Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (156)

Search Parameters:
Keywords = multifunctional biomaterials

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
32 pages, 2710 KiB  
Review
Polyphosphazene-Based Nanotherapeutics
by Sara Gutierrez-Gutierrez, Rocio Mellid-Carballal, Noemi Csaba and Marcos Garcia-Fuentes
J. Funct. Biomater. 2025, 16(8), 285; https://doi.org/10.3390/jfb16080285 - 2 Aug 2025
Viewed by 223
Abstract
Poly(organo)phosphazenes (PPZs) are increasingly recognized as versatile biomaterials for drug delivery applications in nanomedicine. Their unique hybrid structure—featuring an inorganic backbone and highly tunable organic side chains—confers exceptional biocompatibility and adaptability. Through precise synthetic methodologies, PPZs can be engineered to exhibit a wide [...] Read more.
Poly(organo)phosphazenes (PPZs) are increasingly recognized as versatile biomaterials for drug delivery applications in nanomedicine. Their unique hybrid structure—featuring an inorganic backbone and highly tunable organic side chains—confers exceptional biocompatibility and adaptability. Through precise synthetic methodologies, PPZs can be engineered to exhibit a wide spectrum of functional properties, including the formation of multifunctional nanostructures tailored for specific therapeutic needs. These attributes enable PPZs to address several critical challenges associated with conventional drug delivery systems, such as poor pharmacokinetics and pharmacodynamics. By modulating solubility profiles, enhancing drug stability, enabling targeted delivery, and supporting controlled release, PPZs offer a robust platform for improving therapeutic efficacy and patient outcomes. This review explores the fundamental chemistry, biopharmaceutical characteristics, and biomedical applications of PPZs, particularly emphasizing their role in zero-dimensional nanotherapeutic systems, including various nanoparticle formulations. PPZ-based nanotherapeutics are further examined based on their drug-loading mechanisms, which include electrostatic complexation in polyelectrolytic systems, self-assembly in amphiphilic constructs, and covalent conjugation with active pharmaceutical agents. Together, these strategies underscore the potential of PPZs as a next-generation material for advanced drug delivery platforms. Full article
(This article belongs to the Special Issue Nanomaterials for Drug Targeting and Drug Delivery (2nd Edition))
Show Figures

Graphical abstract

25 pages, 9193 KiB  
Article
Antibiotic-Loaded Bioglass 45S5 for the Treatment and Prevention of Staphylococcus aureus Infections in Orthopaedic Surgery: A Novel Strategy Against Antimicrobial Resistance
by Humera Sarwar, Richard A. Martin, Heather M. Coleman, Aaron Courtenay and Deborah Lowry
Pathogens 2025, 14(8), 760; https://doi.org/10.3390/pathogens14080760 (registering DOI) - 1 Aug 2025
Viewed by 227
Abstract
This study explores the potential of biodegradable Bioglass 45S5 formulations as a dual-function approach for preventing and treating Staphylococcus aureus infections in orthopaedic surgery while addressing the growing concern of antimicrobial resistance (AMR). The research focuses on the development and characterisation of antibiotic-loaded [...] Read more.
This study explores the potential of biodegradable Bioglass 45S5 formulations as a dual-function approach for preventing and treating Staphylococcus aureus infections in orthopaedic surgery while addressing the growing concern of antimicrobial resistance (AMR). The research focuses on the development and characterisation of antibiotic-loaded BG45S5 formulations, assessing parameters such as drug loading efficiency, release kinetics, antimicrobial efficacy, and dissolution behaviour. Key findings indicate that the F2l-BG45S5-T-T-1.5 and F2l-BG45S5-T-V-1.5 formulations demonstrated controlled antibiotic release for up to seven days, with size distributions of D(10): 7.11 ± 0.806 µm, 4.96 ± 0.007 µm; D(50): 25.34 ± 1.730 µm, 25.20.7 ± 0.425 µm; and D(90): 53.7 ± 7.95 µm, 56.10 ± 0.579 µm, respectively. These formulations facilitated hydroxyapatite formation on their surfaces, indicative of osteogenic potential. The antimicrobial assessments revealed zones of inhibition against methicillin-susceptible Staphylococcus aureus (MSSA, ATCC-6538) measuring 20.3 ± 1.44 mm and 24.6 ± 1.32 mm, while for methicillin-resistant Staphylococcus aureus (MRSA, ATCC-43300), the inhibition zones were 21.6 ± 1.89 mm and 22 ± 0.28 mm, respectively. Time-kill assay results showed complete bacterial eradication within eight hours. Additionally, biocompatibility testing via MTT assay confirmed cell viability of >75%. In conclusion, these findings highlight the promise of antibiotic-loaded BG45S5 as a multifunctional biomaterial capable of both combating bone infections and supporting bone regeneration. These promising results suggest that in vivo studies should be undertaken to expedite these materials into clinical applications. Full article
(This article belongs to the Special Issue Antimicrobial Resistance in the Post-COVID Era: A Silent Pandemic)
Show Figures

