Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 317
Filtrar
1.
ACS Biomater Sci Eng ; 10(5): 3164-3172, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38671385

RESUMO

Intestinal adhesion is one of the complications that occurs more frequently after abdominal surgery. Postsurgical intestinal adhesion (PIA) can lead to a series of health problems, including abdominal pain, intestinal obstruction, and female infertility. Currently, hydrogels and nanofibrous films as barriers are often used for preventing PIA formation; however, these kinds of materials have their intrinsic disadvantages. Herein, we developed a dual-structure drug delivery patch consisting of poly lactic-co-glycolic acid (PLGA) nanofibers and a chitosan hydrogel (NHP). PLGA nanofibers loaded with deferoxamine mesylate (DFO) were incorporated into the hydrogel; meanwhile, the hydrogel was loaded with anti-inflammatory drug dexamethasone (DXMS). The rapid degradation of the hydrogel facilitated the release of DXMS at the acute inflammatory stage of the early injury and provided effective anti-inflammatory effects for wound sites. Moreover, PLGA composite nanofibers could provide sustained and stable release of DFO for promoting the peritoneal repair by the angiogenesis effects of DFO. The in vivo results indicated that NHP can effectively prevent PIA formation by restraining inflammation and vascularization, promoting peritoneal repair. Therefore, we believe that our NHP has a great potential application in inhibition of PIA.


Assuntos
Dexametasona , Sistemas de Liberação de Medicamentos , Hidrogéis , Nanofibras , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Nanofibras/química , Nanofibras/uso terapêutico , Hidrogéis/química , Hidrogéis/farmacologia , Hidrogéis/administração & dosagem , Aderências Teciduais/prevenção & controle , Animais , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Dexametasona/farmacologia , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Quitosana/química , Quitosana/farmacologia , Intestinos/efeitos dos fármacos , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Complicações Pós-Operatórias/prevenção & controle , Ratos Sprague-Dawley , Camundongos , Feminino , Ratos
2.
Burns ; 50(6): 1586-1596, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38641499

RESUMO

BACKGROUND: The purpose of dermal substitutes is to mimic the basic properties of the extracellular matrix of human skin. The application of dermal substitutes to the defect reduces the formation of hypertrophic scars and improves the scar quality. This study aims to develop an original dermal substitute enriched with stable fibroblast growth factor 2 (FGF2-STAB®) and test it in an animal model. METHODS: Dermal substitutes based on collagen/chitosan scaffolds or collagen/chitosan scaffolds with nanofibrous layer were prepared and enriched with FGF2-STAB® at concentrations of 0, 0.1, 1.0, and 10.0 µg ‧ cm-2. The performance of these dermal substitutes was tested in vivo on artificially formed skin defects in female swine. The outcomes were evaluated using cutometry at 3 and 6 months. In addition, visual appearance was assessed based on photos of the scars at 1-month, 3-month and 6-month follow-ups using Yeong scale and Visual Analog Scale. RESULTS: The dermal substitute was fully integrated into all defects and all wounds healed successfully. FGF2-STAB®-enriched matrices yielded better results in cutometry compared to scaffolds without FGF2. Visual evaluation at 1, 3, and 6 months follow-ups detected no significant differences among groups. The FGF2-STAB® effectiveness in improving the elasticity of scar tissues was confirmed in the swine model. This effect was independently observed in the scaffolds with nanofibres as well as in the scaffolds without nanofibres. CONCLUSION: The formation of scars with the best elasticity was exhibited by addition 1.0 µg ‧ cm-2of FGF2-STAB® into the scaffolds, although it had no significant effect on visual appearance at longer follow-ups. This study creates the basis for further translational studies of the developed product and its progression into the clinical phase of the research.


Assuntos
Quitosana , Elasticidade , Fator 2 de Crescimento de Fibroblastos , Pele Artificial , Animais , Suínos , Feminino , Alicerces Teciduais , Colágeno , Viscosidade , Cicatriz Hipertrófica , Queimaduras , Cicatrização/efeitos dos fármacos , Nanofibras/uso terapêutico , Modelos Animais de Doenças , Pele
3.
Adv Sci (Weinh) ; 11(17): e2306577, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38441409

