Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (8,432)

Search Parameters:
Keywords = extra-cellular matrix

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 1193 KB  
Article
Effect of Short-Term and Long-Term Non-Physiological T3 Concentrations on Cardiac Stromal Cells: From Cellular Response to In Vivo Adaptation
by Ahmad Alhamid, Yoshishige Urata, Kodai Nishi, Hiroshi Kurazumi, Ryo Suzuki, Koji Ueno, Akihito Mikamo, Kimikazu Hamano and Tao-Sheng Li
Med. Sci. 2026, 14(1), 66; https://doi.org/10.3390/medsci14010066 (registering DOI) - 31 Jan 2026
Abstract
Background/Objectives: Epidemiological and clinical studies have linked both hypothyroidism and hyperthyroidism to adverse cardiac outcomes, including heart failure and myocardial fibrosis. Triiodothyronine (T3), a biologically active thyroid hormone, is important for cardiovascular homeostasis. While the effects of physiological and non-physiological T3 levels on [...] Read more.
Background/Objectives: Epidemiological and clinical studies have linked both hypothyroidism and hyperthyroidism to adverse cardiac outcomes, including heart failure and myocardial fibrosis. Triiodothyronine (T3), a biologically active thyroid hormone, is important for cardiovascular homeostasis. While the effects of physiological and non-physiological T3 levels on cardiomyocytes have been extensively investigated, the impact of hypothyroidism and hyperthyroidism on cardiac stromal cells (CSCs), which constitute the majority of the cells in the heart, remains understudied. Given CSCs’ essential role in extracellular matrix (ECM) remodeling and paracrine signaling, understanding their response to altered T3 states is necessary to fully elucidate the thyroid hormone-induced cardiac responses. Methods: Cardiac stromal cells were isolated from human atrial appendages and cultured under hypothyroid (0 nM T3), euthyroid (2.5 nM T3), and hyperthyroid (25 nM T3) conditions for 24 (short term) and 120 h (long term). The cells were harvested after 24 h of treatment using trypsin and automatically counted, and their ECM-related gene and growth factor expression levels were assessed using quantitative RT-PCR. Cardiac glucose uptake in hypothyroid, euthyroid, and hyperthyroid mice was monitored using [18F]-FDG PET/CT at acute (7 days) and chronic (42 days) time points. Results: Both hypo- and hyperthyroidism significantly increased the number of CSCs harvested after 24 h. There were acute alterations in the expression of the ECM-related genes COL1A1, COL3A1, TIMP3 (p < 0.05), and TIMP1 (p < 0.01). Similarly, growth factors such as PDGF-A (p < 0.001), TGF-b, and IGF1 (p < 0.05) were transiently upregulated under non-physiological T3 conditions, especially hypothyroidism. Most of these alterations were attenuated or reversed at the 120 h time point. In vivo PET imaging revealed significant increases in cardiac glucose uptake under acute hypothyroidism (p < 0.05) and decreases under acute hyperthyroidism (p < 0.05). However, these metabolic shifts normalized with chronic exposure, paralleling the transient nature of the gene expression changes observed in vitro. Conclusions: Non-physiological T3 concentrations induce proliferation and changes in ECM-related and growth factor gene expression in CSCs. Most of these changes are acute and return to normal levels after chronic exposure. These transient cellular responses correlate closely with the cardiac metabolic response patterns to acute and chronic hypothyroidism and hyperthyroidism. Full article
18 pages, 862 KB  
Review
Tetranectin and Paraoxonase-1 as Markers of Heart Failure
by Paula Alexandra Vulciu, Nicolae Catalin Valea, Dana Zdremtan, Chioreanu Alexandru, Norberth-Istvan Varga, Imola Donath-Miklos, Maria-Daniela Mot and Maria Puschita
Medicina 2026, 62(2), 284; https://doi.org/10.3390/medicina62020284 (registering DOI) - 31 Jan 2026
Abstract
Background and Objectives: This narrative review evaluates the potential of Tetranectin (TN) and Paraoxonase-1 (PON1) to bridge the gap between biological pathology and clinical risk stratification by mapping the “Fibrosis-Oxidative Axis”. Materials and Methods: A targeted literature search was conducted using [...] Read more.
Background and Objectives: This narrative review evaluates the potential of Tetranectin (TN) and Paraoxonase-1 (PON1) to bridge the gap between biological pathology and clinical risk stratification by mapping the “Fibrosis-Oxidative Axis”. Materials and Methods: A targeted literature search was conducted using Scopus, PubMed, and Google Scholar to identify studies examining the diagnostic and prognostic value of TN and PON1 in heart failure (HF). Evidence was synthesized qualitatively to analyze their roles in structural fibrosis and oxidative defense. Results: Tetranectin functions as a structural indicator, where its dynamics reflect fibroblast activation, extracellular matrix (ECM) deposition, and protein sequestration during tissue remodeling. On the other hand, PON1 serves as a functional metabolic barometer; its reduced activity correlates with systemic oxidative burden, loss of endothelial protection, and pro-inflammatory signaling. These markers capture a bidirectional pathology where oxidative injury drives fibrotic remodeling, which subsequently continue metabolic dysfunction. A dual-biomarker profile is proposed to stratify disease activity: early-stage metabolic stress (reduced PON1) precedes structural changes, while progressive HF involves active fibrosis (altered TN) alongside persistent oxidative injury. Conclusions: The combined assessment of TN and PON1 offers a complementary approach to HF profiling, potentially refining risk stratification beyond hemodynamic parameters. However, clinical implementation requires large-scale validation to address standardization issues and specificity limitations regarding multimorbidity. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

