Pre-Culture of Scaffolds with Vasculogenic Cells Improves Stem Cell-Derived β Cell Transplantation
Transplantation of stem cell-derived β cells is a promising treatment for type-1 diabetes, increasing the supply of insulin-producing cells beyond that of cadaveric islet transplantation. Transplant success is limited by cell death after transplantation, with insufficient oxygen and nutrient accessibility strongly contributing to apoptosis and de-differentiation. Herein, we investigate cotransplantation of endothelial cells and fibroblasts with stem cell-derived β cells to enhance survival and function posttransplantation. A microporous poly (lactide coglycolide) scaffold was used for culture and transplantation of stem cell-derived β cells. Coculture of the stem cell-derived β cells with endothelial cells and fibroblasts generated vascular networks during culture, which persisted through transplantation and enhanced vascularization. 7-days of culture supported enhanced survival of transplanted cells, though function in terms of insulin secretion and reduction of hyperglycemia was compromised. However, 3-days of preculture led to both improved survival and function of the stem cell-derived β cells, with transplant recipients demonstrating reduced fasting blood glucose levels. These studies demonstrate the potential and some constraints on the application of vascularization strategies to enhance function of stem cell-derived β cells with transplantation to extrahepatic sites.
Promoting Human Intestinal Organoid Formation and Stimulation Using Piezoelectric Nanofiber Matrices
Human organoid model systems have changed the landscape of developmental biology and basic science. They serve as a great tool for human-specific interrogation. In order to advance our organoid technology, we aimed to test the compatibility of a piezoelectric material with organoid generation, because it will create a new platform with the potential for sensing and actuating organoids in physiologically relevant ways. We differentiated human pluripotent stem cells into spheroids following the traditional human intestinal organoid (HIO) protocol atop a piezoelectric nanofiber scaffold. We observed that exposure to the biocompatible piezoelectric nanofibers promoted spheroid morphology 3 days sooner than with the conventional methodology. At day 28 of culture, HIOs grown on the scaffold appeared similar. Both groups were readily transplantable and developed well-organized laminated structures. Graft sizes between groups were similar. Upon characterizing the tissue further, we found no detrimental effects of the piezoelectric nanofibers on intestinal patterning or maturation. Furthermore, to test the practical feasibility of the material, HIOs were also matured on the nanofiber scaffolds and treated with ultrasound, which lead to increased cellular proliferation which is critical for organoid development and tissue maintenance. This study establishes a proof of concept for integrating piezoelectric materials as a customizable platform for on-demand electrical stimulation of cells using remote ultrasonic waveforms in regenerative medicine.
Evaluation of Carboxymethyl Chitosan Hydrogel Containing Caviar Extract Effect on Skin Regeneration
Skin aging involves changes in extracellular matrix components, such as wrinkles and pigmentation. Caviar extract (CE) is a promising compound for skin rejuvenation, but effective topical delivery requires optimized carriers. This study evaluated polyvinyl alcohol/carboxymethyl chitosan (PVA/CMC) hydrogels loaded with CE at concentrations of 2%, 3.5%, and 5% as scaffolds to influence the epithelial differentiation of adipose-derived mesenchymal stem cells (ADMSCs). Hydrogels were synthesized using a freeze-thaw method and characterized by Fourier-transform infrared spectroscopy (FTIR), scanning electron microscopy, swelling and degradation tests, and mechanical analysis. Biocompatibility and cell migration were assessed using MTT and scratch assays; at the same time, expression of cytokeratin-18 () and pan-cytokeratin (pan-CK) was measured via reverse transcription-quantitative polymerase chain reaction and immunocytochemistry (ICC), respectively. FTIR confirmed successful CE incorporation, and SEM revealed a porous structure. Hydrogels with 3.5% and 5% CE demonstrated a good balance between swelling and degradation over 336 h. The biocompatibility tests showed that 5% CE supported enhanced long-term cell growth. The scratch assay indicated improved cell migration, and transcriptional analysis revealed significantly higher levels in ADMSCs treated with PVA/CMC/CE 5% ( < 0.001). ICC results showed significantly higher pan-CK expression at 3.5% CE (41.82%) and 5% CE (48.16%), suggesting that CE promotes repair processes. These findings suggest that 5% CE-loaded PVA/CMC hydrogel could be an effective option for skin regeneration and antiaging. Impact Statement Caviar extract (CE) was considered a bioactive ingredient, along with polyvinyl alcohol (PVA) and carboxymethyl chitosan (CMC) polymers, to prepare a functional and practical hydrogel without hazardous components for anti-aging and cosmetic applications. In the present study, the PVA/CMC hydrogel contains various concentrations of CE (3.5% and 5%), is biocompatible, and enhances cellular viability and migration of adipose-derived mesenchymal stem cell. Our results demonstrated that the synergistic effect of CE and CMC could promote the expression of cytokeratin-18 gene and pan-cytokeratin protein and play a critical role in stimulating skin regeneration.
