Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (19,145)

Search Parameters:
Keywords = in vitro modeling

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 2227 KB  
Article
Cinnarizine, a Calcium Channel Blocker, Partially Prevents the Striatal Dopamine Decline and Loss of Nigral Dopamine Neurons in the Lactacystin-Induced Rat Model of Parkinson’s Disease
by Elżbieta Lorenc-Koci, Tomasz Lenda, Jolanta Konieczny, Danuta Jantas and Helena Domin
Int. J. Mol. Sci. 2025, 26(18), 8833; https://doi.org/10.3390/ijms26188833 - 10 Sep 2025
Abstract
Selective proteasome inhibitors, used to model Parkinsonian-like pathology, are known to disrupt calcium homeostasis, but the role of calcium ions in dopaminergic neuron degeneration remains unclear. The present in vivo study examined the effects of a 7-day intraperitoneal administration of cinnarizine (10 or [...] Read more.
Selective proteasome inhibitors, used to model Parkinsonian-like pathology, are known to disrupt calcium homeostasis, but the role of calcium ions in dopaminergic neuron degeneration remains unclear. The present in vivo study examined the effects of a 7-day intraperitoneal administration of cinnarizine (10 or 30 mg/kg), a voltage-gated calcium channel blocker, in rats unilaterally injected into the substantia nigra compacta (SNc) with lactacystin (Lac; 1 µg/2 µL) or vehicle. Dopamine (DA) and its metabolites were quantified in striatal homogenates via high-performance liquid chromatography. The SN of rats treated with 10 mg/kg cinnarizine was used for Western blot analysis of tyrosine hydroxylase (TH), while tissue from animals receiving 30 mg/kg was processed for histological analysis of TH-immunoreactive (TH-ir) and cresyl violet (CV)-stained neurons. Significant reductions in striatal DA and its metabolites were observed one week after Lac injection, along with increased DA catabolism. Cinnarizine at both doses partially prevented DA loss and attenuated enhanced DA turnover. Moreover, 10 mg/kg cinnarizine partially preserved TH protein levels, while 30 mg/kg provided histological protection of TH-ir neurons in the SN. Cinnarizine was also tested in vitro in human SH-SY5Y neuroblastoma cells and primary mouse cortical neurons exposed to Lac or rotenone to further assess its neuroprotective potential. In SH-SY5Y cells, cinnarizine (1–10 µM) significantly increased cell viability and reduced lactate dehydrogenase release after toxin exposure. Cinnarizine failed to counteract lactacystin-induced toxicity in primary cortical neurons but markedly reduced rotenone-evoked cell death at similar concentrations. These findings indicate that cinnarizine exerts dose-dependent neuroprotective effects in vivo and selective protective actions in vitro, supporting the potential utility of voltage-gated calcium channel blockers in treating Parkinson’s disease. Full article
(This article belongs to the Special Issue Neuroprotective Effects of Food Ingredients)
10 pages, 1488 KB  
Article
Electromigration of Aquaporins Controls Water-Driven Electrotaxis
by Pablo Sáez and Sohan Kale
Mathematics 2025, 13(18), 2936; https://doi.org/10.3390/math13182936 - 10 Sep 2025
Abstract
Cell motility is a process central to life and is undoubtedly influenced by mechanical and chemical signals. Even so, other stimuli are also involved in controlling cell migration in vivo and in vitro. Among these, electric fields have been shown to provide a [...] Read more.
Cell motility is a process central to life and is undoubtedly influenced by mechanical and chemical signals. Even so, other stimuli are also involved in controlling cell migration in vivo and in vitro. Among these, electric fields have been shown to provide a powerful and programmable cue to manipulate cell migration. There is now a clear consensus that the electromigration of membrane components represents the first response to an external electric field, which subsequently activates downstream signals responsible for controlling cell migration. Here, we focus on a specific mode of electrotaxis: frictionless, amoeboid-like migration. We used the Finite Element Method to solve an active gel model coupled with a mathematical model of the electromigration of aquaporins and investigate the effect of electric fields on ameboid migration. We demonstrate that an electric field can polarize aquaporins in a cell and, consequently, that the electromigration of aquaporins can be exploited to regulate water flux across the cell membrane. Our findings indicate that controlling these fluxes allows modulation of cell migration velocity, thereby reducing the cell’s migratory capacity. Our work provides a mechanistic framework to further study the impact of electrotaxis and to add new insights into specific modes by which electric fields modify cell motility. Full article
(This article belongs to the Special Issue Advances in Biological Systems with Mathematics)
Show Figures