22 pages, 851 KB  
Perspective
Embodied Neuroplasticity: Exploring Biological and Molecular Pathways of Inner Development for Planetary Health
by Karen B. Kirkness
Challenges 2026, 17(1), 6; https://doi.org/10.3390/challe17010006 - 30 Jan 2026
Abstract
Understanding how inner development capacities are embodied at biological levels remains an underexplored dimension of planetary health research. The aim of this viewpoint is to provide transdisciplinary integration across neuroscience, cell biology, education, and social systems toward addressing planetary health challenges. Despite growing [...] Read more.
Understanding how inner development capacities are embodied at biological levels remains an underexplored dimension of planetary health research. The aim of this viewpoint is to provide transdisciplinary integration across neuroscience, cell biology, education, and social systems toward addressing planetary health challenges. Despite growing recognition of the Inner Development Goals (IDG) framework as complementary to the UN Sustainable Development Goals, the biophysical dynamics underlying personal and collective transformation remain largely unexplored. This viewpoint presents key molecular pathways that may underpin the Embodied Neuroplastic Resilience Model (ENRM) via calcium signaling and hyaluronan (the CHA axis). This viewpoint explores educational and therapeutic implications while simultaneously illuminating how socioeconomic inequalities constrain access to neuroplasticity-supporting practices. Four key conclusions emerge: (1) The CHA axis provides a compelling mechanistic framework for understanding how bodily experiences can reshape neural circuits through calcium signaling and hyaluronic acid matrix dynamics; (2) Mapping molecular mechanisms to complex human inner development capacities remains provisional, requiring further interdisciplinary investigation; (3) Socioeconomic inequality creates structural barriers to neuroplasticity and inner development, necessitating an integrated approach that connects mechanistic understanding with equitable access to transformative practices; (4) Enhanced understanding of embodied neuroplasticity must serve compassion and systemic transformation, moving beyond individual optimization toward collective well-being. By bridging neuroscience and sustainability frameworks, this viewpoint calls for a nuanced understanding of inner development that transcends individual optimization and emphasizes collective transformation. Full article
20 pages, 22998 KB  
Article
N-(2-Hydroxyphenyl)-2-Propylpentanamide Modulates HDAC1 and GPER1 Expression in a Rodent Model of Triple-Negative Breast Cancer
by Cynthia Ramírez-Farías, Javier Ventura-Juárez, Argelia Calvillo-Robledo, Manuel Enrique Ávila-Blanco, Daniel González-Blas, José Correa-Basurto and Andrés Quintanar Stephano
Biomedicines 2026, 14(2), 322; https://doi.org/10.3390/biomedicines14020322 - 30 Jan 2026
Abstract
Background: Triple-negative breast cancer (TNBC) is one of the most aggressive breast cancer subtypes due to its rapid growth, poor prognosis, and low response to chemotherapies owing to a lack of therapeutic targets and drug resistance. Histone deacetylases (HDACs) induce stromal changes that [...] Read more.
Background: Triple-negative breast cancer (TNBC) is one of the most aggressive breast cancer subtypes due to its rapid growth, poor prognosis, and low response to chemotherapies owing to a lack of therapeutic targets and drug resistance. Histone deacetylases (HDACs) induce stromal changes that increase extracellular matrix density through the activity of cancer-associated fibroblasts (CAFs). HDACs are overexpressed in TNBC and have been linked to the activation and sustained activity of CAFs. Additionally, HDAC inhibitors decrease the fibroblastic activity. Objectives: We aimed to analyze the antifibrotic effect of the N-(2′-hydroxyphenyl)-2-propylpentanamide (HO-AAVPA), an inhibitor of the HDAC1, 6, and 8 (iHDAC) on TNBC. Methods: The TNBC (4T1) cell line was inoculated under the dorsal skin in mice to develop a TNBC tumor. CAF’s activation was determined by measuring collagen-1 and alpha-smooth muscle actin (α-SMA), as well as their association with the G-protein-coupled estrogenic receptor (GPER1) and HDAC1 expression. Results: Dose of 20 mg/kg of HO-AAVPA decreased tumor fibrosis by inducing decreased collagen-1 and alpha-smooth muscle actin (α-SMA) levels and increased GPER1 expression. Moreover, HO-AAVPA reduced the activation and activity of CAFs. Conclusion: Our results support the notion that HDAC1 inhibition may be a novel approach to sensitizing resistant tumor cells to chemotherapy and radiotherapy by increasing GPER1 expression, and thus the use of antiproliferative GPER1 agonists/antagonists, at least in the early stages, without causing significant changes in liver function or morphological alterations. Full article
(This article belongs to the Special Issue Advances in Novel Drug Discovery, Synthesis, and Evaluation)
Show Figures

