Plants possess tremendous medicinal properties which have been supposed to be promising candidates for biomedical applications, especially in the field of nanobiotechnology. To analyze one such view, the current study was adopted to synthesize gold nanoparticles (Au*nps) by employing the extract of Murraya koenigii (EMk) for the evaluation of phenolics, antioxidant, antimicrobial, hemolytic, and biocompatible activities.
The synthesis process was carried out in a single step by mixing EMk and gold salt (Au salt) solution and monitored using UV/Visible spectroscopy. The process was optimized via variation in environmental variables. Characterization techniques such as Fourier transform infrared (FTIR) spectroscopy, transmission electron microscopy (TEM), scanning electron microscopy (SEM), X-ray diffractometer (XRD), and energy dispersive X-rays (EDX) were employed. In vitro biological activities (total phenolic, antioxidant, antimicrobial, and hemolytic) using different concentrations of Au*nps along with EMk were assessed. An in vivo histopathology study on Wistar rats was analyzed.
The band of Au*nps was observed at 540 nm, which showed successful synthesis. The FTIR spectra of Au*nps indicated the role of different functional groups (alkane, aromatic ester, thiol, nitro, and aldehyde) of EMk during synthesis. The TEM analysis illustrated a 50 nm size of Au*nps; SEM showed the presence of some aggregates; EDX represented elemental nature, and XRD proved the crystalline nature of these Au*nps. The Au*nps possessed significant phenolic content and displayed prominent antioxidant activities by quenching free radicals. Similarly, momentous inhibitory action was observed against microbial strains of Escherichia coli and Bacillus subtilis. The hemolytic study showed the least to non-toxic effect of these nanoparticles on red blood cells (RBCs) even at enhanced concentration. Histopathology study showed fair compatibility without inducing any apparent pathological lesions on the liver tissues of Wistar rats.
Plausibly, all the above investigations strongly emphasized the use of medicinal plant-based Au*nps for biological applications.
Plants possess tremendous medicinal properties which have been supposed to be promising candidates for biomedical applications, especially in the field of nanobiotechnology. To analyze one such view, the current study was adopted to synthesize gold nanoparticles (Au*nps) by employing the extract of Murraya koenigii (EMk) for the evaluation of phenolics, antioxidant, antimicrobial, hemolytic, and biocompatible activities.
The synthesis process was carried out in a single step by mixing EMk and gold salt (Au salt) solution and monitored using UV/Visible spectroscopy. The process was optimized via variation in environmental variables. Characterization techniques such as Fourier transform infrared (FTIR) spectroscopy, transmission electron microscopy (TEM), scanning electron microscopy (SEM), X-ray diffractometer (XRD), and energy dispersive X-rays (EDX) were employed. In vitro biological activities (total phenolic, antioxidant, antimicrobial, and hemolytic) using different concentrations of Au*nps along with EMk were assessed. An in vivo histopathology study on Wistar rats was analyzed.
The band of Au*nps was observed at 540 nm, which showed successful synthesis. The FTIR spectra of Au*nps indicated the role of different functional groups (alkane, aromatic ester, thiol, nitro, and aldehyde) of EMk during synthesis. The TEM analysis illustrated a 50 nm size of Au*nps; SEM showed the presence of some aggregates; EDX represented elemental nature, and XRD proved the crystalline nature of these Au*nps. The Au*nps possessed significant phenolic content and displayed prominent antioxidant activities by quenching free radicals. Similarly, momentous inhibitory action was observed against microbial strains of Escherichia coli and Bacillus subtilis. The hemolytic study showed the least to non-toxic effect of these nanoparticles on red blood cells (RBCs) even at enhanced concentration. Histopathology study showed fair compatibility without inducing any apparent pathological lesions on the liver tissues of Wistar rats.
Plausibly, all the above investigations strongly emphasized the use of medicinal plant-based Au*nps for biological applications.
Polymer-based nanoparticles have emerged as powerful multifunctional platforms in cancer theranostics, offering the ability to integrate diagnostic imaging and targeted therapy within a single system. These nanocarriers enable improved tumor localization, enhanced contrast agent delivery, and controlled therapeutic release, addressing limitations associated with conventional contrast agents such as poor specificity, rapid clearance, and systemic toxicity. Advances in polymer chemistry and nanoparticle fabrication methods, including solvent evaporation, nanoprecipitation, emulsion-diffusion, and emulsion polymerization, have allowed precise control over particle size, surface charge, and drug-loading efficiency, optimizing biodistribution and imaging performance. Hybrid polymer-inorganic nanoparticles further expand functionality by incorporating magnetic, optical, or radiopaque components, enabling multimodal imaging and stimuli-responsive drug release while maintaining biocompatibility. Key factors influencing the efficiency of polymer nanoparticle-based contrast agents include physicochemical properties such as particle size, morphology, surface functionalization, and responsiveness to tumor microenvironmental stimuli. These attributes collectively govern circulation time, cellular uptake, and accumulation in tumor tissues via passive and active targeting strategies. While promising, the clinical translation of these systems faces challenges including immunogenicity, pharmacokinetic variability, long-term safety concerns, and manufacturing scalability. Recent innovations in ligand functionalization, biomimetic coatings, and multifunctional nanoparticle design continue to advance therapeutic specificity and imaging precision, positioning polymer nanoparticles as versatile candidates for personalized oncologic care. This review provides a comprehensive synthesis of current methods for contrast agent integration, the role of physicochemical properties in performance, biological interactions, safety considerations, recent design innovations, translational barriers, and future research directions for polymer nanoparticle-based cancer theranostics.
Polymer-based nanoparticles have emerged as powerful multifunctional platforms in cancer theranostics, offering the ability to integrate diagnostic imaging and targeted therapy within a single system. These nanocarriers enable improved tumor localization, enhanced contrast agent delivery, and controlled therapeutic release, addressing limitations associated with conventional contrast agents such as poor specificity, rapid clearance, and systemic toxicity. Advances in polymer chemistry and nanoparticle fabrication methods, including solvent evaporation, nanoprecipitation, emulsion-diffusion, and emulsion polymerization, have allowed precise control over particle size, surface charge, and drug-loading efficiency, optimizing biodistribution and imaging performance. Hybrid polymer-inorganic nanoparticles further expand functionality by incorporating magnetic, optical, or radiopaque components, enabling multimodal imaging and stimuli-responsive drug release while maintaining biocompatibility. Key factors influencing the efficiency of polymer nanoparticle-based contrast agents include physicochemical properties such as particle size, morphology, surface functionalization, and responsiveness to tumor microenvironmental stimuli. These attributes collectively govern circulation time, cellular uptake, and accumulation in tumor tissues via passive and active targeting strategies. While promising, the clinical translation of these systems faces challenges including immunogenicity, pharmacokinetic variability, long-term safety concerns, and manufacturing scalability. Recent innovations in ligand functionalization, biomimetic coatings, and multifunctional nanoparticle design continue to advance therapeutic specificity and imaging precision, positioning polymer nanoparticles as versatile candidates for personalized oncologic care. This review provides a comprehensive synthesis of current methods for contrast agent integration, the role of physicochemical properties in performance, biological interactions, safety considerations, recent design innovations, translational barriers, and future research directions for polymer nanoparticle-based cancer theranostics.
