Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 548
Filter
1.
Int J Biol Macromol ; 272(Pt 2): 132830, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825264

ABSTRACT

Artificial graft serves as the primary grafts used in the clinical management of sports-related injuries. Until now, optimizing its graft-host integration remains a great challenge due to the excessive inflammatory response during the inflammatory phase, coupled with an absence of tissue-inductive capacity during the regeneration phase. Here, a multi-layered regenerated silk fibroin (RSF) coating loaded with curcumin (Cur) and Zn2+ on the surface of the PET grafts (Cur@Zn2+@PET) was designed and fabricated for providing time-matched regulation specifically tailored to address issues arising at both inflammatory and regeneration phases, respectively. The release of Cur and Zn2+ from the Cur@Zn2+@PET followed a time-programmed pattern in vitro. Specifically, cellular assays revealed that Cur@Zn2+@PET initially released Cur during the inflammatory phase, thereby markedly inhibit the expression of inflammatory cytokines TNF-a and IL-1ß. Meanwhile, a significant release of Zn2+ was major part during the regeneration phase, serving to induce the osteogenic differentiation of rBMSC. Furthermore, rat model of anterior cruciate ligament reconstruction (ACLR) showed that through time-programmed drug release, Cur@Zn2+@PET could suppress the formation of fibrous interface (FI) caused by inflammatory response, combined with significant new bone (NB) formation during regeneration phase. Consequently, the implementation of the Cur@Zn2+@PET characterized by its time-programmed release patterns hold considerable promise for improving graft-host integration for sports-related injuries.


Subject(s)
Curcumin , Fibroins , Zinc , Curcumin/pharmacology , Curcumin/chemistry , Animals , Zinc/chemistry , Zinc/pharmacology , Rats , Fibroins/chemistry , Fibroins/pharmacology , Drug Liberation , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Male , Osteogenesis/drug effects , Rats, Sprague-Dawley
2.
Transl Res ; 271: 26-39, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38734063

ABSTRACT

Peptide drug discovery for the treatment of chronic kidney disease (CKD) has attracted much attention in recent years due to the urge to find novel drugs and mechanisms to delay the progression of the disease. In this study, we identified a novel short peptide (named YR-7, primary sequence 'YEVEDYR') from the natural Fibroin protein, and demonstrated that it significantly alleviated pathological renal changes in ADR-induced nephropathy. PANX1 was identified as the most notably upregulated component by RNA-sequencing. Further analysis showed that YR-7 alleviated the accumulation of lipid droplets via regulation of the lipid metabolism-related proteins PPAR α and PANK1. Using chemical proteomics, fluorescence polarization, microscale thermophoresis, surface plasmon resonance, and molecular docking, YR-7 was proven to directly bind to ß-barrel domains of TGM2 protein to inhibit lipid accumulation. TGM2 knockdown in vivo increased the protein levels of PPAR α and PANK1 while decreased the levels of fibrotic-related proteins to alleviate nephropathy. In vitro, overexpression TGM2 reversed the protective effects of YR-7. Co-immunoprecipitation indicated that TGM2 interacted with PANX1 to promote lipid deposition, and pharmacological inhibition or knockdown of PANX1 decreased the levels of PPAR α and PANK1 induced by ADR. Taken together, our findings revealed that TGM2-PANX1 interaction in promoting lipid deposition may be a new signaling in promoting ADR-induced nephropathy. And a novel natural peptide could ameliorate renal fibrosis through TGM2-PANX1-PPAR α/PANK1 pathway, which highlight the potential of it in the treatment of CKD.


Subject(s)
Doxorubicin , Fibroins , Lipid Metabolism , PPAR alpha , Protein Glutamine gamma Glutamyltransferase 2 , Animals , PPAR alpha/metabolism , PPAR alpha/genetics , Lipid Metabolism/drug effects , Male , Fibroins/chemistry , Fibroins/pharmacology , Signal Transduction/drug effects , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Peptides/pharmacology , Peptides/chemistry , Rats , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Rats, Sprague-Dawley
3.
J Mater Chem B ; 12(22): 5439-5454, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38726947