Graphical abstract

16 pages, 449 KB  
Review
Applications of Food-Associated Lactobacillaceae in Fermented Foods, Health, and Emerging Biotechnologies
by Shazia Pathan, Veronika Karlegan and David Q. Shih
Fermentation 2026, 12(2), 75; https://doi.org/10.3390/fermentation12020075 - 30 Jan 2026
Abstract
The family Lactobacillaceae, reclassified in 2020 into 25 genera comprising 261 species, remains one of the most extensively studied groups of lactic acid bacteria (LAB) due to its wide distribution in fermented products, commensal presence in the gastrointestinal tract, and studied health [...] Read more.
The family Lactobacillaceae, reclassified in 2020 into 25 genera comprising 261 species, remains one of the most extensively studied groups of lactic acid bacteria (LAB) due to its wide distribution in fermented products, commensal presence in the gastrointestinal tract, and studied health effects. Long classified as “generally recognized as safe (GRAS)” by the U.S. Food and Drug Administration (FDA), these organisms not only contribute to the flavor, texture, and preservation of fermented foods and beverages but also provide important health benefits as probiotics. Their metabolic versatility allows them to produce lactic acid, bacteriocins, and other bioactive compounds that inhibit pathogenic microorganisms and enhance food quality. This review provides a comprehensive overview of the functional roles of members of the Lactobacillaceae family in the context of the food matrix in fermentation, health, and biotechnology, and examines recent advances in functional genomics, metabolomics, and extracellular vesicle research to highlight future directions for leveraging these microorganisms in sustainable and innovative applications. Full article
Show Figures