Figure 1

15 pages, 1513 KB  
Article
Nitrofurantoin–Aminoglycoside Synergy Against Common Uropathogens Evaluated by Disc Diffusion: A Pilot Study
by Filip Bielec, Monika Łysakowska, Małgorzata Brauncajs, Adrian Bekier, Stanisław Klimaszewski and Dorota Pastuszak-Lewandoska
Microorganisms 2025, 13(9), 2117; https://doi.org/10.3390/microorganisms13092117 - 10 Sep 2025
Abstract
The emergence of multidrug-resistant uropathogens requires the development of novel therapeutic strategies. This pilot study assessed the in vitro synergy between nitrofurantoin and aminoglycosides (amikacin, gentamicin, and tobramycin) against three major uropathogens: Escherichia coli, Klebsiella pneumoniae, and Enterococcus faecalis. Ninety [...] Read more.
The emergence of multidrug-resistant uropathogens requires the development of novel therapeutic strategies. This pilot study assessed the in vitro synergy between nitrofurantoin and aminoglycosides (amikacin, gentamicin, and tobramycin) against three major uropathogens: Escherichia coli, Klebsiella pneumoniae, and Enterococcus faecalis. Ninety clinical isolates were tested using the disk diffusion and double-disk synergy methods. Statistical analysis included Kruskal–Wallis and Mann–Whitney U tests, as well as logistic regression models to assess associations between inhibition zone diameters and synergy occurrence. While synergy was observed in all bacterial species, it was neither universally present nor species-specific. Significant associations were identified between nitrofurantoin inhibition zone size and synergy with amikacin and tobramycin in E. coli, and with amikacin in K. pneumoniae. In E. faecalis, synergy was more likely with larger aminoglycoside inhibition zones, particularly tobramycin. These findings underscore the potential of nitrofurantoin–aminoglycoside combinations in treatment of multidrug-resistant urinary tract infections, while emphasizing the need for further studies incorporating quantitative synergy assays and clinical validation. Full article
(This article belongs to the Special Issue Research on Relevant Clinical Infections: 2nd Edition)
Show Figures

