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

Journals

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

Search Results (453)

Search Parameters:
Keywords = endothelial cell network

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
30 pages, 51386 KB  
Article
Aspirin Eugenol Ester Alleviates Vascular Endothelial Ferroptosis by Enhancing Antioxidant Ability and Inhibiting the JNK/c-Jun/NCOA4/FTH Signaling Pathway
by Ji Feng, Qi Tao, Zhi-Jie Zhang, Qin-Fang Yu, Ya-Jun Yang and Jian-Yong Li
Antioxidants 2025, 14(10), 1220; https://doi.org/10.3390/antiox14101220 - 10 Oct 2025
Abstract
Oxidative stress occurs within bovine when exposed to harmful stimuli, accompanied by substantial accumulation of reactive oxygen species. Without timely clearance, these reactive oxygen species attack vascular endothelial cells, concurrently inducing extensive production of lipid peroxides within the vascular endothelium, and thereby triggering [...] Read more.
Oxidative stress occurs within bovine when exposed to harmful stimuli, accompanied by substantial accumulation of reactive oxygen species. Without timely clearance, these reactive oxygen species attack vascular endothelial cells, concurrently inducing extensive production of lipid peroxides within the vascular endothelium, and thereby triggering ferroptosis. Aspirin eugenol ester (AEE) showed pharmacological activity against oxidative stress-induced vascular endothelial damage. However, whether it could alleviate vascular endothelial damage by inhibiting ferroptosis remains unclear. This study aimed to evaluate the effects of AEE on vascular endothelial ferroptosis and elucidate its underlying molecular mechanisms. This study established vascular endothelial damage models in vitro and in vivo to explore the ability of AEE to inhibit ferroptosis and oxidative stress by measuring ferroptosis- and oxidative stress-related biomarkers. Transcriptomic and network pharmacology analyses were performed to identify AEE-regulated pathways and key targets. Validation of the pathways were conducted using molecular docking, cellular thermal shift assay, and specific protein agonists/inhibitors. AEE inhibited oxidative stress and ferroptosis in bovine aortic endothelial cells induced by hydrogen peroxide (H2O2) or RSL3 via suppressing the upregulation of ferroptosis-related genes and enhancing the expression of antioxidant genes. Transcriptomic and network pharmacology analyses identified JNK as a core target of AEE in regulating ferroptosis. JNK agonists enhanced H2O2-induced ferritinophagy; on the contrary, JNK inhibitors alleviated it. AEE suppressed H2O2-induced phosphorylation of JNK/c-Jun and ferritinophagy. In a carrageenan-induced rat aortic vascular endothelial damage model, AEE alleviated vascular endothelial damage and ferroptosis-related gene changes, promoted antioxidant gene expression, and inhibited JNK/c-Jun phosphorylation and ferritinophagy. AEE inhibited vascular endothelial ferroptosis by enhancing antioxidant ability, blocking downstream ferritinophagy, and reducing ferrous ion release. Full article
(This article belongs to the Section Aberrant Oxidation of Biomolecules)
Show Figures

Graphical abstract

26 pages, 6743 KB  
Article
Matrix-Guided Vascular-like Cord Formation by MRC-5 Lung Fibroblasts: Evidence of Structural and Transcriptional Plasticity
by Nikoleta F. Theodoroula, Alexandros Giannopoulos-Dimitriou, Aikaterini Saiti, Aliki Papadimitriou-Tsantarliotou, Androulla N. Miliotou, Giannis Vatsellas, Yiannis Sarigiannis, Eleftheria Galatou, Christos Petrou, Dimitrios G. Fatouros and Ioannis S. Vizirianakis
Cells 2025, 14(19), 1519; https://doi.org/10.3390/cells14191519 - 29 Sep 2025
Viewed by 694
Abstract
The role of mesenchymal-to-endothelial transition in the angiogenic response remains controversial. In this study, we investigated whether human fetal lung fibroblasts (MRC-5 cells) exhibit morphological plasticity in a biomimetic extracellular matrix environment. To this end, MRC-5 cells were first cultured on and within [...] Read more.
The role of mesenchymal-to-endothelial transition in the angiogenic response remains controversial. In this study, we investigated whether human fetal lung fibroblasts (MRC-5 cells) exhibit morphological plasticity in a biomimetic extracellular matrix environment. To this end, MRC-5 cells were first cultured on and within Matrigel hydrogel and then studied with tube formation assays, confocal/fluorescence microscopy, invasion assays, and transcriptomic profiling. In addition, quantitative assessment for cord formation and gene expression was conducted via qPCR and RNA sequencing. In this study, MRC-5 cells quickly self-organized into cord-like networks, resembling early stages of vascular patterning, and at higher densities, invaded the hydrogel and formed spheroid-like aggregates. Transcriptomic analysis revealed upregulation of genes related to nervous system development and synaptic signaling in Matrigel-grown MRC-5 cultures. Collectively, these findings suggest that MRC-5 fibroblasts display structural and transcriptional plasticity in 3D Matrigel cultures, forming vascular-like cords that are more likely to resemble early developmental morphologies or neuroectodermal-like transcriptional signatures than definitive endothelial structures. This work underscores the potential of fibroblasts as an alternative cell source for vascular tissue engineering and highlights a strategy to overcome current limitations in autologous endothelial cell availability for regenerative applications. Full article
(This article belongs to the Collection Advances in Epithelial-Mesenchymal Transition (EMT))
Show Figures