Figure 1

28 pages, 2087 KB  
Review
Harnessing Microbiome-Mediated and Macrophage-Driven Mechanisms for Oral Wound Healing
by Keerthi Priya Chinniampalayam Sekar, Bianca Schmiliver, Paige Elizabeth Pieterick, Tim Cha, Helly A. Patel, Hope Robinson, Prashant Kumar, David T. Wu, Rheinallt Jones and Steven Goudy
Microorganisms 2026, 14(2), 330; https://doi.org/10.3390/microorganisms14020330 - 30 Jan 2026
Abstract
Oral mucosa healing is a complex process that involves the innate wound healing system, including the coagulation cascade, extracellular matrix remodeling, immune cell responses, and fibroblast and epithelial responses, within the context of a dynamic resident microbiome. Unlike cutaneous wounds, oral wounds heal [...] Read more.
Oral mucosa healing is a complex process that involves the innate wound healing system, including the coagulation cascade, extracellular matrix remodeling, immune cell responses, and fibroblast and epithelial responses, within the context of a dynamic resident microbiome. Unlike cutaneous wounds, oral wounds heal rapidly with minimal scarring despite constant exposure to diverse microbial communities, saliva, and mechanical stress. Emerging evidence highlights the critical interplay between microbiome-mediated signaling and macrophage plasticity in shaping wound outcomes, suggesting that similar mechanisms operate within the oral cavity. Inflammation is an essential component of wound repair, and its resolution is necessary to promote tissue remodeling and functional regeneration. Macrophages play a central role in this transition through phenotype switching from a pro-inflammatory (M1) to a pro-resolving, anti-inflammatory (M2) state. This review synthesizes current understanding of the oral microbiome’s influence on macrophage polarization across distinct stages of oral wound healing and examines microbial-based strategies that modulate the immune response to enhance repair. Significant knowledge gaps remain, including limited clinical translation, inter-individual variability in microbiome composition, and complete mechanistic insight into host–microbe immune interaction. Addressing these challenges enables the development of precision microbiome-based therapeutics that restore microbial balance, direct macrophage-driven regeneration, and improve outcomes in oral wounds and chronic inflammatory conditions. Full article
(This article belongs to the Section Microbiomes)
Show Figures

Graphical abstract

19 pages, 10797 KB  
Article
Integrative Multi-Omics and Machine Learning Identify ID1 as a Candidate Gene Associated with Abdominal Aortic Aneurysm
by Feng Guo, Michael Keese, Yu Zhao and Qining Fu
Curr. Issues Mol. Biol. 2026, 48(2), 156; https://doi.org/10.3390/cimb48020156 - 30 Jan 2026
Abstract
Abdominal aortic aneurysm (AAA) is a fatal vascular disorder driven by immune dysregulation and extracellular matrix (ECM) degradation, yet its molecular mechanisms remain unclear. This study investigated the mechanistic role of ID1 in AAA using an integrative multi-omics and machine learning approach. Two [...] Read more.
Abdominal aortic aneurysm (AAA) is a fatal vascular disorder driven by immune dysregulation and extracellular matrix (ECM) degradation, yet its molecular mechanisms remain unclear. This study investigated the mechanistic role of ID1 in AAA using an integrative multi-omics and machine learning approach. Two bulk transcriptomic datasets (GSE232911 and GSE183464) were analyzed through differential expression, WGCNA, and three machine learning algorithms (LASSO, Random Forest, and SVM-RFE), followed by immune infiltration analysis via ssGSEA and CIBERSORT. ID1 and CYP4B1 were identified by all three machine learning algorithms, but only ID1 showed stable downregulation and consistent discriminatory ability across independent datasets. (AUC = 0.939 and 0.868). Functional enrichment and immune deconvolution linked low ID1 expression to enhanced adaptive immune signaling, increased M1 macrophages, γδ T cells, and memory B cells, and reduced neutrophil and mast cell activity. Single-cell RNA sequencing (GSE226492) confirmed endothelial- and fibroblast-specific ID1 downregulation in AAA. These findings identify ID1 as a candidate gene associated with vascular immune remodeling and extracellular matrix–related pathways, providing a basis for future mechanistic investigation in AAA. Full article
(This article belongs to the Special Issue Featured Papers in Bioinformatics and Systems Biology)
Show Figures