ABSTRACT

Powder-based hemostatic technology has offered unprecedented opportunities in surgical sealing and repair of irregularly shaped and noncompressible wounds. Despite their routine use, existing clinical hemostatic powders are challenged either by poor mechanical properties or inadequate adhesion to bleeding tissues in biological environments. Here, inspired by the mussel foot proteins' fusion assembly strategy, a novel silk fibroin-based hemostatic powder (named as SF/PEG/TA) with instant and robust adhesion performance is developed. Upon absorbing interfacial liquids, the SF/PEG/TA powders rapidly swell into micro-gels and subsequently contact with each other to transform into a macroscopically homogeneous hydrogel in situ, strengthening its interfacial bonding with various substrates in fluidic environments. The in vitro and in vivo results show that the SF/PEG/TA powder possesses ease of use, good biocompatibility, strong antibacterial activities, and effective blood clotting abilities. The superior hemostatic sealing capability of the SF/PEG/TA powder is demonstrated in the rat liver, heart, and gastrointestinal injury models. Moreover, in vivo investigation of rat skin incision and gastrointestinal perforation models validates that the SF/PEG/TA powder promotes wound healing and tissue regeneration. Taken together, compared to existing clinical hemostatic powders, the proposed SF/PEG/TA powder with superior wound treatment capabilities has high potential for clinical hemostasis and emergency rescue.


Subject(s)
Fibroins , Hemostatics , Powders , Rats, Sprague-Dawley , Fibroins/chemistry , Fibroins/pharmacology , Animals , Hemostatics/chemistry , Hemostatics/pharmacology , Rats , Wound Healing/drug effects , Male , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Humans , Polyethylene Glycols/chemistry
4.
Nat Commun ; 15(1): 4160, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755128

ABSTRACT

The regeneration of critical-size bone defects, especially those with irregular shapes, remains a clinical challenge. Various biomaterials have been developed to enhance bone regeneration, but the limitations on the shape-adaptive capacity, the complexity of clinical operation, and the unsatisfied osteogenic bioactivity have greatly restricted their clinical application. In this work, we construct a mechanically robust, tailorable and water-responsive shape-memory silk fibroin/magnesium (SF/MgO) composite scaffold, which is able to quickly match irregular defects by simple trimming, thus leading to good interface integration. We demonstrate that the SF/MgO scaffold exhibits excellent mechanical stability and structure retention during the degradative process with the potential for supporting ability in defective areas. This scaffold further promotes the proliferation, adhesion and migration of osteoblasts and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro. With suitable MgO content, the scaffold exhibits good histocompatibility, low foreign-body reactions (FBRs), significant ectopic mineralisation and angiogenesis. Skull defect experiments on male rats demonstrate that the cell-free SF/MgO scaffold markedly enhances bone regeneration of cranial defects. Taken together, the mechanically robust, personalised and bioactive scaffold with water-responsive shape-memory may be a promising biomaterial for clinical-size and irregular bone defect regeneration.


Subject(s)
Biocompatible Materials , Bone Regeneration , Fibroins , Magnesium , Mesenchymal Stem Cells , Osteogenesis , Tissue Scaffolds , Fibroins/chemistry , Fibroins/pharmacology , Bone Regeneration/drug effects , Animals , Tissue Scaffolds/chemistry , Male , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Rats , Magnesium/chemistry , Magnesium/pharmacology , Biocompatible Materials/chemistry , Osteoblasts/drug effects , Cell Differentiation/drug effects , Rats, Sprague-Dawley , Water/chemistry , Cell Proliferation/drug effects , Tissue Engineering/methods , Skull/drug effects , Cell Adhesion/drug effects , Bombyx
5.
Biofabrication ; 16(3)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38776895

ABSTRACT

Silk fibroin (SF) is a natural protein extracted fromBombyx morisilkworm thread. From its common use in the textile industry, it emerged as a biomaterial with promising biochemical and mechanical properties for applications in the field of tissue engineering and regenerative medicine. In this study, we evaluate for the first time the effects of SF on cardiac bioink formulations containing cardiac spheroids (CSs). First, we evaluate if the SF addition plays a role in the structural and elastic properties of hydrogels containing alginate (Alg) and gelatin (Gel). Then, we test the printability and durability of bioprinted SF-containing hydrogels. Finally, we evaluate whether the addition of SF controls cell viability and function of CSs in Alg-Gel hydrogels. Our findings show that the addition of 1% (w/v) SF to Alg-Gel hydrogels makes them more elastic without affecting cell viability. However, fractional shortening (FS%) of CSs in SF-Alg-Gel hydrogels increases without affecting their contraction frequency, suggesting an improvement in contractile function in the 3D cultures. Altogether, our findings support a promising pathway to bioengineer bioinks containing SF for cardiac applications, with the ability to control mechanical and cellular features in cardiac bioinks.