RESUMO

Spinal cord injury (SCI) leads to massive cell death, disruption, and demyelination of axons, resulting in permanent motor and sensory dysfunctions. Stem cell transplantation is a promising therapy for SCI. However, owing to the poor microenvironment that develops following SCI, the bioactivities of these grafted stem cells are limited. Cell implantation combined with biomaterial therapies is widely studied for the development of tissue engineering technology. Herein, an insulin-like growth factor-1 (IGF-1)-bioactive supramolecular nanofiber hydrogel (IGF-1 gel) is synthesized that can activate IGF-1 downstream signaling, prevent the apoptosis of neural stem cells (NSCs), improve their proliferation, and induce their differentiation into neurons and oligodendrocytes. Moreover, implantation of NSCs carried out with IGF-1 gels promotes neurite outgrowth and myelin sheath regeneration at lesion sites following SCI. In addition, IGF-1 gels can enrich extracellular vesicles (EVs) derived from NSCs or from nerve cells differentiated from these NSCs via miRNAs related to axonal regeneration and remyelination, even in an inflammatory environment. These EVs are taken up by autologous endogenous NSCs and regulate their differentiation. This study provides adequate evidence that combined treatment with NSCs and IGF-1 gels is a potential therapeutic strategy for treating SCI.


Assuntos
Hidrogéis , Fator de Crescimento Insulin-Like I , Nanofibras , Células-Tronco Neurais , Traumatismos da Medula Espinal , Animais , Ratos , Diferenciação Celular , Modelos Animais de Doenças , Hidrogéis/química , Fator de Crescimento Insulin-Like I/metabolismo , Nanofibras/química , Nanofibras/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/métodos , Feminino
4.
Carbohydr Polym ; 334: 122025, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38553224

RESUMO

Diabetic foot ulcers (DFUs) are one of the most serious and devastating complication of diabetes, manifesting as foot ulcers and impaired wound healing in patients with diabetes mellitus. To solve this problem, sulfated hyaluronic acid (SHA)/collagen-based nanofibrous biomimetic skins was developed and used to promote the diabetic wound healing and skin remodeling. First, SHA was successfully synthetized using chemical sulfation and incorporated into collagen (COL) matrix for preparing the SHA/COL hybrid nanofiber skins. The polyurethane (PU) was added into those hybrid scaffolds to make up the insufficient mechanical properties of SHA/COL nanofibers, the morphology, surface properties and degradation rate of hybrid nanofibers, as well as cell responses upon the nanofibrous scaffolds were studied to evaluate their potential for skin reconstruction. The results demonstrated that the SHA/COL, SHA/HA/COL hybrid nanofiber skins were stimulatory of cell behaviors, including a high proliferation rate and maintaining normal phenotypes of specific cells. Notably, SHA/COL and SHA/HA/COL hybrid nanofibers exhibited a significantly accelerated wound healing and a high skin remodeling effect in diabetic mice compared with the control group. Overall, SHA/COL-based hybrid scaffolds are promising candidates as biomimetic hybrid nanofiber skin for accelerating diabetic wound healing.


Assuntos
Diabetes Mellitus Experimental , Nanofibras , Humanos , Camundongos , Animais , Nanofibras/uso terapêutico , Nanofibras/química , Ácido Hialurônico/química , Biomimética/métodos , Sulfatos/farmacologia , Cicatrização , Colágeno/química , Alicerces Teciduais/química
5.
Adv Sci (Weinh) ; 11(14): e2309993, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38326085

RESUMO

To address current challenges in effectively treating large skin defects caused by trauma in clinical medicine, the fabrication, and evaluation of a novel radially aligned nanofiber scaffold (RAS) with dual growth factor gradients is presented. These aligned nanofibers and the scaffold's spatial design provide many all-around "highways" for cell migration from the edge of the wound to the center area. Besides, the chemotaxis induced by two growth factor gradients further promotes cell migration. Incorporating epidermal growth factor (EGF) aids in the proliferation and differentiation of basal layer cells in the epidermis, augmenting the scaffold's ability to promote epidermal regeneration. Concurrently, the scaffold-bound vascular endothelial growth factor (VEGF) recruits vascular endothelial cells at the wound's center, resulting in angiogenesis and improving blood supply and nutrient delivery, which is critical for granulation tissue regeneration. The RAS+EGF+VEGF group demonstrates superior performance in wound immune regulation, wound closure, hair follicle regeneration, and ECM deposition and remodeling compared to other groups. This study highlights the promising potential of hierarchically assembled nanofiber scaffolds with dual growth factor gradients for wound repair and tissue regeneration applications.


