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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (352)

Search Parameters:
Keywords = endothelial heterogeneity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 935 KB  
Review
Cancer-Associated Fibroblasts Arising from Endothelial-to-Mesenchymal Transition: Induction Factors, Functional Roles, and Transcriptomic Evidence
by Junyeol Han, Eung-Gook Kim, Bo Yeon Kim and Nak-Kyun Soung
Biology 2025, 14(10), 1403; https://doi.org/10.3390/biology14101403 (registering DOI) - 13 Oct 2025
Abstract
Cancer-associated fibroblasts (CAFs) are key components of the tumor microenvironment (TME) that influence cancer progression via extracellular matrix (ECM) remodeling and secretion of growth factors and cytokines. Endothelial-to-mesenchymal transition (EndMT) is emerging as an important axis among the heterogeneous origins of CAFs. This [...] Read more.
Cancer-associated fibroblasts (CAFs) are key components of the tumor microenvironment (TME) that influence cancer progression via extracellular matrix (ECM) remodeling and secretion of growth factors and cytokines. Endothelial-to-mesenchymal transition (EndMT) is emerging as an important axis among the heterogeneous origins of CAFs. This review introduces the diverse methods used to induce EndMT in cancer—mouse tumor models, conditioned-medium treatment, co-culture, targeted gene perturbation, ligand stimulation, exosome exposure, irradiation, viral infection, and three-dimensional (3D) culture systems—and summarizes EndMT cell-type evidence uncovered using transcriptomic and proteomic technologies. Hallmark EndMT features include spindle-like morphology, increased motility, impaired angiogenesis and barrier function, decreased endothelial markers (CD31, VE-cadherin), and increased mesenchymal markers (α-SMA, FN1). Reported mechanisms include signaling via TGF-β, cytoskeletal/mechanical stress, reactive oxygen species, osteopontin, PAI-1, IL-1β, GSK-3β, HSP90α, Tie1, TNF-α, HSBP1, and NOTCH. Cancer-induced EndMT affects tumors and surrounding TME—promoting tumor growth and metastasis, expanding cancer stem cell-like cells, driving macrophage differentiation, and redistributing pericytes—and is closely associated with poor survival and therapy resistance. Finally, we indicate each study’s stance: some frame cancer-induced EndMT as a source of CAFs, whereas others, from an endothelial perspective, emphasize barrier weakening and promotion of metastasis. Full article
(This article belongs to the Special Issue Recent Advances in Tumor Microenvironment Biology)
Show Figures