Figure 1

52 pages, 4770 KiB  
Review
Biomaterial-Based Nucleic Acid Delivery Systems for In Situ Tissue Engineering and Regenerative Medicine
by Qi-Xiang Wu, Natalia De Isla and Lei Zhang
Int. J. Mol. Sci. 2025, 26(15), 7384; https://doi.org/10.3390/ijms26157384 - 30 Jul 2025
Viewed by 438
Abstract
Gene therapy is a groundbreaking strategy in regenerative medicine, enabling precise cellular behavior modulation for tissue repair. In situ nucleic acid delivery systems aim to directly deliver nucleic acids to target cells or tissues to realize localized genetic reprogramming and avoid issues like [...] Read more.
Gene therapy is a groundbreaking strategy in regenerative medicine, enabling precise cellular behavior modulation for tissue repair. In situ nucleic acid delivery systems aim to directly deliver nucleic acids to target cells or tissues to realize localized genetic reprogramming and avoid issues like donor cell dependency and immune rejection. The key to success relies on biomaterial-engineered delivery platforms that ensure tissue-specific targeting and efficient intracellular transport. Viral vectors and non-viral carriers are strategically modified to enhance nucleic acid stability and cellular uptake, and integrate them into injectable or 3D-printed scaffolds. These scaffolds not only control nucleic acid release but also mimic native extracellular microenvironments to support stem cell recruitment and tissue regeneration. This review explores three key aspects: the mechanisms of gene editing in tissue repair; advancements in viral and non-viral vector engineering; and innovations in biomaterial scaffolds, including stimuli-responsive hydrogels and 3D-printed matrices. We evaluate scaffold fabrication methodologies, nucleic acid loading–release kinetics, and their biological impacts. Despite progress in spatiotemporal gene delivery control, challenges remain in balancing vector biocompatibility, manufacturing scalability, and long-term safety. Future research should focus on multifunctional “smart” scaffolds with CRISPR-based editing tools, multi-stimuli responsiveness, and patient-specific designs. This work systematically integrates the latest methodological advances, outlines actionable strategies for future investigations and advances clinical translation perspectives beyond the existing literature. Full article
(This article belongs to the Section Materials Science)
Show Figures

Figure 1

33 pages, 2265 KiB  
Review
From Sea to Therapy: Marine Biomaterials for Drug Delivery and Wound Healing
by Mansi Chilwant, Valentina Paganini, Mariacristina Di Gangi, Sofia Gisella Brignone, Patrizia Chetoni, Susi Burgalassi, Daniela Monti and Silvia Tampucci
Pharmaceuticals 2025, 18(8), 1093; https://doi.org/10.3390/ph18081093 - 23 Jul 2025
Viewed by 516
Abstract
Marine biomass represents a valuable yet underexploited resource for the development of high-value biomaterials. Recent advances have highlighted the significant potential of marine-derived polysaccharides, proteins, and peptides in biomedical applications, most notably in drug delivery and wound healing. This review provides a comprehensive [...] Read more.
Marine biomass represents a valuable yet underexploited resource for the development of high-value biomaterials. Recent advances have highlighted the significant potential of marine-derived polysaccharides, proteins, and peptides in biomedical applications, most notably in drug delivery and wound healing. This review provides a comprehensive synthesis of current research on the extraction, processing and pharmaceutical valorization of these biopolymers, with a focus on their structural and functional properties that allow these materials to be engineered into nanocarriers, hydrogels, scaffolds, and smart composites. Key fabrication strategies such as ionic gelation, desolvation, and 3D bioprinting are critically examined for their role in drug encapsulation, release modulation, and scaffold design for regenerative therapies. The review also covers preclinical validation, scale-up challenges, and relevant regulatory frameworks, offering a practical roadmap from sustainable sourcing to clinical application. Special attention is given to emerging technologies, including stimuli-responsive biomaterials and biosensor-integrated wound dressings, as well as to the ethical and environmental implications of marine biopolymer sourcing. By integrating materials science, pharmaceutical technology and regulatory insight, this review aims to provide a multidisciplinary perspective for researchers and industrial stakeholders seeking sustainable and multifunctional pharmaceutical platforms for precision medicine and regenerative therapeutics. Full article
(This article belongs to the Collection Feature Review Collection in Pharmaceutical Technology)
Show Figures