Figure 1

20 pages, 2967 KB  
Article
Effect of Precipitated Extracellular Marennine on Angiogenesis and Tumour Cell Proliferation
by Mostefa Fodil, Javier Muñoz-Garcia, Amel-Khitem Benali, Jasmina Rogozarski, Virginie Mignon, Honora Labrana, Anna Lokajczyk, Pamela Pasetto, Jean-Luc Mouget, Catherine Boisson-Vidal and Dominique Heymann
Mar. Drugs 2025, 23(9), 364; https://doi.org/10.3390/md23090364 - 19 Sep 2025
Viewed by 448
Abstract
Angiogenesis is a fundamental biological process involved in the formation of new blood vessels from the pre-existing vascular network. In addition to physiological processes, angiogenesis is also implicated in pathological conditions such as tumour growth and metastatic progression. Research on marennine, a water-soluble [...] Read more.
Angiogenesis is a fundamental biological process involved in the formation of new blood vessels from the pre-existing vascular network. In addition to physiological processes, angiogenesis is also implicated in pathological conditions such as tumour growth and metastatic progression. Research on marennine, a water-soluble blue-green pigment produced by the marine diatom Haslea ostrearia, has highlighted various promising biological activities. In vivo studies have suggested the potential of marennine in cancer treatment. However, these studies were conducted with crude extracts, the exact composition of which remained poorly defined. In this context, our study aimed to explore the effects of marennine on angiogenesis and tumour proliferation by using a Precipitated Extracellular Marennine (PEMn) extract. Our results confirmed the antiproliferative properties of PEMn on several cancer cell lines associated with angiogenic tumours. We then analysed its impact on the key steps of the angiogenic process, including Endothelial Colony-Forming Cells (ECFCs) proliferation, migration, and tubulogenesis. In parallel, we investigated the underlying mechanisms of its action, notably by assessing its effects on cell cycle regulation, senescence, and apoptosis. PEMn significantly inhibited tumour cell proliferation, induced ECFC senescence and apoptosis, impaired migration and tubulogenesis, and downregulated VEGFR-1 expression, highlighting its potential as a novel marine-derived antiangiogenic compound. These findings provide deeper insights into the mechanisms of action of marennine, identifying this bioactive natural compound as a novel bioactive compound in cancer treatment. Full article
(This article belongs to the Collection Bioactive Compounds from Marine Plankton)
Show Figures

Figure 1

28 pages, 3457 KB  
Review
Alveolar Epithelial Cell Dysfunction in Acute Respiratory Distress Syndrome: Mechanistic Insights and Targeted Interventions
by Jing Wang and Jie Chao
Biomedicines 2025, 13(9), 2299; https://doi.org/10.3390/biomedicines13092299 - 19 Sep 2025
Viewed by 828
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening condition with high mortality. A central driver in its pathogenesis is alveolar epithelial cell (AEC) dysfunction, which leads to disruption of the epithelial barrier, impaired fluid clearance, and dysregulated inflammatory responses. This review summarizes the [...] Read more.
Acute respiratory distress syndrome (ARDS) is a life-threatening condition with high mortality. A central driver in its pathogenesis is alveolar epithelial cell (AEC) dysfunction, which leads to disruption of the epithelial barrier, impaired fluid clearance, and dysregulated inflammatory responses. This review summarizes the key mechanisms underlying AEC injury, including programmed cell death (apoptosis, pyroptosis, necroptosis, ferroptosis), oxidative stress, mitochondrial dysfunction, epigenetic reprogramming (DNA methylation, histone modifications), metabolic rewiring (succinate accumulation), and spatiotemporal heterogeneity revealed by single-cell sequencing and spatial transcriptomics. Multicellular crosstalk involving epithelial–immune–endothelial networks and the gut-lung axis further shapes disease progression. Building on these mechanistic foundations, we evaluate emerging AEC-targeted interventions such as pharmacologic agents (antioxidants, anti-inflammatories), biologics (mesenchymal stem cells and engineered exosomes), and gene-based approaches (adeno-associated virus and CRISPR-Cas9 systems delivered via smart nanocarriers). Complementary strategies include microbiome modulation through probiotics, short-chain fatty acids, or fecal microbiota transplantation, and biomarker-guided precision medicine (e.g., sRAGE, exosomal miRNAs) to enable promise individualized regimens. We also discuss translational hurdles, including nanotoxicity, mesenchymal stem cell (MSC) heterogeneity, and gene-editing safety, and highlight future opportunities involving AI-driven multi-omics, lung-on-chip platforms, and epithelium-centered regenerative therapies. By integrating mechanistic insights with innovative therapeutic strategies, this review aims to outline a roadmap toward epithelium-targeted, precision-guided therapies for ARDS. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Graphical abstract