Figure 1

57 pages, 1382 KB  
Article
Bidirectional Endothelial Feedback Drives Turing-Vascular Patterning and Drug-Resistance Niches: A Hybrid PDE-Agent-Based Study
by Zonghao Liu, Louis Shuo Wang, Jiguang Yu, Jilin Zhang, Erica Martel and Shijia Li
Bioengineering 2025, 12(10), 1097; https://doi.org/10.3390/bioengineering12101097 - 12 Oct 2025
Abstract
We present a hybrid partial differential equation-agent-based model (PDE-ABM). In our framework, tumor cells secrete tumor angiogenic factor (TAF), while endothelial cells chemotactically migrate and branch in response. Reaction–diffusion PDEs for TAF, oxygen, and cytotoxic drug are coupled to discrete stochastic dynamics of [...] Read more.
We present a hybrid partial differential equation-agent-based model (PDE-ABM). In our framework, tumor cells secrete tumor angiogenic factor (TAF), while endothelial cells chemotactically migrate and branch in response. Reaction–diffusion PDEs for TAF, oxygen, and cytotoxic drug are coupled to discrete stochastic dynamics of tumor cells and endothelial tip cells, ensuring multiscale integration. Motivated by observed perfusion heterogeneity in tumors and its pharmacokinetic consequences, we conduct a linear stability analysis for a reduced endothelial–TAF reaction–diffusion subsystem and derive an explicit finite-domain threshold for Turing instability. We demonstrate that bidirectional coupling, where endothelial cells both chemotactically migrate along TAF gradients and secrete TAF, is necessary and sufficient to generate spatially periodic vascular clusters and inter-cluster hypoxic regions. These emergent patterns produce heterogeneous drug penetration and resistant niches. Our results identify TAF clearance, chemotactic sensitivity, and endothelial motility as effective levers to homogenize perfusion. The model is two-dimensional and employs simplified kinetics, and we outline necessary extensions to three dimensions and saturable kinetics required for quantitative calibration. The study links reaction–diffusion mechanisms with clinical principles and suggests actionable strategies to mitigate resistance by targeting endothelial–TAF feedback. Full article
(This article belongs to the Special Issue Applications of Partial Differential Equations in Bioengineering)
19 pages, 5979 KB  
Article
Improving the Biocompatibility of Plant-Derived Scaffolds for Tissue Engineering Using Heat Treatment
by Arvind Ramsamooj, Nicole Gorbenko, Cristian Olivares, Sashane John and Nick Merna
J. Funct. Biomater. 2025, 16(10), 380; https://doi.org/10.3390/jfb16100380 - 10 Oct 2025
Viewed by 318
Abstract
Small-diameter vascular grafts often fail due to thrombosis and compliance mismatch. Decellularized plant scaffolds are a biocompatible, sustainable alternative. Leatherleaf viburnum leaves provide natural architecture and mechanical integrity suitable for tissue-engineered vessels. However, the persistence of immunogenic plant biomolecules and limited degradability remain [...] Read more.
Small-diameter vascular grafts often fail due to thrombosis and compliance mismatch. Decellularized plant scaffolds are a biocompatible, sustainable alternative. Leatherleaf viburnum leaves provide natural architecture and mechanical integrity suitable for tissue-engineered vessels. However, the persistence of immunogenic plant biomolecules and limited degradability remain barriers to clinical use. This study tested whether mild heat treatment improves scaffold biocompatibility without compromising mechanical performance. Decellularized leatherleaf viburnum scaffolds were treated at 30–40 °C in 5% NaOH for 15–60 min and then evaluated via tensile testing, burst pressure analysis, scanning electron microscopy, histology, and in vitro assays with white blood cells and endothelial cells. Scaffold properties were compared to those of untreated controls. Heat treatment did not significantly affect scaffold thickness but decreased fiber area fraction and diameter across all anatomical layers. Scaffolds treated at 30–35 °C for ≤30 min retained >90% of tensile strength and achieved burst pressures ≥820 mmHg, exceeding physiological arterial pressures. Heat treatment reduced surface fractal dimension while increasing entropy and lacunarity, producing a smoother but more heterogeneous microarchitecture. White blood cell viability increased up to 2.5-fold and endothelial cell seeding efficiency improved with treatment duration, with 60 min producing near-confluent monolayers. Mild alkaline heat treatment therefore improved immune compatibility and endothelialization while preserving mechanical integrity, offering a simple, scalable modification to advance plant-derived scaffolds for grafting. Full article
Show Figures

Graphical abstract

20 pages, 632 KB  
Review
Cardioprotective Mechanisms of Beta-Blockers in Myocardial Ischemia and Reperfusion: From Molecular Targets to Clinical Implications
by Athina Nasoufidou, Marios G. Bantidos, Barbara Fyntanidou, Christos Kofos, Panagiotis Stachteas, Alexandra Arvanitaki, Paschalis Karakasis, Marios Sagris, George Kassimis, Nikolaos Fragakis and Efstratios Karagiannidis
Int. J. Mol. Sci. 2025, 26(20), 9843; https://doi.org/10.3390/ijms26209843 - 10 Oct 2025
Viewed by 107
Abstract
Ischemic heart disease remains the leading cause of death despite substantial advances in diagnosis, revascularization therapies, and risk-factor control. Beta-adrenergic receptor blockers (Beta-Blockers, BBs), long used to control heart rate, blood pressure, and reduce arrhythmic risk, may also confer cardioprotection through mechanisms beyond [...] Read more.
Ischemic heart disease remains the leading cause of death despite substantial advances in diagnosis, revascularization therapies, and risk-factor control. Beta-adrenergic receptor blockers (Beta-Blockers, BBs), long used to control heart rate, blood pressure, and reduce arrhythmic risk, may also confer cardioprotection through mechanisms beyond hemodynamic unloading. This review integrates an extensive range of preclinical, translational, and clinical studies to present a comprehensive overview of the cardioprotective effects of BBs in the context of myocardial ischemia and reperfusion injury. Mechanistic domains include modulation of redox homeostasis, attenuation of inflammation and neutrophil activation, preservation of mitochondrial integrity and anti-apoptotic signaling, improvement of endothelial function, and stabilization of calcium handling. Third-generation compounds, carvedilol and nebivolol, demonstrate additional antioxidant and vasodilatory benefits compared with first- and second-generation agents; however, no consistent class-wide effect exists across most pathways. The evidence base remains fragmented, often derived from agent- or context-specific studies in heterogeneous populations, with uncertainty surrounding optimal timing of intervention. By bridging mechanistic understanding with clinical outcomes, this review highlights the importance of standardized assessment of BB effects, the development of personalized treatment approaches, and the pursuit of future research to address ongoing translational gaps. Full article
Show Figures