Graphical abstract

35 pages, 1038 KiB  
Review
Hydrogels in Cardiac Surgery: Versatile Platforms for Tissue Repair, Adhesion Prevention, and Localized Therapeutics
by Seok Beom Hong, Jin-Oh Jeong and Hoon Choi
Gels 2025, 11(7), 564; https://doi.org/10.3390/gels11070564 - 21 Jul 2025
Viewed by 494
Abstract
Hydrogels have emerged as multifunctional biomaterials in cardiac surgery, offering promising solutions for myocardial regeneration, adhesion prevention, valve engineering, and localized drug and gene delivery. Their high water content, biocompatibility, and mechanical tunability enable close emulation of the cardiac extracellular matrix, supporting cellular [...] Read more.
Hydrogels have emerged as multifunctional biomaterials in cardiac surgery, offering promising solutions for myocardial regeneration, adhesion prevention, valve engineering, and localized drug and gene delivery. Their high water content, biocompatibility, and mechanical tunability enable close emulation of the cardiac extracellular matrix, supporting cellular viability and integration under dynamic physiological conditions. In myocardial repair, injectable and patch-forming hydrogels have been shown to be effective in reducing infarct size, promoting angiogenesis, and preserving contractile function. Hydrogel coatings and films have been designed as adhesion barriers to minimize pericardial adhesions after cardiotomy and improve reoperative safety. In heart valve and patch engineering, hydrogels contribute to scaffold design by providing bio-instructive, mechanically resilient, and printable matrices that are compatible with 3D fabrication. Furthermore, hydrogels serve as localized delivery platforms for small molecules, proteins, and nucleic acids, enabling sustained or stimuli-responsive release while minimizing systemic toxicity. Despite these advances, challenges such as mechanical durability, immune compatibility, and translational scalability persist. Ongoing innovations in smart polymer chemistry, hybrid composite design, and patient-specific manufacturing are addressing these limitations. This review aims to provide an integrated perspective on the application of hydrogels in cardiac surgery. The relevant literature was identified through a narrative search of PubMed, Scopus, Web of Science, Embase, and Google Scholar. Taken together, hydrogels offer a uniquely versatile and clinically translatable platform for addressing the multifaceted challenges of cardiac surgery. Hydrogels are poised to redefine clinical strategies in cardiac surgery by enabling tailored, bioresponsive, and functionally integrated therapies. Full article
(This article belongs to the Special Issue Recent Advances in Hydrogels for Tissue Engineering Applications)
Show Figures

