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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (730)

Search Parameters:
Keywords = endothelial cells (ECs)

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 8033 KB  
Article
Luteolin Enhances Endothelial Barrier Function and Attenuates Myocardial Ischemia–Reperfusion Injury via FOXP1-NLRP3 Pathway
by Hanyan Xie, Xinyi Zhong, Nan Li, Mijia Zhou, Miao Zhang, Xiaomin Yang, Hui Wang, Yu Yan, Pengrong Gao, Tianhua Liu, Qiyan Wang and Dongqing Guo
Int. J. Mol. Sci. 2026, 27(2), 874; https://doi.org/10.3390/ijms27020874 - 15 Jan 2026
Viewed by 36
Abstract
As a natural flavonoid, the flavonoid luteolin is characterized by its powerful antioxidant and anti-inflammatory effects. While its precise mechanisms require further elucidation, existing evidence confirms its efficacy in ameliorating myocardial ischemia–reperfusion injury (MIRI). This research was designed to investigate the mechanism through [...] Read more.
As a natural flavonoid, the flavonoid luteolin is characterized by its powerful antioxidant and anti-inflammatory effects. While its precise mechanisms require further elucidation, existing evidence confirms its efficacy in ameliorating myocardial ischemia–reperfusion injury (MIRI). This research was designed to investigate the mechanism through which luteolin protects against MIRI. We established MIRI rat models through the ligation of left anterior descending coronary artery (LAD). To evaluate the cardioprotective effects of luteolin, echocardiographic analysis was performed, Hematoxylin and Eosin (HE) staining, and serum cardiac injury markers creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH). Cardiac vascular permeability was determined using Evans blue staining. To mimic ischemia–reperfusion injury, endothelial cells (ECs) were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) in vitro. Endothelial cell barrier function was evaluated through F-actin phalloidin staining and FITC-Dextran fluorescence leakage experiments. To elucidate the molecular mechanism, FOXP1 small interfering RNA (siRNA) and NLRP3 inhibitor MCC950 were administered. In MIRI rats, luteolin significantly improved cardiac function and preserved endothelial barrier integrity. These effects were associated with upregulation of FOXP1 and suppression of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. In OGD/R-treated endothelial cells, luteolin restored barrier function and cell viability. The protective effects of luteolin were abolished after FOXP1 silencing. Pharmacological NLRP3 inhibition (MCC950) mirrored luteolin’s protection. Our study indicates that luteolin enhances endothelial barrier function and attenuates MIRI via the FOXP1-NLRP3 pathway. The current study provides a potential drug for MIRI treatment. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

19 pages, 723 KB  
Review
iPSC-Derived Endothelial Cells as Experimental Models for Predictive and Personalized Strategies in Cardiovascular and Cerebrovascular Disease
by Lorenzo Fontanelli, Alessio Castronovo, Carolina Ferri, Federico Vozzi, Fabio A. Recchia and Andrea Borghini
Int. J. Mol. Sci. 2026, 27(2), 780; https://doi.org/10.3390/ijms27020780 - 13 Jan 2026
Viewed by 118
Abstract
Endothelial cells (ECs) regulate vascular homeostasis, and their dysfunction is a key driver of many cardiovascular and cerebrovascular diseases. Human-induced pluripotent stem cell-derived endothelial cells (hiPSC-ECs) provide access to patient-specific vascular cells that can be directed toward arterial, venous, or organotypic phenotypes, enabling [...] Read more.
Endothelial cells (ECs) regulate vascular homeostasis, and their dysfunction is a key driver of many cardiovascular and cerebrovascular diseases. Human-induced pluripotent stem cell-derived endothelial cells (hiPSC-ECs) provide access to patient-specific vascular cells that can be directed toward arterial, venous, or organotypic phenotypes, enabling personalized in vitro modeling of endothelial pathology. In this review, we discuss how patient-specific iPSC-ECs are used as predictive and personalized two- and three-dimensional models to dissect disease mechanisms and prioritize targeted therapies. We highlight some limitations of this methodology and outline future directions for integrating iPSC-EC-based assays into individualized treatment algorithms. Full article
(This article belongs to the Special Issue Cellular and Molecular Progression of Cardiovascular Diseases)
Show Figures

