Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 294
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nano Lett ; 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38833276

RESUMEN

Inspired by the imbalance between extrinsic and intrinsic tendon healing, this study fabricated a new biofilter scaffold with a hierarchical structure based on a melt electrowriting technique. The outer multilayered fibrous structure with connected porous characteristics provides a novel passageway for vascularization and isolates the penetration of scar fibers, which can be referred to as a biofilter process. In vitro experiments found that the porous architecture in the outer layer can effectively prevent cell infiltration, whereas the aligned fibers in the inner layer can promote cell recruitment and growth, as well as the expression of tendon-associated proteins in a simulated friction condition. It was shown in vivo that the biofilter process could promote tendon healing and reduce scar invasion. Herein, this novel strategy indicates great potential to design new biomaterials for balancing extrinsic and intrinsic healing and realizing scarless tendon healing.

2.
Small ; : e2402040, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38829027

RESUMEN

The extracellular matrix (ECM) engages in regulatory interactions with cell surface receptors through its constituent proteins and polysaccharides. Therefore, nano-sized extracellular matrix conjugated with doxorubicin (DOX) is utilized to produce extracellular matrix-drug conjugates (ECM-DOX) tailored for targeted delivery to cancer cells. The ECM-DOX nanoparticles exhibit rod-like morphology, boasting a commendable drug loading capacity of 4.58%, coupled with acid-sensitive drug release characteristics. Notably, ECM-DOX nanoparticles enhance the uptake by tumor cells and possess the ability to penetrate endothelial cells and infiltrate tumor multicellular spheroids. Mechanistic insights reveal that the internalization of ECM-DOX nanoparticle is facilitated through clathrin-mediated endocytosis and macropinocytosis, intricately involving hyaluronic acid receptors and integrins. Pharmacokinetic assessments unveil a prolonged blood half-life of ECM-DOX nanoparticles at 3.65 h, a substantial improvement over the 1.09 h observed for free DOX. A sustained accumulation effect of ECM-DOX nanoparticles at tumor sites, with drug levels in tumor tissues surpassing those of free DOX by several-fold. The profound therapeutic impact of ECM-DOX nanoparticles is evident in their notable inhibition of tumor growth, extension of median survival time in animals, and significant reduction in DOX-induced cardiotoxicity. The ECM platform emerges as a promising carrier for avant-garde nanomedicines in the realm of cancer treatment.

3.
Small ; : e2403564, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38966875

RESUMEN

Nasal administration can bypass the blood-brain barrier and directly deliver drugs to the brain, providing a non-invasive route for central nervous system (CNS) diseases. Inspired by the appearance that a gate can block the outside world and the characteristics of the sol-gel transition can form a "gate" in the nasal cavity, a Drop to Gate nasal drop (DGND) is designed to set a gate in nose, which achieves protecting role from the influence of nasal environment. The DGND demonstrates the efficiency and application prospect of delivering drugs to the brain through the N-to-B. The effective concentration of single administration is increased through the hydrophobic interaction between C8-GelMA and SRT1720 (SA), and then cross-linked under UV to form nanogel, which can respond to MMP in the inflammatory microenvironment of sepsis-induced cognitive dysfunction. Finally, the SA/nanogel is compounded into the thermogel, which can respond to the nasal cavity temperature to form DGND in situ, increasing the residence time and delivery efficiency of drugs in the nasal cavity. In vitro, the DGND alleviates lipopolysaccharides (LPS)-induced BV2 inflammation. In vivo, DGND effectively targets the nasal mucosa and deliver drugs to the brain, which activate Sirt1 to alleviate inflammation mediated by microglia and improve cognitive dysfunction in sepsis mice.

4.
J Nanobiotechnology ; 22(1): 289, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802863

RESUMEN

By integrating magnetic resonance-visible components with scaffold materials, hydrogel microspheres (HMs) become visible under magnetic resonance imaging(MRI), allowing for non-invasive, continuous, and dynamic monitoring of the distribution, degradation, and relationship of the HMs with local tissues. However, when these visualization components are physically blended into the HMs, it reduces their relaxation rate and specificity under MRI, weakening the efficacy of real-time dynamic monitoring. To achieve MRI-guided in vivo monitoring of HMs with tissue repair functionality, we utilized airflow control and photo-crosslinking methods to prepare alginate-gelatin-based dual-network hydrogel microspheres (G-AlgMA HMs) using gadolinium ions (Gd (III)), a paramagnetic MRI contrast agent, as the crosslinker. When the network of G-AlgMA HMs degrades, the cleavage of covalent bonds causes the release of Gd (III), continuously altering the arrangement and movement characteristics of surrounding water molecules. This change in local transverse and longitudinal relaxation times results in variations in MRI signal values, thus enabling MRI-guided in vivo monitoring of the HMs. Additionally, in vivo data show that the degradation and release of polypeptide (K2 (SL)6 K2 (KK)) from G-AlgMA HMs promote local vascular regeneration and soft tissue repair. Overall, G-AlgMA HMs enable non-invasive, dynamic in vivo monitoring of biomaterial degradation and tissue regeneration through MRI, which is significant for understanding material degradation mechanisms, evaluating biocompatibility, and optimizing material design.


