse Carotid Arteries
by Mark S. Taylor, Michael Francis and Chung-Sik Choi
Biomedicines 2024, 12(12), 2900; https://doi.org/10.3390/biomedicines12122900 - 20 Dec 2024
Abstract
Background: Small conductance Ca2+ activated K+ channels (KCa2.3) are important regulators of vascular function. They provide Ca2+-dependent hyperpolarization of the endothelial membrane potential, promoting agonist-induced vasodilation. Another important mechanism of influence may occur through positive feedback regulation [...] Read more.
Background: Small conductance Ca2+ activated K+ channels (KCa2.3) are important regulators of vascular function. They provide Ca2+-dependent hyperpolarization of the endothelial membrane potential, promoting agonist-induced vasodilation. Another important mechanism of influence may occur through positive feedback regulation of endothelial Ca2+ signals, likely via amplification of influx through membrane cation channels. KCa2.3 channels have recently been implicated in flow-mediated dilation of the arterial vasculature and may contribute to the crucial homeostatic role of shear stress in preventing vascular wall remodeling and progressive vascular disease (i.e., atherosclerosis). The impact of KCa2.3 channels on endothelial Ca2+ signaling under physiologically relevant shear stress conditions remains unknown. Methods: In the current study, we employ mice expressing an endothelium-specific Ca2+ fluorophore (cdh5-GCaMP8) to characterize the KCa2.3 channel influence on the dynamic Ca2+ signaling profile along the arterial endothelium in the presence and absence of shear-stress. Results: Our data indicate KCa2.3 channels have a minimal influence on basal Ca2+ signaling in the carotid artery endothelium in the absence of flow, but they contribute substantially to amplification of Ca2+ dynamics in the presence of flow and their influence can be augmented through exogenous positive modulation. Conclusions: The findings suggest a pivotal role for KCa2.3 channels in adjusting the profile of homeostatic dynamic Ca2+ signals along the arterial intima under flow. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

Figure 1
<p>Characterization of dynamic Ca<sup>2+</sup> signaling in cdh5-GCaMP8 mouse carotid artery endothelium. (<b>A</b>) Open carotid arteries were mounted in a flow chamber for fluorescence confocal imaging of the vascular endothelium under no-flow or flow conditions (red arrows). Images show isolation and tracking of basal dynamic Ca<sup>2+</sup> signals from image sequences using S8 (under no-flow conditions). The three dimensional plot shows extrapolation of events in the field (x,y) over time (t). (<b>B</b>) Quantification of the basal Ca<sup>2+</sup> signaling profile based on event frequency, amplitude, duration, and maximal area (18 arteries from 14 animals). (<b>C</b>) Characterization of Ca<sup>2+</sup> dynamics in response to flow (10 dyn/cm<sup>2</sup>). (<b>D</b>) Summary of Ca<sup>2+</sup> dynamics under flow (n = 4).</p>
Full article ">Figure 1 Cont.
<p>Characterization of dynamic Ca<sup>2+</sup> signaling in cdh5-GCaMP8 mouse carotid artery endothelium. (<b>A</b>) Open carotid arteries were mounted in a flow chamber for fluorescence confocal imaging of the vascular endothelium under no-flow or flow conditions (red arrows). Images show isolation and tracking of basal dynamic Ca<sup>2+</sup> signals from image sequences using S8 (under no-flow conditions). The three dimensional plot shows extrapolation of events in the field (x,y) over time (t). (<b>B</b>) Quantification of the basal Ca<sup>2+</sup> signaling profile based on event frequency, amplitude, duration, and maximal area (18 arteries from 14 animals). (<b>C</b>) Characterization of Ca<sup>2+</sup> dynamics in response to flow (10 dyn/cm<sup>2</sup>). (<b>D</b>) Summary of Ca<sup>2+</sup> dynamics under flow (n = 4).</p>
Full article ">Figure 2
<p>Effect of K<sub>Ca</sub>2.3 inhibition on endothelial Ca<sup>2+</sup> dynamics under no-flow conditions. (<b>A</b>) Panels show binary masks and time-extrapolated plots of Ca<sup>2+</sup> events before and after addition of apamin (0.5 µM) for 10 min. (<b>B</b>) Summary of Ca<sup>2+</sup> dynamics before and after apamin (n = 5).</p>
Full article ">Figure 3
<p>Effect of K<sub>Ca</sub>2.3 inhibition on endothelial Ca<sup>2+</sup> dynamics under flow conditions. (<b>A</b>) Panels show binary masks and time-extrapolated plots of Ca<sup>2+</sup> events at 10 dyn/cm<sup>2</sup> shear stress before and after addition of apamin (0.5 µM) for 10 min. (<b>B</b>) Summary of Ca<sup>2+</sup> dynamics before and after apamin (n = 5).</p>
Full article ">Figure 4
<p>Effect of K<sub>Ca</sub>2.3 stimulation on endothelial Ca<sup>2+</sup> dynamics under no-flow and flow conditions. (<b>A</b>) Panels show binary masks and time-extrapolated plots of Ca<sup>2+</sup> events before and after addition of CyPPA (10 µM) for 10 min. (<b>B</b>) Summary of Ca<sup>2+</sup> dynamics before and after CyPPA (n = 5). (<b>C</b>) Panels show binary masks and time-extrapolated plots of Ca<sup>2+</sup> events at 5 dyn/cm<sup>2</sup> shear stress before and after addition of CyPPA (10 µM) for 10 min. (<b>D</b>) Summary of Ca2+ dynamics before and after CyPPA (n = 4).</p>
Full article ">Figure 4 Cont.