17 pages, 2575 KB  
Article
Murine Cell Line Models for Vascular Mimicry: The Role of YAP/TAZ Signaling
by Matilde Righetti, Ana-Maria Primorac, Janine Terra Erler and Victor Oginga Oria
Int. J. Mol. Sci. 2025, 26(18), 9129; https://doi.org/10.3390/ijms26189129 - 18 Sep 2025
Viewed by 379
Abstract
Vascular mimicry (VM) refers to the formation of vessel-like structures by tumor cells independent of endothelial cells. These VM channels connect to the host’s vascular network and are associated with aggressive tumors and poor patient prognosis. Most VM research has been conducted on [...] Read more.
Vascular mimicry (VM) refers to the formation of vessel-like structures by tumor cells independent of endothelial cells. These VM channels connect to the host’s vascular network and are associated with aggressive tumors and poor patient prognosis. Most VM research has been conducted on melanoma, relying on patient-derived and mouse cell lines. In other solid tumors, VM studies rely on human cell lines, which have certain limitations for in vivo studies. Specifically, most in vivo VM research involving human cells uses subcutaneous mouse models that fail to recapitulate organ-specific tumor microenvironments. As the microenvironment is an essential driver of tumor vascularization, including VM, murine cell lines could facilitate VM investigations in syngeneic mouse models. Here, we present CT26 and KPC, well-characterized murine colorectal and pancreatic cancer cell lines, as cell models for VM investigations. Using in vitro cell-based assays, we demonstrate that CT26 and KPC undergo VM, a cell-intrinsic process that is enhanced by serum deprivation and exposure to hypoxia and is independent of tumor-secreted growth factors. Additionally, we demonstrate the importance of YAP/TAZ signaling in VM formation, as inhibition at non-cytotoxic concentrations attenuated VM formation. Remarkably, CA3, the most potent of the two inhibitors, significantly reduced cell proliferation in both cell lines at the IC50 concentration. This reduction in cell proliferation was associated with the induction of apoptosis in CT26 cells and changes in the cell cycle in both CT26 and KPC cells. Finally, dual YAP/TAZ knockdown in both cell lines significantly abrogated VM formation, validating our initial findings using inhibitors. These results show that CT26 and KPC cells undergo VM, and given their extensive use in cancer research, can be used to investigate VM in vivo using syngeneic models. Full article
Show Figures

Figure 1

18 pages, 604 KB  
Review
MicroRNA (miRNA) in the Pathogenesis of Diabetic Retinopathy: A Narrative Review
by Stamatios Lampsas, Chrysa Agapitou, Alexandros Chatzirallis, Georgios Papavasileiou, Dimitrios Poulakis, Sofia Pegka, Panagiotis Theodossiadis, Vaia Lambadiari and Irini Chatziralli
Genes 2025, 16(9), 1060; https://doi.org/10.3390/genes16091060 - 9 Sep 2025
Viewed by 620
Abstract
Diabetic retinopathy (DR) is the most common microvascular complication associated with diabetes mellitus and represents a leading cause of visual impairment worldwide. Inflammation, endothelial dysfunction, angiogenesis, neurodegeneration, and oxidative stress are key pathogenic processes in the development and progression of DR. Numerous microRNAs [...] Read more.
Diabetic retinopathy (DR) is the most common microvascular complication associated with diabetes mellitus and represents a leading cause of visual impairment worldwide. Inflammation, endothelial dysfunction, angiogenesis, neurodegeneration, and oxidative stress are key pathogenic processes in the development and progression of DR. Numerous microRNAs (miRNAs) show altered expression in DR and modulate critical biological pathways. Pro-inflammatory miRNAs such as miR-155 and miR-21 promote cytokine release and vascular inflammation, while miR-146a acts as a negative regulator of Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. MiR-126 and miR-21 regulate endothelial integrity and angiogenesis through pathways involving Vascular Endothelial Growth Factor (VEGF). MiR-200b and miR-126 are downregulated in DR, leading to increased neovascularization via activation of the VEGF/ Mitogen-Activated Protein Kinase (MAPK) cascade. Apoptotic processes are affected by miR-195, which downregulates Sirtuin 1 (SIRT1) and B-cell lymphoma 2 (Bcl-2), promoting retinal cell death, while miR-29b downregulation permits upregulation of the transcription factor SP1, enhancing caspase-mediated apoptosis in Müller cells and endothelial cells. miRNAs collectively modulate an intricate regulatory network that contributes to the underlying mechanisms of diabetic retinopathy development and progression. This narrative review aims to summarize knowledge regarding the mechanisms miRNAs mediating pathogenetic mechanisms of DR. Full article
(This article belongs to the Section RNA)
Show Figures