Figure 1

24 pages, 3541 KiB  
Review
Towards Intelligent Wound Care: Hydrogel-Based Wearable Monitoring and Therapeutic Platforms
by Yan Niu, Ziyao Zhao, Lihong Yang, Dan Lv, Rui Sun, Ting Zhang, Yuhan Li, Qianqian Bao, Mingqing Zhang, Lanzhong Wang, Wei Yan, Fei Han and Biwei Yan
Polymers 2025, 17(13), 1881; https://doi.org/10.3390/polym17131881 - 6 Jul 2025
Viewed by 1000
Abstract
Chronic wounds present clinical challenges due to persistent inflammation, infection, and dysregulated tissue repair, often exacerbated by the passive nature of conventional wound dressings. Recent advancements in hydrogel-based wearable technologies have transformed these biomaterials into multifunctional platforms capable of integrating real-time monitoring and [...] Read more.
Chronic wounds present clinical challenges due to persistent inflammation, infection, and dysregulated tissue repair, often exacerbated by the passive nature of conventional wound dressings. Recent advancements in hydrogel-based wearable technologies have transformed these biomaterials into multifunctional platforms capable of integrating real-time monitoring and targeted therapy, ushering in a new era of intelligent wound care. In this review, we show innovative diagnostic and therapeutic strategies, including wound-monitoring devices and multifunctional healing-promoted platforms, highlighting integrated closed-loop systems that dynamically adapt treatments to wound microenvironments, thus merging diagnostics and therapeutics. Challenges in fabrication engineering and clinical application are discussed, alongside emerging trends like AI-driven analytics and 3D-bioprinted technology. By bridging fragmented research, this work underscores the potential of hydrogels to enable intelligent wound management. Full article
(This article belongs to the Special Issue New Progress in the Polymer-Based Biomaterials)
Show Figures

Figure 1

26 pages, 905 KiB  
Review
Advancements in Antimicrobial Surface Coatings Using Metal/Metaloxide Nanoparticles, Antibiotics, and Phytochemicals
by Preetha Ebenezer, S. P. S. N. Buddhika Sampath Kumara, S. W. M. A. Ishantha Senevirathne, Laura J. Bray, Phurpa Wangchuk, Asha Mathew and Prasad K. D. V. Yarlagadda
Nanomaterials 2025, 15(13), 1023; https://doi.org/10.3390/nano15131023 - 1 Jul 2025
Viewed by 430
Abstract
The growing prevalence of bacterial infections and the alarming rise of antimicrobial resistance (AMR) have driven the need for innovative antimicrobial coatings for medical implants and biomaterials. However, implant surface properties, such as roughness, chemistry, and reactivity, critically influence biological interactions and must [...] Read more.
The growing prevalence of bacterial infections and the alarming rise of antimicrobial resistance (AMR) have driven the need for innovative antimicrobial coatings for medical implants and biomaterials. However, implant surface properties, such as roughness, chemistry, and reactivity, critically influence biological interactions and must be engineered to ensure biocompatibility, corrosion resistance, and sustained antibacterial activity. This review evaluates three principal categories of antimicrobial agents utilized in surface functionalization: metal/metaloxide nanoparticles, antibiotics, and phytochemical compounds. Metal/metaloxide-based coatings, especially those incorporating silver (Ag), zinc oxide (ZnO), and copper oxide (CuO), offer broad-spectrum antimicrobial efficacy through mechanisms such as reactive oxygen species (ROS) generation and bacterial membrane disruption, with a reduced risk of resistance development. Antibiotic-based coatings enable localized drug delivery but often face limitations related to burst release, cytotoxicity, and diminishing effectiveness against multidrug-resistant (MDR) strains. In contrast, phytochemical-derived coatings—using bioactive plant compounds such as curcumin, eugenol, and quercetin—present a promising, biocompatible, and sustainable alternative. These agents not only exhibit antimicrobial properties but also provide anti-inflammatory, antioxidant, and osteogenic benefits, making them multifunctional tools for implant surface modification. The integration of these antimicrobial strategies aims to reduce bacterial adhesion, inhibit biofilm formation, and enhance tissue regeneration. By leveraging the synergistic effects of metal/metaloxide nanoparticles, antibiotics, and phytochemicals, next-generation implant coatings hold the potential to significantly improve infection control and clinical outcomes in implant-based therapies. Full article
(This article belongs to the Special Issue Nanocoating for Antibacterial Applications)
Show Figures