Assuntos
Nanofibras , Nanofibras/uso terapêutico , Fator A de Crescimento do Endotélio Vascular , Fator de Crescimento Epidérmico/farmacologia , Células Endoteliais , Alicerces Teciduais , Cicatrização
6.
ACS Appl Bio Mater ; 7(2): 1179-1190, 2024 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-38215047

RESUMO

The epidermis of a deep burn wound is entirely absent and the dermal tissue sustains significant damage, accompanied by a substantial amount of tissue exudate. Due to the excessively humid environment, the formation of a scab on the wound becomes challenging, leaving it highly vulnerable to external bacterial invasion. In this work, a core-shell dual-drug-loaded nanofiber dressing was prepared by electrospinning technology for the synergistic treatment of a deep burn. The shell layer consists of polycaprolactone and chitosan encapsulating asiaticoside, with the core layer comprising the clathrate of 2-hydroxypropyl-ß-cyclodextrin and curcumin. Upon application to the wound, the dual-drug-loaded nanofiber dressing exhibited rapid release of asiaticoside, stimulating collagen deposition and promoting tissue repair. The core-shell structure and clathrate configuration ensured sustained release of curcumin, providing antibacterial and anti-inflammatory functions for the wound. The mechanical strength, broad-spectrum antibacterial ability, cell proliferation, and adhesion ability of the nanofiber dressing showed its potential as a medical dressing. This dressing also exhibited excellent wound healing promoting effects in the SD rat burn model. This paper provides a strategy for burn wound healing.


Assuntos
Queimaduras , Curcumina , Nanofibras , Triterpenos , Ratos , Animais , Nanofibras/uso terapêutico , Nanofibras/química , Curcumina/farmacologia , Curcumina/uso terapêutico , Ratos Sprague-Dawley , Queimaduras/tratamento farmacológico , Antibacterianos/uso terapêutico , Bandagens
7.
Adv Healthc Mater ; 13(10): e2303539, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38233357

RESUMO

Peripheral nerve injury (PNI) is a common clinical problem and regenerating peripheral nerve defects remain a significant challenge. Poly(polyol sebacate) (PPS) polymers are developed as promising materials for biomedical applications due to their biodegradability, biocompatibility, elastomeric properties, and ease of production. However, the application of PPS-based biomaterials in nerve tissue engineering, especially in PNI repair, is limited. In this study, PPS-based composite nanofibers poly(l-lactic acid)-poly(polycaprolactone triol-co-sebacic acid-co-N,N-bis(2-hydroxyethyl)-2-aminoethanesulfonic acid sodium salt) (PLLA-PPSB) are aimed to construct through electrospinning and assess their in vitro biocompatibility with Schwann cells (SCs) and in vivo repair capabilities for peripheral nerve defects. For the first time, the biocompatibility and bioactivity of PPS-based nanomaterial are examined at the molecular, cellular, and animal levels for PNI repair. Electrospun PLLA-PPSB nanofibers display favorable physicochemical properties and biocompatibility, providing an effective interface for the proliferation, glial expression, and adhesion of SCs in vitro. In vivo experiments using a 10-mm rat sciatic nerve defect model show that PLLA-PPSB nanofiber nerve conduits enhance myelin formation, axonal regeneration, angiogenesis, and functional recovery. Transcriptome analysis and biological validation indicate that PLLA-PPSB nanofibers may promote SC proliferation by activating the PI3K/Akt signaling pathway. This suggests the promising potential of PLLA-PPSB nanomaterial for PNI repair.


Assuntos
Fatores de Coagulação Sanguínea , Nanofibras , Traumatismos dos Nervos Periféricos , Ratos , Animais , Nanofibras/uso terapêutico , Nanofibras/química , Fosfatidilinositol 3-Quinases , Nervo Isquiático/fisiologia , Alicerces Teciduais/química , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Poliésteres/química , Regeneração Nervosa
8.
Adv Healthc Mater ; 13(9): e2303293, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38060135

RESUMO

Invasion of bacteria and continuous oozing of exudate are significant causes of interference with the healing of infected wounds. Therefore, an exudate-induced gelatinizable and near-infrared (NIR)-responsive nanofiber membrane composed of polyvinyl alcohol (PVA), carboxymethyl chitosan (CMC), and Fe-doped phosphomolybdic acid (Fe-PMA) with exceptional exudate absorption capacity and potent bactericidal efficacy is developed and denoted as the PVA-FP-CMC membrane. After absorbing exudate, the fiber membrane can transform into a hydrogel membrane, forming coordination bonds between the Fe-PMA and CMC. The unique exudate-induced gelation process imparts the membrane with high exudate absorption and retention capability, and the formed hydrogel also traps the bacteria that thrive in the exudate. Moreover, it is discovered for the first time that the Fe-PMA exhibits an enhanced photothermal conversion capability and photocatalytic activity compared to the PMA. Therefore, the presence of Fe-PMA provides the membrane with a photothermal and photodynamic therapeutic effect for killing bacteria. The PVA-FP-CMC membrane is proven with a liquid absorption ratio of 520.7%, a light-heat conversion efficiency of 41.9%, high-level generation of hydroxyl radical (•OH) and singlet oxygen (1O2), and a bacterial killing ratio of 100% for S. aureus and 99.6% for E. coli. The treatment of infected wounds on the backs of rats further confirms the promotion of wound healing by the PVA-FP-CMC membrane with NIR irradiation. Overall, this novel functional dressing for the synergistic management of bacteria-infected wounds presents a promising therapeutic strategy for tissue repair and regeneration.