Figure 1

20 pages, 2378 KB  
Article
Phosphomimetic Thrombospondin-1 Modulates Integrin β1-FAK Signaling and Vascular Cell Functions
by Assala Raya, Bálint Bécsi and Anita Boratkó
Biomolecules 2026, 16(1), 84; https://doi.org/10.3390/biom16010084 - 4 Jan 2026
Viewed by 346
Abstract
Thrombospondin-1 (TSP1) is a multifunctional glycoprotein that plays a crucial role in angiogenesis and vascular remodeling. Ser93 of TSP1 has recently been identified as a novel phosphorylation site, influencing angiogenic properties; however, the underlying signaling mechanism remains unclear. Here, we investigated the functional [...] Read more.
Thrombospondin-1 (TSP1) is a multifunctional glycoprotein that plays a crucial role in angiogenesis and vascular remodeling. Ser93 of TSP1 has recently been identified as a novel phosphorylation site, influencing angiogenic properties; however, the underlying signaling mechanism remains unclear. Here, we investigated the functional impact of Ser93 phosphorylation using phosphomimetic (TSP1S93D) and phosphonull (TSP1S93A) mutants. Endothelial cell (EC) migration was analyzed using scratch assay and electric cell-substrate impedance sensing. Activation of key pathways (Akt, p38, ERK, and FAK) was analyzed by immunoblotting. TSP1 secretion was quantified by ELISA. Downstream effects on smooth muscle cells were examined by Western blot using conditioned media of endothelial cells. Expression of TSP1S93D significantly impaired endothelial migration and wound closure, associated with reduced phosphorylation of FAK and paxillin. Upstream of FAK signaling, TSP1S93D showed enhanced binding to integrin β1 and promoted its clustering. In contrast, TSP1S93D stimulated smooth muscle cell proliferation, migration, cytoskeletal remodeling, and phenotypic switching toward a synthetic, pro-inflammatory state characterized by elevated marker protein expression. Together, these findings demonstrate that the impaired angiogenic properties induced by TSP1S93D result from the modulation of integrin β1-FAK pathways in ECs, suppressing endothelial motility while promoting smooth muscle activation, suggesting a role in early vascular remodeling and inflammation. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

23 pages, 3415 KB  
Article
Exosomal NAMPT from Engineered Mesenchymal Stem Cells Mitigates Aortic Stenosis via Metabolic and Anti-Inflammatory Pathways
by Dipan Kumar Kundu, Matthew Kiedrowski, James Gadd, Min Gao, Madeline Evan, Yang Wang, Liya Yin, Vahagn Ohanyan, William M. Chilian and Feng Dong
Int. J. Mol. Sci. 2026, 27(1), 256; https://doi.org/10.3390/ijms27010256 - 25 Dec 2025
Viewed by 464
Abstract
The aim of this study was to determine whether exosomes from Nicotinamide phosphoribosyltransferase (NAMPT)-overexpressing mesenchymal stem cells (MSC NAMPT-Exo) can attenuate aortic stenosis (AS) and explored the underlying mechanism. NAMPT expression was examined in EC CXCR4 KO (AS) mouse hearts. Six-week-old AS mice [...] Read more.
The aim of this study was to determine whether exosomes from Nicotinamide phosphoribosyltransferase (NAMPT)-overexpressing mesenchymal stem cells (MSC NAMPT-Exo) can attenuate aortic stenosis (AS) and explored the underlying mechanism. NAMPT expression was examined in EC CXCR4 KO (AS) mouse hearts. Six-week-old AS mice received weekly injections of NAMPT-Exo, MSC-Exo, or PBS for three weeks, followed by echocardiography and histological examination of the valves (H&E, Alizarin Red, immunofluorescence). Cardiac ECs from control, AS, and NAMPT-Exo-treated mice were analyzed for miRNA expression (miR-146a-3p/5p, miR-125b-5p, miR-142a-5p). NAMPT expression was decreased in AS hearts. Treatment with NAMPT-Exo reduced aortic valve peak velocity, valvular thickening, and microcalcifications, while improving ejection fraction, fractional shortening, and ventricular dimensions. AS endothelial cells showed elevated levels of miR-146a-3p, miR-146a-5p, and miR-142a-5p, NAMPT-Exo specifically normalized miR-146a-3p. Histology revealed EndMT in AS valves, which was diminished by NAMPT-Exo. In vitro, inhibiting miR-146a-3p suppressed TGF-β-induced EndMT. Our results demonstrate that NAMPT-enriched MSC-derived exosomes effectively slow the progression of AS. Additionally, our findings highlight miR-146a-3p as a key regulator of EndMT, suggesting it as a potential molecular target for future therapies. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Graphical abstract