Graphical abstract

14 pages, 2508 KiB  
Article
Enhancement of Efficiency in an Ex Situ Coprecipitation Method for Superparamagnetic Bacterial Cellulose Hybrid Materials
by Thaís Cavalcante de Souza, Italo José Batista Durval, Hugo Moraes Meira, Andréa Fernanda de Santana Costa, Eduardo Padrón Hernández, Attilio Converti, Glória Maria Vinhas and Leonie Asfora Sarubbo
Membranes 2025, 15(7), 198; https://doi.org/10.3390/membranes15070198 - 1 Jul 2025
Viewed by 465
Abstract
Superparamagnetic magnetite nanoparticles (Fe3O4) have garnered considerable interest due to their unique magnetic properties and potential for integration into multifunctional biomaterials. In particular, their incorporation into bacterial cellulose (BC) matrices offers a promising route for developing sustainable and high-performance [...] Read more.
Superparamagnetic magnetite nanoparticles (Fe3O4) have garnered considerable interest due to their unique magnetic properties and potential for integration into multifunctional biomaterials. In particular, their incorporation into bacterial cellulose (BC) matrices offers a promising route for developing sustainable and high-performance magnetic composites. Numerous studies have explored BC-magnetite systems; however, innovations combining ex situ coprecipitation synthesis within BC matrices, tailored reagent molar ratios, stirring protocols, and purification processes remain limited. This study aimed to optimize the ex situ coprecipitation method for synthesizing superparamagnetic magnetite nanoparticles embedded in BC membranes, focusing on enhancing particle stability and crystallinity. BC membranes containing varying concentrations of magnetite (40%, 50%, 60%, and 70%) were characterized using X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), and vibrating sample magnetometry (VSM). The resulting magnetic BC membranes demonstrated homogenous dispersion of nanoparticles, improved crystallite size (6.96 nm), and enhanced magnetic saturation (Ms) (50.4 emu/g), compared to previously reported methods. The adoption and synergistic optimization of synthesis parameters—unique to this study—conferred greater control over the physicochemical and magnetic properties of the composites. These findings position the optimized BC-magnetite nanocomposites as highly promising candidates for advanced applications, including electromagnetic interference (EMI) shielding, electronic devices, gas sensors, MRI contrast agents, and targeted drug delivery systems. Full article
(This article belongs to the Section Membrane Fabrication and Characterization)
Show Figures

Figure 1

22 pages, 1884 KiB  
Review
Melatonin/Chitosan Biomaterials for Wound Healing and Beyond: A Multifunctional Therapeutic Approach
by Karolina Kulka-Kamińska, Patrycja Brudzyńska, Mayuko Okura, Tatsuyuki Ishii, Marco Skala, Russel J. Reiter, Andrzej T. Slominski, Kazuo Kishi, Kerstin Steinbrink, Alina Sionkowska and Konrad Kleszczyński
Int. J. Mol. Sci. 2025, 26(13), 5918; https://doi.org/10.3390/ijms26135918 - 20 Jun 2025
Viewed by 875
Abstract
Chitosan is increasingly utilized in combination with melatonin in novel formulations for a wide range of therapeutic applications. As a biocompatible and biodegradable polymer, chitosan exhibits notable properties, including antioxidant, antimicrobial, moisturizing, and absorption capabilities, in addition to a high potential for chemical [...] Read more.
Chitosan is increasingly utilized in combination with melatonin in novel formulations for a wide range of therapeutic applications. As a biocompatible and biodegradable polymer, chitosan exhibits notable properties, including antioxidant, antimicrobial, moisturizing, and absorption capabilities, in addition to a high potential for chemical modification due to its functional groups. These characteristics make it a valuable material in biomedical, pharmaceutical, cosmetic, food packaging, and environmental applications. Melatonin, an indoleamine primarily synthesized in the pineal gland but also found in various peripheral organs and in diverse organisms—including plants, bacteria, and fungi—has been extensively investigated for its antioxidant, anti-apoptotic, and anti-inflammatory activities, as well as its roles in immunomodulation, mitochondrial function, and melanin biosynthesis. This review summarizes recent advances in the combined use of chitosan and melatonin, with emphasis on their synergistic effects in wound healing, anti-cancer therapies, tissue engineering (i.e., skin and bone regeneration), and drug delivery systems. Additional potential applications are discussed in the context of cosmetology, aesthetic medicine, and veterinary practice. Full article
(This article belongs to the Special Issue The Chitosan Biomaterials: Advances and Challenges—2nd Edition)
Show Figures