Figure 1

23 pages, 2403 KB  
Review
Decoding the Tumor Microenvironment: Insights and New Targets from Single-Cell Sequencing and Spatial Transcriptomics
by Shriya Pattabiram, Prakash Gangadaran, Sanjana Dhayalan, Gargii Chatterjee, Danyal Reyaz, Kruthika Prakash, Raksa Arun, Ramya Lakshmi Rajendran, Byeong-Cheol Ahn and Kandasamy Nagarajan Aruljothi
Curr. Issues Mol. Biol. 2025, 47(9), 730; https://doi.org/10.3390/cimb47090730 - 9 Sep 2025
Viewed by 738
Abstract
The field of oncology has been extensively studied to design more effective and efficient treatments. This review explores the advanced techniques that are transforming our comprehension of cancer and its constituents. Specifically, it highlights the signaling pathways that drive tumor progression, angiogenesis, and [...] Read more.
The field of oncology has been extensively studied to design more effective and efficient treatments. This review explores the advanced techniques that are transforming our comprehension of cancer and its constituents. Specifically, it highlights the signaling pathways that drive tumor progression, angiogenesis, and resistance to therapy, as well as the modern approaches used to identify and characterize these pathways within the tumor microenvironment (TME). Key pathways discussed in this review include vascular endothelial growth factor (VEGF), programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and various extracellular matrix (ECM) pathways. Conventional methods of diagnosis have yielded sufficient knowledge but have failed to reveal the heterogeneity that exists within the TME, resulting in gaps in our understanding of the cellular interaction and spatial dynamics. Single-cell sequencing (SCS) and spatial transcriptomics (ST) are effective tools that can enable the dissection of the TME with the resolution capacity of a single cell. SCS allows the capture of the unique genetic and transcriptomic profiles of individual cells along with rare cell types and new therapeutic targets. ST complements this by providing a spatial map of gene expression, showing the gene expression profiles within the tumor tissue at specific sites with good accuracy. By mapping gene expression patterns at a single cell level and correlating them with the spatial locations, researchers can uncover the intricate networks and microenvironmental influences that contribute to tumor heterogeneity. Full article
(This article belongs to the Special Issue Technological Advances Around Next-Generation Sequencing Application)
Show Figures

Figure 1

29 pages, 994 KB  
Review
Angiogenic microRNAs in Systemic Sclerosis: Insights into Microvascular Dysfunction and Therapeutic Implications
by Marta Rusek
Genes 2025, 16(9), 1057; https://doi.org/10.3390/genes16091057 - 9 Sep 2025
Viewed by 582
Abstract
Systemic sclerosis (SSc) is a complex connective tissue disease that affects the skin and internal organs and is characterized by immune dysregulation, progressive fibrosis, and microvascular dysfunction. Chronic tissue ischemia, accompanied by impaired angiogenesis, leads to the gradual loss of small vessels, resulting [...] Read more.
Systemic sclerosis (SSc) is a complex connective tissue disease that affects the skin and internal organs and is characterized by immune dysregulation, progressive fibrosis, and microvascular dysfunction. Chronic tissue ischemia, accompanied by impaired angiogenesis, leads to the gradual loss of small vessels, resulting in clinical complications, such as Raynaud’s phenomenon, digital ulcers, pulmonary arterial hypertension, and renal crisis. Emerging evidence highlights the crucial regulatory role of microRNAs (miRNAs) in vascular homeostasis through the modulation of key signaling pathways and endothelial cell activity. Dysregulated miRNAs influence fibroblast proliferation, inflammatory responses, and immune cell activity in SSc, contributing to disease progression. Current knowledge is still limited, highlighting the need for further research to elucidate the miRNAs network involved in the etiopathogenesis of SSc. The use of miRNA-based biomarkers is gaining tremendous attention for early diagnosis, risk stratification, classification, and the prediction of therapeutic responses. This review provides insights into angiogenesis-related miRNAs involved in SSc pathogenesis, discusses their relevance as biomarkers, and explores their promise as therapeutic targets. Advancing our knowledge of miRNAs-mediated regulatory networks may open new possibilities for personalized approaches to SSc management. Full article
(This article belongs to the Section RNA)
Show Figures