41 pages, 20290 KB  
Article
Palmitic but Not Oleic Acid Induces Pro-Inflammatory Dysfunction of Human Endothelial Cells from Different Vascular Beds In Vitro
by Daria Shishkova, Victoria Markova, Yulia Yurieva, Alexey Frolov, Anastasia Lazebnaya, Maxim Sinitsky, Anna Sinitskaya, Vera Matveeva, Evgenia Torgunakova, Alexander Stepanov, Anna Malashicheva, Asker Khapchaev, Nikita Podkuychenko, Alexander Vorotnikov, Vladimir Shirinsky and Anton Kutikhin
Int. J. Mol. Sci. 2025, 26(24), 12148; https://doi.org/10.3390/ijms262412148 - 17 Dec 2025
Viewed by 466
Abstract
Palmitic acid (PA) is the most common dietary saturated fatty acid, and is abundant in palm and cottonseed oil, butter, and cheese, whereas oleic acid (OA) is a monounsaturated omega-9 fatty acid found in olive oil. The differences in the cytotoxic and pro-inflammatory [...] Read more.
Palmitic acid (PA) is the most common dietary saturated fatty acid, and is abundant in palm and cottonseed oil, butter, and cheese, whereas oleic acid (OA) is a monounsaturated omega-9 fatty acid found in olive oil. The differences in the cytotoxic and pro-inflammatory effects of PA and OA across endothelial cells (ECs) isolated from different vascular beds have not been investigated in detail. Here, we incubated primary human aortic valve (HAVEC), saphenous vein (HSaVEC), internal thoracic artery (HITAEC), and microvascular (HMVEC) ECs with albumin-bound PA or OA for 24 h and found that PA induced a considerable cytotoxic response, accompanied by an elevated expression of the genes encoding cell adhesion molecules (VCAM1, ICAM1, SELE, and SELP) and pro-inflammatory cytokines (MIF, PTX3, CSF2, CSF3, IL1A, IL6, CCL2, CCL5, CCL20, CSF2, CSF3, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL8, and CXCL10), followed by an increased release of interleukin-6 and interleukin-8. HAVEC and HSaVEC were more susceptible to PA, whereas OA had mild-to-moderate cytotoxic effects on HAVEC and HMVEC but did not induce generalized EC activation. Compared with other EC types, HITAEC was the most resistant to PA and OA treatment. Collectively, these results indicate considerable heterogeneity across the ECs of distinct origin in response to PA. Full article
(This article belongs to the Special Issue Endothelial Cells in Health and Disease)
Show Figures

Figure 1

13 pages, 2114 KB  
Communication
Nanomechanical Characterization of E-Cigarette-Induced Lung Endothelial Dysfunction: Roles of Cortactin and Mitochondrial Reactive Oxygen Species
by Mounica Bandela, Xue Geng, Joe G. N. Garcia, James C. Lee and Steven M. Dudek
Int. J. Mol. Sci. 2025, 26(24), 12104; https://doi.org/10.3390/ijms262412104 - 16 Dec 2025
Viewed by 298
Abstract
E-cigarettes (E-cigs) are increasing in popularity and are considered a potentially safer alternative to traditional cigarettes. However, prior studies have demonstrated that inhalation of nicotine-containing e-cigs can cause substantial pathophysiologic changes, and “vaping” of some substances has led to severe lung damage. Our [...] Read more.
E-cigarettes (E-cigs) are increasing in popularity and are considered a potentially safer alternative to traditional cigarettes. However, prior studies have demonstrated that inhalation of nicotine-containing e-cigs can cause substantial pathophysiologic changes, and “vaping” of some substances has led to severe lung damage. Our group recently described the role of cortactin (CTTN), a cytoskeletal actin-binding regulatory protein, in mediating cigarette smoke (CS) and E-cig-induced lung endothelial apoptosis and mitochondrial dysfunction. In the current study, we advance this work by characterizing the effects of E-cig on lung endothelial nanomechanical properties and barrier function. Lung EC exposure to E-cig extract (50 µg/mL) resulted in disruption of endothelial barrier properties as assessed by Electric Cell–Substrate Impedance Sensing (ECIS). Since excess mitochondrial reactive oxygen species (mitoROS) is an important marker of mitochondrial dysfunction, we next assessed the effect of Mito-TEMPO (10 µM, 3 h), a cell-permeable antioxidant, on E-cig-induced endothelial permeability. Pretreatment with Mito-TEMPO provided EC barrier protection after E-cig challenge, suggesting a key role of mitoROS in E-cig-induced EC permeability. E-cig exposure induces cytoskeleton rearrangement, leading to gap formation in lung EC, and significantly alters EC elastic properties as assessed by atomic force microscopy (AFM). Reduction in CTTN expression by siRNA further augmented the injurious effects of E-cig on EC permeability and elastic properties. This is the first study to explore the role of CTTN in evaluating the effect of E-cigarette exposure on the lung endothelium using AFM and provides novel mitochondrial and biophysical characterization of the effects of E-cig exposure on human lung EC. This work advances our understanding of the pathophysiologic effects of E-cig exposure. Full article
(This article belongs to the Special Issue Molecular Research on Endothelial Cell Injury and Repair)
Show Figures