Figure 1

41 pages, 2919 KB  
Review
Organoids as Next-Generation Models for Tumor Heterogeneity, Personalized Therapy, and Cancer Research: Advancements, Applications, and Future Directions
by Ayush Madan, Ramandeep Saini, Nainci Dhiman, Shu-Hui Juan and Mantosh Kumar Satapathy
Organoids 2025, 4(4), 23; https://doi.org/10.3390/organoids4040023 - 8 Oct 2025
Viewed by 389
Abstract
Organoid technology has emerged as a revolutionary tool in cancer research, offering physiologically accurate, three-dimensional models that preserve the histoarchitecture, genetic stability, and phenotypic complexity of primary tumors. These self-organizing structures, derived from adult stem cells, induced pluripotent stem cells, or patient tumor [...] Read more.
Organoid technology has emerged as a revolutionary tool in cancer research, offering physiologically accurate, three-dimensional models that preserve the histoarchitecture, genetic stability, and phenotypic complexity of primary tumors. These self-organizing structures, derived from adult stem cells, induced pluripotent stem cells, or patient tumor biopsies, recapitulate critical aspects of tumor heterogeneity, clonal evolution, and microenvironmental interactions. Organoids serve as powerful systems for modeling tumor progression, assessing drug sensitivity and resistance, and guiding precision oncology strategies. Recent innovations have extended organoid capabilities beyond static culture systems. Integration with microfluidic organoid-on-chip platforms, high-throughput CRISPR-based functional genomics, and AI-driven phenotypic analytics has enhanced mechanistic insight and translational relevance. Co-culture systems incorporating immune, stromal, and endothelial components now permit dynamic modeling of tumor–host interactions, immunotherapeutic responses, and metastatic behavior. Comparative analyses with conventional platforms, 2D monolayers, spheroids, and patient-derived xenografts emphasize the superior fidelity and clinical potential of organoids. Despite these advances, several challenges remain, such as protocol variability, incomplete recapitulation of systemic physiology, and limitations in scalability, standardization, and regulatory alignment. Addressing these gaps with unified workflows, synthetic matrices, vascularized and innervated co-cultures, and GMP-compliant manufacturing will be crucial for clinical integration. Proactive engagement with regulatory frameworks and ethical guidelines will be pivotal to ensuring safe, responsible, and equitable clinical translation. With the convergence of bioengineering, multi-omics, and computational modeling, organoids are poised to become indispensable tools in next-generation oncology, driving mechanistic discovery, predictive diagnostics, and personalized therapy optimization. Full article
Show Figures