Graphical abstract

25 pages, 2568 KiB  
Review
Emerging Multifunctional Biomaterials for Addressing Drug Resistance in Cancer
by Mohamed El-Tanani, Syed Arman Rabbani, Rasha Babiker, Yahia El-Tanani, Shakta Mani Satyam and Thantrira Porntaveetus
Biology 2025, 14(5), 497; https://doi.org/10.3390/biology14050497 - 2 May 2025
Cited by 1 | Viewed by 887
Abstract
Drug resistance remains a major barrier to effective cancer treatment, contributing to poor patient outcomes. Multifunctional biomaterials integrating electrical and catalytic properties offer a transformative strategy to target diverse resistance mechanisms. This review explores their ability to modulate cellular processes, remodel the tumor [...] Read more.
Drug resistance remains a major barrier to effective cancer treatment, contributing to poor patient outcomes. Multifunctional biomaterials integrating electrical and catalytic properties offer a transformative strategy to target diverse resistance mechanisms. This review explores their ability to modulate cellular processes, remodel the tumor microenvironment (TME), and enhance drug delivery. Electrically active biomaterials enhance drug uptake and apoptotic sensitivity by altering membrane potentials, ion channels, and intracellular signaling, synergizing with chemotherapy. Catalytic biomaterials generate reactive oxygen species (ROS), activate prodrugs, reprogram hypoxic and acidic TME, and degrade the extracellular matrix (ECM) to improve drug penetration. Hybrid nanomaterials (e.g., conductive hydrogels, electrocatalytic nanoparticles), synergize electrical and catalytic properties for localized, stimuli-responsive therapy and targeted drug release, minimizing systemic toxicity. Despite challenges in biocompatibility and scalability, future integration with immunotherapy, personalized medicine, and intelligent self-adaptive systems capable of real-time tumor response promises to accelerate clinical translation. The development of these adaptive biomaterials, alongside advancements in nanotechnology and AI-driven platforms, represents the next frontier in precision oncology. This review highlights the potential of multifunctional biomaterials to revolutionize cancer therapy by addressing multidrug resistance at cellular, genetic, and microenvironmental levels, offering a roadmap to improve therapeutic outcomes and reshape oncology practice. Full article
Show Figures

Figure 1

22 pages, 2398 KiB  
Review
Current State of Knowledge Regarding the Treatment of Cranial Bone Defects: An Overview
by Jagoda Kurowiak, Krystian Piesik and Tomasz Klekiel
Materials 2025, 18(9), 2021; https://doi.org/10.3390/ma18092021 - 29 Apr 2025
Viewed by 1030
Abstract
In this article, an analysis of the problem of treating bone defects using cranial bone disorders as an example is presented. The study was performed in the context of the development of various implant biomaterials used to fill bone defects. An analysis of [...] Read more.
In this article, an analysis of the problem of treating bone defects using cranial bone disorders as an example is presented. The study was performed in the context of the development of various implant biomaterials used to fill bone defects. An analysis of the requirements for modern materials is undertaken, indicating the need for their further development. The article focuses particular attention on these biomaterial properties, which have an influence on bioresorbability and promote osteointegration and bone growth. The analysis showed the need for further development of biomaterials, the characteristics of which may be multifunctionality. Multifunctional scaffolds are those that simultaneously fill and stabilize the defect and contribute to the proper process of regeneration and reconstruction of cranial bones. Due to the complex structure of the skull and special protective functions, there is a need to develop innovative implants. Implants with complex geometries can be successfully manufactured using additive technologies. Full article
(This article belongs to the Special Issue Advances in Implant Materials and Biocompatibility)
Show Figures