Zinc is essential for normal bone growth and can promote bone regeneration. Processed human bone allograft treated with zinc shows improved bone formation activity. Various factors were tested for effects on zinc binding to bone allograft with the long-term goal of developing methods to enhance the bone formation activity and safety of bone allograft in orthopaedic applications.
The amount of zinc bound to allograft was measured using Inductively Coupled Plasma-Mass Spectrometry (ICP-MS). Fluorescent visualization of zinc bound to allograft was accomplished using Zinpyr-1. The potential anti-microbial property of zinc-treated allograft was measured by exposing allograft to Staphylococcus aureus. After washing, the exposed allograft was cultured in bacterial media to measure residual Staphylococcus aureus. Data were analyzed using standard parametric methods.
Rapid binding of zinc to bone allograft (1–15 min) was relatively insensitive to zinc concentration, incubation time, pH, or divalent cation competition. In contrast, zinc salt counter ions had significant effects, with zinc acetate producing more rapid zinc binding than zinc chloride or zinc picolinate. The ability of Staphylococcus aureus to contaminate bone allograft was also significantly reduced by prior zinc treatment.
The study results provide guidelines for modifying the processing of bone allograft to enhance bone formation activity while also improving the resistance of the allograft to bacterial contamination.
Zinc is essential for normal bone growth and can promote bone regeneration. Processed human bone allograft treated with zinc shows improved bone formation activity. Various factors were tested for effects on zinc binding to bone allograft with the long-term goal of developing methods to enhance the bone formation activity and safety of bone allograft in orthopaedic applications.
The amount of zinc bound to allograft was measured using Inductively Coupled Plasma-Mass Spectrometry (ICP-MS). Fluorescent visualization of zinc bound to allograft was accomplished using Zinpyr-1. The potential anti-microbial property of zinc-treated allograft was measured by exposing allograft to Staphylococcus aureus. After washing, the exposed allograft was cultured in bacterial media to measure residual Staphylococcus aureus. Data were analyzed using standard parametric methods.
Rapid binding of zinc to bone allograft (1–15 min) was relatively insensitive to zinc concentration, incubation time, pH, or divalent cation competition. In contrast, zinc salt counter ions had significant effects, with zinc acetate producing more rapid zinc binding than zinc chloride or zinc picolinate. The ability of Staphylococcus aureus to contaminate bone allograft was also significantly reduced by prior zinc treatment.
The study results provide guidelines for modifying the processing of bone allograft to enhance bone formation activity while also improving the resistance of the allograft to bacterial contamination.
Bone tissue engineering (BTE) represents a cutting-edge approach to treating critical-sized bone defects, complex fractures, and degenerative bone diseases by promoting the regeneration of functional bone tissue. A crucial element in this process is the design and optimization of scaffolds that emulate the natural extracellular matrix (ECM), supporting cell adhesion, proliferation, and differentiation necessary for bone regeneration. Polymers are widely used in scaffold fabrication. They offer versatility, biocompatibility, and tunable properties that are essential for tissue engineering. This paper provides a comprehensive analysis of polymeric scaffolds in BTE, focusing on synthetic and natural polymers, composite scaffold designs, and the fabrication techniques employed to enhance their performance. Key design criteria, such as scaffold porosity, mechanical properties, and biodegradability, are discussed in the context of facilitating optimal bone regeneration. Additionally, we explore functionalization strategies to improve biological interactions, such as the incorporation of growth factors and surface modifications, and evaluate in vivo performance to highlight clinical potential. The paper also addresses current challenges, including the need for enhanced mechanical strength and controlled degradation, while offering insights into future directions for the development of polymeric scaffolds in bone tissue regeneration therapies.
Bone tissue engineering (BTE) represents a cutting-edge approach to treating critical-sized bone defects, complex fractures, and degenerative bone diseases by promoting the regeneration of functional bone tissue. A crucial element in this process is the design and optimization of scaffolds that emulate the natural extracellular matrix (ECM), supporting cell adhesion, proliferation, and differentiation necessary for bone regeneration. Polymers are widely used in scaffold fabrication. They offer versatility, biocompatibility, and tunable properties that are essential for tissue engineering. This paper provides a comprehensive analysis of polymeric scaffolds in BTE, focusing on synthetic and natural polymers, composite scaffold designs, and the fabrication techniques employed to enhance their performance. Key design criteria, such as scaffold porosity, mechanical properties, and biodegradability, are discussed in the context of facilitating optimal bone regeneration. Additionally, we explore functionalization strategies to improve biological interactions, such as the incorporation of growth factors and surface modifications, and evaluate in vivo performance to highlight clinical potential. The paper also addresses current challenges, including the need for enhanced mechanical strength and controlled degradation, while offering insights into future directions for the development of polymeric scaffolds in bone tissue regeneration therapies.
Acute cutaneous injuries and refractory chronic wounds represent prevalent clinical challenges in daily life. To address the impediments to wound healing, we propose a novel hydrogel-based therapeutic approach designed to prevent bacterial invasion, mitigate infection-induced persistent inflammatory responses, and reduce excessive oxidative stress, thereby enhancing the wound healing process.
This study presents a method for preparing hyaluronic acid/silk fibroin (HA/SF) composite hydrogels via photo-crosslinking. HA and SF were respectively modified via methacrylation and glycidyl methacrylate to synthesize HAMA and SFMA. Mussel-inspired catechol groups were then grafted onto HAMA chain segments to prepare precursor HAC. Under photo-initiator LAP, polymerization was triggered to ultimately form a hydrogel network integrating mechanical toughness and tissue adhesiveness. Composite hydrogels with varying degrees of crosslinking are synthesized by adjusting the SFMA content.
The results demonstrate that this hydrogel can effectively achieve hemostasis within 20 seconds. Lap shear testing revealed that the HASF-gel-2 hydrogel exhibited the highest maximum adhesive strength of 160.3 kPa among all experimental groups. Furthermore, while the cell viability of the control group was normalized to 1, the composite hydrogel groups displayed values of 1.015, 1.085, 1.136, and 1.263, respectively, indicating favorable biocompatibility. The appropriate incorporation of SF was shown to enhance cellular proliferation. On day 3 post-wounding, the HASF hydrogel group demonstrated a wound closure rate of 41.7%, outperforming commercial products under identical conditions.
In rat wound models, the HASF composite hydrogel significantly accelerated wound healing progression.
Acute cutaneous injuries and refractory chronic wounds represent prevalent clinical challenges in daily life. To address the impediments to wound healing, we propose a novel hydrogel-based therapeutic approach designed to prevent bacterial invasion, mitigate infection-induced persistent inflammatory responses, and reduce excessive oxidative stress, thereby enhancing the wound healing process.
This study presents a method for preparing hyaluronic acid/silk fibroin (HA/SF) composite hydrogels via photo-crosslinking. HA and SF were respectively modified via methacrylation and glycidyl methacrylate to synthesize HAMA and SFMA. Mussel-inspired catechol groups were then grafted onto HAMA chain segments to prepare precursor HAC. Under photo-initiator LAP, polymerization was triggered to ultimately form a hydrogel network integrating mechanical toughness and tissue adhesiveness. Composite hydrogels with varying degrees of crosslinking are synthesized by adjusting the SFMA content.