Bonegraft Using Octacalcium Phosphate Collagen Composite with Teriparatide and Polylactic Acid Cage
Octacalcium phosphate (OCP) is a bone grafting material known for its biocompatibility, osteoconductive, and osteogenic properties. Current treatments for extensive jaw defects often involve vascularized bone grafts or titanium mesh-based osteogenesis, which have limitations such as graft resorption, infections, and reoperation needs. In this study, a new bone regeneration therapy was explored, in which OCP combined with collagen (Col), treated with teriparatide (TPTD), was encased in a polylactic acid (PLA) cage to enhance structural stability and promote controlled bone formation. The therapeutic effects of this approach were evaluated using a rat model for calvarial regeneration, employing immunohistochemical staining. TPTD-treated OCP/Col composites were encased in cylindrical PLA cages, which were created using a 3D printer, and implanted into rat skulls. Three cage designs were tested: no holes, one large hole, and several small holes. Following implantation, the specimens underwent microcomputed tomography (micro-CT), histological, and immunohistochemical analyses to assess bone regeneration. In the micro-CT analysis, radiopacity at the OCP/Col graft site was higher in the "no hole" cage group than in the other groups from 4 to 12 weeks after implantation, particularly in the marginal area and region adjacent to the bone. Histological analysis revealed that, in all groups, new bone formation was observed along the surface of the skull 12 weeks postimplantation. In the "no hole" cage group, bone formation extended to the upper middle section, and bone matrix was present in areas where mature bone formation was lacking. In the other two groups, fibrous tissue filled the holes in the PLA cage, and no bone formation was observed directly beneath the holes. Immunohistochemical analysis revealed the expression of osteopontin, osteocalcin, runt-related transcription factor 2, vascular endothelial growth factor, and collagen I in all groups. The "no hole" cage group exhibited uniform and successful bone formation, with these cell markers consistently observed throughout all regions. These results suggest that using PLA cages to cover TPTD-treated OCP/Col discs effectively promotes bone regeneration. This approach provides a promising alternative to conventional bone grafting techniques and may help overcome the limitations associated with free or autologous bone grafts in oral and maxillofacial reconstruction. Impact Statement This study demonstrates that encasing teriparatide-treated octacalcium phosphate/collagen composites in polylactic acid (PLA) cages enhances bone regeneration. Using a rat model, microcomputed tomography, histological, and immunohistochemical analyses revealed that the "no hole" PLA cage design promoted uniform and successful bone formation, whereas perforated cages led to fibrous tissue infiltration. These findings highlight the potential of PLA cages in optimizing bone grafting strategies, offering a promising approach for tailored bone regeneration therapies.