Assuntos
Nanofibras , Infecção dos Ferimentos , Ratos , Animais , Nanofibras/uso terapêutico , Nanofibras/química , Escherichia coli , Staphylococcus aureus , Antibacterianos/farmacologia , Antibacterianos/química , Álcool de Polivinil/farmacologia , Álcool de Polivinil/química , Infecção dos Ferimentos/tratamento farmacológico , Hidrogéis/química , Exsudatos e Transudatos
9.
World Neurosurg ; 181: 96-106, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37852475

RESUMO

Nanofibers made by electrospinning have been used as bridging materials in animal models to regenerate nerves after spinal cord injury (SCI). In this meta-analysis study, we investigated the effect of these nanofibers on the motor function of animals after SCI. An extensive search in databases was performed. After primary and secondary screening, data included functional behavior, expression of glial fibrillary acidic protein, neurofilament-200 (NF-200), and ß-tubulin III were taken from the articles. The quality control of the articles, statistical analysis, and subgroup analysis were performed. The results from 14 articles and 16 separate experiments showed that electrospun nanofibers used alone could improve motor behavior and reduce glial injury after SCI.


Assuntos
Nanofibras , Traumatismos da Medula Espinal , Ratos , Animais , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Nanofibras/uso terapêutico , Traumatismos da Medula Espinal/complicações , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Medula Espinal
10.
Int J Biol Macromol ; 258(Pt 1): 128482, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38042326

RESUMO

Utilizing electrospun nanofibers and microneedle arrays in wound regeneration has been practiced for several years. Researchers have recently asserted that using multiple methods concurrently might enhance efficiency, despite the inherent strengths and weaknesses of each individual approach. The combination of microneedle arrays with electrospun nanofibers has the potential to create a drug delivery system and wound healing method that offer improved efficiency and accuracy in targeting. The use of microneedles with nanofibers allows for precise administration of pharmaceuticals due to the microneedles' capacity to pierce the skin and the nanofibers' role as a drug reservoir, resulting in a progressive release of drugs over a certain period of time. Electrospun nanofibers have the ability to imitate the extracellular matrix and provide a framework for cellular growth and tissue rejuvenation, while microneedle arrays show potential for enhancing tissue regeneration and enhancing the efficacy of wound healing. The integration of electrospun nanofibers with microneedle arrays may be customized to effectively tackle particular obstacles in the fields of wound healing and drug delivery. However, some issues must be addressed before this paradigm may be fully integrated into clinical settings, including but not limited to ensuring the safety and sterilization of these products for transdermal use, optimizing manufacturing methods and characterization of developed products, larger-scale production, optimizing storage conditions, and evaluating the inclusion of multiple therapeutic and antimicrobial agents to increase the synergistic effects in the wound healing process. This research examines the combination of microneedle arrays with electrospun nanofibers to enhance the delivery of drugs and promote wound healing. It explores various kinds of microneedle arrays, the materials and processes used, and current developments in their integration with electrospun nanofibers.


Assuntos
Nanofibras , Nanofibras/uso terapêutico , Pele , Cicatrização , Polissacarídeos/farmacologia , Sistemas de Liberação de Medicamentos/métodos
11.
ACS Nano ; 18(1): 245-263, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38117780

RESUMO

Poor clinical efficacy associated with postoperative hepatocellular carcinoma (HCC) often results from recurrence and metastasis. Hence, research has focused on establishing an effective multimodal therapy. However, complex combinations of active ingredients require multiple functions in therapeutic systems. Herein, a portable nanofiber patch composing germanium phosphorus (GeP) and anlotinib (AL) was designed to form a versatile platform for molecularly targeted photothermal-immune checkpoint blockade (ICB) trimodal combination therapy. The patches possess hydrophilic, satisfactory mechanical, and excellent photothermal conversion properties. Moreover, they achieve a penetrating and sustained drug release. The near-infrared light-assisted GeP-induced temperature increase regulates AL release, downregulating the expression of vascular-related factor receptors, triggering immunogenic cell death of tumor cells, and inducing dendritic cell maturation. Simultaneously, ICB therapy (programmed cell death ligand 1, PD-L1) was introduced to improve treatment outcomes. Notably, this trimodal combination therapy significantly inhibits vascular hypergrowth, enhances effector T-cell infiltration, and sensitizes the PD-L1 antibody response, boosting immunotherapy to suppress residual HCC recurrence and metastasis. Further validation of the genome sequencing results revealed cell pathways related primarily to regulatory immune effects. This study demonstrates the use of an effective and practical nanofiber patch to improve multimodal therapy of postoperative HCC, with high clinical translation value.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanofibras , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Antígeno B7-H1 , Nanofibras/uso terapêutico , Terapia Combinada , Imunoterapia/métodos , Microambiente Tumoral
12.
Int J Nanomedicine ; 18: 6763-6779, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38026517