Figure 1

22 pages, 1358 KB  
Review
Beyond Viral Assembly: The Emerging Role of HIV-1 p17 in Vascular Inflammation and Endothelial Dysfunction
by Ylenia Pastorello, Nicoleta Arnaut, Mihaela Straistă, Francesca Caccuri, Arnaldo Caruso and Mark Slevin
Int. J. Mol. Sci. 2025, 26(24), 11949; https://doi.org/10.3390/ijms262411949 - 11 Dec 2025
Viewed by 339
Abstract
p17, the human immunodeficiency virus type 1 (HIV-1) matrix protein traditionally associated with viral assembly, has been recently investigated for its extracellular functions linked to vascular damage. This review examines the molecular and pathogenic signatures by which p17 and its variants (vp17s) contribute [...] Read more.
p17, the human immunodeficiency virus type 1 (HIV-1) matrix protein traditionally associated with viral assembly, has been recently investigated for its extracellular functions linked to vascular damage. This review examines the molecular and pathogenic signatures by which p17 and its variants (vp17s) contribute to endothelial activation, aberrant angiogenesis, and vascular inflammation, highlighting their relevance even under effective antiretroviral therapy (ART). Specifically, p17 exerts chemokine-like activities by binding to chemokine (C-X-C motif) receptor-1 and 2 (CXCR-1/2) on endothelial cells (ECs). This interaction triggers key signaling cascades, including the protein kinase B (Akt)-dependent extracellular signal-regulated kinase (ERK) pathway and endothelin-1/endothelin receptor B axis, driving EC motility, capillary formation, and lymphangiogenesis. Variants such as S75X demonstrate enhanced lymphangiogenic potency, associating them with tumorigenic processes involved in non-Hodgkin lymphoma (NHL) pathogenesis. Importantly, p17 promotes endothelial von Willebrand factor (vWF) storage and secretion, implicating a pro-coagulant state that may trigger the increased thromboembolic risks observed in HIV-positive patients. Furthermore, p17 crosses the blood–brain barrier (BBB) via CXCR-2-mediated pathways, contributing to neuroinflammation by activating microglia and astrocytes and amplifying monocyte chemoattractant protein-1 (MCP-1) levels, therefore playing a critical role in the development of HIV-associated neurocognitive disorders. Hence, the elaboration of potential therapeutic strategies finalized at inhibiting p17/vp17s’ interaction with their receptors could complement ART by addressing HIV-related neurovascular morbidity. Full article
(This article belongs to the Special Issue Advances in HIV Research: Molecular Basis and Potential Therapies)
Show Figures