Experimental Study of Umbilical Cord Graft and Established Sling Materials for Frontalis Suspension
Frontalis suspension surgery is the preferred treatment option for patients with poor levator function ptosis. This procedure connects the affected eyelid to the brow using sling material, harnessing the action of the frontalis muscle to elevate the upper eyelid. Various sling materials have been used, most commonly silicone rods and fascia lata. However, both have notable limitations: silicone rods carry a relatively high risk of postoperative inflammation and ptosis recurrence, while fascia lata, due to its low elasticity, may cause blinking dysfunction and exposure keratopathy. Additionally, fascia lata harvesting poses challenges in young children. Therefore, there is a need for an alternative human tissue sling material that is both readily available and capable of overcoming the limitations of established sling materials. This study aimed to evaluate the viability of human umbilical cord grafts as a novel sling material for frontalis suspension surgery in ptosis patients. We developed a new method for dissecting and dehydrating umbilical cord tissue and assessed its mechanical and histological properties using uniaxial tensile testing and histological analysis. Untreated umbilical cord grafts exhibited mechanical strength (15.9546 ± 2.6117 N) and strain (96.8674 ± 3.6707%) values intermediate between those of silicone rod and fascia lata. Alcohol dehydration significantly increased ultimate tensile strength and maximum strain, ultimate strength values exceeding those of silicone rod. These grafts withstood forces exceeding those generated during forced blinking, outperforming silicone rod in strength and exhibiting greater elasticity than fascia lata. Histological analysis revealed abundant collagen and glycosaminoglycans within Wharton's jelly, alongside elastic fiber-rich regions in vessel walls. The presence of these extracellular matrix components likely underlies the grafts' favorable mechanical properties. Overall, umbilical cord grafts may emerge as a promising alternative to conventional sling materials in ptosis surgery, potentially addressing limitations in material availability. Impact Statement This study introduces human umbilical cord grafts as a novel sling material for frontalis suspension surgery in patients with ptosis. We developed a new method for dissecting and dehydrating umbilical cord tissue. Our results suggest that umbilical cord graft may offer sufficient tensile strength and strain, potentially reducing recurrence rates and minimizing postoperative complications. This work lays the groundwork for future studies exploring the clinical application of umbilical cord-derived biomaterials in surgical procedures.
Bile Duct Regeneration Using a Gelatin Hydrogel Nonwoven Fabric-Based Artificial Bile Duct in Pigs
Bile duct jejunal anastomosis is a standard reconstruction method following bile duct resection. Nevertheless, this procedure is technically intricate and carries significant postoperative risks. This study evaluated bile duct regeneration in pigs using artificial bile ducts (ABDs) made of gelatin hydrogel nonwoven fabric (GHNF). An ABD composed of polyglycolic acid (PGA) as the inner layer and GHNF as the outer layer was implanted in the defect of the bile duct in pigs. After a 105-day implantation period, tissue samples were analyzed via histology, immunohistochemistry, and RNA sequencing. The implantation of the ABD promoted fibroblast infiltration, extracellular matrix (ECM) formation, and bile duct epithelial regeneration in the site of the bile duct defect by postoperative day 105. Histological analysis revealed complete absorption and replacement of GHNF by collagen-rich ECM. Immunohistochemistry studies indicated the presence of CK19-positive bile duct epithelial cells in the ABD area, suggesting the successful regeneration of the entire bile duct structure. Furthermore, RNA sequencing revealed gene expression patterns analogous to those observed in native bile ducts, showing a similarity with a significant correlation coefficient between the regenerated and the native bile ducts. Differentially expressed genes related to ECM formation, such as COL3A1, SPARC, and COL1A1, were highly expressed, along with growth factors such as FGF1, FGF7, FGF18, FGF22, TGFβ1, and TGFβ3. The experimental findings demonstrated the successful regeneration of bile duct tissue by the ABD made of GHNF implanted in pigs, thereby signifying its potential for future clinical applications.