Figure 1

42 pages, 3184 KB  
Review
The β-1,4 GalT-V Interactome—Potential Therapeutic Targets and a Network of Pathways Driving Cancer and Cardiovascular and Inflammatory Diseases
by Subroto Chatterjee, Dhruv Kapila, Priya Dubey, Swathi Pasunooti, Sruthi Tatavarthi, Claire Park and Caitlyn Ramdat
Int. J. Mol. Sci. 2025, 26(16), 8088; https://doi.org/10.3390/ijms26168088 - 21 Aug 2025
Viewed by 1110
Abstract
UDP-Gal-β-1,4 galactosyltransferase-V (GalT-V) is a member of a large family of galactosyltransferases whose function is to transfer galactose from the nucleotide sugar UDP-galactose to a glycosphingolipid glucosylceramide, to generate lactosylceramide (LacCer). It also causes the N and O glycosylation of proteins in the [...] Read more.
UDP-Gal-β-1,4 galactosyltransferase-V (GalT-V) is a member of a large family of galactosyltransferases whose function is to transfer galactose from the nucleotide sugar UDP-galactose to a glycosphingolipid glucosylceramide, to generate lactosylceramide (LacCer). It also causes the N and O glycosylation of proteins in the Trans Golgi area. LacCer is a bioactive lipid second messenger that activates an “oxidative stress pathway”, leading to critical phenotypes, e.g., cell proliferation, migration angiogenesis, autophagy, and apoptosis. It also activates an “inflammatory pathway” that contributes to the progression of disease pathology. β-1,4-GalT-V gene expression is regulated by the binding of the transcription factor Sp-1, one of the most O-GlcNAcylated nuclear factors. This review elaborates the role of the Sp-1/GalT-V axis in disease phenotypes and therapeutic approaches targeting not only Sp-1 but also Notch-1, Wnt-1 frizzled, hedgehog, and β-catenin. Recent evidence suggests that β-1,4GalT-V may glycosylate Notch-1 and, thus, regulate a VEGF-independent angiogenic pathway, promoting glioma-like stem cell differentiation into endothelial cells, thus contributing to angiogenesis. These findings have significant implications for cancer and cardiovascular disease, as tumor vascularization often resumes aggressively following anti-VEGF therapy. Moreover, LacCer can induce angiogenesis independent of VEGF and its level are reported to be high in tumor tissues. Thus, targeting both VEGF-dependent and VEGF-independent pathways may offer novel therapeutic strategies. This review also presents an up-to-date therapeutic approach targeting the β-1,4-GalT-V interactome. In summary, the β-1,4-GalT-V interactome orchestrates a broad network of signaling pathways essential for maintaining cellular homeostasis. Conversely, its dysregulation can promote unchecked proliferation, angiogenesis, and inflammation, contributing to the initiation and progression of multiple diseases. Environmental factors and smoking can influence β-1,4-GalT-V expression and its interactome, whereas elevated β-1,4-GalT-V expression may serve as a diagnostic biomarker of colorectal cancer, inflammation—exacerbated by factors that may worsen pre-existing cancer malignancies, such as smoking and a Western diet—and atherosclerosis, amplifying disease progression. Increased β-1,4-GalT-V expression is frequently associated with tumor aggressiveness and chronic inflammation, underscoring its potential as both a biomarker and therapeutic target in colorectal and other β-1,4-GalT-V-driven cancers, as well as in cardiovascular and inflammatory diseases. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