Figure 1

12 pages, 1094 KB  
Article
A20 and TNIP-3 Reduce NF-κB-Mediated Paracrine Responses to Hypoxia/Hyperglycemia-Induced Endothelial Senescence
by Lara Russo, Serena Babboni, Serena Del Turco and Giuseppina Basta
Cells 2025, 14(23), 1908; https://doi.org/10.3390/cells14231908 - 2 Dec 2025
Viewed by 682
Abstract
Background: Hypoxia and ageing both involve impaired oxygen delivery, leading to oxidative damage, and endothelial cell (EC) dysfunction. In the presence of chronic hyperglycemia, these effects are amplified, accelerating EC senescence and vascular impairment. Methods: We assessed key mediators of inflammatory signalling and [...] Read more.
Background: Hypoxia and ageing both involve impaired oxygen delivery, leading to oxidative damage, and endothelial cell (EC) dysfunction. In the presence of chronic hyperglycemia, these effects are amplified, accelerating EC senescence and vascular impairment. Methods: We assessed key mediators of inflammatory signalling and senescence, as well as transcriptional regulators responsive to oxidative stress in ECs exposed to high glucose (30.5 mmol/L) for 72 h under either normoxia (21% O2) or prolonged (16 h) hypoxia (2% O2) followed by 2 h of reoxygenation. Results: ECs exposed to high glucose and hypoxia developed a senescent phenotype, as indicated by increased expression of p21 and p16, and elevated β-galactosidase staining. Interestingly, hypoxia-induced senescence did not coincide with the classical senescence-associated secretory phenotype (SASP). Compared to normoxia, ECs exposed to hypoxia, particularly under high-glucose conditions, showed reduced NF-κB-driven proinflammatory secretome (MCP-1, IL-6, IL-8), downregulation of the NF-κB p50 subunit, and simultaneous upregulation of the angiogenic factor VEGF-A with downregulation of YAP-1, a key regulator of cell survival. Notably, we observed a strong upregulation of A20 and TNIP-3, two well-characterized negative regulators of NF-κB signalling. Conclusions: Hypoxia-induced senescence did not trigger a typical inflammatory SASP. Although ECs enter a senescent state, they activate an anti-inflammatory response, suppressing NF-κB signalling and increasing the expression of its inhibitors, A20 and TNIP-3. This may reflect a non-canonical senescence response whose functional significance remains to be determined. Full article
Show Figures

Figure 1

23 pages, 9680 KB  
Article
Integrative Analysis of Single-Cell and Spatial Transcriptomics Reveals Intratumor Heterogeneity Shaping the Tumor Microenvironment in Hepatocellular Carcinoma
by Yue Liu, Guoping Dong, Jie Yu and Ping Liang
Livers 2025, 5(4), 62; https://doi.org/10.3390/livers5040062 - 1 Dec 2025
Viewed by 1418
Abstract
Background: Intratumour heterogeneity (ITH) is one of the key characteristics of cancer and is closely associated with patient prognosis, treatment resistance, and tumor metastasis. Nevertheless, the study of ITH in hepatocellular carcinoma (HCC) remains limited. Methods: The present study elucidated the [...] Read more.
Background: Intratumour heterogeneity (ITH) is one of the key characteristics of cancer and is closely associated with patient prognosis, treatment resistance, and tumor metastasis. Nevertheless, the study of ITH in hepatocellular carcinoma (HCC) remains limited. Methods: The present study elucidated the influence of ITH on the tumor microenvironment (TME) in HCC. We applied Non-negative Matrix Factorization (NMF) analysis to a cohort of 78 single-cell RNA sequencing (scRNA-seq) HCC samples to systematically characterize ITH. Furthermore, by integrating spatial transcriptomics (ST) data from five HCC patients, we comprehensively analyzed the spatial organization and functional properties of distinct niches within HCC. We conducted a detailed analysis of the cell-type co-localization relationships within the TME and constructed a comprehensive atlas of HCC spatial organization. Results: We observed a co-localization relationship between hypoxia tumor cells, plasmalemma vesicle-associated protein (PLVAP+) endothelial cells (EC), and vascular endothelial growth factor A (VEGFA+) cancer-associated fibroblasts (CAF), suggesting a key role for hypoxia tumor cells in VEGFA+ CAF transformation and tumor angiogenesis. We identified a unique boundary region enriched with dendritic cells1 (DC1), interferon-expressing tumor cells, lymphatic EC, C–X–C Motif Chemokine Ligand 10 (CXCL10+) macrophages (Mac), and secreted phosphoprotein 1 (SPP1+) Mac located between the tumor-infiltrating immune cells and tumor regions. Furthermore, we found that CXCL10+ Mac and SPP1+ Mac, despite co-localizing in the boundary region, exhibit distinct functions, which may be attributed to their unique spatial locations, with the former being closer to the immune-infiltrated region and the latter more proximal to the tumor area. Conclusions: Our study highlights the critical role of spatial interactions between tumor cells and the microenvironment in HCC. The findings offer new insights into ITH and underscore the importance of spatial organization in understanding cancer biology and designing future precision therapies. Full article
Show Figures