Comparative Structural and Functional Analysis of Micronized Collagen-Based Scaffolds
Micronized collagen-based bioscaffolds are increasingly used in clinical applications for wound repair and soft tissue regeneration. This study compared the structural properties of four different commercially available micronized products derived from either reconstituted collagen (pRC), urinary bladder matrix (pUBM), or ovine forestomach matrix (mOFM, mOFMµ). The test articles were characterized by laser diffraction analysis, scanning electron microscopy (SEM), micro-computed tomography (micro-CT), packing density, differential scanning calorimetry, rheometry, proteolytic stability, agarose gel electrophoresis, and blood clotting index. Particle size and surface morphology, assessed by laser diffraction, SEM, and micro-CT, revealed marked differences in particle size, shape, and aggregation. Packing density ranged from 80.3 ± 2.7 mg/cm (mOFM) to 484.7 ± 17.8 mg/cm (pRC). Thermal analysis demonstrated the native structure of the OFM-based test articles (T, 59.80 ± 0.11°C and 58.15 ± 0.15°C) relative to pUBM and pRC (T, 41.06 ± 0.06°C and 40.59 ± 0.23°C). Rheological testing revealed that mOFM and mOFMµ had increased cohesive energy, indicating better mechanical resilience when the micronized materials were rehydrated to form a paste. The OFM-based test articles exhibited the greatest resistance to proteolytic digestion (T, 12.730 ± 1.232 and 5.759 ± 0.1296). All the test articles, except for the reconstituted collagen product, demonstrated hemostasis in whole blood. Micronized reconstituted collagen showed immediate dissolution and no fluid absorption, hemostasis, or resistance to proteolytic digestion, whereas micronized OFM showed the greatest proteolytic stability and packing density. Substantial differences among the micronized bioscaffolds were revealed from the analysis, most likely due to their different source materials and manufacturing processes. Careful consideration of these parameters is warranted when selecting a micronized product for soft tissue applications.
Extracellular Matrix Degradation Products Inhibit Esophageal Cancer Cell Proliferation and Migration
Biological materials composed of extracellular matrix (ECM) or its components have been successfully used for tissue repair and reconstruction. Preclinical studies, along with a cohort study following stage T1A esophageal adenocarcinoma (EAC) resection, have shown that ECM biomaterials can restore esophageal mucosa and submucosa without cancer recurrence. However, the molecular mechanisms underlying these effects remain largely unexplored. The present study investigates the effects of ECM degradation products from nonmalignant esophageal (eECM) and urinary bladder (ubECM) sources on EAC cell proliferation, migration, and associated signaling pathways. Both eECM and ubECM significantly inhibited OE33 cell proliferation, with eECM exhibiting a stronger effect-reducing proliferation to 25% at 24 h and 7% at 72 h compared with pepsin control ( < 0.001). A high-throughput cell surface marker screen followed by gene and protein expression analysis revealed that both ECM sources downregulated CD164 and CXCR4, reducing CXCR4 protein levels by approximately 50% ( = 0.006 for eECM, = 0.007 for ubECM). Notably, only eECM significantly suppressed OE33 cell migration ( ≤ 0.0001) and downregulated bone morphogenetic protein 4 expression, along with its downstream targets pSMAD1/5/8, , and , thereby reducing epithelial-mesenchymal transition. These findings support the concept that biochemical cues from nonmalignant ECM modulate neoplastic cell behavior. Given the involvement of PI3K-Akt and BMP4 signaling in EAC progression, ECM-based strategies may warrant further investigation as potential therapeutic approaches following esophageal cancer resection.
Cost of Stem Cell-Based Tissue-Engineered Airway Transplants in the United Kingdom: Case Series
Keratin 3D Printing: Sustainable Biomaterials for Regenerative Medicine and Biomanufacturing
Keratin as an abundantly available natural protein from sources such as hair, wool, and feathers possesses excellent biocompatibility, biodegradability, and bioactivity that support cell growth. Recent advances in extracting, purifying, and characterizing keratin have led to the development of various keratin-based biomaterials, such as fibers, gels, films, and nanoparticles via conventional fabrication methods. However, these biomaterials are often limited by simple geometries, weak mechanical strength, and limited reproducibility. Emerging 3D printing technologies offer a promising alternative, allowing the creation of keratin-based scaffolds with precise architecture, tunable mechanical strength, and reproducible geometries. Despite keratin's abundance and biological advantages, the use of keratin in 3D printing remains relatively underexplored. This review provides a comprehensive overview of keratin's molecular structure and biochemistry, its diverse natural sources, extraction and purification methodologies, and the cross-linking mechanisms (chemical, UV, and enzymatic) used to formulate printable keratin-based inks. Furthermore, it discusses the biomedical applications of keratin-derived bioinks in tissue engineering and additive biomanufacturing, with emphasis on skin and bone regeneration. Combining keratin's biological functionality with the design flexibility of 3D printing offers a sustainable and cost-effective pathway toward next-generation biomaterials for regenerative medicine.