Figure 1

21 pages, 4834 KiB  
Article
A Multifunctional PEEK Composite Scaffold with Immunomodulatory, Angiogenic, and Osteogenic Properties for Enhanced Bone Regeneration
by Mengen Zhao, Han Yang, Qianwen Yang, Chao Zhang, Jie Liu, Zhaoying Wu, Lijun Wang, Wei Zhang, Bing Wang and Wenliang Liu
Polymers 2025, 17(9), 1206; https://doi.org/10.3390/polym17091206 - 28 Apr 2025
Viewed by 546
Abstract
Polyetheretherketone (PEEK) is a widely used material in bone tissue engineering due to its favorable mechanical properties and radiolucency. However, its bioinert nature and lack of osteogenic activity restrict its ability to support effective bone regeneration. In this study, a novel APS-coated plasma-treated [...] Read more.
Polyetheretherketone (PEEK) is a widely used material in bone tissue engineering due to its favorable mechanical properties and radiolucency. However, its bioinert nature and lack of osteogenic activity restrict its ability to support effective bone regeneration. In this study, a novel APS-coated plasma-treated sulfonated bioactive PEEK scaffold (APS/PSBPK) was developed to overcome these limitations. The scaffold integrates strontium-doped bioactive glass (SrBG) to enhance biocompatibility and osteogenic potential, while astragalus polysaccharide (APS) was incorporated via plasma cleaning to modulate immune responses and promote vascularization. In vitro studies demonstrated that the APS/PSBPK scaffold facilitates M2 macrophage polarization, reduces pro-inflammatory cytokines, and enhances the secretion of anti-inflammatory factors. It also promotes endothelial cell migration and angiogenesis while supporting the adhesion, proliferation, and osteogenic differentiation of rBMSCs. In vivo experiments revealed that the scaffold effectively regulates the immune microenvironment, promotes vascularization, and accelerates bone regeneration. Thus, the APS/PSBPK composite scaffold serves as a multifunctional biomaterial with significant potential for applications in bone repair and regeneration by combining immunomodulation, angiogenesis, and osteogenesis. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Graphical abstract

22 pages, 25653 KiB  
Article
Synergistic Antioxidant and Anti-Ferroptosis Therapy via BPNS-Encapsulated Thermoresponsive Chitosan Hydrogel for Spinal Cord Injury Regeneration
by Yang Liu, Yingkai Wang, Xiangzi Wang, Wanchen Zeng, Zehong Zhang, Zhengmian Zhang and Zhongquan Qi
Pharmaceutics 2025, 17(5), 573; https://doi.org/10.3390/pharmaceutics17050573 - 26 Apr 2025
Viewed by 737
Abstract
Background: Spinal cord injury (SCI) is a devastating neurological condition with limited therapeutic options. Current clinical interventions predominantly rely on prolonged or high-dose pharmacological regimens, often causing systemic toxicity and adverse events. Although black phosphorus nanosheets (BPNSs) exhibit remarkable reactive oxygen species [...] Read more.
Background: Spinal cord injury (SCI) is a devastating neurological condition with limited therapeutic options. Current clinical interventions predominantly rely on prolonged or high-dose pharmacological regimens, often causing systemic toxicity and adverse events. Although black phosphorus nanosheets (BPNSs) exhibit remarkable reactive oxygen species (ROS)-scavenging capacity to mitigate oxidative damage, their rapid degradation severely compromises their therapeutic efficacy. Methods: This study presents a thermosensitive hydrogel with rapid gelation properties by incorporating different proportions and concentrations of sodium alginate (SA) into a chitosan/β-glycerophosphate (CS/β-GP) hydrogel and loading it with BPNS for the treatment of SCI in rats. In vitro, the physical properties of the composite were characterized and the cytotoxicity and ROS scavenging abilities were assessed using PC12 cells; in vivo, behavioral tests, histopathological analysis, transcriptomics, immunohistochemistry, and Western blotting were performed to explore the therapeutic effects and mechanisms. Results: The results demonstrate that this hydrogel effectively slows BPNS degradation, exhibits a high ROS scavenging capacity, reduces lipid peroxidation, and thereby inhibits ferroptosis and apoptosis, offering neuroprotective effects and promoting motor function recovery. Conclusions: Our findings establish the CS/β-GP/SA-BPNS hydrogel as a multifunctional therapeutic platform for SCI, synergizing sustained drug release with ROS–ferroptosis–apoptosis axis modulation to achieve neuroprotection and functional restoration. This strategy provides a translatable paradigm for combining nanotechnology and biomaterial engineering in neural repair. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