Figure 1

23 pages, 10345 KB  
Article
A Patient-Derived Scaffold-Based 3D Culture Platform for Head and Neck Cancer: Preserving Tumor Heterogeneity for Personalized Drug Testing
by Alinda Anameriç, Emilia Reszczyńska, Tomasz Stankiewicz, Adrian Andrzejczak, Andrzej Stepulak and Matthias Nees
Cells 2025, 14(19), 1543; https://doi.org/10.3390/cells14191543 - 2 Oct 2025
Viewed by 283
Abstract
Head and neck cancer (HNC) is highly heterogeneous and difficult to treat, underscoring the need for rapid, patient-specific models. Standard three-dimensional (3D) cultures often lose stromal partners that influence therapy response. We developed a patient-derived system maintaining tumor cells, cancer-associated fibroblasts (CAFs), and [...] Read more.
Head and neck cancer (HNC) is highly heterogeneous and difficult to treat, underscoring the need for rapid, patient-specific models. Standard three-dimensional (3D) cultures often lose stromal partners that influence therapy response. We developed a patient-derived system maintaining tumor cells, cancer-associated fibroblasts (CAFs), and cells undergoing partial epithelial–mesenchymal transition (pEMT) for drug sensitivity testing. Biopsies from four HNC patients were enzymatically dissociated. CAFs were directly cultured, and their conditioned medium (CAF-CM) was collected. Cryopreserved primary tumor cell suspensions were later revived, screened in five different growth media under 2D conditions, and the most heterogeneous cultures were re-embedded in 3D hydrogels with varied gel mixtures, media, and seeding geometries. Tumoroid morphology was quantified using a perimeter-based complexity index. Viability after treatment with cisplatin or Notch modulators (RIN-1, recombination signal-binding protein for immunoglobulin κ J region (RBPJ) inhibitor; FLI-06, inhibitor) was assessed by live imaging and the water-soluble tetrazolium-8 (WST-8) assay. Endothelial Cell Growth Medium 2 (ECM-2) medium alone produced compact CAF-free spheroids, whereas ECM-2 supplemented with CAF-CM generated invasive aggregates that deposited endogenous matrix. Matrigel with this medium and single-point seeding gave the highest complexity scores. Two of the three patient tumoroids were cisplatin-sensitive, and all showed significant growth inhibition with the FLI-06 Notch inhibitor, while the RBPJ inhibitor RIN-1 induced minimal change. The optimized scaffold retains tumor–stroma crosstalk and provides patient-specific drug response data within days after operation, supporting personalized treatment selection in HNC. Full article
(This article belongs to the Special Issue 3D Cultures and Organ-on-a-Chip in Cell and Tissue Cultures)
Show Figures

Figure 1

20 pages, 1335 KB  
Review
Advances in Epicardial Biology: Insights from Development, Regeneration, and Human Cardiac Organoids
by Shasha Lyu, Alvin Gea Chen Yao, Yu Xia and Jingli Cao
J. Cardiovasc. Dev. Dis. 2025, 12(10), 389; https://doi.org/10.3390/jcdd12100389 - 2 Oct 2025
Viewed by 377
Abstract
The epicardium plays a pivotal role in heart development, regeneration, and disease response through its contributions to multiple cardiac lineages and its dynamic paracrine signaling. Recent advances in lineage tracing, single-cell technologies, and, particularly, human pluripotent stem cell (hPSC)-derived cardiac organoid models have [...] Read more.
The epicardium plays a pivotal role in heart development, regeneration, and disease response through its contributions to multiple cardiac lineages and its dynamic paracrine signaling. Recent advances in lineage tracing, single-cell technologies, and, particularly, human pluripotent stem cell (hPSC)-derived cardiac organoid models have illuminated the cellular heterogeneity, developmental plasticity, and intercellular crosstalk of epicardial cells with other cardiac cell types. These models have revealed conserved and divergent mechanisms of epicardial function across species, offering new insights into epicardial–myocardial–endothelial–immune interactions and the regulation of cardiac repair. This review highlights recent key findings from developmental and regenerative studies, integrating them with emerging data from human cardiac organoids to provide an updated framework for understanding epicardial biology and its therapeutic potential. Full article
(This article belongs to the Section Cardiac Development and Regeneration)
Show Figures