Figure 1

18 pages, 3942 KB  
Article
Preparation of Uniform PEG-PLLA Microspheres via Membrane Emulsification for Soft Tissue Filling Applications
by Siqi Zhang, Yuan Gao, Danyang Wang, Yongjie Chi, Fang Wu, Lianyan Wang and Hailan Jin
J. Funct. Biomater. 2026, 17(2), 71; https://doi.org/10.3390/jfb17020071 - 30 Jan 2026
Abstract
Skin aging could lead to dermal collagen loss and elastic fiber degradation, ultimately manifesting as skin laxity. We aimed to counteract this by using poly-L-lactic acid (PLLA) microsphere (MS)-based fillers to facilitate long-term volume restoration through collagen regeneration. However, conventional MSs exhibit limitations [...] Read more.
Skin aging could lead to dermal collagen loss and elastic fiber degradation, ultimately manifesting as skin laxity. We aimed to counteract this by using poly-L-lactic acid (PLLA) microsphere (MS)-based fillers to facilitate long-term volume restoration through collagen regeneration. However, conventional MSs exhibit limitations such as broad size distribution and surface irregularities, which are frequently associated with significant adverse reactions. This study employed shirasu porous glass (SPG) membrane emulsification to fabricate uniform and well-shaped polyethylene glycol-block-poly (L-lactic acid) (PEG-PLLA) MSs. A single-factor experiment was employed to optimize the parameters. The optimal preparation conditions for PEG-PLLA MSs were as follows: PEG-PLLA concentration of 40 mg/mL, polyvinyl alcohol (PVA) concentration of 0.5%, and magnetic stirring speed of 200 rpm. Under the optimal conditions, the average particle size of PEG-PLLA MSs was 58.982 μm, and the span value (SPAN) was 1.367. In addition, a cytotoxicity assay was performed, and the results revealed no significant toxicity of the MSs toward L929 mouse fibroblasts at concentrations below 500 μg/mL. Furthermore, PEG-PLLA MSs significantly enhanced the production of key extracellular matrix (ECM) components—type I collagen (Col-I), type III collagen (Col-III), and hyaluronic acid (HA)—while simultaneously alleviating cellular oxidative stress responses. This work offers a reliable and reproducible fabrication strategy for developing biocompatible MS fillers with controllable particle sizes. Full article
Show Figures

Figure 1

2 pages, 148 KB  
Editorial
Editorial for the Special Issue “Targeting Tumor Microenvironment for Cancer Therapy, 3rd Edition”
by Cheng-Che Lee and Ming-Wei Lin
Curr. Issues Mol. Biol. 2026, 48(2), 155; https://doi.org/10.3390/cimb48020155 - 30 Jan 2026
Abstract
The tumor microenvironment (TME) is a highly dynamic and heterogeneous ecosystem composed of cancer cells, stromal components, immune cells, and extracellular matrix [...] Full article
(This article belongs to the Special Issue Targeting Tumor Microenvironment for Cancer Therapy, 3rd Edition)
12 pages, 3011 KB  
Article
Heparan Sulfates Regulate Vascular Reactivity in Syndecan 1 Knockout Mice
by Simone R. Potje, Aishwarya Katiki, Paulo W. Pires and Andreia Zago Chignalia
Int. J. Mol. Sci. 2026, 27(3), 1386; https://doi.org/10.3390/ijms27031386 - 30 Jan 2026
Abstract
Heparan sulfates (HS) are polysaccharides abundantly expressed in the extracellular matrix and the glycocalyx of endothelial cells, having a putative role in vascular function. The role of HS in vascular reactivity remains unclear. Herein, we sought to determine whether HS regulate the vascular [...] Read more.
Heparan sulfates (HS) are polysaccharides abundantly expressed in the extracellular matrix and the glycocalyx of endothelial cells, having a putative role in vascular function. The role of HS in vascular reactivity remains unclear. Herein, we sought to determine whether HS regulate the vascular tone in physiological conditions. Using male, 6–8-weeks-old, CD1, C57BL/6, syndecan 1 (Sdc1−/−) and glypican 1 (Gpc1−/−) knockout mice, we investigated if the degradation of HS with heparinase III altered vascular reactivity to norepinephrine (NE), acetylcholine (ACh) and potassium chloride (KCl). Our findings indicate that HS are crucial players in the vascular response to NE and ACh in CD1, C57BL/6, and Sdc1−/− but not in Gpc1−/− mice. Both Sdc1−/− and Gpc1−/− showed increased compensatory expression of syndecan 2 and syndecan 4. However, while Sdc1−/− showed decreased expression of glypican 1, Gpc1−/− showed increased expression of syndecan 1 in aortic homogenates. The lack of response to the vascular reactivity effects of heparinase III in Gpc1−/− suggests a differential role of HS to proteoglycan function in the regulation of the vascular tone. Our data demonstrate a physiological role for HS in the regulation of the vascular tone in physiological conditions. Full article
(This article belongs to the Special Issue Glycobiology in Human Health and Disease)
Show Figures