<p>Effect of K<sub>Ca</sub>2.3 stimulation on endothelial Ca<sup>2+</sup> dynamics under no-flow and flow conditions. (<b>A</b>) Panels show binary masks and time-extrapolated plots of Ca<sup>2+</sup> events before and after addition of CyPPA (10 µM) for 10 min. (<b>B</b>) Summary of Ca<sup>2+</sup> dynamics before and after CyPPA (n = 5). (<b>C</b>) Panels show binary masks and time-extrapolated plots of Ca<sup>2+</sup> events at 5 dyn/cm<sup>2</sup> shear stress before and after addition of CyPPA (10 µM) for 10 min. (<b>D</b>) Summary of Ca2+ dynamics before and after CyPPA (n = 4).</p>
Full article ">
10 pages, 899 KiB  
Article
The Influence of Bariatric Surgery on Matrix Metalloproteinase Plasma Levels in Patients with Type 2 Diabetes Mellitus
by João Kleber de Almeida Gentile, Renato Migliore, Jaques Waisberg and Marcelo Augusto Fontenelle Ribeiro Junior
Biomolecules 2024, 14(12), 1633; https://doi.org/10.3390/biom14121633 - 19 Dec 2024
Abstract
Background: Bariatric surgery is a safe and effective procedure for treating obesity and metabolic conditions such as type 2 diabetes mellitus (T2DM). Remodeling of the extracellular matrix (ECM) supports adipose tissue expansion and its metabolic activity, where matrix metalloproteinases (MMPs) play a key [...] Read more.
Background: Bariatric surgery is a safe and effective procedure for treating obesity and metabolic conditions such as type 2 diabetes mellitus (T2DM). Remodeling of the extracellular matrix (ECM) supports adipose tissue expansion and its metabolic activity, where matrix metalloproteinases (MMPs) play a key role in ECM regulation. The MMPs, particularly MMP-2 and MMP-9, are elevated in patients with morbid obesity, metabolic syndrome, and T2DM. Objectives: To evaluate the effect of weight loss in bariatric surgery patients using oxidative stress markers and to compare MMP levels in patients undergoing bariatric surgery. Methods: This was a prospective, controlled study including 45 morbidly obese patients with T2DM (BMI > 35 kg/m2) who underwent RYGB (n = 24) or VG (n = 21). Weight loss was assessed through anthropometric measurements (weight, height, BMI). MMP-2 and MMP-9 levels were measured preoperatively and at 3 and 12 months postoperatively. Results: Significant and sustained weight loss was observed after surgery in both groups, with reductions in BMI. MMP-2 and MMP-9 levels decreased significantly after one year of follow-up. Conclusions: Bariatric surgery is an effective long-term intervention for weight loss and associated comorbidities, including T2DM. MMP-2 and MMP-9 proved to be effective markers of extracellular matrix remodeling, with significant reductions following surgery. Full article
(This article belongs to the Special Issue Role of Matrix Metalloproteinase in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Median, 1st quartile, and 3rd quartile BMI (kg/m<sup>2</sup>) at M0, M3, and M12 in diabetic patients, according to surgical technique. RYGB (n = 24) and VG (n = 21). (a, b): indicate a statistically significant difference when comparing RYGB and VG at each time point, using the Mann–Whitney U-test. (A, B, C): comparison within the same group, considering the differences between M0, M3, and M12, using the Friedman test.</p>
Full article ">Figure 2
<p>Median, 1st quartile, and 3rd quartile of MMP2 (ng/mL) at M0, M3, and M12 in diabetic patients, according to surgical technique. RYGB (n = 24) and VG (n = 21). (A, B, C): comparison within the same group, considering the differences between M0, M3, and M12, using the Friedman test.</p>
Full article ">Figure 3
<p>Median, 1st quartile, and 3rd quartile of MMP9 (ng/mL) at M0, M3, and M12 in diabetic patients, according to surgical technique. RYGB (n = 24) and VG (n = 21). (A, B, C): comparison within the same group, considering the differences between M0, M3, and M12, using the Friedman test.</p>
Full article ">
19 pages, 1218 KiB  
Review
The Potential Regulatory Role of Ferroptosis in Orthodontically Induced Inflammatory Root Resorption
by Leilei Wang, Chuan Wang and Hong He
Int. J. Mol. Sci. 2024, 25(24), 13617; https://doi.org/10.3390/ijms252413617 - 19 Dec 2024
Abstract
People, in increasing numbers, are seeking orthodontic treatment to correct malocclusion, while some of them are suffering from orthodontically induced inflammatory root resorption (OIIRR). Recent evidence suggests that the immune-inflammatory response occurring during bone remodeling may be responsible for OIIRR. Ferroptosis, a new [...] Read more.