Graphical abstract

32 pages, 34159 KB  
Article
Isolation of Primary Human Saphenous Vein Endothelial Cells, Human Internal Thoracic Artery Endothelial Cells, and Human Adipose Tissue-Derived Microvascular Endothelial Cells from Patients Undergoing Coronary Artery Bypass Graft Surgery
by Daria Shishkova, Yulia Yurieva, Alexey Frolov, Vera Matveeva, Evgenia Torgunakova, Victoria Markova, Anastasia Lazebnaya and Anton Kutikhin
Int. J. Mol. Sci. 2025, 26(18), 9217; https://doi.org/10.3390/ijms26189217 - 21 Sep 2025
Viewed by 452
Abstract
Primary human endothelial cells represent an essential tool to model endothelial dysfunction and to screen interventions for its treatment. Here, we developed a protocol for the synchronous isolation of primary human saphenous vein endothelial cells (HSaVEC), human internal thoracic artery endothelial cells (HITAEC), [...] Read more.
Primary human endothelial cells represent an essential tool to model endothelial dysfunction and to screen interventions for its treatment. Here, we developed a protocol for the synchronous isolation of primary human saphenous vein endothelial cells (HSaVEC), human internal thoracic artery endothelial cells (HITAEC), and human microvascular endothelial cells (HMVEC) from SV and ITA utilized as conduits during coronary artery bypass graft surgery and from subcutaneous adipose tissue excised while providing an access to the heart. Treatment by collagenase type IV and magnetic separation with anti-CD31-antibody-coated beads ensured relatively high efficiency of the isolation (≈60% for HSaVEC, ≈50% for HITAEC, and ≈20% for HMVEC) and high purity (≥99%) of isolated ECs within ≈2 weeks (HSaVEC), ≈2–3 weeks (HITAEC), and ≈3–4 weeks (HMVEC). A colorimetric assay of cell viability and proliferation, as well as real-time bioimpedance monitoring using the xCELLigence instrument, demonstrated high proliferative activity in HSaVEC, HITAEC, and HMVEC, whilst the in vitro tube formation assay indicated their angiogenic potential. The isolation of HSaVEC, HITAEC, and HMVEC from patients undergoing coronary artery bypass graft surgery is a promising option to investigate endothelial heterogeneity, to interrogate endothelial responses to various stresses, and to pinpoint the optimal approaches for restoring endothelial homeostasis, thereby reproducing them within the bedside-to-bench-to-bedside concept. Full article
(This article belongs to the Special Issue Endothelial Cells in Health and Disease)
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 896
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

30 pages, 1944 KB  
Review
Interactions of Hematopoietic and Associated Mesenchymal Stem Cell Populations in the Bone Marrow Microenvironment, In Vivo and In Vitro Model
by Darina Bačenková, Marianna Trebuňová, Erik Dosedla, Jana Čajková and Jozef Živčák
Int. J. Mol. Sci. 2025, 26(18), 9036; https://doi.org/10.3390/ijms26189036 - 17 Sep 2025
Viewed by 725
Abstract
Multipotent hematopoietic stem cells (HSC) reside in specialized niches of the bone marrow (BM). The maintenance of their stemness requires a precisely regulated bone marrow microenvironment (BMM), supported by mesenchymal stem cells (MSCs), stromal reticular cells, and endothelial and nerve cells located within [...] Read more.
Multipotent hematopoietic stem cells (HSC) reside in specialized niches of the bone marrow (BM). The maintenance of their stemness requires a precisely regulated bone marrow microenvironment (BMM), supported by mesenchymal stem cells (MSCs), stromal reticular cells, and endothelial and nerve cells located within the vascular and endosteal niches. The heterogeneity of the niche environment is caused by the diversity of cell populations from HSCs to more mature hematopoietic cell types and MSCs, which collectively influence the complex intercellular interactions involved in hematopoiesis. MSC subclusters in BM are characterized by the phenotypes of CXC-chemokine ligand 12, leptin receptor, neuron-glial antigen 2, and Nestin+ cells. The article presents a detailed characterization of individual stem cell types in the BM, their reciprocal interaction, and the possibility of in vitro simulation of the bone marrow niche as a dynamic structure. Development of a suitable simulation of the BMM is essential for advancing research into both physiological and pathological processes of hematopoiesis. The main goal is to simulate 3D cell culture using biomaterials that mimic the BM niche in the form of hydrogels and scaffolds, in combination with extracellular matrix components. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