Figure 1

19 pages, 6001 KB  
Article
Identification of Variable Lymphocyte Receptors That Target the Human Blood–Brain Barrier
by Moriah E. Katt, Elizabeth A. Waters, Benjamin D. Gastfriend, Brantley R. Herrin, Max D. Cooper and Eric V. Shusta
Pharmaceutics 2025, 17(9), 1179; https://doi.org/10.3390/pharmaceutics17091179 - 10 Sep 2025
Abstract
Background/Objectives: Receptor-mediated transcytosis utilizing the native transporters at the blood–brain barrier (BBB) is a growing strategy for the delivery of therapeutics to the brain. One of the major challenges in identifying appropriate human transcytosis targets is that there is a species-specific transporter [...] Read more.
Background/Objectives: Receptor-mediated transcytosis utilizing the native transporters at the blood–brain barrier (BBB) is a growing strategy for the delivery of therapeutics to the brain. One of the major challenges in identifying appropriate human transcytosis targets is that there is a species-specific transporter expression profile at the BBB, complicating translation of successful preclinical candidates into humans. In an effort to overcome this obstacle and identify proteins capable of binding human-relevant BBB ligands, we generated and screened a BBB-targeting library against human-induced pluripotent stem cell-derived brain microvascular endothelial-like cells (iPSC-derived BMEC-like cells). As targeting molecules, we used lamprey antibodies, known as variable lymphocyte receptors (VLRs), and generated a VLR library by immunizing lamprey with iPSC-derived BMEC-like cells, and inserting the resultant VLR repertoire into the yeast surface display system. Methods: The yeast displayed VLR library was then panned against human iPSC-derived BMEC-like cells and lead VLRs were validated using human in vitro models and mouse and human ex vivo brain tissue sections. Results: Finally, brain uptake for a set of VLRs was validated in mice. Of the 15 lead VLR candidates, 14 bound to human BBB antigens, and 10 bound to the murine BBB. Pharmacodynamic testing using the neuroactive peptide neurotensin indicated that the lead candidate, VLR2G, could cross the mouse BBB after intravenous injection and deliver sufficient neurotensin payload to generate a pharmacological response and lower systemic body temperature. Conclusions: Together, these results demonstrate the application of a novel screening technique capable of identifying a VLR with human relevance that can cross the BBB and deliver a payload. Full article
(This article belongs to the Special Issue Advancements and Innovations in Antibody Drug Conjugates)
14 pages, 6366 KB  
Article
A Simple Three-Dimensional Compartmentalized Co-Culture Model for Basal Forebrain and Hippocampal Neurons
by Xiaoman Luo, Jing Li, Zhiyu Deng, Yali Xu, Xixi Li, Miao Ren and Xiangning Li
Biology 2025, 14(9), 1238; https://doi.org/10.3390/biology14091238 - 10 Sep 2025
Abstract
The basal forebrain (BF)-hippocampus (HPC) circuit is indispensable for learning and memory, and in vitro models are essential for dissecting its age-related decline. Nonetheless, current culture methods endure brief survival or confine cells to two dimensions, leaving the circuit’s progressive degeneration refractory to [...] Read more.
The basal forebrain (BF)-hippocampus (HPC) circuit is indispensable for learning and memory, and in vitro models are essential for dissecting its age-related decline. Nonetheless, current culture methods endure brief survival or confine cells to two dimensions, leaving the circuit’s progressive degeneration refractory to long-term investigation. Here, we developed a simple, three-dimensional (3D) compartmentalized co-culture model that mimics the anatomical organization of BF and HPC neurons. Results demonstrate that basal forebrain cholinergic neurons (BFCNs) co-cultured with primary HPC neurons remain viable for more than two months without exogenous growth factors, significantly promoting BFCNs growth, polarity development, and functional maturation. In this system, BFCNs somata were confined within the hydrogel, whereas cholinergic axons extended toward adjacent hippocampal area, reaching 1681.9 ± 351.8 μm by week 5—significantly longer than in BFCNs monocultures. This model can successfully recapitulate age-dependent progressive neuronal degeneration during long-term culture, validating this long-term co-culture as a platform for studying circuit aging and degeneration. Therefore, this low-cost and highly physiological platform provides a new avenue for in-depth investigations into the mechanisms of neurodegenerative diseases. Full article
Show Figures

Figure 1

17 pages, 9548 KB  
Communication
Evaluating Leucine, Isoleucine, and Valine Ratios in Mixed Cortical Cell Cultures Following Cortical Trauma: An In Vitro Assessment
by Ezek Mathew, Nathan Jones, Katherine Hernandez, Sterling B. Ortega and Rob Dickerman
Int. J. Transl. Med. 2025, 5(3), 42; https://doi.org/10.3390/ijtm5030042 - 10 Sep 2025
Abstract
Background/Objectives: Traumatic brain injury (TBI) remains the most common cause of morbidity and mortality in adolescents and adults. Although numerous animal and human studies have demonstrated the beneficial effects of branched-chain amino acids (BCAA) treatment on various models of brain injury, the optimal [...] Read more.
Background/Objectives: Traumatic brain injury (TBI) remains the most common cause of morbidity and mortality in adolescents and adults. Although numerous animal and human studies have demonstrated the beneficial effects of branched-chain amino acids (BCAA) treatment on various models of brain injury, the optimal concentration and mechanism of action have not been elucidated. Methods: Based on our prior work, we hypothesized that a 2:1:1 ratio of BCAAs promotes neuronal regrowth and repair. Using in vitro mixed cortical cultures (composed of CNS cells, including neuronal and glial cells), we recapitulated the mechanical damage induced by TBI using the scratch assay model. We evaluated various concentrations of BCAA to promote the regrowth of CNS cells after mechanical damage. Results: A 2:1:1 ratio of leucine: isoleucine: valine was observed to yield superior regrowth rates at the 48 h time point across various concentrations when compared to a 1:1:1 ratio and even a 4:1:1 ratio. In addition, both 2:1:1 and 4:1:1 ratios offered multiple instances of accelerated regrowth, where less than 5% of the wound remained unhealed. Conclusions: The importance of leucine ratios in the context of BCAA treatment for TBI was demonstrated by the superior CNS cell regrowth offered by the 2:1:1 ratio. Full article
Show Figures