People, in increasing numbers, are seeking orthodontic treatment to correct malocclusion, while some of them are suffering from orthodontically induced inflammatory root resorption (OIIRR). Recent evidence suggests that the immune-inflammatory response occurring during bone remodeling may be responsible for OIIRR. Ferroptosis, a new type of programmed cell death (PCD), has been found to have a close interrelation with inflammation during disease progression. While ferroptosis has been extensively studied in bone-related diseases, its role in OIIRR is poorly understood. Considering that the tooth root shares a lot of similar characteristics with bone, it is reasonable to hypothesize that ferroptosis contributes to the development of OIIRR. Nevertheless, direct evidence supporting this theory is currently lacking. In this review, we introduced ferroptosis and elucidated the mechanisms underlying orthodontic tooth movement (OTM) and OIIRR, with a special focus on the pivotal role inflammation plays in these processes. Additionally, we covered recent research exploring the connections between inflammation and ferroptosis. Lastly, we emphasized the important regulatory function of ferroptosis in bone homeostasis. Further investigations are required to clarify the modulation mechanisms of ferroptosis in OIIRR and to develop novel and potential therapeutic strategies for the management of OIIRR. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Orthodontic force-induced inflammation may cause root resorption by inducing ferroptosis. The inflammation reactions commence immediately after orthodontic force is applied to the teeth. Inflammatory cytokines, including TNF-α and IL-1β secreted by immune cells, can cause lipid ROS accumulation through stat3 or p53, ultimately inducing ferroptosis. In addition, the NLRP3 inflammasome can trigger the release of IL-1β to induce ferroptosis. Iron and lipid peroxidation can also induce ferroptosis. Ferroptosis of different types of bone cells can result in alveolar bone remodeling, resulting in tooth resorption. Black arrows represent promotion, and red arrows represent the progress of OIIRR.</p>
Full article ">Figure 2
<p>The signaling pathways that trigger ferroptosis in different kinds of bone cells. Bone cells including osteoblasts, osteoclasts, bone marrow mesenchymal stem cells (BMSCs), and osteocytes can undergo ferroptosis through modulating different signaling pathways. Up arrows represent upregulation (↑), and down arrows represent downregulation (↓).</p>
Full article ">
15 pages, 1853 KiB  
Article
SNPs in GPCR Genes and Impaired Osteogenic Potency in Osteoporotic Patient Lines-Based Study
by Julia Sopova, Olga Krasnova, Giomar Vasilieva, Anna Zhuk, Olga Lesnyak, Vitaliy Karelkin and Irina Neganova
Int. J. Mol. Sci. 2024, 25(24), 13594; https://doi.org/10.3390/ijms252413594 - 19 Dec 2024
Viewed by 62
Abstract
G-protein-coupled receptors (GPCRs) have emerged as critical regulators of bone development and remodeling. In this study, we aimed to identify specific GPCR mutations in osteoporotic patients via next-generation sequencing (NGS). We performed NGS sequencing of six genomic DNA samples taken from osteoporotic patients [...] Read more.
G-protein-coupled receptors (GPCRs) have emerged as critical regulators of bone development and remodeling. In this study, we aimed to identify specific GPCR mutations in osteoporotic patients via next-generation sequencing (NGS). We performed NGS sequencing of six genomic DNA samples taken from osteoporotic patients and two genomic DNA samples from healthy donors. Next, we searched for single-nucleotide polymorphisms (SNPs) in GPCR genes that are associated with osteoporosis. For three osteoporotic patients and one healthy donor, bone biopsies were used to generate patient-specific mesenchymal stem cell (MSC) lines, and their ability to undergo osteodifferentiation was analyzed. We found that MSCs derived from osteoporotic patients have a different response to osteoinductive factors and impaired osteogenic differentiation using qPCR and histochemical staining assays. The NGS analysis revealed specific combinations of SNPs in GPCR genes in these patients, where SNPs in ADRB2 (rs1042713), GIPR (rs1800437), CNR2 (rs2501431, rs3003336), and WLS (rs3762371) were associated with impaired osteogenic differentiation capacity. By integrating NGS data with functional assessments of patient-specific cell lines, we linked GPCR mutations to impaired bone formation, providing a foundation for developing personalized therapeutic strategies. SNP analysis is recognized as a proactive approach to osteoporosis management, enabling earlier interventions and targeted preventive measures for individuals at risk. Furthermore, SNP analysis contributes to the development of robust, holistic risk prediction models that enhance the accuracy of risk assessments across the population. This integration of genetic data into public health strategies facilitates healthcare initiatives. This approach could guide treatment decisions tailored to the patient’s genetic profile and provide a foundation for developing personalized therapeutic strategies. Full article
(This article belongs to the Special Issue Diagnosis and Treatment of Osteoporosis)
Show Figures

Figure 1

Figure 1
<p>Combination of bioinformatics analysis and patient-specific cell lines for the determination of the patient-specific SNPs involved in the development of osteoporosis. Figure created with Biorender.</p>
Full article ">Figure 2
<p>Osteoporotic patients have different sets of SNPs, potentially involved in osteoporosis progression. (<b>A</b>) The occurrence of known SNPs in GPCRs linked to BMD and osteoporosis progression in osteoporotic patients listed in this study. Missense mutations are shown in orange, intron variants in green, and 3′prime UTR variants in violet. (<b>B</b>) The distribution of SNPs in GPCRs found in four or more osteoporotic patients and absent in control patients. Intron variants shown in green, upstream gene variants in blue, 3′prime UTR variants in violet, and downstream gene variants in blue. Multicolored rectangles present different SNP types.</p>
Full article ">Figure 3
<p>Characteristics of mesenchymal stem cells (MSCs) derived from patients’ bone samples. (<b>A</b>) Phase contrast representative images of MSCs derived from bone samples of #8 (control), #9, #10, and #14 patients. Scale bar 400 μm. (<b>B</b>) Immunophenotype of MSCs derived from bone samples of #8 (control), #9, #10, and #14 patients. Cells are positive for mesenchymal stem cell markers CD73, CD90, and CD105, while they are negative for CD34 and CD45. (<b>C</b>) <span class="html-italic">RUNX2</span>, <span class="html-italic">COL1A1</span>, <span class="html-italic">POSTN</span>, <span class="html-italic">BGLAP</span> gene expression in MSCs of #8, #9, #10, and #14 patients after 7 days of osteogenic differentiation. Data shown as mean SD, n = 3, with significant differences indicated with asterisks (ns—not significant, **—<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). (<b>D</b>) Alkaline phosphatase activity in MSCs of #8 (control), #9, #10, and #14 patients after 14 days of osteogenic differentiation. Cells were grown on 24-well plate. (<b>E</b>) Alizarin Red staining of MSCs of #8, #9, #10, and #14 patients after 21 days of osteogenic differentiation. Cells were grown on 24-well plate. Abbreviations: BM—basal medium; OM—osteogenic medium.</p>