Figure 1

17 pages, 4742 KB  
Article
Multimodal Imaging of the Corneal Endothelial Transition Zone Reveals Progenitor Cell Population
by Sonika Rathi, Patricia Hülse, Susanne Staehlke, Marcus Walckling, Mahmoud Anwar, Peter Trosan, Sebastian Bohn, Oliver Stachs, Gary S. L. Peh, Gary Hin-Fai Yam, Jodhbir S. Mehta, Nicola Hofmann, Martin Börgel and Thomas A. Fuchsluger
Cells 2025, 14(23), 1851; https://doi.org/10.3390/cells14231851 - 25 Nov 2025
Viewed by 544
Abstract
The assessment of donor corneas is currently based solely on central endothelial cell (EC) density, which potentially overlooks the transition zone (TZ) regenerative potential. Therefore, the present study characterizes TZ using multimodal imaging techniques to understand its regenerative potential and refine the assessment [...] Read more.
The assessment of donor corneas is currently based solely on central endothelial cell (EC) density, which potentially overlooks the transition zone (TZ) regenerative potential. Therefore, the present study characterizes TZ using multimodal imaging techniques to understand its regenerative potential and refine the assessment of donor tissue. Ex vivo donor corneas (n = 41) were examined using phase-contrast microscopy for EC counting and reflectance confocal microscopy (HRTII/RCM) for non-invasive visualization of the TZ. A subset of eight of these corneas underwent ultrastructural analysis using field-emission scanning electron microscopy (SEM) and immunostaining analysis using confocal microscopy. We observed a significant decrease in central EC density (p < 0.001) with increasing storage duration and donor age, while TZ width and TZ surface cell count remained stable. HRTII/RCM and SEM revealed distinct morphological differences (small, polygonal cells, irregular arrangement) in the TZ compared to the peripheral endothelium (PE). Immunostaining revealed elevated expression of progenitor markers (Nestin, ABCG2, SOX2, Lgr5, Vimentin) and reduced expression of endothelial markers (ZO1 and Na/K-ATPase) in the TZ compared to the PE, indicating the presence of a stem cell-like population. These findings suggest that TZ may contribute to endothelial cell regeneration, and HRTII/RCM could serve as a novel tool for TZ evaluation in low EC count donor corneas. Full article
(This article belongs to the Special Issue Advanced Technology for Cellular Imaging)
Show Figures

Figure 1

29 pages, 3761 KB  
Review
SHP2: A Redox-Sensitive Regulator Linking Immune Checkpoint Inhibitor Therapy to Cancer Treatment and Vascular Risk
by Silvia Fernanda López Moreno, Stefania Assunto Lenz, Bernardo Casso-Chapa, Angelica Paniagua-Bojorges, Jung Hyun Kim, Nicolas L. Palaskas, Kevin T. Nead, Venkata S. K. Samanthapudi, Gilbert Mejia, Oanh Hoang, Jonghae Lee, Steven H. Lin, Joerg Herrmann, Guangyu Wang, Syed Wamique Yusuf, Cezar A. Iliescu, Noah I. Beinart, Charlotte Manisty, Masuko Ushio-Fukai, Tohru Fukai, Pietro Ameri, Roza I. Nurieva, Michelle A. T. Hildebrandt, Keri Schadler, Efstratios Koutroumpakis, Sivareddy Kotla, Nhat-Tu Le and Jun-ichi Abeadd Show full author list remove Hide full author list
Antioxidants 2025, 14(12), 1388; https://doi.org/10.3390/antiox14121388 - 21 Nov 2025
Viewed by 1220
Abstract
Src homology 2-domain containing protein tyrosine phosphatase 2 (SHP2), encoded by the Ptpn11 gene (Tyrosine-protein phosphatase non-receptor type 11), is a key downstream effector of PD-1/PD-L1 signaling and is likely important, in addition to immune modulation, in tumor development and vascular homeostasis. SHP2 [...] Read more.
Src homology 2-domain containing protein tyrosine phosphatase 2 (SHP2), encoded by the Ptpn11 gene (Tyrosine-protein phosphatase non-receptor type 11), is a key downstream effector of PD-1/PD-L1 signaling and is likely important, in addition to immune modulation, in tumor development and vascular homeostasis. SHP2 conveys PD-1 mediated inhibitory signaling in T cells, and is emerging as a therapeutic target. Importantly, there is an association between immune checkpoint inhibitors (ICIs), immune-related adverse events (irAEs), and cardiovascular complications, underscoring the need to understand SHP2’s role in these processes. This review aims to summarize current knowledge on SHP2/PTPN11 biology, its role in immune regulation, cancer progression, and vascular homeostasis, and to discuss emerging therapeutic strategies targeting this pathway. The concept of using SHP2 inhibitors with immune checkpoint inhibitors (ICIs) is being investigated to address ICI resistance and to improve anti-tumor efficacy substantially. SHP2 is also being studied in non-cancer cell contexts, and signaling responses can differ by large magnitudes depending on the biological context and stimuli. Under normal circumstances, SHP2 promotes vascular homeostasis in endothelial cells (ECs) and myeloid cells and inhibits inflammation, and the reduction in SHP2 activity by oxidative stress, such as in atherosclerosis or diabetes, upregulates inflammation. In contrast, in response to radiation, the fibrotic response and subsequent lung injury were increased by endothelial SHP2 induction via Notch-Jag1 signaling. Vascular smooth muscle cells SHP2 act as a pro-atherogenic effector by enhancing ERK/MAPK signaling, and the upregulation of mitochondria localized SHP2 can also induce cellular senescence-associated inflammation by upregulating mitochondrial reactive oxygen species. Taken together, the two opposite signaling effects of SHP2 suggest that both the immune and vascular system responses appear to be more modulated by the redox, cell, and compartment-specific signaling of SHP2. More studies are needed for mitigating cardiovascular toxicity to patients, particularly with ICI-based treatment regimens. Full article
Show Figures