Figure 1

11 pages, 1679 KB  
Article
Use of 4-Nitroquinoline 1-Oxide (4NQO) in Dysplastic and Malignant Induction: In Vitro and In Vivo Studies
by Daniela Oliveira Meneses, Brunna da Silva Nobrega Souza, Mateus José Dutra, Isabella Souza Malta, Bruna Oliveira Silva, Isis Moraes Cançado, Nathan Stevan Cezar Conceição, Maria Leticia de Almeida Lança, Luana Marotta Reis de Vasconcellos and Estela Kaminagakura
Biomedicines 2025, 13(9), 2223; https://doi.org/10.3390/biomedicines13092223 - 10 Sep 2025
Abstract
Objectives: Tobacco has been associated with the development of oral leukoplakia (OL) and oral squamous cell carcinoma (OSCC). This study aimed to evaluate the in vitro and in vivo changes caused by carcinogen 4-nitroquinoline 1-oxide (4NQO), simulating smoking conditions. Materials and Methods: In [...] Read more.
Objectives: Tobacco has been associated with the development of oral leukoplakia (OL) and oral squamous cell carcinoma (OSCC). This study aimed to evaluate the in vitro and in vivo changes caused by carcinogen 4-nitroquinoline 1-oxide (4NQO), simulating smoking conditions. Materials and Methods: In the in vitro study, normal keratinocytes were exposed to 1.3 µM and 2.6 µM concentrations of 4NQO to induce dysplastic transformation (H-DISP) and malignant transformation (H-SCC), respectively. The cells were collected and subjected to hematoxylin and eosin (H&E) staining and immunocytochemistry with Ki-67. For the in vivo study, female C57BL/6J mice were divided into a pure control (PC) group and experimental groups exposed to 50 µg/mL (NQ) and 100 µg/mL (CM) of 4NQO in autoclaved drinking water. Each group was euthanized after 8, 12, 16, and 20 weeks of exposure. The tongues were collected, processed, stained with H&E, and analyzed using conventional light microscopy. Results: In vitro, significant morphological changes were observed in the H-DISP and H-SCC groups, with a cell proliferation index exceeding 30% in the H-DISP group. In vivo, the CM group showed greater progression to severe dysplasia/carcinoma within a shorter treatment period compared to the NQ group. Conclusions: We established critical doses and exposure durations for 4NQO, both in vitro and in vivo, to induce cellular changes and the formation of OL and OSCC, providing a standardized model for studies related to oral carcinogenesis. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutic Approaches for Oral Disorders)
Show Figures

Figure 1

19 pages, 6504 KB  
Article
CLEC5A Activation in Inflammatory Monocytes: A Mechanism for Enhanced Adaptive Immunity Following COVID-19 mRNA Vaccination in a Preclinical Study
by Renan Galuzo, Thiago Lazari Machado, Ryann de Souza Nascimento, Jorvan Ramos de Medeiros, Luciana Neves Tubarão, Jane Silva, Vanessa Pimenta Rocha, Tamiris Azamor, Felipe Soares Coelho, Andrea Marques Vieira da Silva, Lorenna Carvalho da Rosa, Juliana Fernandes Amorim da Silva, Renata Tourinho Santos, Rodrigo Müller, Carolina Baeta Salvador Várady, Ana Paula Dinis Ano Bom, Patricia Cristina da Costa Neves and Juliana Gil Melgaço
Viruses 2025, 17(9), 1233; https://doi.org/10.3390/v17091233 - 10 Sep 2025
Abstract
Background: CLEC5A is a C-type lectin expressed by monocytes and neutrophils, playing an important role in innate immunity. Although it has been shown to interact with the spike protein of SARS-CoV-2, its role during vaccination remains poorly understood. Methods: To address this question, [...] Read more.
Background: CLEC5A is a C-type lectin expressed by monocytes and neutrophils, playing an important role in innate immunity. Although it has been shown to interact with the spike protein of SARS-CoV-2, its role during vaccination remains poorly understood. Methods: To address this question, we combined in vitro assays to characterize CLEC5A and spike expression and their impact on monocyte differentiation and T-cell activation; in vivo studies to evaluate CLEC5A expression, immune responses, and vaccine efficacy in a murine model; and in silico analyses to identify potential spike epitopes and CLEC5A interaction sites. Results: The Pfizer-BioNTech bivalent mRNA vaccine induced spike expression and CLEC5A upregulation in THP-1 monocytes, promoting M1-like differentiation and CD86+ activation. In PBMC co-cultures, CLEC5A+ monocytes acted as antigen-presenting cells, releasing inflammatory chemokines and activating both CD4+ and CD8+ T cells, thereby linking CLEC5A expression to adaptive immunity. In mice, CLEC5A expression was observed on inflammatory monocytes (CCR2+CX3CR1low) within two days of vaccination. In vivo, CLEC5A expression increased during SARS-CoV-2 infection and after immunization, but declined following viral challenge in vaccinated animals. Consistently, robust humoral and cellular responses were detected post-immunization. In silico analysis further suggested differential CLEC5A binding across B- and T-cell epitopes within the spike glycoprotein. Conclusions: These findings suggest that CLEC5A may play a role in bridging innate and adaptive immune responses during SARS-CoV-2 vaccination. Although further studies with different vaccine platforms are necessary to confirm and expand these observations, our results provide preliminary evidence supporting the potential of CLEC5A as an exploratory biomarker of vaccine-induced immunity. Full article
(This article belongs to the Section Coronaviruses)
Show Figures