From Chemistry to Clinic: Polysaccharide-Bioceramic Composites for Tissue Engineering Applications
Composite scaffolds combining polysaccharides and bioceramics represent next-generation scaffolds extensively investigated in tissue engineering (TE) and biomedical applications. Polysaccharides such as chitosan, hyaluronic acid, and pectin mimic the extracellular matrix components with their tunable physicochemical properties, enabling a favorable microenvironment for cell adhesion, proliferation, and cell-matrix interactions. On the other hand, bioceramics, including calcium phosphate, hydroxyapatite, and bioactive glasses, enhance the mechanical properties of the material and offer structural integrity and osteoconductive properties. While they have generally been preferred to be used in bone TE and dental applications, various studies have also demonstrated their potential in cartilage regeneration, wound healing, and broader biomedical applications. Recent advancements in material design and scaffold fabrication techniques, particularly 3D printing and electrospinning, have provided precise engineering of materials and fabrication of scaffolds for desirable mechanical properties and biological performance. These innovations foster the development of patient-specific scaffolds, thereby paving the way for applications in personalized medicine. This review critically summarizes alternative polysaccharides, bioceramics, and composite materials used in TE and biomedical applications. It also highlights advanced fabrication strategies and finally explores the translational potential of these biocomposites. By integrating emerging technologies, this review aims to provide alternative and sustainable materials for the development of next-generation scaffolds that meet clinical needs. Impact Statement This study introduces polysaccharide-bioceramic composites with enhanced mechanical and biological properties for tissue engineering. Beyond bone and dental repair, their applications increasingly extend to wound healing, cartilage, cardiac, and muscle regeneration with drug delivery, angiogenesis, and neurogenesis. By mimicking the native extracellular matrix, these composites support cell growth and tissue regeneration, offering a versatile platform for advanced regenerative therapies.
Inhibition of TGF-β in Tubule Cells Increases Respiration via Upregulation of Respiratory Genes
Renal tubule cells lose differentiated characteristics in artificial culture, limiting their application in medical research and cell therapy. We previously showed that adding inhibitors of transforming growth factor-β (TGF-β) signaling to cell culture media increased specific transport functions characteristic of differentiated tubule cells. Transport in proximal tubule cells is energetically demanding; , these cells rely primarily on oxidative phosphorylation of fatty acids for adenosine triphosphate (ATP) generation. We examined whether TGF-β inhibition, with or without metformin, altered glycolysis and oxidative phosphorylation compared with standard culture conditions. Primary renal tubule cells (PRTC) were cultured with or without an inhibitor of TGF-β receptor I and with or without metformin in a 2 × 2 factorial design. First, expression of proteins in fatty acid transport and the electron transport chain was compared between conditions. The relative contributions of glycolysis and oxidative phosphorylation to ATP generation were assessed by extracellular acidification rate (ECAR) and oxygen consumption rate (OCR). We also tested substrate-specific contributions using inhibitors of pyruvate, glutamine, and carnitine mitochondrial entry. Finally, OCR and transport were measured after 48 weeks in culture to determine durability of culture phenotype. Metformin and SB431542 increased expression and phosphorylation of proteins in the electron transport chain and involved in fatty acid transport. Metformin and TGF-β inhibition increased oxidative phosphorylation. Metformin decreased glucose dependency, while combination with TGF-β inhibition increased fatty acid dependency. Differences in OCR and transport between treatment conditions persisted at 48 weeks in culture. Renal tubule cell transport is energetically demanding, so cellular differentiation requires matching increases in energetic machinery. We found that metformin and inhibition of TGF-β increased oxygen consumption and utilization of fatty acids in cultured primary tubule cells. These data support the hypothesis that TGF-β inhibition not only increases expression of a broad array of transporters characteristic of the proximal tubule, as we previously showed, but also improves the supply of energy to support active transport.