Subject(s)
Alginates , Elasticity , Fibroins , Gelatin , Hydrogels , Myocytes, Cardiac , Alginates/chemistry , Alginates/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Gelatin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Bioprinting , Cell Survival/drug effects , Tissue Engineering , Ink , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Rats , Myocardial Contraction/drug effects
6.
Int J Artif Organs ; 47(5): 338-346, 2024 May.
Article in English | MEDLINE | ID: mdl-38693724

ABSTRACT

In the present study, porous silk fibroin sponges (SFS) were prepared using silk fibroin (SF), fish bone collagen (FBC), and olive oil (OO). The study investigates the potential use of using this sponge as skin tissue regeneration. The sponge was characterized for its physicochemical, mechanical, antimicrobial, and drug release properties. An in vitro study was carried out using human keratinocyte cell line (HaCaT). Biodegradation study using enzymatic method was carried out. The results showed that the mechanical properties such as tensile strength (23.40 ± 0.05 MPa), elongation at break (14.25 ± 0.02%), and water absorption (30.23 ± 0.01%) of the SFS were excellent, indicating promising performance. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays proved the biocompatible nature of the SFS. The SFS exhibited outstanding antibacterial properties against E. coli (4.72 ± 0.05 mm) and S. aureus (4.98 ± 0.07 mm). The developed SFS promote a promising solution for skin tissue regeneration and wound dressing.


Subject(s)
Anti-Bacterial Agents , Collagen , Fibroins , Regeneration , Skin , Staphylococcus aureus , Tissue Scaffolds , Wound Healing , Fibroins/chemistry , Fibroins/pharmacology , Wound Healing/drug effects , Humans , Collagen/metabolism , Animals , Regeneration/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Skin/drug effects , Skin/metabolism , Staphylococcus aureus/drug effects , HaCaT Cells , Escherichia coli/drug effects , Keratinocytes/drug effects , Olive Oil , Bone and Bones/drug effects , Bone and Bones/metabolism , Fishes , Tensile Strength , Porosity , Biocompatible Materials , Cell Line
7.
J Colloid Interface Sci ; 668: 646-657, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38696992

ABSTRACT

Severe spinal cord injury (SCI) leads to dysregulated neuroinflammation and cell apoptosis, resulting in axonal die-back and the loss of neuroelectric signal transmission. While biocompatible hydrogels are commonly used in SCI repair, they lack the capacity to support neuroelectric transmission. To overcome this limitation, we developed an injectable silk fibroin/ionic liquid (SFMA@IL) conductive hydrogel to assist neuroelectric signal transmission after SCI in this study. The hydrogel can form rapidly in situ under ultraviolet (UV) light. The mechanical supporting and neuro-regenerating properties are provided by silk fibroin (SF), while the conductive capability is provided by the designed ionic liquid (IL). SFMA@IL showed attractive features for SCI repair, such as anti-swelling, conductivity, and injectability. In vivo, SFMA@IL hydrogel used in rats with complete transection injuries was found to remodel the microenvironment, reduce inflammation, and facilitate neuro-fiber outgrowth. The hydrogel also led to a notable decrease in cell apoptosis and the achievement of scar-free wound healing, which saved 45.6 ± 10.8 % of spinal cord tissue in SFMA@IL grafting. Electrophysiological studies in rats with complete transection SCI confirmed SFMA@IL's ability to support sensory neuroelectric transmission, providing strong evidence for its signal transmission function. These findings provide new insights for the development of effective SCI treatments.


Subject(s)
Electric Conductivity , Fibroins , Hydrogels , Rats, Sprague-Dawley , Spinal Cord Injuries , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/pathology , Animals , Rats , Hydrogels/chemistry , Hydrogels/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Injections , Apoptosis/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Particle Size
8.
ACS Appl Mater Interfaces ; 16(17): 21582-21594, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38634578

ABSTRACT

Excessive blood loss and infections are the prominent risks accounting for mortality and disability associated with acute wounds. Consequently, wound dressings should encompass adequate adhesive, hemostatic, and bactericidal attributes, yet their development remains challenging. This investigation presented the benefits of incorporating a perfluorocarbon nanoemulsion (PPP NE) into a silk-fibroin (SF)-based hydrogel. By stimulating the ß-sheet conformation of the SF chains, PPP NEs drastically shortened the gelation time while augmenting the elasticity, mechanical stability, and viscosity of the hydrogel. Furthermore, the integration of PPP NEs improved hemostatic competence by boosting the affinity between cells and biomacromolecules. It also endowed the hydrogel with ultrasound-controlled bactericidal ability through the inducement of inner cavitation by perfluorocarbon and reactive oxygen species (ROS) generated by the sonosensitizer protoporphyrin. Ultimately, we employed a laparotomy bleeding model and a Staphylococcus aureus-infected trauma wound to demonstrate the first-aid efficacy. Thus, our research suggested an emulsion-incorporating strategy for managing emergency wounds.