Graphical abstract

23 pages, 929 KB  
Systematic Review
Scaffolds and Stem Cells Show Promise for TMJ Regeneration: A Systematic Review
by Miljana Nedeljkovic, Gvozden Rosic, Dragica Selakovic, Jovana Milanovic, Aleksandra Arnaut, Milica Vasiljevic, Nemanja Jovicic, Lidija Veljkovic, Pavle Milanovic and Momir Stevanovic
Bioengineering 2026, 13(2), 169; https://doi.org/10.3390/bioengineering13020169 - 29 Jan 2026
Viewed by 50
Abstract
Temporomandibular joint (TMJ) disorders represent chronic degenerative musculoskeletal conditions with a high prevalence in the general population and limited regenerative treatment options. Owing to the insufficient efficacy of current conservative and surgical therapies, there is a growing clinical need for biologically based regenerative [...] Read more.
Temporomandibular joint (TMJ) disorders represent chronic degenerative musculoskeletal conditions with a high prevalence in the general population and limited regenerative treatment options. Owing to the insufficient efficacy of current conservative and surgical therapies, there is a growing clinical need for biologically based regenerative approaches. Tissue engineering (TE), particularly scaffold-based strategies, has emerged as a promising avenue for TMJ regeneration. This systematic review analyzed preclinical in vivo studies investigating scaffold-based interventions for TMJ disc and osteochondral repair. A structured literature search of PubMed and Scopus databases identified 39 eligible studies. Extracted data included scaffold composition, use of cellular and bioactive components, animal models, and reported histological, radiological, and functional outcomes. Natural scaffolds, such as decellularized extracellular matrix and collagen-based hydrogels, demonstrated favorable biocompatibility and support for fibrocartilaginous regeneration, whereas synthetic materials including polycaprolactone, poly (lactic-co-glycolic acid), and polyvinyl alcohol provided superior mechanical stability and structural tunability. Cells were used in 17/39 studies (43%); quantitative improvements were variably reported across these studies. Bioactive molecule delivery, including transforming growth factor-β, histatin-1, and platelet-rich plasma, further enhanced tissue regeneration, while emerging drug- and gene-delivery approaches showed potential for modulating local inflammation. Despite encouraging results, the reviewed studies exhibited substantial heterogeneity in experimental design, outcome measures, and animal models, limiting direct comparison and translational interpretation. Scaffold-based approaches show preclinical promise but heterogeneity in design and incomplete quantitative reporting limit definitive conclusions. Future research should emphasize standardized methodologies, long-term functional evaluation, and the use of clinically relevant large-animal models to facilitate translation toward clinical application. However, functional and biomechanical outcomes were inconsistently reported and rarely standardized, preventing robust conclusions regarding the relationship between structural regeneration and restoration of TMJ function. Full article
(This article belongs to the Section Biomedical Engineering and Biomaterials)
Show Figures