Differentiation of iPS Cells into Periodontal Ligament Cells
Periodontal ligament (PDL) is a thin connective tissue that connects the tooth to the bony socket and plays a crucial role in the regeneration and maintenance of homeostasis of periodontal tissues by supplying stem/progenitor cells. Induced pluripotent stem cells (iPSCs) are highly anticipated in regenerative medicine because of their differentiation potential into a wide variety of cell types. In this study, we investigated the effects of humoral factors on iPSC differentiation by culturing iPSCs in the presence of PDL cell-derived culture supernatants. Changes in gene expression were analyzed using quantitative real-time PCR, reverse-transcription PCR, and RNA sequencing. The marker protein expression on the cell surface was assessed using flow cytometry. Periodontal regeneration was verified by microcomputed tomography and histomorphological observation in a periodontal defect model using male F344/NJcl-/ rats. When iPSCs were cultured in the PDL culture supernatant, some cells formed clumps, and spindle-shaped cells grew out from them. Upon passaging, spindle cells increased further, and by the fifth passage, these cells occupied the entire culture. These cells (iPS-PDLs) expressed genes such as periostin and Asporin/PLAP1, and their comprehensive gene expression patterns resembled those of PDL cells. iPS-PDL cells exhibited a cell surface antigen profile of CD90+, CD73+, CD105+, CD44+, CD29+, CD14-, CD34-, CD45-, and CD19- and differentiation potential into osteoblasts, adipocytes, and chondrocytes. Transplantation of iPS-PDLs into rat periodontal defects increased the height of newly formed bone and enhanced periodontal tissue regeneration after 4 weeks. Our results showed that iPSCs differentiated into cells with properties similar to those of PDL cells in the presence of humoral factors of cultured PDL cells. Additionally, the transplantation of iPS-PDL cells into periodontal defects induces periodontal tissue regeneration. These findings provide valuable insights for developing novel periodontal regenerative therapies using iPSCs.
Evaluating Seeding Density Effects on Cardiac Organoid Health and Functionality for Toxicity Studies
Development of relevant human induced pluripotent stem cell-derived cardiac organoids is essential to recapitulate myocardium physiology and functionality for the assessment of drug-induced toxicity evaluations. However, the optimal conditions for culturing self-aggregating multicellular cardiac organoids are not well-elucidated, particularly the impact of noncardiomyocytes. In this study, we generated cardiac organoids at varying seeding densities to formulate organoids that meet or exceed the biological diffusion limit. We assessed their morphology, gene expression profiles, beating functionality, viability, and mitochondrial activity over time. Our results show that organoid sizes stabilize by 7 days of culture, regardless of seeding density. However, organoids seeded with 20,000 cells retained a more optimal cardiac signature that promotes cardiac maturity and minimizes fibrotic tendencies, especially when cultured for longer than 7 days. While all organoid populations maintained their beating functionalities, those seeded with 80,000 cells exhibited greater cell shedding and increased apoptosis at long-term culture. In contrast, minimal apoptosis was observed in organoids seeded with 20,000 cells after 7 days. Mitochondrial staining further revealed that organoids seeded with 20,000 cells consistently demonstrated higher metabolic activity. Taken together, organoids seeded with 20,000 cells and cultured for 7 days yielded the healthiest morphology, transcriptional signature, and viability while maintaining robust beating kinetics. Importantly, the organoid model identified in this study demonstrated a selectivity index (SI) that is over an order of magnitude larger than that of two-dimensional cultures, showing improved sensitivity to clinically relevant doxorubicin-induced cardiotoxicity, enabling more accurate dose-response evaluations that better reflect therapeutic conditions.