Full article ">Figure 4
<p>Simplified scheme summarizing osteogenic differentiation phases with schematic evaluation of osteogenic differentiation potency of cells derived from control and osteoporotic patients. Green arrows indicate increase in gene expression and specific staining, (+)—differentiation phase detected, (x)—differentiation phase absent.</p>
Full article ">
21 pages, 31149 KiB  
Article
Biomechanics of Osseointegration of a Dental Implant in the Mandible Under Shock Wave Therapy: In Silico Study
by Alexey Smolin, Galina Eremina, Irina Martyshina and Jing Xie
Materials 2024, 17(24), 6204; https://doi.org/10.3390/ma17246204 - 19 Dec 2024
Viewed by 109
Abstract
The most time-consuming aspect of dental prosthesis installation is the osseointegration of a metal implant with bone tissue. The acceleration of this process may be achieved through the use of extracorporeal shock wave therapy. The objective of this study is to investigate the [...] Read more.
The most time-consuming aspect of dental prosthesis installation is the osseointegration of a metal implant with bone tissue. The acceleration of this process may be achieved through the use of extracorporeal shock wave therapy. The objective of this study is to investigate the conditions for osseointegration of the second premolar implant in the mandibular segment through the use of a poroelastic model implemented in the movable cellular automaton method. The mandibular segment under consideration includes a spongy tissue layer, 600 µm in thickness, covered with a cortical layer, 400 µm in thickness, and a gum layer, 400 µm in thickness. Furthermore, the periodontal layers of the roots of the first premolar and first molar were considered, while the implant of the second premolar was situated within a shell of specific tissue that corresponded to the phase of osseointegration. The model was subjected to both physiological loading and shock wave loading across the three main phases of osseointegration. The resulting fields of hydrostatic pressure and interstitial fluid pressure were then subjected to analysis in accordance with the mechanobiological principles. The results obtained have indicated that low-intensity shock wave therapy can accelerate and promote direct osseointegration: 0.05–0.15 mJ/mm2 in the first and second phases and less than 0.05 mJ/mm2 in the third phase. In comparison to physiological loads (when bone tissue regeneration conditions are observed only around the implant distal end), shock waves offer the primary advantage of creating conditions conducive to osseointegration along the entire surface of the implant simultaneously. This can significantly influence the rate of implant integration during the initial osteoinduction phase and, most crucially, during the longest final phase of bone remodeling. Full article
(This article belongs to the Special Issue Latest Materials and Technologies in Dentistry)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the study.</p>
Full article ">Figure 2
<p>Model of the mandibular segment with an implant of the second premolar and its different parts with corresponding materials.</p>
Full article ">Figure 3
<p>Scheme of loading the segment of the mandible in modeling: (<b>a</b>) therapeutic shock wave loading; (<b>b</b>) physiological loading.</p>
Full article ">Figure 4
<p>Stiffness versus number of automata in the model sample.</p>
Full article ">Figure 5
<p>Distribution of the von Mises stress in the mandibular segment with an implant under a physiological load of 100 N.</p>
Full article ">Figure 6
<p>Distribution of hydrostatic pressure in the peri-implant zone in the first phase of osseointegration under physiological loads of 100 N (<b>a</b>) and 200 N (<b>b</b>).</p>
Full article ">Figure 7
<p>Distribution of pore fluid pressure in the peri-implant zone in the first phase of osseointegration under physiological loads of 100 N (<b>a</b>) and 200 N (<b>b</b>).</p>
Full article ">Figure 8
<p>Distribution of hydrostatic pressure in the mandibular segment in the first phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 9
<p>Distribution of hydrostatic pressure in the peri-implant zone of the mandibular segment in the first phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 10
<p>Distribution of biological fluid pressure in the mandibular segment in the first phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 11
<p>Distribution of biological fluid pressure in the peri-implant zone of the mandibular segment in the first phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 12
<p>Distribution of hydrostatic pressure in the mandibular segment in the second phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 13
<p>Distribution of biological fluid pressure in the mandibular segment in the second phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 14
<p>Distribution of biological fluid pressure in the peri-implant zone of the mandibular segment in the second phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 15
<p>Distribution of hydrostatic pressure in the peri-implant zone during the third phase of implant osseointegration under physiological loads of 100 N (<b>a</b>) and 200 N (<b>b</b>).</p>
Full article ">Figure 16
<p>Distribution of pore fluid pressure in the peri-implant zone during the third phase of implant osseointegration under physiological loads of 100 N (<b>a</b>) and 200 N (<b>b</b>).</p>
Full article ">Figure 17
<p>Distribution of hydrostatic pressure in the mandibular segment in the third phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.15, and (<b>c</b>) 0.26 mJ/mm<sup>2</sup>.</p>
Full article ">Figure 18
<p>Distribution of biological fluid pressure in the mandibular segment in the third phase of implant osseointegration under shock wave exposure with energy flux densities of (<b>a</b>) 0.02, (<b>b</b>) 0.05, (<b>c</b>) 0.15 mJ/mm<sup>2</sup>.</p>
Full article ">
20 pages, 1322 KiB  
Review
Atrial Fibrillation and Cancer—Epidemiology, Mechanisms, and Management
by Nathaniel E. Davis, Narut Prasitlumkum and Nicholas Y. Tan
J. Clin. Med. 2024, 13(24), 7753; https://doi.org/10.3390/jcm13247753 - 19 Dec 2024
Viewed by 98
Abstract
Atrial fibrillation (AF) and cancer are increasingly recognized as interrelated conditions, with cancer patients showing elevated incidences of AF, and there is evidence that AF may sometimes precede cancer diagnoses. This comprehensive review investigates the epidemiology, pathophysiology, and management challenges associated with AF [...] Read more.