Graphical abstract

22 pages, 6540 KB  
Article
The Role of Gap Junctions in MSC-EA.hy926 (An Endothelial Cell Model) Crosstalk Under Hypoxic Stress: Regulation of the Angiogenic Response
by Mariia Ezdakova, Diana Matveeva and Margarita Lobanova
Int. J. Mol. Sci. 2025, 26(22), 11239; https://doi.org/10.3390/ijms262211239 - 20 Nov 2025
Viewed by 669
Abstract
Effective communication between multipotent mesenchymal stromal cells (MSCs) and endothelial cells (ECs) plays a critical role in the regulation of angiogenesis, especially under conditions of hypoxia. In addition to paracrine stimulation, direct intercellular contacts play an important role in the angiogenic interaction between [...] Read more.
Effective communication between multipotent mesenchymal stromal cells (MSCs) and endothelial cells (ECs) plays a critical role in the regulation of angiogenesis, especially under conditions of hypoxia. In addition to paracrine stimulation, direct intercellular contacts play an important role in the angiogenic interaction between MSCs and ECs, making them an important target for modulating vascular network restoration under ischemic conditions. The aim of this study was to determine the contribution of gap junctions (GJs) to the angiogenic response of MSCs and the EA.hy926 cell line (an Endothelial Cell Model) under acute hypoxic stress. In a cell co-culture model at 0.1% O2 using a specific GJ inhibitor (carbenoxolone), molecular, cellular, and functional tests were performed: assessment of viability, proliferation, migration, secretion of angiogenic mediators, and expression of crucial genes. GJ blockade was accompanied by decreases in the proliferation and migration activity and angiogenic potential of the conditioned medium in in vitro and in ovo tests. These data highlight the importance of the GJ in coordinating the angiogenic response in conditions of acute hypoxia and can be used to develop protocols for regenerative medicine. Full article
Show Figures

Figure 1

19 pages, 7795 KB  
Article
Endothelial Cells Differentiated from Human Induced Pluripotent Stem Cells Form Aligned Network Structures in Engineered Neural Tissue
by Poppy O. Smith, Parmjit Jat and James B. Phillips
J. Funct. Biomater. 2025, 16(11), 425; https://doi.org/10.3390/jfb16110425 - 20 Nov 2025
Viewed by 1048
Abstract
Background/Objectives: Endothelial cells play a key role in peripheral nerve regeneration, forming aligned vasculature which bridges the gap in the injured nerve tissue and guides the regrowing tissue. This work aimed to mimic key features of this aligned vasculature by differentiating endothelial cells [...] Read more.
Background/Objectives: Endothelial cells play a key role in peripheral nerve regeneration, forming aligned vasculature which bridges the gap in the injured nerve tissue and guides the regrowing tissue. This work aimed to mimic key features of this aligned vasculature by differentiating endothelial cells from human induced pluripotent stem cells (hiPSCs) and incorporating them into engineered neural tissue (EngNT). Methods: hiPSCs were differentiated into endothelial cells with the temporal addition of growth factors and biomolecules. These hiPSC-derived endothelial cells (hiPSC-ECs) were incorporated into EngNT fabricated from collagen hydrogels using the gel aspiration-ejection (GAE) technique and maintained in vitro to allow endothelial network formation. Results: At the mRNA and protein level, pluripotency marker expression decreased and endothelial cell marker expression increased over the course of hiPSC differentiation to endothelial cells. The derived endothelial cells expressed CD31, CD144, ENG, VEGFR2, and VWF, and formed network structures in the matrix tubulogenesis assay. hiPSC-ECs incorporated into EngNT were viable and aligned. They formed highly aligned tube-like structures containing lumens after four days in culture and the EngNT constructs supported neurite growth in vitro when co-cultured with rat dorsal root ganglion (DRG) neurons. Conclusions: This work rapidly generated engineered nerve tissue containing highly aligned endothelial tube-like structures, resembling key features of the early nerve regeneration bridge. Therefore, this 3D engineered tissue provides a platform to study the effects of endothelial cell structures in nerve repair treatment and translational development. Full article
Show Figures