The results demonstrate that this hydrogel can effectively achieve hemostasis within 20 seconds. Lap shear testing revealed that the HASF-gel-2 hydrogel exhibited the highest maximum adhesive strength of 160.3 kPa among all experimental groups. Furthermore, while the cell viability of the control group was normalized to 1, the composite hydrogel groups displayed values of 1.015, 1.085, 1.136, and 1.263, respectively, indicating favorable biocompatibility. The appropriate incorporation of SF was shown to enhance cellular proliferation. On day 3 post-wounding, the HASF hydrogel group demonstrated a wound closure rate of 41.7%, outperforming commercial products under identical conditions.
In rat wound models, the HASF composite hydrogel significantly accelerated wound healing progression.
Metal 3D printing has revolutionized the fabrication of biometallic prostheses and implants, offering unprecedented design flexibility, patient-specific customization, and enhanced biomechanical performance. This review explores the current advancements in metal additive manufacturing (AM) techniques, including selective laser melting (SLM), electron beam melting (EBM), fused deposition modeling (FDM), directed energy deposition (DED), sheet lamination, stereolithography (SLA), and binder jetting, for processing biocompatible metals such as titanium, cobalt-chromium, and stainless steel. The article discusses major benefits, such as enhanced osseointegration, complex lattice architectures for weight saving, and optimized mechanical properties. The challenges of residual stresses, surface finish, and regulatory issues are also discussed. The review concludes by defining future research avenues in material design, process development, and clinical translation to increase the efficacy and reliability of 3D-printed biometal implants.
Metal 3D printing has revolutionized the fabrication of biometallic prostheses and implants, offering unprecedented design flexibility, patient-specific customization, and enhanced biomechanical performance. This review explores the current advancements in metal additive manufacturing (AM) techniques, including selective laser melting (SLM), electron beam melting (EBM), fused deposition modeling (FDM), directed energy deposition (DED), sheet lamination, stereolithography (SLA), and binder jetting, for processing biocompatible metals such as titanium, cobalt-chromium, and stainless steel. The article discusses major benefits, such as enhanced osseointegration, complex lattice architectures for weight saving, and optimized mechanical properties. The challenges of residual stresses, surface finish, and regulatory issues are also discussed. The review concludes by defining future research avenues in material design, process development, and clinical translation to increase the efficacy and reliability of 3D-printed biometal implants.
This review highlights the challenges of current wound healing methods, such as scar formation and limited regeneration, and emphasizes the potential of tissue engineering to address these issues. Chitosan, a biopolymer derived from chitin, has garnered significant attention in epidermal-dermal wound healing due to its exceptional biocompatibility, biodegradability, and versatile functional properties. This review article delves into the diverse roles of chitosan, with a particular focus on its use as a scaffold material with fine-tunable physicochemical and biological properties for accelerated wound healing. While bare chitosan provides a suitable microenvironment for cell adhesion and proliferation, it exhibits limited mechanical strength and drug-delivery properties. However, combining it with other natural and synthetic polymers and nanoparticles facilitates drug and biosignal delivery and enhances biocompatibility and antibacterial activity. Furthermore, the review covers various chemical modifications of chitosan, including quaternization and methacrylation, to improve biocompatibility, water solubility and mechanical strength, for developing advanced wound dressings for effective skin regeneration. The review also discusses various types of smart chitosan hydrogels and the clinical translation of chitosan based scaffolds for wound healing and tissue regeneration applications. Finally, it discusses the integration of 3D bioprinting techniques for creating complex, cell-incorporated scaffolds for advanced wound healing therapies.
This review highlights the challenges of current wound healing methods, such as scar formation and limited regeneration, and emphasizes the potential of tissue engineering to address these issues. Chitosan, a biopolymer derived from chitin, has garnered significant attention in epidermal-dermal wound healing due to its exceptional biocompatibility, biodegradability, and versatile functional properties. This review article delves into the diverse roles of chitosan, with a particular focus on its use as a scaffold material with fine-tunable physicochemical and biological properties for accelerated wound healing. While bare chitosan provides a suitable microenvironment for cell adhesion and proliferation, it exhibits limited mechanical strength and drug-delivery properties. However, combining it with other natural and synthetic polymers and nanoparticles facilitates drug and biosignal delivery and enhances biocompatibility and antibacterial activity. Furthermore, the review covers various chemical modifications of chitosan, including quaternization and methacrylation, to improve biocompatibility, water solubility and mechanical strength, for developing advanced wound dressings for effective skin regeneration. The review also discusses various types of smart chitosan hydrogels and the clinical translation of chitosan based scaffolds for wound healing and tissue regeneration applications. Finally, it discusses the integration of 3D bioprinting techniques for creating complex, cell-incorporated scaffolds for advanced wound healing therapies.
Multifunctional nanomaterials with photodynamic-sonodynamic therapy (PSDT) potential offer significant advantages in cancer treatment. However, designing and preparing single-component “two-in-one” multifunctional nanomaterials remains challenging. Hematoporphyrin monomethyl ether (HMME), a second-generation porphyrin-related sonosensitizer, is a porphyrin derivative with two asymmetric carboxyl groups. Notably, the carboxyl groups in HMME can coordinate with metal ions to construct metal-organic coordination nanomaterials (MCPs). Titanium (Ti), a biocompatible metal element, is commonly used in medical devices such as implantable metal alloys. Therefore, this study reported the synthesis of “two-in-one” type Ti-HMME coordination nanomaterials (TiCPs) as efficient nanoscale photo/sonosensitizers.
Under a nitrogen atmosphere, TiCPs were synthesized via self-assembly between HMME and Ti4+ ions.
The average particle size of TiCPs was approximately 70 nm. Additionally, TiCPs contained the photo/sonosensitizer HMME, which could convert O2 into cytotoxic reactive oxygen species (ROS) under light and ultrasound (US) excitation. The generation of ROS could be detected using 1,3-diphenylisobenzofuran (DPBF). When the mixed solution (TiCPs + DPBF) was irradiated with light, the DPBF peak rapidly decreased with increasing irradiation time, indicating the production of ROS by TiCPs under light. Similarly, the absorbance of TiCPs + DPBF significantly decreased with increasing US time, demonstrating the sonodynamic effect of TiCPs + US. After 10 min of light or US excitation, 49.4% (Light) and 38.1% (US) of DPBF were oxidized by ROS generated by TiCPs, showcasing excellent photodynamic/sonodynamic effects. In vitro cell experiments further demonstrated that TiCPs had excellent biocompatibility, could be effectively internalized by cells, and significantly reduced cell viability under light and US excitation, effectively killing tumor cells.
This study not only demonstrated TiCPs as “two-in-one” type multifunctional nanomaterials for PSDT but also provided insights into designing other photo/sonosensitizer molecules with similar HMME structures for tumor theranostics.
Multifunctional nanomaterials with photodynamic-sonodynamic therapy (PSDT) potential offer significant advantages in cancer treatment. However, designing and preparing single-component “two-in-one” multifunctional nanomaterials remains challenging. Hematoporphyrin monomethyl ether (HMME), a second-generation porphyrin-related sonosensitizer, is a porphyrin derivative with two asymmetric carboxyl groups. Notably, the carboxyl groups in HMME can coordinate with metal ions to construct metal-organic coordination nanomaterials (MCPs). Titanium (Ti), a biocompatible metal element, is commonly used in medical devices such as implantable metal alloys. Therefore, this study reported the synthesis of “two-in-one” type Ti-HMME coordination nanomaterials (TiCPs) as efficient nanoscale photo/sonosensitizers.
Under a nitrogen atmosphere, TiCPs were synthesized via self-assembly between HMME and Ti4+ ions.
The average particle size of TiCPs was approximately 70 nm. Additionally, TiCPs contained the photo/sonosensitizer HMME, which could convert O2 into cytotoxic reactive oxygen species (ROS) under light and ultrasound (US) excitation. The generation of ROS could be detected using 1,3-diphenylisobenzofuran (DPBF). When the mixed solution (TiCPs + DPBF) was irradiated with light, the DPBF peak rapidly decreased with increasing irradiation time, indicating the production of ROS by TiCPs under light. Similarly, the absorbance of TiCPs + DPBF significantly decreased with increasing US time, demonstrating the sonodynamic effect of TiCPs + US. After 10 min of light or US excitation, 49.4% (Light) and 38.1% (US) of DPBF were oxidized by ROS generated by TiCPs, showcasing excellent photodynamic/sonodynamic effects. In vitro cell experiments further demonstrated that TiCPs had excellent biocompatibility, could be effectively internalized by cells, and significantly reduced cell viability under light and US excitation, effectively killing tumor cells.
This study not only demonstrated TiCPs as “two-in-one” type multifunctional nanomaterials for PSDT but also provided insights into designing other photo/sonosensitizer molecules with similar HMME structures for tumor theranostics.
This study evaluated the impact of retinal extracellular matrix (ECM) and key biomaterial substrates on the motility of transplantable retinal cells with genomic manipulation, using the therapeutic molecule, Topoisomerase II beta (Top2b), as a model.
Tests first applied in ovo electroporation to examine the effects of a pharmacological Top2b inhibitor (ICRF-193) on progenitor motility and development of embryonic retina. Complementary qRT-PCR tests measured changes in select cadherin molecules in response to treatment. In vitro transfection produced cultured retinal progenitor cell groups with Top2b overexpression and Top2b knockdown. Differences in the adhesion and motility of Top2b altered groups, compared to wildtype cells, were measured upon biomaterial substrates used in emerging transplantation matrixes.
Data illustrated significant differences in the number and spacing of retinal ganglion cells when retina was treated with ICRF-193, as well as downregulation of several key cadherin molecules. Cultured retinal progenitors with Top2b knockdown and Top2b overexpression exhibited different expression of chemotactic receptors, adhesion parameters, and modalities of migration upon substrates of laminin, poly-L-lysine, and collagen IV. Significant changes in cell morphology and surface area were also measured compared to wildtype cells.
Corroborating in vivo and in vitro data support Top2b as a therapeutic target for retinal progenitor motility but indicate significant differences in the migration of Top2b altered cells upon substrates used in transplantation. These data highlight the therapeutic advantages of bioinspired materials developed to aid the motility of replacement cells with modified genetic expression to improve transplantation outcomes across the nervous system.
This study evaluated the impact of retinal extracellular matrix (ECM) and key biomaterial substrates on the motility of transplantable retinal cells with genomic manipulation, using the therapeutic molecule, Topoisomerase II beta (Top2b), as a model.
Tests first applied in ovo electroporation to examine the effects of a pharmacological Top2b inhibitor (ICRF-193) on progenitor motility and development of embryonic retina. Complementary qRT-PCR tests measured changes in select cadherin molecules in response to treatment. In vitro transfection produced cultured retinal progenitor cell groups with Top2b overexpression and Top2b knockdown. Differences in the adhesion and motility of Top2b altered groups, compared to wildtype cells, were measured upon biomaterial substrates used in emerging transplantation matrixes.
Data illustrated significant differences in the number and spacing of retinal ganglion cells when retina was treated with ICRF-193, as well as downregulation of several key cadherin molecules. Cultured retinal progenitors with Top2b knockdown and Top2b overexpression exhibited different expression of chemotactic receptors, adhesion parameters, and modalities of migration upon substrates of laminin, poly-L-lysine, and collagen IV. Significant changes in cell morphology and surface area were also measured compared to wildtype cells.
Corroborating in vivo and in vitro data support Top2b as a therapeutic target for retinal progenitor motility but indicate significant differences in the migration of Top2b altered cells upon substrates used in transplantation. These data highlight the therapeutic advantages of bioinspired materials developed to aid the motility of replacement cells with modified genetic expression to improve transplantation outcomes across the nervous system.
The purpose of this study is to investigate the deformability and strength of silicone breast implant shells from different manufactures as a function of implantation time.
The strength properties of about 200 shells of Eurosilicone, Mentor, Motiva, Allergan, Arion, PIP silicone breast implants removed for various reasons during repeated surgeries with a period of stay in the body from 6 months to 29 years were measured and compared with the corresponding properties of four shells of unused Eurosilicone and Motiva implants. Deformation was measured using a videoXtens extensometer.
The mechanical properties of the Allergan implant shell are almost completely consistent with the properties of the Eurosilicone shell after 9 years of use. The Mentor implants showed greater strength and stiffness. The Motiva implant shells initially had higher ultimate properties—rupture stress and rupture strain—in comparison with the Eurosilicone implant shells.
The strength and deformation properties of all examined breast implant shells decrease in the course of time. After 13 years of use, the strength of breast implants is halved and their rupture strain is reduced by one third. The main mechanism responsible for loss of strength is the accumulation of microdamages during long-term use of breast implants. The thickness of the nano-textured shells of Motiva implants is twice less of the Eurosilicone implant shell thickness. This was possible due to a significant increase in the mechanical properties of Motiva shells.
The purpose of this study is to investigate the deformability and strength of silicone breast implant shells from different manufactures as a function of implantation time.
The strength properties of about 200 shells of Eurosilicone, Mentor, Motiva, Allergan, Arion, PIP silicone breast implants removed for various reasons during repeated surgeries with a period of stay in the body from 6 months to 29 years were measured and compared with the corresponding properties of four shells of unused Eurosilicone and Motiva implants. Deformation was measured using a videoXtens extensometer.
The mechanical properties of the Allergan implant shell are almost completely consistent with the properties of the Eurosilicone shell after 9 years of use. The Mentor implants showed greater strength and stiffness. The Motiva implant shells initially had higher ultimate properties—rupture stress and rupture strain—in comparison with the Eurosilicone implant shells.
The strength and deformation properties of all examined breast implant shells decrease in the course of time. After 13 years of use, the strength of breast implants is halved and their rupture strain is reduced by one third. The main mechanism responsible for loss of strength is the accumulation of microdamages during long-term use of breast implants. The thickness of the nano-textured shells of Motiva implants is twice less of the Eurosilicone implant shell thickness. This was possible due to a significant increase in the mechanical properties of Motiva shells.
To show that a wireless-powered thrombolytic filter can be used in the treatment of venous thromboembolism (VTE) as an alternative to the existing VTE therapies, which have serious side effects.
The wireless-powered thrombolytic filter that we propose combines the positive attributes of anticoagulants and thrombolytics, through the capture and dissolution of blood clots, without the associated adverse effects of existing treatments. The filter absorbs radio-frequency energy from a source and converts it into heat at the thrombolytic filter.
We used computer simulations with COMSOL and lab experiments to demonstrate that a wireless-powered thrombolytic filter can be heated up through the absorption of radio-frequency energy from an external source.
We demonstrate that a wireless-powered thrombolytic filter has the potential to be used in the treatment of VTE, since it can be designed to absorb energy from an external radio-frequency source and convert it to heat that is sufficient to dissolve blood clots captured by the thrombolytic filter.
To show that a wireless-powered thrombolytic filter can be used in the treatment of venous thromboembolism (VTE) as an alternative to the existing VTE therapies, which have serious side effects.
The wireless-powered thrombolytic filter that we propose combines the positive attributes of anticoagulants and thrombolytics, through the capture and dissolution of blood clots, without the associated adverse effects of existing treatments. The filter absorbs radio-frequency energy from a source and converts it into heat at the thrombolytic filter.
We used computer simulations with COMSOL and lab experiments to demonstrate that a wireless-powered thrombolytic filter can be heated up through the absorption of radio-frequency energy from an external source.
We demonstrate that a wireless-powered thrombolytic filter has the potential to be used in the treatment of VTE, since it can be designed to absorb energy from an external radio-frequency source and convert it to heat that is sufficient to dissolve blood clots captured by the thrombolytic filter.
Mineral nanoparticles and osteoinductive biomaterials are essential in advancing bone regeneration by addressing skeletal conditions and injuries that compromise structural integrity and functionality. These biomaterials stimulate the differentiation of precursor cells into osteoblasts, creating biocompatible environments conducive to bone tissue regeneration. Among the most promising innovations, mineral-based nanoparticles and nanocomposite hydrogels have emerged as effective strategies for enhancing osteoinductive potential. This review explores the diverse types of osteoinductive biomaterials, including natural sources, synthetic compounds, and hybrid designs that incorporate mineralized nanoparticles. Emphasis is placed on polymeric hydrogels as delivery platforms for these materials, highlighting their dual role as structural supports and bioactive agents that promote osteogenesis. Challenges such as immune rejection, biodegradability, mechanical stability, and short in vivo residence time are critically discussed, alongside their impact on clinical translation. By presenting a comprehensive analysis of mechanisms, applications, and limitations, this review identifies opportunities for integrating osteoinductive biomaterials with emerging fields like immunology and biomechanics. Ultimately, this work aims to provide actionable insights and advance the development of novel, clinically relevant solutions that improve patient outcomes and address the growing global need for effective bone repair and regeneration.
Mineral nanoparticles and osteoinductive biomaterials are essential in advancing bone regeneration by addressing skeletal conditions and injuries that compromise structural integrity and functionality. These biomaterials stimulate the differentiation of precursor cells into osteoblasts, creating biocompatible environments conducive to bone tissue regeneration. Among the most promising innovations, mineral-based nanoparticles and nanocomposite hydrogels have emerged as effective strategies for enhancing osteoinductive potential. This review explores the diverse types of osteoinductive biomaterials, including natural sources, synthetic compounds, and hybrid designs that incorporate mineralized nanoparticles. Emphasis is placed on polymeric hydrogels as delivery platforms for these materials, highlighting their dual role as structural supports and bioactive agents that promote osteogenesis. Challenges such as immune rejection, biodegradability, mechanical stability, and short in vivo residence time are critically discussed, alongside their impact on clinical translation. By presenting a comprehensive analysis of mechanisms, applications, and limitations, this review identifies opportunities for integrating osteoinductive biomaterials with emerging fields like immunology and biomechanics. Ultimately, this work aims to provide actionable insights and advance the development of novel, clinically relevant solutions that improve patient outcomes and address the growing global need for effective bone repair and regeneration.
A potential solution for prosthetic heart valves is tissue-engineered heart valves. Tissue-engineered heart valves (TEHVs) are designed to replicate the complex properties found in natural tissues, such as stiffness, anisotropy, and composition and organization of cells and extracellular matrix (ECM). Electrospinning is regarded as a highly versatile and innovative approach for fabricating numerous fibrous designs. In this review, we discuss recent developments in electrospun heart valve scaffolds, including scaffold materials, cell types, and electrospinning setups used to prepare aligned nanofibers. Despite the fact that natural biomaterials provided excellent biocompatibility, nanofibers from synthetic materials provided the required mechanical compatibility. Accordingly, most studies highlighted the benefits of designing composite heart valves using biological and synthetic polymers. Various strategies, such as the application of motorized mandrel and micropatterned collector in electrospinning were effective in controlling nanofiber alignment. Studies also showed that aligned nanofiber’s mechanical strength and anisotropic structure promote cell proliferation, and differentiation, and promote attachment. Numerous studies have reported that multiple cell sources are suitable for producing heart valves. Successful results were obtained with human umbilical vein endothelial cells (HUVECs), since they provide a convenient cell source for cellularization of valve leaflets. A higher conductivity of scaffolds was achieved by using biomaterials that conduct electricity, such as polyaniline, polypyrrole, and carbon nanotubes, which resulted in better differentiation of precursor cells to cardiomyocytes and higher cell beating rates. In light of these attributes, nanofibrous scaffolds produced through electrospinning are expected to offer numerous advantages for tissue engineering and medical applications in the near future. However, multiple challenges were identified as cell infiltration and 2D nature of nanofiber mats necessitate further engineering approaches in electrospinning procedure leaflet production.
A potential solution for prosthetic heart valves is tissue-engineered heart valves. Tissue-engineered heart valves (TEHVs) are designed to replicate the complex properties found in natural tissues, such as stiffness, anisotropy, and composition and organization of cells and extracellular matrix (ECM). Electrospinning is regarded as a highly versatile and innovative approach for fabricating numerous fibrous designs. In this review, we discuss recent developments in electrospun heart valve scaffolds, including scaffold materials, cell types, and electrospinning setups used to prepare aligned nanofibers. Despite the fact that natural biomaterials provided excellent biocompatibility, nanofibers from synthetic materials provided the required mechanical compatibility. Accordingly, most studies highlighted the benefits of designing composite heart valves using biological and synthetic polymers. Various strategies, such as the application of motorized mandrel and micropatterned collector in electrospinning were effective in controlling nanofiber alignment. Studies also showed that aligned nanofiber’s mechanical strength and anisotropic structure promote cell proliferation, and differentiation, and promote attachment. Numerous studies have reported that multiple cell sources are suitable for producing heart valves. Successful results were obtained with human umbilical vein endothelial cells (HUVECs), since they provide a convenient cell source for cellularization of valve leaflets. A higher conductivity of scaffolds was achieved by using biomaterials that conduct electricity, such as polyaniline, polypyrrole, and carbon nanotubes, which resulted in better differentiation of precursor cells to cardiomyocytes and higher cell beating rates. In light of these attributes, nanofibrous scaffolds produced through electrospinning are expected to offer numerous advantages for tissue engineering and medical applications in the near future. However, multiple challenges were identified as cell infiltration and 2D nature of nanofiber mats necessitate further engineering approaches in electrospinning procedure leaflet production.
In this study, the physicochemical properties of stearyl glycyrrhetinate/β-cyclodextrin (SG/βCD) and SG/γCD complexes were characterized, and the complexes were prepared using the co-milling method. The molecular interactions within the SG/CD complexes were also investigated using nuclear magnetic resonance (NMR) measurements to determine the mode of interaction.
Here, we evaluated the physicochemical properties of SG complexes with CDs prepared by ground mixtures (GM SG/βCD or γCD = 1/1, 1/2).
Powder X-ray diffraction (PXRD) showed that the characteristic SG and CD peaks disappeared after co-grinding with GM (SG/CD = molar ratio of 1/2), indicating a halo pattern. Differential scanning calorimetry (DSC) measurements showed that after co-grinding, the endothermic peak due to SG melting, as well as the dehydration peak and the endothermic peak due to SG melting, disappeared. Near-infrared (NIR) spectroscopy results showed that the peaks of SG-derived –CH moieties and CD-derived –OH and –CH moieties broadened in GM, suggesting their involvement in complex formation through SG/CDs intermolecular interactions. In GM (SG/CDs), NMR measurements showed broadened H-A and H-F peaks of the steroid skeleton derived from SG. In GM (SG/βCD = 1/2), the doublet peak, especially OH-3 at the wide edge of CD, shifted to a singlet peak. In GM (SG/γCD = 1/2), the H-3 peak, which is the wide edge of γCD, and the H-6 peak, which is the narrow edge, shifted to broad peaks, suggesting that γCD is deeply encapsulated in the steroidal structure.
These findings suggest that complex formation occurred in SG/CDs and that inclusion behavior is different between GM (SG/βCD = 1/2) and GM (SG/γCD = 1/2).
In this study, the physicochemical properties of stearyl glycyrrhetinate/β-cyclodextrin (SG/βCD) and SG/γCD complexes were characterized, and the complexes were prepared using the co-milling method. The molecular interactions within the SG/CD complexes were also investigated using nuclear magnetic resonance (NMR) measurements to determine the mode of interaction.
Here, we evaluated the physicochemical properties of SG complexes with CDs prepared by ground mixtures (GM SG/βCD or γCD = 1/1, 1/2).
Powder X-ray diffraction (PXRD) showed that the characteristic SG and CD peaks disappeared after co-grinding with GM (SG/CD = molar ratio of 1/2), indicating a halo pattern. Differential scanning calorimetry (DSC) measurements showed that after co-grinding, the endothermic peak due to SG melting, as well as the dehydration peak and the endothermic peak due to SG melting, disappeared. Near-infrared (NIR) spectroscopy results showed that the peaks of SG-derived –CH moieties and CD-derived –OH and –CH moieties broadened in GM, suggesting their involvement in complex formation through SG/CDs intermolecular interactions. In GM (SG/CDs), NMR measurements showed broadened H-A and H-F peaks of the steroid skeleton derived from SG. In GM (SG/βCD = 1/2), the doublet peak, especially OH-3 at the wide edge of CD, shifted to a singlet peak. In GM (SG/γCD = 1/2), the H-3 peak, which is the wide edge of γCD, and the H-6 peak, which is the narrow edge, shifted to broad peaks, suggesting that γCD is deeply encapsulated in the steroidal structure.
These findings suggest that complex formation occurred in SG/CDs and that inclusion behavior is different between GM (SG/βCD = 1/2) and GM (SG/γCD = 1/2).
This study aimed to synthesize, characterize, and evaluate the antifungal efficacy of green-synthesized silver nanoparticles (AgNPs) against Verticillium dahliae Kleb., a soil-borne fungal pathogen that affects numerous crops.
AgNPs were synthesized using Laurus nobilis L. (laurel) leaf extract. The synthesized AgNPs were characterized using UV-VIS spectroscopy, Fourier-Transform Infrared Spectroscopy (FTIR), zeta potential, particle size analysis (PSA), and scanning electron microscopy (SEM). In vitro antifungal assays were conducted to assess the impact of AgNPs on V. dahliae mycelial growth, and SEM was used to examine the morphological changes in treated mycelium.
UV-VIS spectroscopy confirmed AgNP synthesis with a characteristic SPR peak between 400–450 nm. FTIR analysis identified the presence of phenolic compounds on the nanoparticle surface. Zeta potential analysis (–27.7 mV) indicated stable dispersion. Zeta size analysis indicated an average diameter of approximately 100 nm and a polydispersity index (PdI) of 0.229. SEM imaging confirmed a predominantly spherical morphology and PSA revealed a size range of 14–34 nm, with an average diameter of 24 nm. In vitro antifungal assays showed significant inhibition of V. dahliae mycelial growth, with radial mycelial growth reduced to 2.75 cm compared to 4.8–6.4 cm in the control group after 14 days. SEM imaging of treated mycelium revealed pronounced morphological damage, including collapse and shrinkage of hyphae and spores.
Green-synthesized AgNPs using L. nobilis leaf extract demonstrated significant antifungal activity against V. dahliae. The observed inhibition of mycelial growth and morphological damage suggests the potential of these AgNPs as a sustainable and eco-friendly alternative for managing this fungal pathogen. The antifungal mechanism may involve membrane disruption, increased permeability, oxidative stress, and the inactivation of cellular components.
This study aimed to synthesize, characterize, and evaluate the antifungal efficacy of green-synthesized silver nanoparticles (AgNPs) against Verticillium dahliae Kleb., a soil-borne fungal pathogen that affects numerous crops.
AgNPs were synthesized using Laurus nobilis L. (laurel) leaf extract. The synthesized AgNPs were characterized using UV-VIS spectroscopy, Fourier-Transform Infrared Spectroscopy (FTIR), zeta potential, particle size analysis (PSA), and scanning electron microscopy (SEM). In vitro antifungal assays were conducted to assess the impact of AgNPs on V. dahliae mycelial growth, and SEM was used to examine the morphological changes in treated mycelium.
UV-VIS spectroscopy confirmed AgNP synthesis with a characteristic SPR peak between 400–450 nm. FTIR analysis identified the presence of phenolic compounds on the nanoparticle surface. Zeta potential analysis (–27.7 mV) indicated stable dispersion. Zeta size analysis indicated an average diameter of approximately 100 nm and a polydispersity index (PdI) of 0.229. SEM imaging confirmed a predominantly spherical morphology and PSA revealed a size range of 14–34 nm, with an average diameter of 24 nm. In vitro antifungal assays showed significant inhibition of V. dahliae mycelial growth, with radial mycelial growth reduced to 2.75 cm compared to 4.8–6.4 cm in the control group after 14 days. SEM imaging of treated mycelium revealed pronounced morphological damage, including collapse and shrinkage of hyphae and spores.
Green-synthesized AgNPs using L. nobilis leaf extract demonstrated significant antifungal activity against V. dahliae. The observed inhibition of mycelial growth and morphological damage suggests the potential of these AgNPs as a sustainable and eco-friendly alternative for managing this fungal pathogen. The antifungal mechanism may involve membrane disruption, increased permeability, oxidative stress, and the inactivation of cellular components.
This work aimed to evaluate the antiproliferative activity of silver nanoparticles (AgNPs) biosynthesized with aqueous extract of Stenocereus queretaroensis peel (SAgNPs) in pancreatic ductal cancer cells PANC-1.
Nanoparticles were synthesized using 2 mM silver nitrate (AgNO3) and a 1% aqueous extract of Stenocereus queretaroensis peel. SAgNPs were characterized by ultraviolet-visible spectroscopy (UV-Vis) light spectroscopy, dynamic light scattering analysis, and transmission electron microscopy. The antiproliferative activity was evaluated in the PANC-1 cell line by measuring the viability percentage with the 3'-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method and subsequently the IC50 of SAgNPs.
The presence of AgNPs was confirmed by silver surface plasmon resonance at 420 nm. The average size obtained by dynamic light scattering analysis was 98.96 nm, with a spherical and uniform shape according to transmission electron microscopy analysis. SAgNPs were tested at concentrations from 10 µg/mL to 0.3125 µg/mL and presented inhibition percentages from 3.76% to 90.09% with an IC50 value of 3.04 µg/mL (p-value of 0.02, 95% confidence level) in PANC-1 cells.
The biologically synthesized nanoparticles with Stenocereus queretaroensis peel showed antiproliferative activity in PANC-1 pancreatic cancer cells. Therefore, these results suggest their potential use in the prevention and treatment of pancreatic cancer with further investigation.
This work aimed to evaluate the antiproliferative activity of silver nanoparticles (AgNPs) biosynthesized with aqueous extract of Stenocereus queretaroensis peel (SAgNPs) in pancreatic ductal cancer cells PANC-1.
Nanoparticles were synthesized using 2 mM silver nitrate (AgNO3) and a 1% aqueous extract of Stenocereus queretaroensis peel. SAgNPs were characterized by ultraviolet-visible spectroscopy (UV-Vis) light spectroscopy, dynamic light scattering analysis, and transmission electron microscopy. The antiproliferative activity was evaluated in the PANC-1 cell line by measuring the viability percentage with the 3'-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method and subsequently the IC50 of SAgNPs.
The presence of AgNPs was confirmed by silver surface plasmon resonance at 420 nm. The average size obtained by dynamic light scattering analysis was 98.96 nm, with a spherical and uniform shape according to transmission electron microscopy analysis. SAgNPs were tested at concentrations from 10 µg/mL to 0.3125 µg/mL and presented inhibition percentages from 3.76% to 90.09% with an IC50 value of 3.04 µg/mL (p-value of 0.02, 95% confidence level) in PANC-1 cells.
The biologically synthesized nanoparticles with Stenocereus queretaroensis peel showed antiproliferative activity in PANC-1 pancreatic cancer cells. Therefore, these results suggest their potential use in the prevention and treatment of pancreatic cancer with further investigation.
The design of effective treatments for critical size bone defects, which result from various conditions such as trauma, infection, injury, or tumor resection, presents a significant challenge in clinical practice. While autologous grafts are commonly regarded as gold standard treatments in these complex healing scenarios, they are often associated with notable limitations, including donor site morbidity and limited graft volume. As a result, recent research trends have shifted towards developing biomaterials that better emulate the inherent complexity of the native bone structure and function through implementation of a “Diamond Concept” polytherapy strategy. Central to this approach is the utilization of biomaterials, increasingly composed of composite materials that integrate bioactive osteoinductive factors and cell sources to enhance healing outcomes. The usage of Wnt signaling specific agonists as osteoinductive mediators has been recently shown to be a promising strategy for promoting healing, as this pathway is well established to have an important role in both osteogenic differentiation and bone formation processes. Implementation of a localized delivery system through scaffold incorporation is necessary in this scenario, however, to minimize any potential off-target effects caused by the Wnt signaling cascade’s non-specificity to bone. Findings in the literature clearly show that this approach holds promise to improve clinical healing outcomes, paving the way for more effective treatment options. In this review, we will generally discuss the design of biomaterials, specifically bulk materials and composites, for the treatment of critical size bone defects. Additionally, we will highlight recent work on the design of chitosan-based scaffolds modified with purine crosslinking, to overcome cytotoxicity issues associated with other chemical crosslinkers. In this context, we focus on optimizing material design for this bone healing application and discuss the benefits of localized Wnt agonist as mediators to improve the scaffold’s osteoinductive behavior.
The design of effective treatments for critical size bone defects, which result from various conditions such as trauma, infection, injury, or tumor resection, presents a significant challenge in clinical practice. While autologous grafts are commonly regarded as gold standard treatments in these complex healing scenarios, they are often associated with notable limitations, including donor site morbidity and limited graft volume. As a result, recent research trends have shifted towards developing biomaterials that better emulate the inherent complexity of the native bone structure and function through implementation of a “Diamond Concept” polytherapy strategy. Central to this approach is the utilization of biomaterials, increasingly composed of composite materials that integrate bioactive osteoinductive factors and cell sources to enhance healing outcomes. The usage of Wnt signaling specific agonists as osteoinductive mediators has been recently shown to be a promising strategy for promoting healing, as this pathway is well established to have an important role in both osteogenic differentiation and bone formation processes. Implementation of a localized delivery system through scaffold incorporation is necessary in this scenario, however, to minimize any potential off-target effects caused by the Wnt signaling cascade’s non-specificity to bone. Findings in the literature clearly show that this approach holds promise to improve clinical healing outcomes, paving the way for more effective treatment options. In this review, we will generally discuss the design of biomaterials, specifically bulk materials and composites, for the treatment of critical size bone defects. Additionally, we will highlight recent work on the design of chitosan-based scaffolds modified with purine crosslinking, to overcome cytotoxicity issues associated with other chemical crosslinkers. In this context, we focus on optimizing material design for this bone healing application and discuss the benefits of localized Wnt agonist as mediators to improve the scaffold’s osteoinductive behavior.
The chronicity of injuries is also a public health problem, and it is necessary to develop and apply new materials to promote more satisfactory results in the wound healing. Thus, this study aims to develop natural polymer films based on a combination of κ-carrageenan and sodium alginate, crosslinked with Zn2+, for the controlled delivery of mupirocin (MUP).
Vibrational spectroscopy (Raman and infrared spectroscopies) was used to characterize the chemical structure and crosslinking process. Micro-Raman imaging and scanning electron microscopy were employed to observe the spatial distribution of the polymers and morphology of the samples, respectively. The uniformity (in terms of mass, thickness, and MUP concentration) of the films, MUP release kinetics, and their bactericidal activity were subjected to analysis.
The films exhibited good uniformity in terms of thickness, mass, and quantity of MUP. However, the percentage of antibiotics was lower than that added, indicating losses during the film production process. Swelling and release kinetic studies indicated good swelling capacity of the films and controlled drug delivery process. The antibacterial activity of the films was determined against Staphylococcus aureus, Escherichia coli, Staphylococcus epidermidis, and Pseudomonas aeruginosa using the zone of inhibition method. All films produced showed activity against the growth of these bacteria.
The results illustrate the potential of employing κ-carrageenan and sodium alginate in the fabrication of polymeric films for the regulated release of MUP, with the aim of developing wound dressings that can improve wound healing outcomes.
The chronicity of injuries is also a public health problem, and it is necessary to develop and apply new materials to promote more satisfactory results in the wound healing. Thus, this study aims to develop natural polymer films based on a combination of κ-carrageenan and sodium alginate, crosslinked with Zn2+, for the controlled delivery of mupirocin (MUP).
Vibrational spectroscopy (Raman and infrared spectroscopies) was used to characterize the chemical structure and crosslinking process. Micro-Raman imaging and scanning electron microscopy were employed to observe the spatial distribution of the polymers and morphology of the samples, respectively. The uniformity (in terms of mass, thickness, and MUP concentration) of the films, MUP release kinetics, and their bactericidal activity were subjected to analysis.
The films exhibited good uniformity in terms of thickness, mass, and quantity of MUP. However, the percentage of antibiotics was lower than that added, indicating losses during the film production process. Swelling and release kinetic studies indicated good swelling capacity of the films and controlled drug delivery process. The antibacterial activity of the films was determined against Staphylococcus aureus, Escherichia coli, Staphylococcus epidermidis, and Pseudomonas aeruginosa using the zone of inhibition method. All films produced showed activity against the growth of these bacteria.
The results illustrate the potential of employing κ-carrageenan and sodium alginate in the fabrication of polymeric films for the regulated release of MUP, with the aim of developing wound dressings that can improve wound healing outcomes.
Nanoparticles (NPs) are at the forefront as they are providing unprecedented solutions to obstacles and issues in treating neurodegenerative diseases. Due to their size, surface characteristics, and ability to be functionalized, these carriers can directly deliver therapeutics across what is considered one of the main barriers to central nervous system (CNS) treatment, the blood-brain barrier (BBB). Through nano-technology, anti-disease agents such as Alzheimer’s, Parkinson’s, and Huntington’s therapies become more bioavailable, specific in action, and with fewer side effects. The NPs serve as molecular carriers that facilitate transport across the BBB by receptor-mediated transcytosis or by disruption of the barrier with a view to properly delivering drugs to the neural tissues. Some of the therapeutic applications of nanotechnology also present the concept of molecular medicine since the NPs are designed to deliver drugs in accordance with specific biomolecule signals. Besides the therapeutic applications, NPs replace the traditional contrast media for magnetic resonance imaging (MRI) and positron emission tomography (PET) scans for better diagnosis as well as disease tracking in the early stages. In addition, their effects on solubility increase the therapeutic potential of earlier useless compounds, and the preservation of bioactive molecules from degradation increases the therapeutic capacity of medications. Neurodegenerative disorders are marked by oxidative stress and inflammation that contribute to the disease severity; thus, liposomes, dendrimers, and polymeric NPs encapsulate antioxidants and anti-inflammatory compounds, so they target the areas most affected by the disease. Such sophisticated systems minimize the extension of neuronal deterioration and enhance the lot of such patients. The “theranostic” NPs allow for continuous diagnosis and treatment by containing both diagnostic and therapeutic features. These have created unprecedented opportunities to meet the unmet needs in CNS disorders and may revolutionize the evolution of managing neurodegenerative diseases and innovative neuroimaging procedures in the future.
Nanoparticles (NPs) are at the forefront as they are providing unprecedented solutions to obstacles and issues in treating neurodegenerative diseases. Due to their size, surface characteristics, and ability to be functionalized, these carriers can directly deliver therapeutics across what is considered one of the main barriers to central nervous system (CNS) treatment, the blood-brain barrier (BBB). Through nano-technology, anti-disease agents such as Alzheimer’s, Parkinson’s, and Huntington’s therapies become more bioavailable, specific in action, and with fewer side effects. The NPs serve as molecular carriers that facilitate transport across the BBB by receptor-mediated transcytosis or by disruption of the barrier with a view to properly delivering drugs to the neural tissues. Some of the therapeutic applications of nanotechnology also present the concept of molecular medicine since the NPs are designed to deliver drugs in accordance with specific biomolecule signals. Besides the therapeutic applications, NPs replace the traditional contrast media for magnetic resonance imaging (MRI) and positron emission tomography (PET) scans for better diagnosis as well as disease tracking in the early stages. In addition, their effects on solubility increase the therapeutic potential of earlier useless compounds, and the preservation of bioactive molecules from degradation increases the therapeutic capacity of medications. Neurodegenerative disorders are marked by oxidative stress and inflammation that contribute to the disease severity; thus, liposomes, dendrimers, and polymeric NPs encapsulate antioxidants and anti-inflammatory compounds, so they target the areas most affected by the disease. Such sophisticated systems minimize the extension of neuronal deterioration and enhance the lot of such patients. The “theranostic” NPs allow for continuous diagnosis and treatment by containing both diagnostic and therapeutic features. These have created unprecedented opportunities to meet the unmet needs in CNS disorders and may revolutionize the evolution of managing neurodegenerative diseases and innovative neuroimaging procedures in the future.
Silver iodide (AgI) nanostructures have been considered as promising candidates for optical biosensors owing to their optical characteristics of optical properties, including tunable surface plasmon resonance (SPR) and fluorescence enhancement. Such properties let one analyze biomolecules with high sensitivity, which makes them ultra-useful in diagnostics. The formed AgI nanostructures can be synthesized using chemical precipitation and template methods that enable fine-tuning of the morphology and crystallinity of the final nanostructure. The presence of SPR enhances optical signals potentially, and fluorescence enhancement helps visualize biomolecule interactions easier as the analyte concentration is usually low. Such uses of biosensors include applications in proteins, nucleic acids, and other biomolecules for progress in disease diagnosis and pharmacogenomics. Moreover, the good biocompatibility level of the created AgI nanostructures makes it possible to integrate them into biological systems safely, increasing their usage in medicine. This integration of their appealing optics, biosensing operating principles, and biocompatibility establishes their centrality in the creation of future photonic biosensors for faster, intuitive, and painless detection.
Silver iodide (AgI) nanostructures have been considered as promising candidates for optical biosensors owing to their optical characteristics of optical properties, including tunable surface plasmon resonance (SPR) and fluorescence enhancement. Such properties let one analyze biomolecules with high sensitivity, which makes them ultra-useful in diagnostics. The formed AgI nanostructures can be synthesized using chemical precipitation and template methods that enable fine-tuning of the morphology and crystallinity of the final nanostructure. The presence of SPR enhances optical signals potentially, and fluorescence enhancement helps visualize biomolecule interactions easier as the analyte concentration is usually low. Such uses of biosensors include applications in proteins, nucleic acids, and other biomolecules for progress in disease diagnosis and pharmacogenomics. Moreover, the good biocompatibility level of the created AgI nanostructures makes it possible to integrate them into biological systems safely, increasing their usage in medicine. This integration of their appealing optics, biosensing operating principles, and biocompatibility establishes their centrality in the creation of future photonic biosensors for faster, intuitive, and painless detection.