Subject(s)
Anti-Bacterial Agents , Emulsions , Fibroins , Fluorocarbons , Hydrogels , Staphylococcus aureus , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Emulsions/chemistry , Emulsions/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Fibroins/chemistry , Fibroins/pharmacology , Mice , Hemostatics/chemistry , Hemostatics/pharmacology , Nanoparticles/chemistry , Staphylococcal Infections/drug therapy , Ultrasonic Waves , Male , Rats , Humans
9.
Mater Horiz ; 11(13): 3157-3165, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38629215

ABSTRACT

Magnetic propulsion of nano-/micro-robots is an effective way to treat implant-associated infections by physically destroying biofilm structures to enhance antibiotic killing. However, it is hard to precisely control the propulsion in vivo. Magnetic-nanoparticle coating that can be magnetically pulled off does not need precise control, but the requirement of adhesion stability on an implant surface restricts its magnetic responsiveness. Moreover, whether the coating has been fully pulled-off or not is hard to ensure in real-time in vivo. Herein, composited silk fibroins (SFMA) are optimized to stabilize Fe3O4 nanoparticles on a titanium surface in a dry environment; while in an aqueous environment, the binding force of SFMA on titanium is significantly reduced due to hydrophilic interaction, making the coating magnetically controllable by an externally-used magnet but still stable in the absence of a magnet. The maximum working distance of the magnet can be calculated using magnetomechanical simulation in which the yielding magnetic traction force is strong enough to pull Fe3O4 nanoparticles off the surface. The pulling-off removes the biofilms that formed on the coating and enhances antibiotic killing both in vitro and in a rat sub-cutaneous implant model by up to 100 fold. This work contributes to the practical knowledge of magnetic propulsion for biofilm treatment.


Subject(s)
Biofilms , Fibroins , Titanium , Biofilms/drug effects , Animals , Rats , Titanium/pharmacology , Titanium/chemistry , Fibroins/chemistry , Fibroins/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Magnetite Nanoparticles/therapeutic use , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Prostheses and Implants , Rats, Sprague-Dawley , Surface Properties , Staphylococcus aureus/drug effects
10.
Biomater Adv ; 160: 213856, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640877

ABSTRACT

Large bone defects cause significant clinical challenges due to the lack of optimal grafts for effective regeneration. The tissue engineering way that requires the combination of biomaterials scaffold, stem cells and proper bioactive factors is a prospective method for large bone repair. Here, we synthesized a three-arm host-guest supramolecule (HGSM) to covalently crosslinking with the naturally derived polymer methacrylated silk fibroin (SFMA). The combination of HGSM and SFMA can form a high strength double-crosslinked hydrogel HGSFMA, that serve as the hydrogel scaffold for bone marrow mesenchymal stem cells (BMSCs) growing. Icariin (ICA) loaded in the HGSFMA hydrogel can promote the osteogenesis efficiency of BMSCs and inhibit the osteoclasts differentiation. Our findings demonstrated that the HGSFMA/ICA hydrogel effectively promoted the in vitro adhesion, proliferation, and osteogenic differentiation of BMSCs. Rat femoral defects model show that this hydrogel can completely repair femoral damage within 4 weeks and significantly promote the secretion of osteogenesis-related proteins. In summary, we have prepared an effective biomimetic bone carrier, offering a novel strategy for bone regeneration and the treatment of large-scale bone defects.


Subject(s)
Bone Regeneration , Cell Differentiation , Fibroins , Flavonoids , Hydrogels , Mesenchymal Stem Cells , Osteoclasts , Osteogenesis , Fibroins/chemistry , Fibroins/pharmacology , Animals , Osteogenesis/drug effects , Flavonoids/pharmacology , Flavonoids/chemistry , Flavonoids/administration & dosage , Cell Differentiation/drug effects , Bone Regeneration/drug effects , Mesenchymal Stem Cells/drug effects , Osteoclasts/drug effects , Rats , Hydrogels/chemistry , Hydrogels/pharmacology , Rats, Sprague-Dawley , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Cell Proliferation/drug effects
11.
Int J Biol Macromol ; 266(Pt 2): 131357, 2024 May.
Article in English | MEDLINE | ID: mdl-38580010