Atrial fibrillation (AF) and cancer are increasingly recognized as interrelated conditions, with cancer patients showing elevated incidences of AF, and there is evidence that AF may sometimes precede cancer diagnoses. This comprehensive review investigates the epidemiology, pathophysiology, and management challenges associated with AF in cancer patients. Epidemiologically, several cancers are more closely related to increased rates of AF, including lung, colorectal, gastrointestinal, and hematologic malignancies. Mechanistically, both AF and cancer share pathophysiological pathways centered on inflammation, oxidative stress, and common cardiovascular risk factors, such as hypertension, obesity, and diabetes. The inflammatory microenvironment in tumors, marked by increased cytokines and growth factors, promotes atrial remodeling and AF susceptibility. Elevated reactive oxygen species (ROS) levels, driven by the metabolic demands of cancer, further contribute to atrial fibrosis and structural changes. Moreover, many anticancer treatments exacerbate AF risk. Management of AF in cancer patients presents many unique challenges and requires a multidisciplinary approach. Rate and rhythm control strategies are complicated by potential drug–drug interactions and limited data surrounding early implementation of rhythm control strategies in cancer patients. Interventional approaches such as catheter ablation, though effective in maintaining sinus rhythm, carry significant perioperative risk in patients with malignancy. Stroke prevention with anticoagulants is essential but requires cautious administration to avoid heightened bleeding risks, particularly in patients undergoing chemotherapy. Further, the limited applicability of standard risk stratification tools like CHA2DS2-VASc in this population complicate decisions regarding anticoagulation. This review highlights the bidirectional relationship between AF and cancer, the difficulties in management, and the critical need for further research in this field. Full article
(This article belongs to the Special Issue Current Perspectives on the Management of Atrial Fibrillation)
Show Figures

Figure 1

Figure 1
<p>Considerations for atrial fibrillation management in cancer patients.</p>
Full article ">Figure 2
<p>Suggested treatment algorithm of atrial fibrillation in cancer patients.</p>
Full article ">Figure 3
<p>Central illustration.</p>
Full article ">
12 pages, 1491 KiB  
Article
Overlapping Systemic Proteins in COVID-19 and Lung Fibrosis Associated with Tissue Remodeling and Inflammation
by Barbora Svobodová, Anna Löfdahl, Annika Nybom, Jenny Wigén, Gabriel Hirdman, Franziska Olm, Hans Brunnström, Sandra Lindstedt, Gunilla Westergren-Thorsson and Linda Elowsson
Biomedicines 2024, 12(12), 2893; https://doi.org/10.3390/biomedicines12122893 - 19 Dec 2024
Viewed by 161
Abstract
Background/Objectives: A novel patient group with chronic pulmonary fibrosis is emerging post COVID-19. To identify patients at risk of developing post-COVID-19 lung fibrosis, we here aimed to identify systemic proteins that overlap with fibrotic markers identified in patients with idiopathic pulmonary fibrosis (IPF) [...] Read more.