Figure 1

14 pages, 40755 KB  
Article
Triticum vulgare Extract Treatment in UVB-Exposed Human Dermal Fibroblasts Modulates Inflammation, Fibrosis and Oxidative Stress Markers
by Concetta Sozio, Stefano Caccavale, Eugenia Veronica Di Brizzi, Margherita Auriemma, Maddalena Nicoletti, Giuseppe Argenziano, Ciro Menale and Anna Balato
Pharmaceuticals 2026, 19(2), 232; https://doi.org/10.3390/ph19020232 - 29 Jan 2026
Viewed by 26
Abstract
Background/Objectives: UVB radiation triggers oxidative stress, inflammation and extracellular matrix (ECM) remodeling in dermal fibroblasts, contributing to skin aging and fibrosis. Plant-derived extracts with antioxidant and anti-inflammatory activity may counteract these effects. This study evaluated the protective role of Damor Triticum vulgare [...] Read more.
Background/Objectives: UVB radiation triggers oxidative stress, inflammation and extracellular matrix (ECM) remodeling in dermal fibroblasts, contributing to skin aging and fibrosis. Plant-derived extracts with antioxidant and anti-inflammatory activity may counteract these effects. This study evaluated the protective role of Damor Triticum vulgare Aqueous Extract (DTVE) in human dermal fibroblasts (HDFs) exposed to UVB. Methods: Primary HDFs were irradiated with UVB (1.50 J/m2) and treated with DTVE either after irradiation (post-ir) or before and after irradiation (pre-ir). Cell viability was assessed by Trypan Blue and MTT assays. Inflammatory cytokines, fibrosis-related genes, p21 expression, mitochondrial ROS (MitoSOX) and αSMA accumulation were quantified by qRT-PCR, ELISA and immunofluorescence. Results: DTVE was not cytotoxic and preserved HDF viability under UVB exposure. UVB significantly increased pro-inflammatory cytokines, profibrotic markers, αSMA, mitochondrial ROS and p21. DTVE reduced all these UVB-induced alterations, with the pre-ir regimen providing the strongest protection. The extract attenuated early inflammatory activation, limited fibroblast-to-myofibroblast transition and decreased mitochondrial oxidative stress while reducing p21 upregulation. Conclusions: DTVE exerts protective antioxidant, anti-inflammatory and antifibrotic effects in UVB-exposed fibroblasts, particularly when used as pretreatment. These findings support DTVE as a promising candidate to mitigate UVB-induced dermal damage and warrant further investigation for potential therapeutic and cosmetic applications. Full article
Show Figures

Figure 1

21 pages, 2353 KB  
Review
Mechano-Organ-on-Chip for Cancer Research
by Luyang Wang, James Chung Wai Cheung, Xia Zhao, Bee Luan Khoo and Siu Hong Dexter Wong
Int. J. Mol. Sci. 2026, 27(3), 1330; https://doi.org/10.3390/ijms27031330 - 29 Jan 2026
Viewed by 57
Abstract
Mechano-Organ-on-Chip (Mechano-OoC) platforms are emerging as powerful microphysiological systems that place mechanical cues at the center of tumor modeling, providing a scalable and human-relevant approach to recapitulate the biophysical complexity of the tumor microenvironment. Mechanical factors such as matrix stiffness, viscoelasticity, solid stress, [...] Read more.
Mechano-Organ-on-Chip (Mechano-OoC) platforms are emerging as powerful microphysiological systems that place mechanical cues at the center of tumor modeling, providing a scalable and human-relevant approach to recapitulate the biophysical complexity of the tumor microenvironment. Mechanical factors such as matrix stiffness, viscoelasticity, solid stress, interstitial flow, confinement, and shear critically regulate cancer progression, metastasis, immune interactions, and treatment response, yet remain poorly captured by conventional in vitro models and are often studied separately in tumor-on-chip and mechanobiology research. In this review, we summarize recent advances in mechano-OoC technologies for cancer research, highlighting strategies that integrate engineered mechanical cues with microfluidics, tunable extracellular matrices, vascular and stromal interfaces, and dynamic loading to model tumor invasion, vascular transport, immune trafficking, and drug delivery. We also discuss emerging approaches for real-time, multimodal readouts, including sensor-integrated platforms and artificial intelligence-assisted data analysis, and outline key challenges that limit translation, such as device complexity, limited throughput, insufficient standardization, and inadequate validation against in vivo and clinical data. By organizing progress across platform engineering, sensing and readout, data standardization, and AI-driven analytics, this review provides a unified framework for advancing mechanobiology-aware tumor models and guiding the development of predictive preclinical platforms for precision cancer therapy. Full article
(This article belongs to the Special Issue Organoids and Organs-on-Chip for Medical Research)
Show Figures