ABSTRACT

The microenvironment of bone defect site is vital for bone regeneration. Severe bone defect is often accompanied with severe inflammation and elevated generation of reactive oxygen species (ROS) during bone repair. In recent years, the unfriendly local microenvironment has been paid more and more attention. Some bioactive materials with the ability to regulate the microenvironment to promote bone regeneration urgently need to be developed. Here, we develop a multifunctional composite hydrogel composed of photo-responsive methacrylate silk fibroin (SFMA), laponite (LAP) nanocomposite and tannic acid (TA), aiming to endow hydrogel with antioxidant, anti-inflammatory and osteogenic induction ability. Characterization results confirmed that the SFMA-LAP@TA hydrogel could significantly improve the mechanical properties of hydrogel. The ROS-Scavenging ability of the hydrogel enabled bone marrow mesenchymal stem cells (BMSCs) to survive against H2O2-induced oxidative stress. In addition, the SFMA-LAP@TA hydrogel effectively decreased the expression of pro-inflammatory factors in RAW264.7. More importantly, the SFMA-LAP@TA hydrogel could enhance the expression of osteogenic markers of BMSCs under inflammatory condition and greatly promote new bone formation in a critical-sized cranial defect model. Above all, the multifunctional hydrogel could effectively promote bone regeneration in vitro and in vivo by scavenging ROS and reducing inflammation, providing a prospective strategy for bone regeneration.


Subject(s)
Bone Regeneration , Fibroins , Hydrogels , Inflammation , Mesenchymal Stem Cells , Nanocomposites , Osteogenesis , Polyphenols , Reactive Oxygen Species , Tannins , Bone Regeneration/drug effects , Animals , Fibroins/chemistry , Fibroins/pharmacology , Reactive Oxygen Species/metabolism , Tannins/chemistry , Tannins/pharmacology , Mice , Inflammation/drug therapy , Nanocomposites/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , RAW 264.7 Cells , Osteogenesis/drug effects , Methacrylates/chemistry , Methacrylates/pharmacology , Rats , Oxidative Stress/drug effects , Free Radical Scavengers/pharmacology , Free Radical Scavengers/chemistry
12.
Molecules ; 29(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38675654

ABSTRACT

Diabetic wound healing is a significant clinical challenge because abnormal immune cells in the wound cause chronic inflammation and impair tissue regeneration. Therefore, regulating the behavior and function of macrophages may be conducive to improving treatment outcomes in diabetic wounds. Herein, sulfated chitosan (26SCS)-containing composite sponges (26SCS-SilMA/Col-330) with well-arranged layers and high porosity were constructed based on collagen and silk fibroin, aiming to induce an appropriate inflammatory response and promote angiogenesis. The results indicated that the ordered topological structure of composite sponges could trigger the pro-inflammatory response of Mφs in the early stage, and rapid release of 26SCS in the early and middle stages (within the concentration range of 1-3 mg/mL) induced a positive inflammatory response; initiated the pro-inflammatory reaction of Mφs within 3 days; shifted M1 Mφs to the M2 phenotype within 3-7 days; and significantly up-regulated the expression of two typical angiogenic growth factors, namely VEGF and PDGF-BB, on day 7, leading to rapid HUVEC migration and angiogenesis. In vivo data also demonstrated that on the 14th day after surgery, the 26SCS-SilMA/Col-330-implanted areas exhibited less inflammation, faster re-epithelialization, more abundant collagen deposition and a greater number of blood vessels in the skin tissue. The composite sponges with higher 26SCS contents (the (5.0) 26SCS-SilMA/Col-330 and the (7.5) 26SCS-SilMA/Col-330) could better orchestrate the phenotype and function of Mφs and facilitate wound healing. These findings highlight that the 26SCS-SilMA/Col-330 sponges developed in this work might have great potential as a novel dressing for the treatment of diabetic wounds.