Background/Objectives: A novel patient group with chronic pulmonary fibrosis is emerging post COVID-19. To identify patients at risk of developing post-COVID-19 lung fibrosis, we here aimed to identify systemic proteins that overlap with fibrotic markers identified in patients with idiopathic pulmonary fibrosis (IPF) and may predict COVID-19-induced lung fibrosis. Methods: Ninety-two proteins were measured in plasma samples from hospitalized patients with moderate and severe COVID-19 in Sweden, before the introduction of the vaccination program, as well as from healthy individuals. These measurements were conducted using proximity extension assay (PEA) technology with a panel including inflammatory and remodeling proteins. Histopathological alterations were evaluated in explanted lung tissue. Results: Connecting to IPF pathology, several proteins including decorin (DCN), tumor necrosis factor receptor superfamily member 12A (TNFRSF12A) and chemokine (C-X-C motif) ligand 13 (CXCL13) were elevated in COVID-19 patients compared to healthy subjects. Moreover, we found incrementing expression of monocyte chemotactic protein-3 (MCP-3) and hepatocyte growth factor (HGF) when comparing moderate to severe COVID-19. Conclusions: Both extracellular matrix- and inflammation-associated proteins were identified as overlapping with pulmonary fibrosis, where we found DCN, TNFRSF12A, CXCL13, CXCL9, MCP-3 and HGF to be of particular interest to follow up on for the prediction of disease severity. Full article
Show Figures

Figure 1

Figure 1
<p>Elevated protein amount of DCN, TNFRSF12A, MCP-3, HGF, CXCL13 and CXCL9 in plasma from patients with moderate and severe COVID-19 in comparison to healthy subjects. NPX = normalized protein expression. Patients with moderate (n = 8) and severe (n = 8) COVID-19; healthy individuals (n = 7). One-way ANOVA with Tukey’s multiple comparison 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, **** <span class="html-italic">p</span> &lt; 0.0001, ns=not significant.</p>
Full article ">Figure 2
<p>The overlapping protein patterns of DCN and POSTN in post-COVID-19 and IPF. In distal lung tissue, the expression of POSTN was mainly localized to the subepithelial regions of bronchioles in healthy (<b>A</b>,<b>D</b>), post-COVID-19 (<b>B</b>,<b>E</b>) and IPF (<b>C</b>,<b>F</b>) patients, enclosed upon magnification in the basement membrane zone (arrow). POSTN was highly expressed in fibroblastic foci in IPF (<b>G</b>,<b>H</b>, encircled area) and in similar structures in post-COVID-19 patients (<b>I</b>,<b>J</b>). Similarly, DCN was found to be intensely expressed in the subepithelial regions of bronchioles (arrowhead) and in vascular adventitia (arrow) (healthy, <b>K</b>–<b>M</b>). Increased DCN expression was also seen in pleura (arrows) and subpleural regions in healthy (<b>N</b>, including HE staining), post-COVID-19 (<b>O</b>, including HE staining) and IPF (<b>P</b>, including HE staining) patients. Scale bar: 500 µm (<b>N</b>–<b>P</b>); 100 µm (<b>A</b>–<b>F</b>, <b>K</b>–<b>M</b>; enlargements <b>N</b>–<b>P</b>); 20 µm (enlargement (<b>D</b>–<b>F</b>), <b>G</b>–<b>J</b>). * = bronchiole; v = vessel.</p>
Full article ">
28 pages, 734 KiB  
Protocol
A Protocol Investigation Comparing Transcatheter Repair with the Standard Surgical Procedure for Secondary Mitral Regurgitation
by Francesco Nappi, Sanjeet Singh Avtaar Singh, Antonio Salsano, Aubin Nassif, Yasushige Shingu, Satoru Wakasa, Antonio Fiore, Cristiano Spadaccio and Zein EL-Dean
J. Clin. Med. 2024, 13(24), 7742; https://doi.org/10.3390/jcm13247742 - 18 Dec 2024
Viewed by 258
Abstract
Background: Secondary mitral regurgitation (SMR) is characterized by a pathological process impacting the left ventricle (LV) as opposed to the mitral valve (MV). In the absence of structural alterations to the MV, the expansion of the LV or impairment of the papillary muscles [...] Read more.
Background: Secondary mitral regurgitation (SMR) is characterized by a pathological process impacting the left ventricle (LV) as opposed to the mitral valve (MV). In the absence of structural alterations to the MV, the expansion of the LV or impairment of the papillary muscles (PMs) may ensue. A number of technical procedures are accessible for the purpose of determining the optimal resolution for MR. Nevertheless, there is a dearth of rigorous data to facilitate a comparative analysis of MV replacement, MV repair (including subvalvular repair), and transcatheter mitral valve interventions (TMV-Is). The objective of this investigation is to evaluate and compare the efficacy and clinical outcomes of transcatheter mitral valve repair (TMV-r) utilizing the edge-to-edge mitral valve repair (TEER) procedure in comparison to conventional surgical mitral valve interventions (S-SMVis) in patients with secondary mitral regurgitation. Methods and analysis: A consortium of five cardiac surgery institutions from four European states and Japan have joined forces to establish a multicenter observational registry, designated TEERMISO. Patients who underwent technical procedures for SMR between January 2007 and December 2023 will be enrolled consecutively into the TEERMISO registry. The investigation team evaluated the comparative efficacy of replacement and repair techniques, utilizing both the standard surgical methodology and the transcatheter intervention. The primary clinical outcome will be the degree of left ventricular remodeling, as assessed by the left ventricular end-diastolic volume index, at 10 years. The forthcoming research will assess a variety of secondary endpoints, among which all-cause mortality will be the primary endpoint. Subsequent assessments will be made in the following order: functional status, hospitalization, neurocognition, physiological measures (echocardiographic assessment), occurrence of adverse clinical incidents, and reoperation. Ethics and dissemination: The multicenter design of the database is anticipated to reduce the potential for bias associated with institutional caseload and surgical experience. All participating centers possess an established mitral valve protocol that facilitates comprehensive follow-up and management of any delayed mitral complications following replacement surgery or surgical repair of the secondary mitral regurgitation. The data collected will provide insights into the impact of diverse surgical approaches on standard mitral valve surgery and TEER. This will facilitate the evaluation of LV remodeling over the course of long-term post-procedural follow-up. Trial Registration: ClinicalTrials.gov ID: NCT05090540; IRB ID: 202201143 Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Study design schematic.</p>
Full article ">
10 pages, 2120 KiB  
Article
An Artificial Intelligence-Based Automatic Classifier for the Presence of False Lumen Thrombosis After Frozen Elephant Trunk Operation
by Anja Osswald, Konstantinos Tsagakis, Matthias Thielmann, Alan B. Lumsden, Arjang Ruhparwar and Christof Karmonik
Diagnostics 2024, 14(24), 2853; https://doi.org/10.3390/diagnostics14242853 - 18 Dec 2024
Viewed by 210
Abstract
Objective: To develop an unsupervised artificial intelligence algorithm for identifying and quantifying the presence of false lumen thrombosis (FL) after Frozen Elephant Trunk (FET) operation in computed tomography angiographic (CTA) images in an interdisciplinary approach. Methods: CTA datasets were retrospectively collected from eight [...] Read more.