and Evaluation of a Bovine Collagen Matrix for Acute Rotator Cuff Tear Repair
Full-thickness rotator cuff tears (RCTs) represent a musculoskeletal damage that severely affects shoulder function and quality of life. Current surgical interventions are hindered by limited regenerative capacity of rotator cuff repair implants and high retear rates postoperatively. In this study, we investigated a tendon repair matrix (TRM) product prepared from bovine tendon collagen. The TRM was designed as a regenerative scaffold to improve the healing of damaged rotator cuff. results showed excellent cytocompatibility of TRM, with significantly enhanced adhesion, proliferation, and spreading of bone marrow stromal cells and tenocyte-like mouse tendon precursor cells,mouse tendon-derived cell line, clone D6 (TT-D6) cells (mouse tendon-derived cell line, clone D6). In a rabbit model of acute full-thickness supraspinatus tendon tear, TRM promoted type I collagen deposition, improved interface tissue formation, and enhanced tendon-to-bone integration. Furthermore, biomechanical test results revealed load-bearing capacity of the TRM group compared with both the empty and native tissue control groups. These findings support the clinical potential of TRM as a regenerative scaffold for the functional reconstruction of RCTs. Impact Statement This study addresses a critical clinical need in sports medicine by evaluating a novel bovine collagen-based tendon repair matrix (TRM) for the repair of acute full-thickness rotator cuff tears (RCTs). The TRM exhibited excellent biocompatibility and significantly enhanced tendon-to-bone healing, as demonstrated by improved fibrocartilaginous tissue formation and biomechanical strength in a rabbit model. These promising results underscore TRM's potential to reduce postoperative retear rates by promoting effective regeneration of the tendon-bone interface. Consequently, this research represents an important advancement toward improving clinical outcomes for RCT patients, offering substantial potential for translation into clinical practice.
Development of a 3D Wound Healing Model to Assess the Effect of ADSC-EVs on Vascularization
Angiogenesis is critical for effective wound healing and relies on the successful coordination of various cell types, including endothelial cells, macrophages, and fibroblasts. Adipose-derived stem cell extracellular vesicles (ADSC-EVs) have demonstrated proangiogenic properties and have been posited as a novel therapeutic to aid wound healing; however, their functional impact within human-derived multicellular models remains largely uncharacterized. This study explores the development and application of a 3D multicellular model to assess the effects of ADSC-EVs on vascularization in the context of wound healing. 3D multicellular models were developed by coculturing human umbilical vein endothelial cells (HUVECs), monocyte-derived macrophages, and fibroblasts within Matrigel to recapitulate the wound healing microenvironment. A five-color confocal microscopy panel was developed to visualize each cell type and EVs within the models. The optimized models were then treated with ADSC-EVs or control to determine their impact on angiogenesis and cell colocalization. We determined that vessel formation was significantly enhanced when HUVECs were cocultured in multicellular models compared with monocultures, with the greatest effect observed in the full three-cell-type model. This effect was even more pronounced with the addition of ADSC-EVs. ADSC-EV treatment also enhanced macrophage colocalization within endothelial structures. This study developed a multicellular model that can be used for future work assessing wound healing and will be additive to currently used single-cell and models. We have applied these models to demonstrate that ADSC-EVs significantly enhance tube formation in HUVECs and the development of tissue-like structures in multicell systems, highlighting their potential as a promising therapeutic approach for improving wound healing.
Regenerative Healing
Indigenous health and wellness encompasses physical, mental, emotional, and spiritual well-being, with a focus on "the interconnectedness of these aspects and the importance of community and cultural practices." "Regenerative healing," as distinct from "Regenerative medicine," is a similarly wholistic term that has emerged from conversations with selected First Nations and Métis Knowledge Holders from across Canada. Impact Statement Building trust with patients and the broader public who support health and medical research requires continuous engagement with the public. "Regenerative Healing" may be a more welcoming and more humble framework to launch the conversation.
Potential of Low-Dose Carbon Monoxide in Promoting Osseointegration
Successful osseointegration is crucial for dental implant stability, yet it remains challenging due to adverse local microenvironments, particularly infection and inflammation. While carbon monoxide (CO) has been recognized as a promising gaseous signaling molecule with diverse therapeutic properties, its clinical application faces significant limitations due to dose control challenges. To address this issue, we developed a polyetheretherketone (PEEK)-based photo-responsive implant system with surface-immobilized manganese carbonyl nanocrystals, enabling precisely controlled near-infrared light-triggered CO release. The system demonstrated efficient photoresponsiveness, achieving 13.83 ± 1.16 μM CO release within 10 min under optimal illumination conditions. studies revealed that low-dose CO significantly enhanced bone marrow mesenchymal stem cell osteogenic differentiation with upregulated expression of key markers, including Runx2, ALP, and OCN. In a rat femoral defect model, implants with controlled CO release exhibited significantly improved osseointegration. Comprehensive biosafety assessments confirmed the system's excellent biocompatibility without detectable organ toxicity. This research provides compelling evidence for controlled low-dose CO as an innovative strategy to enhance osseointegration, offering new possibilities for dental and orthopedic implant development, particularly for challenging clinical scenarios with compromised bone healing. Impact Statement This study introduces a novel approach for improving implant osseointegration through controlled carbon monoxide delivery, potentially offering a new strategy for enhancing the success rate of dental implant procedures.