18 pages, 4822 KiB  
Article
Co-Encapsulation of Phycocyanin and Albumin-Bound Curcumin in Biopolymeric Hydrogels
by Konstantina Matskou, Ilias Matis, Sotiria Demisli, Konstantinos Rigkos, Eirini Karandrea, Kalliopi Kourioti, Georgios Sotiroudis, Vasiliki Pletsa, Aristotelis Xenakis and Maria Zoumpanioti
Int. J. Mol. Sci. 2025, 26(8), 3805; https://doi.org/10.3390/ijms26083805 - 17 Apr 2025
Viewed by 557
Abstract
Co-encapsulation of hydrophilic and hydrophobic compounds within a single delivery system remains a significant challenge across various scientific and industrial fields. Towards this direction, an encapsulation strategy is proposed, enabling the simultaneous incorporation of both hydrophilic and hydrophobic biomolecules within a hydrogel matrix. [...] Read more.
Co-encapsulation of hydrophilic and hydrophobic compounds within a single delivery system remains a significant challenge across various scientific and industrial fields. Towards this direction, an encapsulation strategy is proposed, enabling the simultaneous incorporation of both hydrophilic and hydrophobic biomolecules within a hydrogel matrix. Specifically, the cyanobacterial protein phycocyanin (hydrophilic), extracted and purified by dry Arthrospira maxima biomass, and curcumin (hydrophobic) bound to bovine serum albumin (BSA) were utilized. This approach facilitates the indirect entrapment of hydrophobic molecules within the hydrophilic hydrogel network. The structural and physicochemical properties of the resulting hydrogels were characterized using optical analysis, scanning electron microscopy (SEM), and confocal laser scanning microscopy (CLSM). Additionally, the antioxidant potential of the encapsulated biomolecules was evaluated to assess their functionality after the encapsulation. Furthermore, a cell viability assay confirmed the hydrogel’s biocompatibility and lack of toxicity, demonstrating its suitability as a multifunctional biomaterial for biomedical and pharmaceutical applications. Full article
(This article belongs to the Special Issue New Trends and Challenges in Polysaccharide Biomaterials)
Show Figures

Figure 1

21 pages, 5152 KiB  
Review
Therapeutic Potential of Nano-Sustained-Release Factors for Bone Scaffolds
by Haoran Jiang, Meng Zhang, Yang Qu, Bohan Xing, Bojiang Wang, Yanqun Liu and Peixun Zhang
J. Funct. Biomater. 2025, 16(4), 136; https://doi.org/10.3390/jfb16040136 - 9 Apr 2025
Cited by 2 | Viewed by 1032
Abstract
Research on nano-sustained-release factors for bone tissue scaffolds has significantly promoted the precision and efficiency of bone-defect repair by integrating biomaterials science, nanotechnology, and regenerative medicine. Current research focuses on developing multifunctional scaffold materials and intelligent controlled-release systems to optimize the spatiotemporal release [...] Read more.
Research on nano-sustained-release factors for bone tissue scaffolds has significantly promoted the precision and efficiency of bone-defect repair by integrating biomaterials science, nanotechnology, and regenerative medicine. Current research focuses on developing multifunctional scaffold materials and intelligent controlled-release systems to optimize the spatiotemporal release characteristics of growth factors, drugs, and genes. Nano slow-release bone scaffolds integrate nano slow-release factors, which are loaded with growth factors, drugs, genes, etc., with bone scaffolds, which can significantly improve the efficiency of bone repair. In addition, these drug-loading systems have also been extended to the fields of anti-infection and anti-tumor. However, the problem of heterotopic ossification caused by high doses has led to a shift in research towards a low-dose multi-factor synergistic strategy. Multiple Phase II clinical trials are currently ongoing, evaluating the efficacy and safety of nano-hydroxyapatite scaffolds. Despite significant progress, this field still faces a series of challenges: the immunity risks of the long-term retention of nanomaterials, the precise matching of multi-factor release kinetics, and the limitations of the large-scale production of personalized scaffolds. Future development directions in this area include the development of responsive sustained-release systems, biomimetic sequential release design, the more precise regeneration of injury sites through a combination of gene-editing technology and self-assembled nanomaterials, and precise drug loading and sustained release through microfluidic and bioprinting technologies to reduce the manufacturing cost of bone scaffolds. The progress of these bone scaffolds has gradually changed bone repair from morphology-matched filling regeneration to functional recovery, making the clinical transformation of bone scaffolds safer and more universal. Full article
(This article belongs to the Special Issue Mesoporous Nanomaterials for Bone Tissue Engineering)
Show Figures

Graphical abstract

Back to TopTop