21 pages, 12853 KB  
Article
Identification of Novel Lactylation-Related Biomarkers for COPD Diagnosis Through Machine Learning and Experimental Validation
by Chundi Hu, Weiliang Qian, Runling Wei, Gengluan Liu, Qin Jiang, Zhenglong Sun and Hui Li
Biomedicines 2025, 13(8), 2006; https://doi.org/10.3390/biomedicines13082006 - 18 Aug 2025
Viewed by 913
Abstract
Objective: This study aims to identify clinically relevant lactylation-related biomarkers in chronic obstructive pulmonary disease (COPD) and investigate their potential mechanistic roles in COPD pathogenesis. Methods: Differentially expressed genes (DEGs) were identified from the GSE21359 dataset, followed by weighted gene co-expression network analysis [...] Read more.
Objective: This study aims to identify clinically relevant lactylation-related biomarkers in chronic obstructive pulmonary disease (COPD) and investigate their potential mechanistic roles in COPD pathogenesis. Methods: Differentially expressed genes (DEGs) were identified from the GSE21359 dataset, followed by weighted gene co-expression network analysis (WGCNA) to detect COPD-associated modules. Least absolute shrinkage and selection operator (LASSO) regression and support vector machine–recursive feature elimination (SVM–RFE) algorithms were applied to screen lactylation-related biomarkers, with diagnostic performance evaluated through the ROC curve. Candidates were validated in the GSE76925 dataset for expression and diagnostic robustness. Immune cell infiltration patterns were exhibited using EPIC deconvolution. Single-cell transcriptomics (from GSE173896) were processed via the ‘Seurat’ package encompassing quality control, dimensionality reduction, and cell type annotation. Cell-type-specific markers and intercellular communication networks were delineated using the ‘FindAllMarkers’ package and the ‘CellChat’ R package, respectively. In vitro validation was conducted using a cigarette smoke extract (CSE)-induced COPD model. Results: Integrated transcriptomic approaches and multi-algorithm screening (LASSO/Boruta/SVM–RFE) revealed carbonyl reductase 1 (CBR1) and peroxiredoxin 1 (PRDX1) as core COPD biomarkers enriched in oxidation–reduction and inflammatory pathways, with high diagnostic accuracy (AUC > 0.85). Immune profiling and scRNA-seq delineated macrophage and cancer-associated fibroblasts (CAFs) infiltration with oxidative-redox transcriptional dominance in COPD. CBR1 was significantly upregulated in T cells, neutrophils, and mast cells; and PRDX1 showed significant upregulation in endothelial, macrophage, and ciliated cells. Experimental validation in CSE-induced models confirmed significant upregulation of both biomarkers via transcription PCR (qRT-PCR) and immunofluorescence. Conclusions: CBR1 and PRDX1 are lactylation-associated diagnostic markers, with lactylation-driven redox imbalance implicated in COPD progression. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

18 pages, 7739 KB  
Article
Construction and Analysis of Immune Infiltration and Competing Endogenous RNA Network in Moyamoya Disease
by Wenhao Liu, Hanhui Fu, Shiyuan Fang, Jun Ni and Bin Peng
Int. J. Mol. Sci. 2025, 26(16), 7957; https://doi.org/10.3390/ijms26167957 - 18 Aug 2025
Cited by 1 | Viewed by 562
Abstract
Moyamoya disease (MMD) is a cerebrovascular condition characterized by progressive stenosis of intracranial arteries, leading to stroke. While MMD was long considered a genetic disorder, emerging evidence suggests autoimmune mechanisms may contribute to its pathogenesis. The role of non-coding RNAs (ncRNAs) in the [...] Read more.
Moyamoya disease (MMD) is a cerebrovascular condition characterized by progressive stenosis of intracranial arteries, leading to stroke. While MMD was long considered a genetic disorder, emerging evidence suggests autoimmune mechanisms may contribute to its pathogenesis. The role of non-coding RNAs (ncRNAs) in the pathogenesis of MMD is under heated discussion, and a competitive endogenous RNA (ceRNA) network involving MMD-related ncRNAs has not been constructed. In this study, we integrated multiple bioinformatic analyses on transcriptomic data from the middle cerebral arteries of MMD patients and controls. Our analysis revealed a significant enrichment of innate immune system pathways, including antigen processing and macrophage activation, in MMD tissue. We constructed a robust ceRNA network centered on the long non-coding RNA MALAT1, identifying 15 core mRNA targets. A classifier built from these MALAT1-related genes accurately distinguished MMD patients from controls, with an area under the curve of 0.869 in independent validation. Furthermore, immune deconvolution analysis showed a marked increase in microvascular endothelial cells and a decrease in CD4+ memory T cells and regulatory T cells in MMD arteries. The expression of the MALAT1 network genes strongly correlated with these shifts in cellular composition, positively with endothelial cells and negatively with T cells. Our findings uncover a MALAT1-driven ceRNA network that links immune dysregulation to vascular changes in MMD, highlighting MALAT1 as a potential biomarker and therapeutic target. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