23 pages, 866 KB  
Review
Circulating Biomarkers in Failing Fontan Circulation: Current Evidence and Future Directions
by Cecilia Vecoli, Lamia Ait-Alì, Simona Storti and Ilenia Foffa
J. Cardiovasc. Dev. Dis. 2025, 12(9), 358; https://doi.org/10.3390/jcdd12090358 - 16 Sep 2025
Viewed by 443
Abstract
Patients with Fontan circulation are at lifelong risk for a range of complications involving multiple organ systems. As survival into adulthood increases, there is an urgent need to refine strategies for long-term follow-up and the early detection of Fontan-related sequelae. This narrative review [...] Read more.
Patients with Fontan circulation are at lifelong risk for a range of complications involving multiple organ systems. As survival into adulthood increases, there is an urgent need to refine strategies for long-term follow-up and the early detection of Fontan-related sequelae. This narrative review aims to provide a comprehensive summary of the current evidence regarding the use of circulating blood biomarkers as non-invasive tools for assessing and monitoring Fontan physiology. We critically analyzed available studies investigating serum biomarkers related to key pathological mechanisms associated with Fontan failure, encompassing not only cardiac dysfunction but also systemic inflammation, endothelial dysfunction, hepatic and renal impairment, and altered bone metabolism. Several biomarkers have shown promise in reflecting global systemic impairments as well as end-organ involvement in Fontan patients. However, current data are insufficient to support evidence-based clinical recommendations for standardized specific biomarkers, mainly due to the small sample sizes, heterogeneous patient populations, and limited longitudinal data in the available studies. Only a large-scale, prospective, multi-center, and multidisciplinary research will permit us to identify a panel of specific biomarkers of clinical utility in this population. Artificial intelligence (AI) and machine learning (ML) approaches could be applied to integrate all these heterogeneous datasets. Furthermore, “omics”-based studies, including proteomics, metabolomics, lipidomics, and microRNA profiling, hold great potential for uncovering novel biomarkers and pathophysiological pathways, ultimately paving the way for precision medicine in the management of Fontan patients. Full article
(This article belongs to the Section Pediatric Cardiology and Congenital Heart Disease)
Show Figures

Figure 1

16 pages, 932 KB  
Review
Hyperuricemia in Chronic Kidney Disease: Emerging Pathophysiology and a Novel Therapeutic Strategy
by Tomoaki Takata, Yukari Mae, Shotaro Hoi, Takuji Iyama and Hajime Isomoto
Int. J. Mol. Sci. 2025, 26(18), 9000; https://doi.org/10.3390/ijms26189000 - 16 Sep 2025
Viewed by 1249
Abstract
Hyperuricemia has been increasingly recognized as a modifiable contributor to chronic kidney disease (CKD) progression. Although the traditional classification of hyperuricemia distinguished between renal underexcretion and renal overload types, recent studies suggest that hyperuricemia in patients with CKD can result from heterogeneous excretory [...] Read more.
Hyperuricemia has been increasingly recognized as a modifiable contributor to chronic kidney disease (CKD) progression. Although the traditional classification of hyperuricemia distinguished between renal underexcretion and renal overload types, recent studies suggest that hyperuricemia in patients with CKD can result from heterogeneous excretory defects, including glomerular under-filtration and tubular over-reabsorption. These distinct phenotypes may drive divergent renal injury mechanisms. Experimental and clinical data reveal that monosodium urate crystals and soluble uric acid independently induce renal damage through oxidative stress, inflammasome activation, and endothelial dysfunction. Furthermore, clinical investigations showed inconsistent associations between serum uric acid levels and renal outcomes, suggesting that serum levels alone may not fully reflect urate-related renal risk. This has prompted increasing interest in uricosuric agents, particularly the selective urate reabsorption inhibitors (SURIs), which target tubular urate handling. Urate transporter 1 inhibitors have shown promise in enhancing urinary uric acid excretion and potentially preserving kidney function, especially in patients with CKD. In this review, we summarize the current evidence linking the emerging pathophysiological classification of hyperuricemia, mechanisms or urate-induced kidney injury, and therapeutic interventions. These insights may inform individualized approaches to urate-lowering therapy in CKD and support future research into phenotype-guided treatment strategies. Full article
(This article belongs to the Special Issue Molecular Advances in Glomerular Diseases)
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 777
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