Graphical abstract

16 pages, 1473 KB  
Review
Lipid-Mediated Assembly of Biomolecular Condensates: Mechanisms, Regulation, and Therapeutic Implications
by Shijie Ma, Zheng Yang, Chang Du, Binjie Gan and Tong Tang
Biology 2025, 14(9), 1232; https://doi.org/10.3390/biology14091232 - 10 Sep 2025
Abstract
Cellular organization relies on both membrane-bound organelles and membraneless biomolecular condensates formed through liquid–liquid phase separation. Recent discoveries reveal intricate coupling between lipid membrane organization and condensate assembly, reshaping our understanding of cellular compartmentalization. This review synthesizes multidisciplinary research using advanced techniques including [...] Read more.
Cellular organization relies on both membrane-bound organelles and membraneless biomolecular condensates formed through liquid–liquid phase separation. Recent discoveries reveal intricate coupling between lipid membrane organization and condensate assembly, reshaping our understanding of cellular compartmentalization. This review synthesizes multidisciplinary research using advanced techniques including super-resolution microscopy, fluorescence recovery after photobleaching, and in vitro reconstitution to examine lipid-condensate interactions. Lipid membranes serve as nucleation platforms that reduce critical concentrations for condensate formation by orders of magnitude through membrane anchoring and thermodynamic coupling, creating specialized microenvironments that substantially enhance enzymatic activities. Key regulatory mechanisms include phosphorylation-driven assembly and disassembly, membrane composition effects from cholesterol content and fatty acid saturation, and environmental factors such as calcium and pH. These interactions drive signal transduction through receptor clustering, membrane trafficking via organized domains, and stress responses through protective condensate formation. Dysregulation of lipid-condensate coupling, including aberrant phase transitions and membrane dysfunction, underlies metabolic disorders and neurodegenerative diseases. This coupling represents a fundamental organizing principle with significant therapeutic potential. Current challenges include developing quantitative methods for characterizing condensate dynamics in complex cellular environments and translating molecular mechanisms into clinical applications. Future progress requires interdisciplinary approaches combining advanced experimental techniques, computational modeling, and standardized protocols to advance both fundamental understanding and therapeutic innovations. Full article
(This article belongs to the Section Biochemistry and Molecular Biology)
Show Figures

Figure 1

20 pages, 5620 KB  
Article
Multispectral Pulsed Photobiomodulation Enhances Re-Epithelialization via Keratinocyte Activation in Full-Thickness Skin Wounds
by Joo Hyun Kim, Delgerzul Baatar, Myung Jin Ban, Ji Won Son, Jihye Choi, Chan Hee Gil, Min-Kyu Kim, Sung Sik Hur, Jung Eun Kim and Yongsung Hwang
Cells 2025, 14(18), 1415; https://doi.org/10.3390/cells14181415 - 10 Sep 2025
Abstract
Chronic wound healing is a complex and tightly regulated process requiring coordinated epithelial and stromal regeneration. Photobiomodulation (PBM) using low-level red light-emitting diode (LED) therapy has emerged as a non-invasive approach to enhancing skin repair. In this study, we evaluated the therapeutic efficacy [...] Read more.
Chronic wound healing is a complex and tightly regulated process requiring coordinated epithelial and stromal regeneration. Photobiomodulation (PBM) using low-level red light-emitting diode (LED) therapy has emerged as a non-invasive approach to enhancing skin repair. In this study, we evaluated the therapeutic efficacy of a pulsed, multi-wavelength LED system on full-thickness excisional wound healing in a normal murine model. Daily LED treatment significantly accelerated wound closure, promoted re-epithelialization, and improved dermal architecture. Histological and immunohistochemical analyses revealed enhanced epidermal stratification, reduced inflammation, and improved collagen organization. Molecular profiling demonstrated increased expression of proliferation marker Ki67, keratins CK14 and CK17, and extracellular matrix-related genes including MMPs, Col1a1, and Col3a1. In vitro assays using HaCaT keratinocytes showed accelerated scratch wound closure and cytoskeletal remodeling following PBM exposure. These findings suggest that pulsed PBM promotes coordinated epithelial regeneration and matrix remodeling, highlighting its potential as a tunable and effective therapeutic modality for accelerating cutaneous wound healing under physiological conditions. Full article
(This article belongs to the Special Issue Cellular and Molecular Mechanisms of Wound Repair)
Show Figures

Figure 1

21 pages, 7662 KB  
Article
Attenuation of Pulmonary Fibrosis by the MyD88 Inhibitor TJ-M2010-5 Through Autophagy Induction in Mice
by Yang Yang, Zeyang Li, Minghui Zhao, Yuanyuan Zhao, Zhimiao Zou, Yalong Xie, Limin Zhang, Dunfeng Du and Ping Zhou
Biomedicines 2025, 13(9), 2214; https://doi.org/10.3390/biomedicines13092214 - 10 Sep 2025
Abstract
Background and Objectives: Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with few effective treatments. In its pathogenesis, damage-associated molecular patterns are released and recognized by Toll-like receptors (TLRs); all TLRs except TLR3 transduce signals through MyD88. Research has shown that [...] Read more.
Background and Objectives: Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with few effective treatments. In its pathogenesis, damage-associated molecular patterns are released and recognized by Toll-like receptors (TLRs); all TLRs except TLR3 transduce signals through MyD88. Research has shown that autophagy participates in the progression of pulmonary fibrosis, and MyD88 is closely associated with autophagy. However, whether targeting MyD88 can affect fibrosis progression by regulating autophagy during lung fibrosis remains unclear. Materials and Methods: TJ-M2010-5 (TJ-5) is a small molecular derivative of aminothiazole that inhibits MyD88 homodimerization. A bleomycin-induced pulmonary fibrosis model in mice was established, and a human lung fibroblast cell line MRC-5 was cultured, and the mechanism of fibrosis induced by TGF-β1 was studied. TJ-5 and the autophagy inhibitor 3-MA were used to intervene. Results: Our study indicated that TJ-5 suppressed fibrosis foci formation and collagen deposition in fibrotic lungs, effectively increased the survival rate of bleomycin-stimulated mice from 40.0% to 80.0%, and repressed lung fibroblast activation in vitro. Subsequently, TJ-5 could trigger autophagy, as indicated by increased autophagosomes, LC3B-II and Beclin-1 promotion, and p62 degradation. Moreover, inhibition of TJ-5-induced autophagy by 3-MA reversed the anti-fibrosis effect of TJ-5. Furthermore, the autophagy-related pathways PI3K/AKT/mTOR and MAPK/mTOR were inhibited under TJ-5 intervention. Conclusions: Our findings demonstrated that the mechanism of TJ-5 in alleviating lung fibrosis was through triggering MyD88-related autophagy, and TJ-5 may be therapeutically useful for the clinical treatment of IPF. Full article
(This article belongs to the Special Issue Advances in Novel Drug Discovery, Synthesis, and Evaluation)
Show Figures

Graphical abstract

13 pages, 4338 KB  
Article
A Nanoformulation of Ubiquinol and Selenium Promotes Proliferation of Human Induced Pluripotent Stem Cells
by Filomain Nguemo, Hai Zhang, Annette Koester, Susan Rohani, Sureshkumar Perumal Srinivasan and Jürgen Hescheler
Antioxidants 2025, 14(9), 1100; https://doi.org/10.3390/antiox14091100 - 10 Sep 2025
Abstract
Human induced pluripotent stem cells (hiPSCs) hold immense promise for regenerative medicine. However, a critical barrier to the clinical application of hiPSCs is the difficulty in promoting robust cell proliferation while preserving their pluripotent state. Efficient hiPSC expansion without loss of pluripotency is [...] Read more.
Human induced pluripotent stem cells (hiPSCs) hold immense promise for regenerative medicine. However, a critical barrier to the clinical application of hiPSCs is the difficulty in promoting robust cell proliferation while preserving their pluripotent state. Efficient hiPSC expansion without loss of pluripotency is crucial for generating high quality cells or therapeutic applications, disease modeling, and drug discovery. In our study, we investigated the effects of QuinoMit Q10® fluid (QMF-Se), a nanoformulated supplement containing Ubiquinol (the active form of Coenzyme Q10) and Selenium, on hiPSC growth and maintenance in vitro. Interesting, QMF-Se supplementation significantly enhances hiPSC proliferation compared to control cultures. This increase in cell number was accompanied by heightened mitochondrial activity, suggesting improved cellular energy metabolism. Importantly, the expression of core pluripotency markers OCT4, NANOG, and SOX2 remained unaltered, confirming that the stem cells retained their undifferentiated status. Moreover, we observed that QMF-Se treatment conferred protective effects during the freeze–thaw process, reducing cell death and supporting post-thaw recovery. These results indicate that QMF-Se may improve both cell culture efficiency and cryopreservation outcomes. Overall, our findings highlight the potential of QMF-Se as a valuable additive for hiPSC culture systems, contributing to more efficient and reliable expansion protocols in regenerative medicine research. Full article
(This article belongs to the Special Issue Unveiling the Essential Role of Coenzyme Q in Health)
Show Figures

Figure 1

18 pages, 1207 KB  
Review
Biochemical, Biological, and Clinical Properties of γ-Oryzanol
by Helena Juricic, Massimiliano Cuccioloni, Laura Bonfili, Mauro Angeletti, Daniela Uberti, Anna Maria Eleuteri, Giulia Abate and Valentina Cecarini
Antioxidants 2025, 14(9), 1099; https://doi.org/10.3390/antiox14091099 - 9 Sep 2025
Abstract
γ-Oryzanol is a complex mixture of ferulic acid esters of phytosterols and triterpene alcohols predominantly found in rice bran. It exhibits a wide range of biological activities, including antioxidant, anti-inflammatory, and lipid-lowering effects, as well as the ability to modulate cellular metabolic pathways [...] Read more.
γ-Oryzanol is a complex mixture of ferulic acid esters of phytosterols and triterpene alcohols predominantly found in rice bran. It exhibits a wide range of biological activities, including antioxidant, anti-inflammatory, and lipid-lowering effects, as well as the ability to modulate cellular metabolic pathways in both in vitro and in vivo models. The composition and concentration of γ-oryzanol vary significantly among rice varieties and are influenced by genetic, environmental, and technological factors. Advances in extraction methods, including traditional solvent extraction and innovative approaches such as supercritical fluid extraction, have improved yield and purity, supporting its use in functional foods, nutraceuticals, and cosmetics. Current research in the biological, biomedical, and cosmetic fields is actively investigating γ-oryzanol’s mechanisms of action in metabolic regulation and inflammation, as well as developing advanced formulation strategies to enhance its antioxidant, skin-protective, and functional properties. These efforts aim to optimize its delivery and efficacy by addressing challenges related to poor water solubility and bioavailability, thereby expanding its role as a multifunctional bioactive compound. This review provides a comprehensive overview on γ-oryzanol, focusing on its extraction techniques, chemical characterization, and biological/pharmacological activities. Additionally, clinical trials investigating its efficacy and safety have been thoroughly dissected, offering valuable insights into its therapeutic potential in human populations. Full article
Show Figures