RESUMO

Peripheral nerve injuries present significant challenges in regenerative medicine, primarily due to inherent limitations in the body's natural healing processes. In response to these challenges and with the aim of enhancing peripheral nerve regeneration, nanofiber scaffolds have emerged as a promising and advanced intervention. However, a deeper understanding of the underlying mechanistic foundations that drive the favorable contributions of nanofiber scaffolds to nerve regeneration is essential. In this comprehensive review, we make an exploration of the latent potential of nanofiber scaffolds in augmenting peripheral nerve regeneration. This exploration includes a detailed introduction to the fabrication methods of nanofibers, an analysis of the intricate interactions between these scaffolds and cellular entities, an examination of strategies related to the controlled release of bioactive agents, an assessment of the prospects for clinical translation, an exploration of emerging trends, and thorough considerations regarding biocompatibility and safety. By comprehensively elucidating the intricate structural attributes and multifaceted functional capacities inherent in nanofiber scaffolds, we aim to offer a prospective and effective strategy for the treatment of peripheral nerve injury.


Assuntos
Nanofibras , Traumatismos dos Nervos Periféricos , Humanos , Alicerces Teciduais/química , Nanofibras/uso terapêutico , Nanofibras/química , Estudos Prospectivos , Traumatismos dos Nervos Periféricos/terapia , Regeneração Nervosa , Engenharia Tecidual
13.
ACS Appl Bio Mater ; 6(11): 4620-4628, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37904513

RESUMO

Wound healing is one of the major global health concerns in diabetic patients. Simvastatin (SMV) is a poorly soluble oral cholesterol-lowering drug that may aid diabetic wound healing. In the current study, a thixotropic peptide hydrogel of Fmoc-diphenylalanine (FmocFF) containing SMV was designed to accelerate skin wound healing effectively and safely in diabetic mice. FmocFF hydrogels were prepared at various concentrations by using the solvent-triggering technique and characterized by spectroscopic methods such as attenuated total reflection Fourier transform infrared (FT-IR) spectroscopy and fluorimetry. Mechanical behaviors were explored by oscillatory rheology. In model mice, the regenerative potential of the FmocFF-SMV hydrogel was evaluated in terms of wound contraction and closure, tissue regeneration, acute and chronic inflammation, granulation, and re-epithelization. The results showed that FmocFF-SMV hydrogels had an entangled nanofibrous microstructure and shear-thinning characteristics. FmocFF-SMV demonstrated a sustained drug release over 7 days. Compared to the unloaded FmocFF hydrogel, treatment with FmocFF-SMV led to superior diabetic wound recovery and reduced inflammation. Therefore, the utilization of the sustained-release FmocFF-SMV hydrogel formulation could become an attractive choice for topical wound therapy in diabetes patients.


Assuntos
Diabetes Mellitus Experimental , Nanofibras , Humanos , Animais , Camundongos , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Nanofibras/uso terapêutico , Espectroscopia de Infravermelho com Transformada de Fourier , Hidrogéis , Inflamação , Peptídeos
14.
Int J Biol Macromol ; 253(Pt 8): 127607, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37871723