25 pages, 3253 KB  
Review
Multisystem Endothelial Inflammation: A Key Driver of Adverse Events Following mRNA-Containing COVID-19 Vaccines
by János Szebeni and Akos Koller
Vaccines 2025, 13(8), 855; https://doi.org/10.3390/vaccines13080855 - 12 Aug 2025
Viewed by 3610
Abstract
mRNA-LNP-based COVID-19 vaccines, namely Pfizer-BioNTech’s Comirnaty and Moderna’s Spikevax, were successfully deployed to help control the SARS-CoV-2 pandemic, and their updated formulations continue to be recommended, albeit only for high-risk populations. One widely discussed aspect of these vaccines is their uniquely broad spectrum [...] Read more.
mRNA-LNP-based COVID-19 vaccines, namely Pfizer-BioNTech’s Comirnaty and Moderna’s Spikevax, were successfully deployed to help control the SARS-CoV-2 pandemic, and their updated formulations continue to be recommended, albeit only for high-risk populations. One widely discussed aspect of these vaccines is their uniquely broad spectrum and increased incidence of adverse events (AEs), collectively referred to as post-vaccination syndrome (PVS). Although the reported PVS rate is low, the high number of administered doses among healthy individuals has resulted in a substantial number of reported vaccine-related injuries. A prominent manifestation of PVS is multisystem inflammation, hypothesized to result from the systemic transfection of organ cells with genetic instructions for a toxin, the spike protein, delivered with lipid nanoparticles (LNPs). In this narrative review, we focus on endothelial cells in the microcirculatory networks of various organs as primary sites of transfection with mRNA-LNP and consequent PVS. We outline the anatomical variations in the microcirculation contributing to the individual variability of symptoms and examine the molecular and cellular responses to vaccine nanoparticle exposure at the endothelial cell level with a focus on the pathways of a sustained cascade of toxic and autoimmune processes. A deeper understanding of the mechanisms underlying mRNA-LNP-induced AEs and PVS at the organ and cellular levels is critical for improving the safety of future vaccines and other therapeutic applications of this groundbreaking technology. Full article
Show Figures

Figure 1

16 pages, 3925 KB  
Communication
Identifying Angiogenic Factors in Pediatric Choroid Plexus Papillomas
by Nurfarhanah Bte Syed Sulaiman, Sofiah M. Y. Sng, Khurshid Z. Merchant, Lee Ping Ng, David C. Y. Low, Wan Tew Seow and Sharon Y. Y. Low
NeuroSci 2025, 6(3), 76; https://doi.org/10.3390/neurosci6030076 - 11 Aug 2025
Viewed by 736
Abstract
(1) Background: Choroid plexus papillomas (CPPs) are rare brain tumors that tend to occur in very young children. Mechanisms of CPP development remain unelucidated. Separately, the process of angiogenesis has been implicated in other primary brain tumors. We hypothesize that angiogenesis is a [...] Read more.
(1) Background: Choroid plexus papillomas (CPPs) are rare brain tumors that tend to occur in very young children. Mechanisms of CPP development remain unelucidated. Separately, the process of angiogenesis has been implicated in other primary brain tumors. We hypothesize that angiogenesis is a hallmark of CPP biology. This study aims to identify and validate angiogenic factors in CPPs. (2) Methods: Cerebrospinal fluid (CSF) and CPP tumor samples are collected. A multiplex immunoassay panel is used to identify differentially expressed cytokines in the CSF samples. Concurrently, patient-derived primary cell cultures and their supernatants are derived from CPP samples. Targeted proteome blot arrays and human umbilical vein endothelial cell (HUVEC) angiogenesis assays are used for validation studies. (3) Results: CSF profiling showed higher expressions of VEGF-A, MCP-1, MMP-1, TNF-α, and CD40L in CPP patient samples versus non-tumor controls. Next, assessment via online protein–protein network platforms reports that these cytokines are associated with endothelial cell regulation. Using an angiogenesis-focused approach, CPP-derived cell lines and supernatants showed similarly higher expressions of VEGF, MCP-1, and MMP-1. Next, sprouting of nodes and tubule formation were observed in HUVEC angiogenesis assay cultures when conditioned CPP cell culture media was added. (4) Conclusions: This proof-of-concept study demonstrates potential to explore angiogenesis in CPP. Full article
Show Figures

Figure 1

29 pages, 14681 KB  
Article
Single-Nucleus RNA Sequencing and Spatial Transcriptomics Reveal Cellular Heterogeneity and Intercellular Communication Networks in the Hypothalamus–Pituitary–Ovarian Axis of Pregnant Mongolian Cattle
by Yanchun Bao, Fengying Ma, Chenxi Huo, Hongxia Jia, Yunhan Li, Xiaoyi Yang, Jiajing Liu, Pengbo Gu, Caixia Shi, Mingjuan Gu, Lin Zhu, Yu Wang, Bin Liu, Risu Na and Wenguang Zhang
Animals 2025, 15(15), 2277; https://doi.org/10.3390/ani15152277 - 4 Aug 2025
Viewed by 914
Abstract
The hypothalamus–pituitary–ovarian (HPO) axis orchestrates reproductive functions through intricate neuroendocrine crosstalk. Here, we integrated single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics (ST) to decode the cellular heterogeneity and intercellular communication networks in the reproductive systems of pregnant Mongolian cattle. We retained a total [...] Read more.
The hypothalamus–pituitary–ovarian (HPO) axis orchestrates reproductive functions through intricate neuroendocrine crosstalk. Here, we integrated single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics (ST) to decode the cellular heterogeneity and intercellular communication networks in the reproductive systems of pregnant Mongolian cattle. We retained a total of 6161 high-quality nuclei from the hypothalamus, 14,715 nuclei from the pituitary, and 26,072 nuclei from the ovary, providing a comprehensive cellular atlas across the HPO axis. In the hypothalamus, neurons exhibited synaptic and neuroendocrine specialization, with glutamatergic subtype Glut4 serving as a TGFβ signaling hub to regulate pituitary feedback, while GABAergic GABA1 dominated PRL signaling, likely adapting maternal behavior. Pituitary stem cells dynamically replenished endocrine populations via TGFβ, and lactotrophs formed a PRLPRLR paracrine network with stem cells, synergizing mammary development. Ovarian luteal cells exhibited steroidogenic specialization and microenvironmental synergy: endothelial cells coregulated TGFβ-driven angiogenesis and immune tolerance, while luteal–stromal PRLPRLR interactions amplified progesterone synthesis and nutrient support. Granulosa cells (GCs) displayed spatial-functional stratification, with steroidogenic GCs persisting across pseudotime as luteinization precursors, while atretic GCs underwent apoptosis. Spatial mapping revealed GCs’ annular follicular distribution, mediating oocyte–somatic crosstalk, and luteal–endothelial colocalization supporting vascularization. This study unveils pregnancy-specific HPO axis regulation, emphasizing multi-organ crosstalk through TGFβ/PRL pathways and stem cell-driven plasticity, offering insights into reproductive homeostasis and pathologies. Full article
(This article belongs to the Section Cattle)
Show Figures

Figure 1

16 pages, 2762 KB  
Article
PriorCCI: Interpretable Deep Learning Framework for Identifying Key Ligand–Receptor Interactions Between Specific Cell Types from Single-Cell Transcriptomes
by Hanbyeol Kim, Eunyoung Choi, Yujeong Shim and Joonha Kwon
Int. J. Mol. Sci. 2025, 26(15), 7110; https://doi.org/10.3390/ijms26157110 - 23 Jul 2025
Viewed by 835
Abstract
Understanding the interactions between specific cell types within tissue environments is essential for elucidating key biological processes, such as immune responses, cancer progression, inflammation, and development, in both physiological and pathological studies. The predominant methods for analyzing cell–cell interactions (CCI) rely primarily on [...] Read more.
Understanding the interactions between specific cell types within tissue environments is essential for elucidating key biological processes, such as immune responses, cancer progression, inflammation, and development, in both physiological and pathological studies. The predominant methods for analyzing cell–cell interactions (CCI) rely primarily on statistical inference using mapping or network-based techniques. However, these approaches often struggle to prioritize meaningful interactions owing to the high sparsity and heterogeneity inherent in single-cell RNA sequencing (scRNA-seq) data, where small but biologically important differences can be easily overlooked. To overcome these limitations, we developed PriorCCI, a deep-learning framework that leverages a convolutional neural network (CNN) alongside Grad-CAM++, an explainable artificial intelligence algorithm. This study aims to provide a scalable, interpretable, and biologically meaningful framework for systematically identifying and prioritizing key ligand–receptor interactions between defined cell-type pairs from single-cell RNA-seq data, particularly in complex environments such as tumors. PriorCCI effectively prioritizes interactions between cancer and other cell types within the tumor microenvironment and accurately identifies biologically significant interactions related to angiogenesis. By providing a visual interpretation of gene-pair contributions, our approach enables robust inference of gene–gene interactions across distinct cell types from scRNA-seq data. Full article
(This article belongs to the Special Issue New Insights in Translational Bioinformatics: Second Edition)
Show Figures

Figure 1

Back to TopTop