Figure 1

17 pages, 3394 KB  
Article
Impact of 45S5-Bioactive Glass on Chondrocytes in Knee Osteoarthritis—In Vitro Study Exploring Cellular Responses
by Max Marinescu, Sébastien Hagmann, Jörg Fellenberg, Elena Tripel, Simone Gantz, Ravikumar Mayakrishnan, Aldo R. Boccaccini, Tobias Renkawitz, Babak Moradi, Fabian Westhauser and Hadrian Platzer
J. Funct. Biomater. 2025, 16(9), 339; https://doi.org/10.3390/jfb16090339 - 9 Sep 2025
Abstract
Osteoarthritis (OA), the most common joint disease, is marked by cartilage degradation and chronic inflammation. While 45S5-bioactive glass (45S5-BG) is well-established in bone regeneration and has been suggested to exert immunomodulatory effects, its impact on OA chondrocytes remains largely unexplored. Therefore, this in [...] Read more.
Osteoarthritis (OA), the most common joint disease, is marked by cartilage degradation and chronic inflammation. While 45S5-bioactive glass (45S5-BG) is well-established in bone regeneration and has been suggested to exert immunomodulatory effects, its impact on OA chondrocytes remains largely unexplored. Therefore, this in vitro study investigated the effects of 45S5-BG microparticles (0.125 mg/mL) on chondrocytes derived from OA patients, evaluating its therapeutic potential in OA. Chondrocytes were cultured with or without 45S5-BG for 1 and 7 days. Gene expression of cartilage markers, cytokines, matrix metalloproteinases (MMPs), and toll-like receptors (TLRs) was analyzed by qPCR. Protein levels were assessed by ELISA. 45S5-BG stimulation significantly altered chondrocyte activity, inducing upregulation of IL-6, IL-1β, TNF-α, MMP-1/-3/-13, and TLR4. Expression of ACAN and COL2A1 was reduced, while COL10A1—a marker of chondrocyte hypertrophy—was significantly increased at day 1. These findings show a catabolic and pro-inflammatory shift in chondrocyte phenotype upon 45S5-BG exposure, showing no therapeutic benefit of 45S5-BG on OA chondrocytes. However, considering the pronounced effects on chondrocyte activity and the well-established bioactivity and biocompatibility of 45S5-BG, our findings suggest that modified BG formulations could be developed to enhance chondroprotective and anti-inflammatory properties, warranting further investigation in co-culture and in vivo models. Full article
(This article belongs to the Special Issue Bioactive Glass in Tissue Engineering Applications)
Show Figures

Figure 1

13 pages, 1097 KB  
Article
Astragalus membranaceus Extract Attenuates Inflammatory Cytokines and Matrix-Degrading Enzymes in Human Chondrocytes: A Novel Nutraceutical Strategy for Joint Health
by Alessia Mariano, Rosario Russo, Anna Scotto d’Abusco and Fabiana Superti
Curr. Issues Mol. Biol. 2025, 47(9), 731; https://doi.org/10.3390/cimb47090731 - 9 Sep 2025
Abstract
The dried root extract of Astragalus membranaceus, also known as Astragali radix, is widely used in traditional Chinese medicine for its multiple health benefits and well-established safety profile. Astragalus root extract exhibits several bioactive properties, including anti-inflammatory, antioxidant, antiviral and hepatoprotective [...] Read more.
The dried root extract of Astragalus membranaceus, also known as Astragali radix, is widely used in traditional Chinese medicine for its multiple health benefits and well-established safety profile. Astragalus root extract exhibits several bioactive properties, including anti-inflammatory, antioxidant, antiviral and hepatoprotective effects. Due to its unique features, it is being investigated in a novel application as a complementary remedy in the management of joint disorders. In this study, we evaluated the effect of Astragalus membranaceus hydroalcoholic root extract (0.01 and 0.1 mg/mL) in vitro on the HTB-94 cell line, a well-known model for studying inflammatory pathways in human chondrocytes. The mRNA modulation levels were measured by quantitative real-time polymerase chain reaction (qRT-PCR), while the protein secretion levels were assessed using an Enzyme-Linked Immunosorbent Assay (ELISA). Results obtained demonstrated that this extract is able to decrease the tumor necrosis factor-α (TNF-α)-induced inflammatory response by downregulating both the mRNA expression and release of the pro-inflammatory mediators Interleukin-6 (IL-6), Interleukin-1β (IL-1β) and Interelukin-8 (IL-8), as well as matrix metalloproteases, including Matrix Metalloprotease-3 (MMP-3), Matrix Metalloprotease-13 (MMP-13) and A disintegrin, and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5). Moreover, the interleukin and matrix metalloprotease production was also assessed in non-TNF-α-stimulated cells, revealing that the extract did not alter the basal levels of these mediators. Finally, our findings highlight the potential benefits of Astragalus membranaceus extract, both in terms of its favorable safety profile and its efficacy mitigating joint inflammatory responses. These results support the potential of this extract as a nutraceutical agent for joint health support. Full article
(This article belongs to the Special Issue Role of Natural Products in Inflammatory Diseases)
Show Figures

Figure 1

Back to TopTop