RESUMO

The significant clinical challenge presented by diabetic wounds is due to their impaired healing process and increased risk of complications. It is estimated that a foot ulcer will develop at some point in the lives of 15-25 % of diabetic patients. Serious complications, including infection and amputation, are often led to by these wounds. In the field of tissue engineering and regenerative medicine, nanofiber-based wound dressings have emerged in recent years as promising therapeutic strategies for diabetic wound healing. Hyaluronic acid (HA), among various nanofiber materials, has gained considerable attention due to its unique properties, including biocompatibility, biodegradability, and excellent moisture retention capacity. By promoting skin hydration and controlling inflammation, a crucial role in wound healing is played by HA. Wounds are also helped to heal faster by HA through the regulation of inflammation levels and signaling the body to build more blood vessels in the damaged area. Great potential in various applications, including wound healing, has been shown by the development and use of nanofiber formulations in medicine. However, challenges and limitations associated with nanofibers in medicine exist, such as reproducibility, proper characterization, and biological evaluation. By providing a biomimetic environment that enhances re-epithelialization and facilitates the delivery of active substances, nanofibers promote wound healing. In accelerating wound healing, promising results have been shown by HA-contained nanofiber formulations in diabetic wounds. Key strategies employed by these formulations include revascularization, modulation of the inflammation microenvironment, delivery of active substances, photothermal nanofibers, and nanoparticle-loaded fabrics. Particularly crucial is revascularization as it restores blood flow to the wound area, promoting healing. Wound healing can also be enhanced by modulating the inflammation microenvironment through controlling inflammation levels. Future perspectives in this field involve addressing the current challenges and limitations of nanofiber technology and further optimizing HA-contained nanofiber formulations for improved efficacy in diabetic wound healing. This includes exploring new fabrication techniques, enhancing the biocompatibility and biodegradability of nanofibers, and developing multifunctional nanofibers for targeted drug delivery. Not only does writing a review in the field of nanofiber-based wound dressings, particularly those containing hyaluronic acid, allow us to consolidate our current knowledge and understanding but also broadens our horizons. An opportunity is provided to delve deeper into the intricacies of this innovative therapeutic strategy, explore its potential and limitations, and envision future directions. By doing so, a contribution can be made to the ongoing advancements in tissue engineering and regenerative medicine, ultimately improving the quality of life for patients with diabetic wounds.


Assuntos
Diabetes Mellitus , Nanofibras , Humanos , Ácido Hialurônico/uso terapêutico , Nanofibras/uso terapêutico , Qualidade de Vida , Reprodutibilidade dos Testes , Cicatrização , Inflamação
15.
Int Immunopharmacol ; 125(Pt A): 111108, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37890380

RESUMO

Currently, there are no effective therapeutic targets for the treatment of chronic cerebral hypoperfusion(CCH)-induced cerebral ischemic injury. Vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are discovered as the inducers of neurogenesis and angiogenesis. We previously made a nanofiber membrane (NFM), maintaining a long-term release of VEGF and bFGF up to 35 days, which might make VEGF and bFGF NFM as the potential protective agents against cerebral ischemic insult. In this study, the effects of VEGF and bFGF delivered by NFM into brain were investigated as well as their underlying mechanismsin a rat model of CCH. VEGF + bFGF NFM application increased the expressions of tight junction proteins, maintained BBB integrity, and alleviated vasogenic cerebral edema. Furthermore, VEGF + bFGF NFM sticking enhanced angiogenesis and elevated CBF. Besides, VEGF + bFGF NFM treatment inhibited neuronal apoptosis and decreased neuronal loss. Moreover, roofing of VEGF + bFGF NFM attenuated microglial activation and blocked the launch of NLRP3/caspase-1/IL-1ß pathway. In addition, VEGF + bFGF NFM administration prevented disruption to the pre/postsynaptic membranes and loss of myelin sheath, relieving synaptic injury and demyelination. Oligodendrogenesis, neurogenesis and PI3K/AKT/mTOR pathway were involved in the treatment of VEGF + bFGF NFM against CCH-induced neuronal injury and hypomyelination. These findings supported that VEGF + bFGF NFM application constitutes a neuroprotective strategy for the treatment of CCH, which may be worth further clinical translational research as a novel neuroprotective approach, benifiting indirect surgical revascularization.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Nanofibras , Ratos , Animais , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Fosfatidilinositol 3-Quinases , Nanofibras/uso terapêutico , Fatores de Crescimento do Endotélio Vascular , Isquemia Encefálica/metabolismo , Isquemia
16.
J Craniomaxillofac Surg ; 51(12): 772-779, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37863731