Graphical abstract

17 pages, 609 KB  
Review
RhoA/Rho-Kinase Signaling in Vascular Smooth Muscle and Endothelium: Mechanistic Insights and Translational Implications in Hypertension
by Stephanie Randar, Diana L. Silva-Velasco, Fernanda Priviero and R. Clinton Webb
Biomolecules 2025, 15(11), 1607; https://doi.org/10.3390/biom15111607 - 16 Nov 2025
Viewed by 1469
Abstract
The small GTPase RhoA and its downstream effector Rho-kinase (ROCK) have emerged as pivotal regulators of vascular smooth muscle cell (VSMC) contraction, endothelial function, and vascular remodeling. Activation of the RhoA/ROCK pathway enhances calcium (Ca2+) sensitivity by inhibiting myosin light chain [...] Read more.
The small GTPase RhoA and its downstream effector Rho-kinase (ROCK) have emerged as pivotal regulators of vascular smooth muscle cell (VSMC) contraction, endothelial function, and vascular remodeling. Activation of the RhoA/ROCK pathway enhances calcium (Ca2+) sensitivity by inhibiting myosin light chain phosphatase (MLCP), thereby promoting sustained vascular tone independent of intracellular Ca2+ levels. In endothelial cells (ECs), RhoA/ROCK signaling contributes to nitric oxide (NO) dysregulation, oxidative stress, cytoskeletal reorganization, and inflammatory activation. Cumulative evidence implicates this pathway in the development and progression of hypertension and other cardiovascular diseases, where maladaptive vascular remodeling, VSMC proliferation, and endothelial dysfunction drive increased vascular resistance. Translational studies have identified ROCK inhibitors and indirect modulators such as statins as promising therapeutic strategies. This review integrates recent mechanistic insights into RhoA/ROCK regulation of vascular function with clinical and translational perspectives on targeting this pathway in hypertension. Full article
Show Figures

Figure 1

48 pages, 2461 KB  
Review
Role of Matricellular Proteins in Endothelial Cell Inflammation and Atherosclerosis
by Ravi Varma Aithabathula, Santosh Kumar and Bhupesh Singla
Antioxidants 2025, 14(11), 1338; https://doi.org/10.3390/antiox14111338 - 6 Nov 2025
Viewed by 1253
Abstract
The vascular endothelium serves as a critical barrier preventing the transmigration of monocytes, circulating lipoproteins, and other molecules into the subendothelial space, and plays a vital role in regulating vascular tone. A dysfunctional and inflamed endothelial layer in response to disturbed blood flow [...] Read more.
The vascular endothelium serves as a critical barrier preventing the transmigration of monocytes, circulating lipoproteins, and other molecules into the subendothelial space, and plays a vital role in regulating vascular tone. A dysfunctional and inflamed endothelial layer in response to disturbed blood flow or other proatherogenic risk factors is the initiating event in the pathogenesis of atherosclerosis, suggesting the importance of an intact and properly functioning endothelium in preventing the onset and progression of this disease. Accumulated evidence demonstrates the significant role of matricellular proteins, which are non-structural and secretory extracellular matrix (ECM) proteins, in the development of atherosclerosis. These proteins exert multifaceted effects on endothelial cells (ECs) ranging from reactive oxygen species (ROS) production, endoplasmic reticulum stress, and expression of adhesion molecules to autophagy and compromised barrier function via stimulating various molecular mechanisms. Given the critical roles of these processes in EC function and atherosclerosis, a better understanding of signaling pathways governed by matricellular proteins in ECs is required to develop therapeutic strategies for suppressing or preventing atherosclerosis and related cardiovascular diseases (CVDs). This review comprehensively summarizes the existing literature on the diverse roles of matricellular proteins in regulating EC inflammation and function, and highlights their potential as viable therapeutic targets for maintaining vascular health and inhibiting the progression of atherosclerosis. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 3rd Edition)
Show Figures

Figure 1

Back to TopTop