Figure 1

18 pages, 1963 KB  
Article
Decellularized Extracellular Matrix/Gellan Gum Hydrogels Enriched with Spermine for Cardiac Models
by Luca Di Nunno, Marcin Wekwejt, Francesco Copes, Francesca Boccafoschi and Diego Mantovani
Gels 2026, 12(2), 118; https://doi.org/10.3390/gels12020118 - 28 Jan 2026
Viewed by 74
Abstract
The physiological relevance of in vitro models is limited because conventional two-dimensional cell culture systems are unable to replicate the structural and functional complexity of native tissues. Extracellular matrix (ECM)-mimetic hydrogels have become important platforms for tissue engineering applications. This work developed hybrid [...] Read more.
The physiological relevance of in vitro models is limited because conventional two-dimensional cell culture systems are unable to replicate the structural and functional complexity of native tissues. Extracellular matrix (ECM)-mimetic hydrogels have become important platforms for tissue engineering applications. This work developed hybrid hydrogels that mimic important biochemical and mechanical characteristics of cardiac tissue by combining decellularized bovine pericardium-derived (dBP) ECM, gellan gum (GG), and spermine (SPM). Although dBP offers tissue-specific biological cues, processing compromises its mechanical integrity. This limitation was overcome by adding GG, whose ionic gelation properties were optimized using DMEM and SPM. The hydrogels’ mechanical, biological, physicochemical, and structural characteristics were all evaluated. Under physiologically simulated conditions, the formulations showed quick gelation and long-term stability; scanning electron microscopy revealed an interconnected, ECM-like porous microarchitecture. While uniaxial compression testing provided Young’s modulus values comparable to native myocardium, rheological analysis revealed a concentration-dependent increase in storage modulus with increasing SPM content. H9C2 cardiomyoblasts were used in cytocompatibility studies to confirm that cell viability, morphology, and cytoskeletal organization were all preserved. All of these findings support the potential application of dBP−GG−SPM hydrogels in advanced in vitro cardiac models by showing that they successfully replicate important characteristics of cardiac ECM. Full article
(This article belongs to the Special Issue Recent Advances in Novel Hydrogels and Aerogels)
19 pages, 8567 KB  
Article
Temporal and Spatial Gene Expression Dynamics in Neonatal HI Hippocampus with Focus on Arginase
by Michael A. Smith, Eesha Natarajan, Carlos Lizama-Valenzuela, Thomas Arnold, David Stroud, Amara Larpthaveesarp, Cristina Alvira, Jeffrey R. Fineman, Donna M. Ferriero, Emin Maltepe, Fernando Gonzalez and Jana K. Mike
Cells 2026, 15(3), 253; https://doi.org/10.3390/cells15030253 - 28 Jan 2026
Viewed by 98
Abstract
Background: Hypoxic–ischemic (HI) brain injury triggers a dynamic, multi-phase response involving early microglial efferocytosis followed by extracellular matrix (ECM) deposition and scar formation. Arginase-1 (ARG1), a key enzyme in tissue repair, is implicated in both processes, yet its role in neonatal microglia remains [...] Read more.
Background: Hypoxic–ischemic (HI) brain injury triggers a dynamic, multi-phase response involving early microglial efferocytosis followed by extracellular matrix (ECM) deposition and scar formation. Arginase-1 (ARG1), a key enzyme in tissue repair, is implicated in both processes, yet its role in neonatal microglia remains poorly defined. We characterize ARG1-linked pathways in neonatal microglia, identifying distinct efferocytic and fibrotic phases post-HI. Methods: HI was induced in P9 mice using the Vannucci model, and brains were collected at 24 h (D1) and 5 days (D5). Spatially resolved single-cell transcriptomics (seqFISH) was performed using a targeted panel enriched for microglial, ARG1-pathway, efferocytosis, and profibrotic genes. Cell segmentation, clustering, and spatial mapping were conducted using Navigator and Seurat. Differential expression, GSEA, and enrichment analyses were used to identify time- and injury-dependent pathways. Results: Spatial transcriptomics identified 12 transcriptionally distinct cell populations with preserved neuroanatomical organization. HI caused the expansion of microglia and astrocytes and the loss of glutamatergic neurons by D5. Microglia rapidly activated regenerative and profibrotic programs—including TGF-β, PI3K–Akt, cytoskeletal remodeling, and migration—driven by early DEGs such as Cd44, Reln, TGF-β1, and Col1a2. By D5, microglia adopted a collagen-rich fibrotic state with an upregulation of Bgn, Col11a1, Anxa5, and Npy. Conclusion: Neonatal microglia transition from early efferocytic responses to later fibrotic remodeling after HI, driven by the persistent activation of PI3K–Akt, TGF-β, and Wnt/FZD4 pathways. These findings identify microglia as central regulators of neonatal scar formation and highlight therapeutic targets within ARG1-linked signaling. Full article
(This article belongs to the Section Cellular Neuroscience)
Show Figures

Figure 1

Back to TopTop