RESUMO

The objective of this randomized clinical trial (RCT) was to assess the effectiveness of electrospun chitosan/polyvinyl alcohol (CS/PVA) nanofibrous scaffolds in preserving the alveolar ridge and enhancing bone remodeling following tooth extraction when compared to a control group. In this split RCT, 24 human alveolar sockets were randomly assigned to two groups, with 12 sockets receiving CS/PVA nanofibrous scaffold grafts (test group) and 12 left to heal by secondary intention as the control group. Cone-beam computed tomography (CBCT) was performed at two different time points: immediately after extraction (T0) and 4 months post-extraction (T4). After 4 months, linear vertical and horizontal radiographic changes and bone density of extraction sockets were assessed in both the test and control groups. The RCT included 12 patients (4 male and 8 female) with a mean age of 24 ± 3.37 years. The test group had a significantly lower mean vertical resorption vs the control group, with a mean difference of 1.1 mm (P < 0.05). Similarly, the control group's mean horizontal bone resorption was -2.01 ± 1.04 mm, while the test group had a significantly lower mean of -0.69 ± 0.41 mm, resulting in a mean difference of 1.35 mm (P < 0.05). Furthermore, the study group exhibited a significant increase in bone density (722.03 ± 131.17 HU) after 4 months compared to the control group (448.73 ± 93.23 HU). In conclusion, we demonstrated within the limitations of this study that CS/PVA nanofibrous scaffold significantly limited alveolar bone resorption horizontally and vertically and enhanced bone density in alveolar sockets after 4 months when compared to results in the control group (TCTR20230526005).


Assuntos
Perda do Osso Alveolar , Aumento do Rebordo Alveolar , Quitosana , Nanofibras , Masculino , Feminino , Humanos , Adulto Jovem , Adulto , Álcool de Polivinil/uso terapêutico , Alvéolo Dental/diagnóstico por imagem , Alvéolo Dental/cirurgia , Aumento do Rebordo Alveolar/métodos , Nanofibras/uso terapêutico , Processo Alveolar/diagnóstico por imagem , Processo Alveolar/cirurgia , Perda do Osso Alveolar/diagnóstico por imagem , Perda do Osso Alveolar/prevenção & controle , Perda do Osso Alveolar/cirurgia , Extração Dentária
17.
Adv Sci (Weinh) ; 10(30): e2302909, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37653617

RESUMO

Retinal ischemia is involved in the occurrence and development of various eye diseases, including glaucoma, diabetic retinopathy, and central retinal artery occlusion. To the best of our knowledge, few studies have reported self-assembling peptide natural products for the suppression of ocular inflammation and oxidative stress. Herein, a self-assembling peptide GFFYE is designed and synthesized, which can transform the non-hydrophilicity of rhein into an amphiphilic sustained-release therapeutic agent, and rhein-based therapeutic nanofibers (abbreviated as Rh-GFFYE) are constructed for the treatment of retinal ischemia-reperfusion (RIR) injury. Rh-GFFYE significantly ameliorates oxidative stress and inflammation in an in vitro oxygen-glucose deprivation (OGD) model of retinal ischemia and a rat model of RIR injury. Rh-GFFYE also significantly enhances retinal electrophysiological recovery and exhibits good biocompatibility. Importantly, Rh-GFFYE also promotes the transition of M1-type macrophages to the M2 type, ultimately altering the pro-inflammatory microenvironment. Further investigation of the treatment mechanism indicates that Rh-GFFYE activates the PI3K/AKT/mTOR signaling pathway to reduce oxidative stress and inhibits the NF-κB and STAT3 signaling pathways to affect inflammation and macrophage polarization. In conclusion, the rhein-loaded nanoplatform alleviates RIR injury by modulating the retinal microenvironment. The findings are expected to promote the clinical application of hydrophobic natural products in RIR injury-associated eye diseases.


Assuntos
Produtos Biológicos , Oftalmopatias , Nanofibras , Traumatismo por Reperfusão , Ratos , Animais , Microglia/metabolismo , Nanofibras/uso terapêutico , Fosfatidilinositol 3-Quinases , Estresse Oxidativo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Oftalmopatias/metabolismo , Produtos Biológicos/metabolismo , Peptídeos/metabolismo , Isquemia
18.
Adv Healthc Mater ; 12(32): e2302484, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37702133

RESUMO

Postoperative recovery of cancer patients can be affected by complications, such as tissue dysfunction or disability caused by tissue resection, and also cancer recurrence resulting from residual cancer cells. Despite impressive progress made for tissue engineering scaffolds that assist tissue regeneration for postoperative cancer patients, the majority of existing tissue engineering scaffolds still lack functions for monitoring and killing residual cancer cells, if there are any, upon their detection. In this study, multifunctional scaffolds that comprise biodegradable nanofibers and core-shell structured microspheres encapsulated with theranostic nanoparticles (NPs) are developed. The multifunctional scaffolds possess an extracellular matrix-like nanofibrous architecture and soft tissue-like mechanical properties, making them excellent tissue engineering patch candidates for assisting in the repair and regeneration of tissues at the cancerous sites after surgery. Furthermore, they are capable of localized delivery of theranostic NPs upon quick degradation of core-shell structured microspheres that contain theranostic NPs. Leveraging on folic acid-mediated ligand-receptor binding, surface-enhanced Raman scattering activity, and near-infrared-responsive photothermal effect of the theranostic gold NPs (AuNPs) delivered locally, the multifunctional scaffolds display excellent active targeting, diagnosis, and photothermal therapy functions for cancer cells, showing great promise for adaptive postoperative cancer management.