Subject(s)
Chitosan , Inflammation , Macrophages , Neovascularization, Physiologic , Wound Healing , Wound Healing/drug effects , Chitosan/chemistry , Animals , Humans , Macrophages/drug effects , Macrophages/metabolism , Neovascularization, Physiologic/drug effects , Inflammation/drug therapy , Inflammation/pathology , Human Umbilical Vein Endothelial Cells , Collagen/metabolism , Collagen/chemistry , Diabetes Mellitus, Experimental , Mice , Rats , Male , Fibroins/chemistry , Fibroins/pharmacology , Angiogenesis
13.
Colloids Surf B Biointerfaces ; 238: 113889, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38574404

ABSTRACT

A novel core-shell with a tetradecyl dimethyl benzyl ammonium chloride-modified montmorillonite (TDMBA/MMT) interlayer silk fibroin (SF)/poly(lactic acid) (PLLA) nanofibrous membrane was fabricated using a simple conventional electrospinning method. Scanning electron microscopy and pore size analyses revealed that this core-shell with TDMBA/MMT interlayer maintained its nanofibrous morphology and larger pore structure more successfully than SF/PLLA nanofibrous membranes after treatment with 75% ethanol vapor. Transmission electron microscopy and energy-dispersive X-ray spectroscopy analyses testified that the SF/PLLA-TDMBA/MMT nanofibers exhibited a core-shell with an interlayer structure, with SF/PLLA in the core-shell layer and TDMBA/MMT in the interlayer. The formation of a core-shell with interlayer nanofibers was primarily attributed to the uniform dispersion of TDMBA/MMT nanosheets in a solution owing to its exfoliation using hexafluoroisopropanol and then preparing a stable spinning solution similar to an emulsion. Compared to SF/PLLA nanofibrous membranes, the core-shell structure with TDMBA/MMT interlayers of SF/PLLA nanofibrous membranes exhibited enhanced hydrophilicity, thermal stability, mechanical properties as well as improved and long-lasting antimicrobial performance against Escherichia coli and Staphylococcus aureus without cytotoxicity.


Subject(s)
Bentonite , Escherichia coli , Nanofibers , Staphylococcus aureus , Bentonite/chemistry , Bentonite/pharmacology , Nanofibers/chemistry , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Polyesters/chemistry , Polyesters/pharmacology , Membranes, Artificial , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Mice , Animals
14.
Biomacromolecules ; 25(4): 2597-2606, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38483111

ABSTRACT

The development of highly effective hemostatic materials with high biocompatibility and outstanding performance is vital to the field of biomaterials. In this study, we develop a hemostatic fiber material that exhibits high biocompatibility and excellent performance. By incorporating polydopamine (PDA) into the alkaline treatment of silk fibroin (SF), we achieve PDA-coated SF fibers with lengths that can be controlled by the alkaline concentration. The PDA coating significantly enhances the hemostatic ability of the silk fibers and exhibits superior performance in both in vitro and ex vivo experiments. By performing animal studies involving a mouse liver puncture model and a femoral vein incision model, we demonstrate the remarkable hemostatic capability of the PDA-coated SF fibers, as evidenced by the lower blood loss compared to that of a commercial hemostat powder. These findings highlight the potential of applying a PDA-assisted alkaline treatment to SF fibers to efficiently create hemostatic fibers with controllable lengths, which would be promising candidates for clinical hemostatic applications.


Subject(s)
Fibroins , Hemostatics , Indoles , Mice , Animals , Silk , Hemostatics/pharmacology , Polymers/pharmacology , Biocompatible Materials , Fibroins/pharmacology
15.
ACS Appl Bio Mater ; 7(4): 2389-2401, 2024 04 15.
Article in English | MEDLINE | ID: mdl-38502100

ABSTRACT

Graphene-based nanomaterials, renowned for their outstanding electrical conductivity, have been extensively studied as electroconductive biomaterials (ECBs) for electrically stimulated tissue regeneration. However, using eco-friendly reducing agents like l-ascorbic acid (l-Aa) can result in lower conductive properties in these ECBs, limiting their full potential for smooth charge transfer in living tissues. Moreover, creating a flexible biomaterial scaffold using these materials that accurately mimics a specific tissue microarchitecture, such as nerves, poses additional challenges. To address these issues, this study developed a microfibrous scaffold of Bombyx mori (Bm) silk fibroin uniformly coated with graphene nanoplatelets (GNPs) through a vacuum coating method. The scaffold's electrical conductivity was optimized by varying the reduction period using l-Aa. The research systematically investigated how different reduction periods impact scaffold properties, focusing on electrical conductivity and its significance on electrically stimulated axonal growth in PC12 cells. Results showed that a 48 h reduction significantly increased surface electrical conductivity by 100-1000 times compared to a shorter or no reduction process. l-Aa contributed to stabilizing the reduced GNPs, demonstrated by a slow degradation profile and sustained conductivity even after 60 days in a proteolytic environment. ß (III) tubulin immunostaining of PC12 cells on varied silk:GNP scaffolds under pulsed electrical stimulation (ES, 50 Hz frequency, 1 ms pulse width, and amplitudes of 100 and 300 mV/cm) demonstrates accelerated axonal growth on scaffolds exhibiting higher conductivity. This is supported by upregulated intracellular Ca2+ dynamics immediately after ES on the scaffolds with higher conductivity, subjected to a prolonged reduction period. The study showcases a sustainable reduction approach using l-Aa in combination with natural Bm silk fibroin to create a highly conductive, mechanically robust, and stable silk:GNP-based aligned fibrous scaffold. These scaffolds hold promise for functional regeneration in electrically excitable tissues such as nerves, cardiac tissue, and muscles.