19 pages, 543 KB  
Review
The Effect of Diabetes Mellitus on Central Corneal Thickness Values: A Systematic Review and Meta-Analysis
by Arda Uzunoglu, Juan José Valenzuela-Fuenzalida, Karin Morales-Calderón, Isidora Aguilar-Aguirre, Alejandro Bruna-Mejias, Pablo Nova-Baeza, Mathias Orellana-Donoso, Gustavo Oyanedel-Amaro, Alejandra Suazo-Santibañez, Juan A. Sanchis-Gimeno, Jose E. León Rojas and Guinevere Granite
Int. J. Mol. Sci. 2025, 26(17), 8695; https://doi.org/10.3390/ijms26178695 - 6 Sep 2025
Viewed by 1149
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder that can induce systemic and ocular complications. Among the latter, an increase in central corneal thickness (CCT) has been reported, potentially affecting endothelial function and increasing the risk of ocular disease. This study aimed to [...] Read more.
Diabetes mellitus (DM) is a chronic metabolic disorder that can induce systemic and ocular complications. Among the latter, an increase in central corneal thickness (CCT) has been reported, potentially affecting endothelial function and increasing the risk of ocular disease. This study aimed to determine the impact of DM on CCT and to assess its correlation with diabetes duration and glycosylated hemoglobin (HbA1c) levels. A systematic literature search was conducted in Web of Science (1980–2025) following a PICO-based strategy. Observational studies evaluating CCT in diabetic patients were included. Data were analyzed using a random-effects model. Statistical heterogeneity was assessed with χ2 test, p values, and I2 index. Publication bias was evaluated using Begg’s funnel plot and Egger’s regression test. Twenty-nine studies were included in the meta-analysis. Diabetic patients showed significantly higher CCT values compared to controls, particularly in those with long-standing DM (p < 0.001) and poor glycemic control (HbA1c, p < 0.001). Egger’s regression suggested an association between increasing CCT, disease duration, and HbA1c levels, while funnel plot asymmetry indicated potential publication bias. CCT appears to increase in patients with long-term DM and inadequate glycemic control. These findings highlight the relevance of CCT assessment as a potential indicator of corneal changes in diabetic patients. Full article
Show Figures

Figure 1

20 pages, 2451 KB  
Article
Precision Medicine Study of Post-Exertional Malaise Epigenetic Changes in Myalgic Encephalomyelitis/Chronic Fatigue Patients During Exercise
by Sayan Sharma, Lynette D. Hodges, Katie Peppercorn, Jemma Davis, Christina D. Edgar, Euan J. Rodger, Aniruddha Chatterjee and Warren P. Tate
Int. J. Mol. Sci. 2025, 26(17), 8563; https://doi.org/10.3390/ijms26178563 - 3 Sep 2025
Viewed by 2894
Abstract
Post-exertional malaise (PEM) is a defining symptom of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), yet its molecular underpinnings remain elusive. This study investigated the temporal–longitudinal DNA methylation changes associated with PEM using a structured two-day maximum repeated effort cardiopulmonary exercise testing (CPET) protocol involving [...] Read more.
Post-exertional malaise (PEM) is a defining symptom of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), yet its molecular underpinnings remain elusive. This study investigated the temporal–longitudinal DNA methylation changes associated with PEM using a structured two-day maximum repeated effort cardiopulmonary exercise testing (CPET) protocol involving pre- and two post-exercise blood samplings from five ME/CFS patients. Cardiopulmonary measurements revealed complex heterogeneous profiles among the patients compared to typical healthy controls, and VO2 peak indicated all patients had poor normative fitness. The switch to anaerobic metabolism occurred at a lower workload in some patients on Day Two of the test. Reduced Representation Bisulphite Sequencing followed by analysis with Differential Methylation Analysis Package-version 2 (DMAP2) identified differentially methylated fragments (DMFs) present in the DNA genomes of all five ME/CFS patients through the exercise test compared with ‘before exercise’. With further filtering for >10% methylation differences, there were early DMFs (0–24 h after first exercise test) and late DMFs between (24–48 h after the second exercise test), as well as DMFs that changed gradually (between 0 and 48 h). Of these, 98% were ME/CFS-specific, compared with the two healthy controls accompanying the longitudinal study. Principal component analysis illustrated the three distinct clusters at the 0 h, 24 h, and 48 h timepoints, but with heterogeneity among the patients within the clusters, highlighting dynamic methylation responses to exertion in individual patients. There were 24 ME/CFS-specific DMFs at gene promoter fragments that revealed distinct patterns of temporal methylation across the timepoints. Functional enrichment of ME-specific DMFs revealed pathways involved in endothelial function, morphogenesis, inflammation, and immune regulation. These findings uncovered temporally dynamic epigenetic changes in stress/immune functions in ME/CFS during PEM and suggest molecular signatures with potential for diagnosis and of mechanistic significance. Full article
Show Figures

Figure 1

Back to TopTop