Assuntos
Nanopartículas Metálicas , Nanofibras , Humanos , Nanofibras/uso terapêutico , Nanofibras/química , Medicina de Precisão , Ouro/química , Neoplasia Residual , Nanopartículas Metálicas/química , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Nanomedicina Teranóstica
19.
J Nanobiotechnology ; 21(1): 313, 2023 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-37661273

RESUMO

The regeneration of cutaneous tissue is one of the most challenging issues in human regenerative medicine. To date, several studies have been done to promote cutaneous tissue healing with minimum side effects. The healing potential of polyurethane (PU)/Poly (caprolactone)-poly (ethylene glycol)-poly (caprolactone) (PCEC)/chitosan (CS) (PCS) nanofibrous mat with cationic photosensitizer meso tetrakis (N-methyl pyridinium-4-yl) porphyrin tetratosylate salt (TMP) was examined. The CS tripolyphosphate nanoparticles (CSNPs) were prepared and loaded by TMP to provide an efficient drug release system (TMPNPs) for delivery of TMP to promote wound healing. In in vitro setting, parameters such as bactericidal effects, cytocompatibility, and hemolytic effects were examined. The healing potential of prepared nanofibrous mats was investigated in a rat model of full-thickness cutaneous injury. PCS/TMP/TMPNPs nanofibers can efficiently release porphyrin in the aqueous phase. The addition of TMPNPs and CS to the PU backbone increased the hydrophilicity, degradation, and reduced mechanical properties. The culture of human fetal foreskin fibroblasts (HFFF2) on PCS/TMP/TMPNPs scaffold led to an increased survival rate and morphological adaptation analyzed by MTT and SEM images. Irradiation with a red laser (635 nm, 3 J/cm2) for the 30 s reduced viability of S. aureus and E. Coli bacteria plated on PCS/TMP and PCS/TMP/TMPNPs nanofibrous mats compared to PU/PCEC (PC) and PU/PCEC/CS (PCS) groups, indicating prominent antibacterial effects of PCS/TMP and PCS/TMP/TMPNPs nanofibrous (p < 0.05). Data indicated that PCS/TMP/TMPNPs mat enhanced healing of the full-thickness excisional wound in a rat model by the reduction of inflammatory response and fibrotic changes compared to the PC, and PCS groups (p < 0.05). Immunofluorescence imaging indicated that levels of Desmoglein were increased in rats that received PCS/TMP/TMPNPs compared to the other groups. It is found that a PU-based nanofibrous mat is an appropriate scaffold to accelerate the healing of injured skin.


Assuntos
Nanofibras , Animais , Ratos , Humanos , Nanofibras/uso terapêutico , Poliuretanos , Escherichia coli , Staphylococcus aureus , Cicatrização , Antibacterianos/farmacologia
20.
Int J Nanomedicine ; 18: 4969-4983, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37693887

RESUMO

Osteoarthritis (OA) is a prevalent global health concern, posing a significant and increasing public health challenge worldwide. Recently, nanotechnology-boosted biomaterials have emerged as a highly promising strategy for OA therapy due to their exceptional physicochemical properties and capacity to regulate pathological processes. However, there is an urgent need for a deeper understanding of the potential therapeutic applications of these biomaterials in the clinical management of diseases, particularly in the treatment of OA. In this comprehensive review, we present an extensive discussion of the current status and future prospects concerning nanotechnology-boosted biomaterials for OA therapy. Initially, we discuss the pathophysiology of OA and the constraints associated with existing treatment modalities. Subsequently, various types of nanomaterials utilized for OA therapy, including nanoparticles, nanofibers, and nanocomposites, are thoroughly discussed and summarized, elucidating their respective advantages and challenges. Furthermore, we analyze recent preclinical and clinical studies that highlight the potential of nanotechnology-boosted biomaterials in OA therapy. Additionally, future research directions in this evolving field are highlighted. By establishing a link between the structural properties of nanotechnology-boosted biomaterials and their therapeutic functions in OA treatment, we aim to foster advances in designing sophisticated nanomaterials for OA, ultimately resulting in improved therapeutic efficacy of OA therapy through translation into clinical setting in the near future.


Assuntos
Nanocompostos , Nanofibras , Osteoartrite , Humanos , Nanotecnologia , Materiais Biocompatíveis/uso terapêutico , Nanofibras/uso terapêutico , Osteoartrite/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA