{"title":"Serum-free endothelial cell culture medium for vascular smooth muscle cells sheet formation.","authors":"Jing Yang, Xuheng Sun, Hongjing Jiang, Jiandong Li, Jierong Liang, Zhanyi Lin","doi":"10.1186/s13036-025-00522-y","DOIUrl":"https://doi.org/10.1186/s13036-025-00522-y","url":null,"abstract":"<p><strong>Background: </strong>Cell sheet technology has been identified as a promising approach for the construction of tissue-engineered vascular grafts (TEVGs). However, concerns regarding immunogenicity and ethical issues, which are raised by the use of fetal bovine serum (FBS) in traditional culture systems, limit its potential for clinical translation. Serum-free medium (SFM) has emerged as a safer and more controllable alternative, but further validation is required to determine its effectiveness and superiority in generating high-quality cell sheets.</p><p><strong>Methods: </strong>This study systematically compared cell sheets generated under SFM and 10% FBS culture conditions in terms of structure, cellular phenotype, and functional properties. The expression levels of α-SMA and SM22, markers of vascular smooth muscle cells(VSMCs), were evaluated using immunofluorescence staining, qRT-PCR, and Western blot analysis to assess cellular phenotype. Histological staining and mechanical testing were employed to compare the morphology and mechanical properties of the cell sheets, while extracellular matrix (ECM) deposition and biochemical characteristics were also analyzed.</p><p><strong>Results: </strong>Under SFM conditions, cells exhibited significantly higher α-SMA and SM22 expression levels (qRT-PCR showed a 1.8-fold and 2-fold increase, respectively; ****p < 0.0001) with clearer cytoskeletal arrangement. Cell sheets formed in SFM displayed comparable area(ns, p > 0.05), thickness(**p < 0.01), and mechanical properties to those cultured in 10% FBS, while ECM deposition was significantly enhanced (collagen content increased by approximately 40%, **p < 0.01). Furthermore, histological analysis revealed that cell sheets generated under SFM conditions were more compact and uniform, exhibiting superior structural organization.</p><p><strong>Conclusion: </strong>SFM facilitates the generation of cell sheets that exhibit structural and functional properties analogous to those cultured in FBS. Additionally, SFM promotes cellular phenotype transition and ECM deposition. Consequently, SFM provides a safer, more controllable, and clinically translatable solution for cell sheet construction.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"51"},"PeriodicalIF":5.7,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144173859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cameron T Roots, Alexis M Hill, Claus O Wilke, Jeffrey E Barrick
{"title":"Codon usage modulates the relationship between the burden and yield of protein overexpression.","authors":"Cameron T Roots, Alexis M Hill, Claus O Wilke, Jeffrey E Barrick","doi":"10.1186/s13036-025-00521-z","DOIUrl":"10.1186/s13036-025-00521-z","url":null,"abstract":"<p><strong>Background: </strong>Excess utilization of translational resources is a critical source of burden on cells engineered to overexpress exogenous proteins. To improve translational efficiency, researchers often modify codon usage in an exogenous gene to more closely match the composition of a host organism's highly expressed genes. Despite empirical data showing the benefits of codon optimization, little is known about the quantitative relationships between codon usage, protein yield, and the burden imposed on a host cell by protein overexpression.</p><p><strong>Results: </strong>We develop and experimentally evaluate a stochastic gene expression model that considers the impact of codon usage bias on the availability of ribosomes and different tRNAs in a cell. In agreement with other studies, our model shows that increasing exogenous protein expression decreases production of native cellular proteins in a linear fashion. We also find that the slope of this relationship is modulated by how well the codon usage bias of the exogenous gene and the host's genes match. Lastly, our model predicts that an overoptimization domain exists where further increasing usage of optimal codons worsens yield and burden. We test our model by expressing sfGFP and mCherry2 from constructs that have a wide range of codon optimization levels in Escherichia coli. The results agree with our model, including for an mCherry2 gene sequence that appears to less efficiently express this gene due to codon overoptimization.</p><p><strong>Conclusions: </strong>Our model reproduces experimentally observed relationships between codon usage bias, gene expression, and burden for overexpressed proteins. Furthermore, it suggests that more nuanced recoding strategies that seek to match a host's overall codon usage bias are less burdensome and will lead to greater protein yields compared to strategies that simply maximize usage of optimal codons. Increasing the level of mechanistic detail in gene expression models can lead to insights that allow researchers to engineer more optimal cellular systems.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"50"},"PeriodicalIF":5.7,"publicationDate":"2025-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12100883/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144127723","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Bio-engineered thermo-sensitive alginate/PNIA-g-CS co-polymeric injectable hydrogel laden with GDF-5 to stimulate nucleus pulposus for IVD regeneration.","authors":"Guangnan Chen, Chong Bian, Xiangyang Cheng, Jun Xu, Kaifeng Zhou, Yiming Zhang","doi":"10.1186/s13036-025-00520-0","DOIUrl":"10.1186/s13036-025-00520-0","url":null,"abstract":"<p><p>Chronic back pain and disability are primarily caused by intervertebral disc degeneration (IDD) that requires novel therapies to regenerate the nucleus pulposus (NP) and restore disc function. In this study, a bioengineered thermo-sensitive injectable hydrogel composed of co-polymeric poly-N-isopropyl acrylamide-grafted-chondroitin sulfate cross-linked with sodium alginate microspheres (PNIA-g-CS-NaA Ms: denote HMs) loaded with growth differentiation factor 5 (GDF-5), to stimulate Nucleus Pulposus cells (NPCs) activity and promote intervertebral disc (IVD) regeneration. The Low critical solution temperature (LCST) of PNIA-g-CS was 31.8 and 32.3 °C at 5% (w/v) and 15% (w/v), respectively. In the in vitro study, GDF-5-loaded hydrogel (1 mg/mL) marginally enhanced NPC proliferation and reduced inflammatory cytokines (TNF-α, IL-6, IL-1β) after 24 h. HMs-GDF-5 combined with Adipose-Derived Mesenchymal Stem Cells (ADMSCs) was delivered to NP tissue using a minimally invasive technique, promoting NP regeneration in rats. At 8 weeks, significant upregulation of COL-II and ACAN proteins and mRNA expressions was observed. X-ray imaging showed disc height recovery and increased water content, while histology revealed partial restoration of NPCs and matrix. The outcomes show that the biodegradable hydrogel could be used as a potential therapeutic agent for IVD repair.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"49"},"PeriodicalIF":5.7,"publicationDate":"2025-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12100944/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144127722","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Parastoo Noory, Ahmad Reza Farmani, Jafar Ai, Naghmeh Bahrami, Mohammad Bayat, Somayeh Ebrahimi-Barough, Ali Farzin, Shima Shojaie, Hamed Hajmoradi, Abdolreza Mohamadnia, Arash Goodarzi
{"title":"Enhancing in vitro osteogenic differentiation of mesenchymal stem cells via sustained dexamethasone delivery in 3D-Printed hybrid scaffolds based on polycaprolactone-nanohydroxyapatite/alginate-gelatin for bone regeneration.","authors":"Parastoo Noory, Ahmad Reza Farmani, Jafar Ai, Naghmeh Bahrami, Mohammad Bayat, Somayeh Ebrahimi-Barough, Ali Farzin, Shima Shojaie, Hamed Hajmoradi, Abdolreza Mohamadnia, Arash Goodarzi","doi":"10.1186/s13036-025-00514-y","DOIUrl":"10.1186/s13036-025-00514-y","url":null,"abstract":"<p><p>Despite the natural ability of bone repair, its limitations have led to advanced organic-inorganic-based biomimetic scaffolds and sustained drug release approaches. Particularly, dexamethasone (DEX), a widely used synthetic glucocorticoid, has been shown to increase the expression of bone-related genes during the osteogenesis process. This study aims to develop a hybrid 3D-printed scaffold for controlled delivery of dexamethasone. Hence, hybrid scaffolds were fabricated using a layer-by-layer 3D-printing of combined materials comprising polycaprolactone (PCL)-nanohydroxyapatite (nHA) composite, and DEX-loaded PCL microparticles embedded in the alginate-gelatin hydrogel. Encapsulation efficiency, loading capacity, and in vitro kinetics of DEX release were evaluated. Osteogenic differentiation of human endometrial mesenchymal stem cells (hEnMSCs) on DEX-loaded hybrid scaffolds was assessed by evaluating osteogenic gene expression levels (collagen I, osteonectin, RUNX2), alkaline phosphatase (ALP) activity, and scaffold mineralization. The hybrid scaffolds exhibited favorable morphology, mechanical-properties, biocompatibility, and biodegradability, enhancing osteogenesis of hEnMSCs. DEX-loaded PCL microparticles within hybrid scaffolds exhibited a controlled release pattern and promoted osteogenic differentiation during the sustained release period through a significant increase in osteonectin and COL1A1 expression. Also, increased mineralization was demonstrated by SEM and alizarin red staining. This study proposes that drug-loaded 3D-printed hybrid organic-inorganic nanocomposite scaffolds are promising for advanced bone tissue engineering applications.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"48"},"PeriodicalIF":5.7,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12093820/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144110808","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Amanda Cimino, Fiona Pat, Omolabake Oyebamiji, Christine T N Pham, Erik D Herzog, Farshid Guilak
{"title":"Dual-responsive synthetic gene circuit for dynamic biologic drug delivery via inflammatory and circadian signaling pathways.","authors":"Amanda Cimino, Fiona Pat, Omolabake Oyebamiji, Christine T N Pham, Erik D Herzog, Farshid Guilak","doi":"10.1186/s13036-025-00519-7","DOIUrl":"10.1186/s13036-025-00519-7","url":null,"abstract":"<p><strong>Background: </strong>Engineered cells provide versatile tools for precise, tunable drug delivery, especially when synthetic stimulus-responsive gene circuits are incorporated. In many complex disease conditions, endogenous pathologic signals such as inflammation can vary dynamically over different time scales. For example, in autoimmune conditions such as rheumatoid arthritis or juvenile idiopathic arthritis, local (joint) and systemic inflammatory signals fluctuate daily, peaking in the early morning, but can also persist over long periods of time, triggering flare-ups that can last weeks to months. However, treatment with disease-modifying anti-rheumatic drugs is typically provided at continuous high doses, regardless of disease activity and without consideration for levels of inflammatory signals. In previous studies, we have developed cell-based drug delivery systems that can automatically address the different scales of flares using either chronogenetic circuits (i.e., clock gene-responsive elements) that can be tuned for optimal drug delivery to dampen circadian variations in inflammatory levels or inflammation-responsive circuits (i.e., NF-κB-sensitive elements) that can respond to sustained arthritis flares on demand with proportional synthesis of drug. The goal of this study was to develop a novel dual-responsive synthetic gene circuit that responds to both circadian and inflammatory inputs using OR-gate logic for both daily timed therapeutic output and enhanced therapeutic output during chronic inflammatory conditions.</p><p><strong>Results: </strong>We developed a synthetic gene circuit driven by tandem inflammatory NF-κB and circadian E'-box response elements. When engineered into induced pluripotent stem cells that were chondrogenically differentiated, the gene circuit demonstrated basal-level circadian output with enhanced stimulus-responsive output during an inflammatory challenge shown by bioluminescence monitoring. Similarly, this system exhibited enhanced therapeutic levels of biologic drug interleukin-1 receptor antagonist (IL-1Ra) during an inflammatory challenge in differentiated cartilage pellets. This dual-responsive therapeutic gene circuit mitigated both the inflammatory response as measured by bioluminescence reporter output and tissue-level degradation during conditions mimicking an arthritic flare.</p><p><strong>Conclusions: </strong>The dual-responsive synthetic gene circuit developed herein responds to input cues from two key homeostatic transcriptional networks, enabling dynamic and tunable output. This proof-of-concept approach has the potential to match drug delivery to disease activity for optimal outcomes that addresses the complex environment of inflammatory arthritis.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"47"},"PeriodicalIF":5.7,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12090394/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144101914","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Core-shell nanofiber dressings with zinc oxide nanoparticles and cell-free fat extract: boosting fibroblast activity and antibacterial efficacy.","authors":"Aydin Mahmoudnezhad, Mahsa Bayrami, Mahdiyeh Saadati, Yavuz Nuri Ertaş, Mozhgan Abasi, Aylar Ebrahimi, Younes Pilehvar","doi":"10.1186/s13036-025-00511-1","DOIUrl":"10.1186/s13036-025-00511-1","url":null,"abstract":"<p><strong>Background: </strong>This study presents the development and characterization of innovative core-shell nanofiber wound dressings incorporating zinc oxide nanoparticles (nZnO) and cell-free fat extract (CEFFE) to enhance fibroblast activity and antibacterial efficacy.</p><p><strong>Results: </strong>CEFFE was prepared and analyzed, revealing high concentrations of essential growth factors, particularly bFGF and TGF-β1, supporting its therapeutic potential in tissue regeneration. The fabricated nanofibers (PLCL, nZnO/PLCL, PLCL-CEFFE/HA, and nZnO/PLCL-CEFFE/HA) were examined using FE-SEM and TEM, demonstrating successful encapsulation and morphological variations due to nZnO incorporation. XRD analysis confirmed the structural integrity and effective loading of nZnO and CEFFE. Hydrophilicity assessment via water contact angle measurements showed that CEFFE/HA significantly enhanced the hydrophilicity of PLCL membranes, crucial for wound exudate management. Mechanical tests indicated that CEFFE/HA addition maintained the scaffold's mechanical robustness, while nZnO slightly reduced mechanical properties. In vitro release studies revealed a biphasic release pattern of Zn²⁺ ions and growth factors from nZnO/PLCL-CEFFE/HA nanofibers, ensuring prolonged antibacterial activity and sustained therapeutic effects. Antibacterial assays demonstrated significant efficacy against E. coli and S. aureus, attributed to nZnO. MTT assays and FE-SEM analysis confirmed enhanced NIH-3T3 cell proliferation and adhesion on PLCL-CEFFE/HA nanofibers due to the controlled release of growth factors. The scratch assay showed superior cell migration and wound healing potential for PLCL-CEFFE/HA formulations.</p><p><strong>Conclusions: </strong>These findings underscore the potential of nZnO/PLCL-CEFFE/HA core-shell nanofibers as multifunctional wound dressings, combining antibacterial properties with enhanced tissue regeneration capabilities. However, further studies are needed to assess long-term stability and in vivo performance, which represent key challenges for future research.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"46"},"PeriodicalIF":5.7,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12090510/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144101913","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Design of a prodrug bispecific antibody masked by a functional molecule for lymphocyte activation for cancer therapy.","authors":"Daimei Miura, Yuki Kato, Masahiro Yasunaga, Izumi Kumagai, Ryutaro Asano","doi":"10.1186/s13036-025-00517-9","DOIUrl":"10.1186/s13036-025-00517-9","url":null,"abstract":"<p><p>Although T cells engaging bispecific antibodies (T-bsAbs) have shown great benefits, their use in treating solid tumors is challenging because of the minimal infiltration of T-cells. We fused an agonistic single-chain variable fragment (scFv) that induces a T cell co-stimulatory signal to the T cell-binding domain of T-bsAb via a linker containing a cancer-specific protease recognition site. With this antibody format, unexpected cytotoxicity to the surrounding normal tissue would be reduced and tumor-specific T cell activation would occur. The scFv-masked T-bsAb was cleaved by collagenase with intrinsic cancer-specific protease activity, releasing agonistic scFv without unwanted fragmentation and restoring the binding ability of the scFv-masked bsAbs to T cells. Compared to the original bsAb, a detectable enhancement of the T cell proliferation and cancer cytotoxicity was observed after the incubation with collagenase or protease-secretory cancer cells, which was suggested to be due to the modest co-stimulation by the released agonistic scFv. Our results provide important insights into an ideal T-bsAb prodrug format, precisely engineered to reduce side effects and exert high cancer cytotoxicity for solid tumor precision medicine.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"45"},"PeriodicalIF":5.7,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12079947/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144077880","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fernando Roberto Paz Cedeno, Olumide Joseph Olubiyo, Sungil Ferreira
{"title":"From microbial proteins to cultivated meat for alternative meat-like products: a review on sustainable fermentation approaches.","authors":"Fernando Roberto Paz Cedeno, Olumide Joseph Olubiyo, Sungil Ferreira","doi":"10.1186/s13036-025-00509-9","DOIUrl":"https://doi.org/10.1186/s13036-025-00509-9","url":null,"abstract":"<p><p>The global demand for protein is rapidly increasing due to population growth and changing dietary preferences, highlighting the need for sustainable alternatives to traditional animal-based proteins. This review explores cultivated meat and microbial alternative proteins, focusing on their potential to meet nutritional needs while mitigating environmental impacts. It also examines the production of cultivated meat as well as various sources of microbial proteins, including mycoproteins, bacterial proteins, and microalgae, highlighting their nutritional profiles, production methods, and commercial applications. This includes an evaluation of the state of commercialization of mycoproteins and the innovative use of agricultural and industrial by-products as substrates for microbial fermentation. The integration of microbial protein production with the bioenergy sector is evaluated as a relevant alternative to attain a synergetic effect between energy and food production systems. Ultimately, this work aims to underscore the importance of microbial proteins in advancing towards a more sustainable protein production system, offering insights into current challenges and future opportunities in the field of fermentation to produce alternative proteins.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"44"},"PeriodicalIF":5.7,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12077041/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144077901","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Effects of antifoam agents on Spodoptera frugiperda 9 cell growth and baculovirus infection dynamics.","authors":"Kristina Worch, Merlin Krause, Antje Burse","doi":"10.1186/s13036-025-00516-w","DOIUrl":"https://doi.org/10.1186/s13036-025-00516-w","url":null,"abstract":"<p><p>The baculovirus expression system is widely used for recombinant protein production. However, its scalability under shaking or stirring cultivation conditions remains a challenge due to foam formation which can negatively affect cell metabolism and viability, complicate process control, and ultimately lower productivity. Compared to other protein expression platforms, the effect of antifoam agents on insect cell culture has been rarely investigated. This study examines the influence of three antifoam agents-Antifoam 204 (AF204), polypropylene glycol (PPG), and a silicone-based compound (SAG471)-on Spodoptera frugiperda (Sf) 9 cell growth, viability, baculovirus infectivity, and infection dynamics. Dose-response experiments in adherent culture showed that high antifoam concentrations inhibited metabolic activity. In suspension culture, low concentrations of AF204 and PPG enhanced cell growth by reducing lag phase and population doubling time, while growth with SAG471 remained comparable to a no-antifoam control. In virus titer experiments, no effects on the plaque-forming ability of baculovirus particles could be observed. However, infection dynamics monitored in suspension cultures improved in the presence of all three antifoam agents, as shown by cell size increase, living cell stagnation, and enhanced single-cell fluorescence. Foam reduction experiments demonstrated that only SAG471 contributed to foam removal within a non-toxic concentration range. The results indicate that antifoam agents, depending on their concentration and composition, can enhance Sf9 cell growth and viability while potentially modulating cell membrane properties that could improve viral infection efficiency and transfection efficiency of exogenous material. This highlights the potential of antifoam agents for optimizing other virus-based expression systems in higher eukaryotic cells.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"43"},"PeriodicalIF":5.7,"publicationDate":"2025-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12065184/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143998474","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Phenolic-loaded nanofiber from Arctium lappa root: a potential therapy for testosterone-induced ovarian oxidative stress.","authors":"Viana Khojasteh, Seyed Amirmohammad Mollanorouzi, Ehsan Karimi, Behrouz Ghorani, Ehsan Oskoueian","doi":"10.1186/s13036-025-00515-x","DOIUrl":"https://doi.org/10.1186/s13036-025-00515-x","url":null,"abstract":"<p><p>Polycystic Ovary Syndrome (PCOS) is a hormonal disorder affecting women of reproductive age, often associated with oxidative stress and inflammation. This study explores the therapeutic potential of Arctium lappa phenolic-rich fraction encapsulated nanofiber (ALPRF-NF) in a testosterone-induced PCOS mouse model. All experiments were performed in triplicate and Duncan's Multiple Range Test was used to assess significant differences between means, with significance determined at p < 0.05. The ALPRF-NF formulation demonstrated favorable physicochemical properties, including a ribbon-like structure (216.9 nm), a zeta potential of -19.3 mV, and a high encapsulation efficiency (93.1%). In vivo findings showed that ALPRF-NF significantly improved body weight, feed intake, and liver enzyme profiles in PCOS-induced mice (p ≤ 0.05). It also enhanced the antioxidant defense system by elevating levels of glutathione peroxidase (GPX), superoxide dismutase (SOD), and catalase (CAT). Mechanistically, ALPRF-NF reduced oxidative stress and inflammation by delivering phenolic compounds that scavenge reactive oxygen species (ROS) and modulate gene expression in ovarian tissue. This included downregulation of inducible nitric oxide synthase (iNOS) and upregulation of SOD expression. These results suggest that ALPRF-NF effectively mitigates testosterone-induced ovarian oxidative damage and inflammation, offering a targeted, nanotechnology-based therapeutic approach for PCOS. The study provides valuable insights into novel strategies for improving women's reproductive health through bioactive compound delivery.</p>","PeriodicalId":15053,"journal":{"name":"Journal of Biological Engineering","volume":"19 1","pages":"42"},"PeriodicalIF":5.7,"publicationDate":"2025-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12065167/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143991402","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}