Tissue-Engineered Endothelial Keratoplasty with Controlled Cell Density: Toward Super TEEKs
Over the past 20 years, endothelial keratoplasty procedures have revolutionized the treatment of corneal endothelial disorders. These conditions have now become the leading indication for corneal transplantation in Western countries and account for half of all donor cornea usage. Despite their undeniable success, the global shortage of donor tissues and major disparities between nations justify the development of alternatives to donor grafts. Cell therapy using injections of suspended endothelial cells has proven effective, and tissue-engineered endothelial keratoplasty (TEEK), comprising a membrane coated with cultured endothelial cells, is under development to better mimic the native endothelial graft. Our team utilizes a femtosecond-laser-cut lens capsule disc as a bioengineering scaffold, taking advantage of this novel tissue's biocompatibility, transparency, curvature, and availability. In the present study, we provide proof of concept, in 12 TEEKs, that it is possible to control the final endothelial cell density (ECD) by varying the seeding density per mm. Cell characterization was performed through morphometric analysis of the endothelial mosaic stained with anti-NCAM (a lateral membrane marker used as a differentiation marker), using the CellPose artificial intelligence algorithm specifically trained for endothelium segmentation. Five criteria related to pleomorphism, polymorphism, and elongation were combined into a single endothelial quality score. The median cell viability at 28 days of culture, assessed by Hoechst 33342 and Calcein-AM staining, reached 98% (range: 83-99%). The median viable ECD (number of live cells per surface unit) in the highest-density group was 3.245 cells/mm (range: 2.778-3.753), paving the way for the bioengineering of supra-physiological TEEKs, or "super TEEKs".
LINC00687 Regulates PRDX2 Expression in High Glucose-Induced Nonunion after Digital Replantation
Diabetic nonunion is a major clinical challenge with unclear molecular mechanisms. This study systematically investigated the key genes and molecular mechanisms of bone nonunion after finger replantation induced by high glucose using Gene Expression Omnibus (GEO), bioinformatics, and experimental analyses. In total, 179 differentially expressed mRNAs and one lncRNA (DElncRNA) were identified using the GEO dataset. Functional enrichment analysis showed that these genes were mainly involved in the regulation of autophagy and metabolism. Protein-protein interaction network analysis identified five core genes (Peroxiredoxin 2 [PRDX2], FK506 binding protein 8 [FKBP8], SHANK-associated RH domain interactor [SHARPIN], WD repeat domain 45 [WDR45], and gamma-aminobutyric acid type A receptor-associated protein like 2 [GABARAPL2]), three of which exhibited good binding affinities for potential therapeutic agents. Immune infiltration analysis revealed significant differences in the CD8+ T cell proportions between nonunion and healthy samples. We constructed a competitive endogenous RNA network (long intergenic non-protein coding RNA 687 [LINC00687]-miR-4443-PRDX2) and verified its direct regulatory interaction using a dual-luciferase reporter assay. FKBP8, PRDX2, SHARPIN, WDR45, and GABARAPL2 were overexpressed in tissue samples from patients with type 2 diabetes mellitus fracture nonunion. Animal experiments further confirmed that LINC00687 upregulated PRDX2 expression by sponging miR-4443 in a hyperglycemic environment, thereby inhibiting bone healing. This study not only identified PRDX2 and other genes as potential biomarkers of diabetic nonunion but also clarified the regulatory role of the LINC00687/miR-4443/PRDX2 axis in hyperglycemia-induced nonunion, providing a new molecular target for clinical prevention and treatment. Impact Statement 1. PRDX2, KBP8, SHARPIN, WDR45, and GABARAPL2 were potential biomarkers for this study. 2. LINC00687-miR-4443-PRDX2 participated in high glucose-induced nonunion in this study. 3. Autophagy process and metabolic pathways contribute to the progression in this study.