Subject(s)
Bombyx , Fibroins , Graphite , Rats , Animals , Silk , Tissue Scaffolds , Graphite/pharmacology , Fibroins/pharmacology , Biocompatible Materials/pharmacology , Electric Conductivity
16.
J Biomed Mater Res B Appl Biomater ; 112(3): e35396, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38433653

ABSTRACT

Development of osteochondral tissue engineering approaches using scaffolds seeded with stem cells in association with mechanical stimulations has been recently considered as a promising technique for the repair of this tissue. In this study, an integrated and biomimetic trilayered silk fibroin (SF) scaffold containing SF nanofibers in each layer was fabricated. The osteogenesis and chondrogenesis of stem cells seeded on the fabricated scaffolds were investigated under a perfusion flow. 3-Dimethylthiazol-2,5-diphenyltetrazolium bromide assay showed that the perfusion flow significantly enhanced cell viability and proliferation. Analysis of gene expression by stem cells revealed that perfusion flow had significantly upregulated the expression of osteogenic and chondrogenic genes in the bone and cartilage layers and downregulated the hypertrophic gene expression in the intermediate layer of the scaffold. In conclusion, applying flow perfusion on the prepared integrated trilayered SF-based scaffold can support osteogenic and chondrogenic differentiation for repairing osteochondral defects.


Subject(s)
Fibroins , Animals , Rabbits , Fibroins/pharmacology , Perfusion , Adipocytes , Biological Assay , Stem Cells
17.
Int J Biol Macromol ; 266(Pt 1): 131167, 2024 May.
Article in English | MEDLINE | ID: mdl-38547948

ABSTRACT

Blend polymers composed of natural polymers are a ubiquitous biomaterial class due to their suitable mechanical and biological characterization. In the present study, composite scaffolds based on bacterial cellulose (BC)/silk fibroin (SF) with bioactive glass nanoparticles (BGNPs) were developed to enhance osteogenesis in human adipose derived stem cells (hASCs). The scanning electron microscopy (SEM) results of BGNPs indicated a spherical morphology and size ranging from 15 to 30 nm. The presence of BC and BGNPs reduced the pore diameter of SF scaffolds to about 210 ± 10 µm and 205 ± 10 µm, respectively, while increasing their compressive strength and compressive modulus. FTIR analyses proved the presence of BGNPs, BC and SF in the scaffolds. Flow cytometry data confirmed the surface markers for hASCs. The results also showed that BC and BGNPs addition to BC/SF scaffolds decreased degradation and swelling rate. The gene expression (Runx2, alkaline phosphatase and osteocalcin) studies signified the osteogenic potential of BGNPs in BC/SF scaffolds on hASCs. Eventually, the increased cell adhesion, viability and differentiation in the BC/SF and BC/SF/BGNPs composite scaffolds drawn from MTT, SEM, Alizarin red staining and alkaline phosphatase activity confirmed that these scaffolds promise to serve as a therapeutic candidate for bone defects.


Subject(s)
Cellulose , Fibroins , Nanoparticles , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Fibroins/chemistry , Fibroins/pharmacology , Osteogenesis/drug effects , Tissue Scaffolds/chemistry , Cellulose/chemistry , Cellulose/pharmacology , Humans , Tissue Engineering/methods , Nanoparticles/chemistry , Glass/chemistry , Cell Differentiation/drug effects , Bone and Bones/drug effects , Bone and Bones/cytology , Bone and Bones/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Cell Adhesion/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Alkaline Phosphatase/metabolism
18.
J Nanobiotechnology ; 22(1): 111, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38486273

ABSTRACT

Brain damage is a common tissue damage caused by trauma or diseases, which can be life-threatening. Stem cell implantation is an emerging strategy treating brain damage. The stem cell is commonly embedded in a matrix material for implantation, which protects stem cell and induces cell differentiation. Cell differentiation induction by this material is decisive in the effectiveness of this treatment strategy. In this work, we present an injectable fibroin/MXene conductive hydrogel as stem cell carrier, which further enables in-vivo electrical stimulation upon stem cells implanted into damaged brain tissue. Cell differentiation characterization of stem cell showed high effectiveness of electrical stimulation in this system, which is comparable to pure conductive membrane. Axon growth density of the newly differentiated neurons increased by 290% and axon length by 320%. In addition, unfavored astrocyte differentiation is minimized. The therapeutic effect of this system is proved through traumatic brain injury model on rats. Combined with in vivo electrical stimulation, cavities formation is reduced after traumatic brain injury, and rat motor function recovery is significantly promoted.


Subject(s)
Bombyx , Brain Injuries, Traumatic , Fibroins , Mesenchymal Stem Cells , Neural Stem Cells , Nitrites , Transition Elements , Rats , Animals , Fibroins/metabolism , Fibroins/pharmacology , Bombyx/metabolism , Hydrogels/pharmacology , Neurons/metabolism , Brain/metabolism , Brain Injuries, Traumatic/metabolism
19.
Carbohydr Polym ; 334: 121972, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38553198

ABSTRACT

Chronic wounds with bacterial infection present formidable clinical challenges. In this study, a versatile hydrogel dressing with antibacterial and angiogenic activity composite of silk fibroin (SF), chondroitin sulfate (CS), and graphene oxide quantum dots (GOQDs) is fabricated. GOQDs@SF/CS (GSC) hydrogel is rapidly formed through the enzyme catalytic action of horseradish peroxidase. With the incorporation of GOQDs both gelation speed and mechanical properties have been enhanced, and the photothermal characteristics of GOQDs in GSC hydrogel enabled bacterial killing through photothermal treatment (PTT) at ∼51 °C. In vitro studies show that the GSC hydrogels demonstrate excellent antibacterial performance and induce type H vessel differentiation of endothelial cells via the activated ERK1/2 signaling pathway and upregulated SLIT3 expression. In vivo results show that the hydrogel significantly promotes type H vessels formation, which is related to the collagen deposition, epithelialization and, ultimately, accelerates the regeneration of infected skin defects. Collectively, this multifunctional GSC hydrogel, with dual action of antibacterial efficacy and angiogenesis promotion, emerges as an innovative skin dressing with the potential for advancing in infected wound healing.


Subject(s)
Fibroins , Graphite , Quantum Dots , Fibroins/pharmacology , Chondroitin Sulfates/pharmacology , Hydrogels/pharmacology , Endothelial Cells , Wound Healing , Anti-Bacterial Agents/pharmacology
20.
Int J Biol Macromol ; 264(Pt 2): 130687, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38462112

ABSTRACT

Silk fibroin derived from silkworm cocoons exhibits excellent mechanical properties, good biocompatibility, and low immunogenicity. Previous studies showed that silk fibroin had an inhibitory effect on cells, suppressing proliferation and inducing apoptosis. However, the source of the toxicity and the mechanism of apoptosis induction are still unclear. In this study, we hypothesized that the toxicity of silk fibroin might originate from the crystalline region of the heavy chain of silk fibroin. We then verified the hypothesis and the specific induction mechanism. A target peptide segment was obtained from α-chymotrypsin. The potentially toxic mixture of silk fibroin peptides (SFPs) was separated by ion exchange, and the toxicity was tested by an MTT assay. The results showed that SFPs obtained after 4 h of enzymatic hydrolysis had significant cytotoxicity, and SFPs with isoelectric points of 4.0-6.8 (SFPα II) had a significant inhibitory effect on cell growth. LC-MS/MS analysis showed that SFPα II contained a large number of glycine-rich and alanine-rich repetitive sequence polypeptides from the heavy-chain crystallization region. A series of experiments showed that SFPα II mediated cell death through the apoptotic pathway by decreasing the expression of Bcl-2 protein and increasing the expression of Bax protein. SFPα II mainly affected the p53 pathway and the AMPK signaling pathway in HepG2 cells. SFPα II may indirectly increase the expression of Cers2 by inhibiting the phosphorylation of EGFR, which activated apoptotic signaling in the cellular mitochondrial pathway and inhibited the Akt/NF-κB pathway by increasing the expression of PPP2R2A.


Subject(s)
Bombyx , Fibroins , Animals , Fibroins/pharmacology , Fibroins/chemistry , Chromatography, Liquid , Tandem Mass Spectrometry , Peptides/pharmacology , Peptides/chemistry , Bombyx/chemistry , Apoptosis , Silk/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...