Objective: To develop an unsupervised artificial intelligence algorithm for identifying and quantifying the presence of false lumen thrombosis (FL) after Frozen Elephant Trunk (FET) operation in computed tomography angiographic (CTA) images in an interdisciplinary approach. Methods: CTA datasets were retrospectively collected from eight patients after FET operation for aortic dissection from a single center. Of those, five patients had a residual aortic dissection with partial false lumen thrombosis, and three patients had no false lumen or thrombosis. Centerlines of the aortic lumen were defined, and images were calculated perpendicular to the centerline. Lumen and thrombosis were outlined and used as input for a variational autoencoder (VAE) using 2D convolutional neural networks (2D CNN). A 2D latent space was chosen to separate images containing false lumen patency, false lumen thrombosis and no presence of false lumen. Classified images were assigned a thrombus score for the presence or absence of FL thrombosis and an average score for each patient. Results: Images reconstructed by the trained 2D CNN VAE corresponded well to original images with thrombosis. Average thrombus scores for the five patients ranged from 0.05 to 0.36 where the highest thrombus scores coincided with the location of the largest thrombus lesion. In the three patients without large thrombus lesions, average thrombus scores ranged from 0.002 to 0.01. Conclusions: The presence and absence of a FL thrombus can be automatically classified by the 2D CNN VAE for patient-specific CTA image datasets. As FL thrombosis is an indication for positive aortic remodeling, evaluation of FL status is essential in follow-up examinations. The presented proof-of-concept is promising for the automated classification and quantification of FL thrombosis. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the Variational Autoencoder (VAE) algorithm, consisting of an encoder and decoder with Conv2D (Convolutional two-dimensional) layer and 2DConvTrans (Transpose two-dimensional Convolutional) layers, respectively. The encoder compresses input images into a 2D latent space to classify false lumen characteristics, while the decoder reconstructs the images by upsampling this representation. Layer names, sizes, depths, and kernel/stride configurations are shown to illustrate the dimensional transformations throughout the process.</p>
Full article ">Figure 2
<p>(<b>A</b>): Conventional three-plane visualization of the aorta. (<b>B</b>): On the left, the centerline is defined by red points and the line inside the aortic lumen; in the center, a 3D curved MPR reconstruction with the centerline and, consequently, the aortic lumen straightened. The perpendicular MPR slices (shown on the right) provide cross-sectional views of the aortic lumen.</p>
Full article ">Figure 3
<p>(<b>A</b>) Latent space visualization of all images used to train the VAE, with areas for reconstructed images outlined in blue. Images are classified based on aortic area and thrombus content, with the red line dividing the thrombus (left) from no thrombus (right). In the upper left are the images with a large aortic area and a large amount of thrombus, and at the bottom on the right are the ones with no thrombus and a small aortic area. (<b>B</b>) Reconstructed images from the VAE decoder mapped to the latent space ranging from −3 to 3, with the red line separating thrombus regions. (<b>C</b>) Sample comparisons of original (bottom) and reconstructed (top) images showing the true lumen with the thrombus (left), true and false lumens (middle), and only the true lumen (right).</p>
Full article ">Figure 4
<p>For each subject, the 3D MPR with the straight aortic lumen is shown together with a panel that contains the thrombus score in pseudo color derived from the classification for each slide perpendicular to the aortic lumen (legend on right). The total thrombus score for each subject is shown below each figure.</p>
Full article ">
30 pages, 3165 KiB  
Review
The RAGE Pathway in Skin Pathology Development: A Comprehensive Review of Its Role and Therapeutic Potential
by Marcin Radziszewski, Ryszard Galus, Krzysztof Łuszczyński, Sebastian Winiarski, Dariusz Wąsowski, Jacek Malejczyk, Paweł Włodarski and Aneta Ścieżyńska
Int. J. Mol. Sci. 2024, 25(24), 13570; https://doi.org/10.3390/ijms252413570 - 18 Dec 2024
Viewed by 260
Abstract
The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, is expressed in various cell types and mediates cellular responses to a wide range of ligands. The activation of RAGE triggers complex signaling pathways that drive inflammatory, oxidative, and proliferative [...] Read more.
The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, is expressed in various cell types and mediates cellular responses to a wide range of ligands. The activation of RAGE triggers complex signaling pathways that drive inflammatory, oxidative, and proliferative responses, which are increasingly implicated in the pathogenesis of skin diseases. Despite its well-established roles in conditions such as diabetes, cancer, and chronic inflammation, the contribution of RAGE to skin pathologies remains underexplored. This review synthesizes current findings on RAGE’s involvement in the pathophysiology of skin diseases, including conditions such as psoriasis, atopic dermatitis, and lichen planus, focusing on its roles in inflammatory signaling, tissue remodeling, and skin cancer progression. Additionally, it examines RAGE-modulating treatments investigated in dermatological contexts, highlighting their potential as therapeutic options. Given RAGE’s significance in a variety of skin conditions, further research into its mediated pathways may uncover new opportunities for targeted interventions in skin-specific RAGE signaling. Full article
(This article belongs to the Special Issue Dermatology: Advances in Pathophysiology and Therapies (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>The cell membrane receptor for advanced glycation end-products (RAGE) consists of a VC1 domain, a C2 domain, and both transmembrane and intracellular regions. RAGE binds a range of endogenous danger-associated molecular patterns (DAMPs) and exogenous pathogen-associated molecular patterns (PAMPs) through its positively charged VC1 domain. Key ligands include advanced glycation end-products (AGEs), advanced oxidation protein products (AOPPs), high-mobility group box 1 (HMGB1), S100 family proteins, beta-amyloid proteins, DNA, and collagen. Created with BioRender.com (accessed on 14 November 2024).</p>
Full article ">Figure 2
<p>RAGE forms oligomers to bind ligands, interacting with negatively charged molecules. Upon ligand binding, conformational changes occur in the receptor’s cytoplasmic tail, activating signaling adaptors such as diaphanous-1 (Dia1), extracellular signal-regulated kinase (ERK1/2), and protein kinase C (PKC). Dia1 subsequently activates small GTPases like Ras, Cdc42, and Rac1, which stimulate NF-κB signaling via the ERK1/2 and p38 MAPK pathways. NF-κB activation may also occur through the PI3K/Akt pathway, often triggered by reactive oxygen species (ROS) generated during RAGE signaling. Additionally, Dia1 can engage the JAK/STAT pathway, activating both NF-κB and interferon-stimulated response elements (ISRE), amplifying the inflammatory response. These cascades result in pro-inflammatory cellular changes and chemotaxis, recruiting additional inflammatory cells. Created with BioRender.com (accessed on 14 November 2024).</p>
Full article ">Figure 3
<p>Comparison of RAGE-deficient and wild-type mice responses to subcutaneous <span class="html-italic">Staphylococcus aureus</span> injection reveals that RAGE facilitates distant bacterial migration and exacerbates local tissue damage during infection (↑—increase, ↓—decrease). Conversely, RAGE activation in infection-related wounds may support the healing process, indicating a dual role for the receptor depending on context. Created with BioRender.com (accessed on 14 November 2024).</p>
Full article ">Figure 4
<p>Overview of RAGE pathway involvement in wound healing (↑—increase, ↓—decrease). The excessive accumulation of AGEs in the skin disrupts fibril formation and scar elasticity, leading to increased tissue contraction. AGEs induce fibroblast apoptosis and cause cell cycle arrest, while also contributing to RAGE overexpression in fibroblasts. This activates signaling pathways such as ERK1/2, MAPK, and NF-κB, promoting pro-inflammatory cytokine secretion (e.g., TNF-α and IL-8). RAGE activation enhances extracellular matrix (ECM) production, including collagen types I and III, and stimulates matrix metalloproteinases (MMPs), impacting tissue remodeling and fibrosis. Created with BioRender.com (accessed on 14 November 2024).</p>
Full article ">Figure 5
<p>The RAGE pathway can be inhibited at three key intervention points: blocking RAGE ligands, inhibiting the receptor itself, and silencing the RAGE gene. Blue boxes highlight these intervention points, while red boxes illustrate various potential inhibitors of the RAGE pathway. Additional therapies can indirectly suppress contributing signaling pathways, providing complementary strategies for RAGE pathway inhibition. Created with BioRender.com (accessed on 14 November 2024).</p>
Full article ">
15 pages, 304 KiB  
Review
Review of Thermal Calculation Methods for Boilers—Perspectives on Thermal Optimization for Improving Ecological Parameters
by Bartosz Ciupek and Andrzej Frąckowiak
Energies 2024, 17(24), 6380; https://doi.org/10.3390/en17246380 - 18 Dec 2024
Viewed by 230
Abstract
This article presents an overview of thermal calculation methods used in boilers powered by fossil fuels (solid, liquid or gas). The analysis was carried out mainly in terms of combustion chamber calculation methods. Changing standards and legal regulations regarding the use of fossil [...] Read more.
This article presents an overview of thermal calculation methods used in boilers powered by fossil fuels (solid, liquid or gas). The analysis was carried out mainly in terms of combustion chamber calculation methods. Changing standards and legal regulations regarding the use of fossil fuels in Europe and the world make it necessary to adapt calculation methods and boiler design to current requirements, and many of them are related to outdated boiler models or for fuels that are no longer so heavily used in industrial solutions. Current research and development trends implemented in the EU and in the world related to the issues of the European Green Deal, the Fit for 55 directive and other ecological trends in the energy sector make it necessary to verify and remodel the calculation methods used so far in terms of the thermal efficiency of the device, fuel consumption or the use of fuels not previously used in their wide range in a wider application. Hence, the knowledge and updating of the state of knowledge in the field of the thermal calculation of boilers in terms of their environmental performance is necessary and strongly sought after by researchers. It is undoubted that in the next few years, boilers will continue to be the main source of thermal energy, especially in the power industry or industry. A reasonable energy transition should be based on the direction of the thermal optimization of already functioning structures and adaptation of their operating parameters to the planned new ecological fuels in the sense of the intensification of energy converted from primary form to thermal energy, and in the last step, it should reorganize the energy and industrial sectors, leaving only these groups of devices treated as a stable and necessary source of energy. Therefore, it should be recognized that activities aimed at improving the thermal parameters of boilers should directly improve the thermal efficiency of the device, and this will translate into fuel savings and reduce their negative impact on the environment. Full article
(This article belongs to the Special Issue Heat Transfer Analysis: Recent Challenges and Applications)
Back to TopTop