Asunto(s)
Alginatos , Medios de Contraste , Gadolinio , Hidrogeles , Imagen por Resonancia Magnética , Microesferas , Imagen por Resonancia Magnética/métodos , Gadolinio/química , Animales , Alginatos/química , Hidrogeles/química , Medios de Contraste/química , Cicatrización de Heridas/efectos de los fármacos , Reactivos de Enlaces Cruzados/química , Gelatina/química , Ratones , Andamios del Tejido/química
5.
Small ; 19(18): e2207248, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36725316

RESUMEN

Glioblastoma (GBM) is the most common lethal brain tumor with dismal treatment outcomes and poor response to chemotherapy. As the regulatory center of cytogenetics and metabolism, most tumor chemotherapeutic molecules exert therapeutic effects in the nucleus. Nanodrugs showing the nuclear aggregation effect are expected to eliminate and fundamentally suppress tumor cells. In this study, a nanodrug delivery system based on polyhedral oligomeric silsesquioxane (POSS) is introduced to deliver drugs into the nuclei of GBM cells, effectively enhancing the therapeutic efficacy of chemotherapy. The nanoparticles are modified with folic acid and iRGD peptides molecules to improve their tumor cell targeting and uptake via receptor-mediated endocytosis. Nuclear aggregation allows for the direct delivery of chemotherapeutic drug temozolomide (TMZ) to the tumor cell nuclei, resulting in more significant DNA damage and inhibition of tumor cell proliferation. Herein, TMZ-loaded POSS nanoparticles can significantly improve the survival of GBM-bearing mice. Therefore, the modified POSS nanoparticles may serve as a promising drug-loaded delivery platform to improve chemotherapy outcomes in GBM patients.


Asunto(s)
Glioblastoma , Nanopartículas , Ratones , Animales , Glioblastoma/patología , Línea Celular Tumoral , Temozolomida/química , Temozolomida/farmacología , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química
6.
Small ; 19(47): e2303615, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37501326

RESUMEN

Current research on hemostatic materials have focused on the inhibition of visible hemorrhage, however, invisible hemorrhage is the unavoidable internal bleeding that occurs after trauma or surgery, leading directly to a dramatic drop in hemoglobin and then to anemia and even death. In this study, bacterial nanocellulose (BNC) was synthesized and oxidized from the primary alcohols to carboxyl groups, and then grafted with tranexamic acid through amide bonds to construct degradable nanoscale short fibers (OBNC-TXA), which rapidly activated the coagulation response. The hemostatic material is made up of nanoscale short fibers that can be constructed into different forms such as emulsions, gels, powders, and sponges to meet different clinical applications. In the hemostatic experiments in vitro, the composites had significantly superior pro-coagulant properties due to the rapid aggregation of blood cells. In the coagulation experiments with rat tail amputation and liver trauma hemorrhage models, the group treated with OBNC-TXA1 sponge showed low hemorrhage and inhibited invisible hemorrhage in rectus abdominis muscle defect hemorrhage models, with a rapid recovery of hemoglobin values from 128±5.5 to 165±2.6 g L-1 within 4 days. In conclusion, the degradable short fibers constructed from bacterial nano-cellulose achieved inhibition of invisible hemorrhage in vivo.


Asunto(s)
Hemostáticos , Hepatopatías , Ácido Tranexámico , Ratas , Animales , Ácido Tranexámico/farmacología , Ácido Tranexámico/uso terapéutico , Hemorragia/tratamiento farmacológico , Hemostáticos/farmacología , Hemostáticos/uso terapéutico , Coagulación Sanguínea , Hemoglobinas/farmacología , Hemoglobinas/uso terapéutico
7.
Small ; 19(30): e2300481, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37035992

RESUMEN

The damaged endometrium and the formation of fibrosis are key barriers to pregnancy and further lead to infertility. However, how to promote endometrium repair is always a challenge. Here, a bioactive injectable and self-healing hydrogel is developed by physically combination of thiolated polyethylene (PEG), Cu2+ and cell-free fat extract (CEFFE, CF) for endometrial regeneration and fertility. By inheriting the advantages of various active proteins contained in CEFFE, it could induce the overall repair of endometrial microenvironment for intrauterine adhesion (IUA). In vitro, CF@Cu-PEG reduces endometrial cell apoptosis by more than 50%, and increases angiogenesis by 92.8%. In the IUA mouse, injection of CF@Cu-PEG significantly reduces the rate of uterine hydrometra and prevents the formation of endometrial fibrosis. Remarkably, CF@Cu-PEG contributes to the repair of endometrial microstructure, especially increases the number of endometrial pinopodes, significantly improves endometrial receptivity, and increases the pregnancy rate of IUA mice from 7.14% to 66.67%. In summary, through the physically combination of CEFFE and Cu-PEG, the construction of loaded bioactive injectable hydrogel not only inhibits the IUA, but also induces the self-repair of endometrial cells in situ and improves fertility, providing a new strategy for IUA repair in clinical application.


Asunto(s)
Hidrogeles , Enfermedades Uterinas , Embarazo , Femenino , Humanos , Ratones , Animales , Hidrogeles/química , Endometrio , Enfermedades Uterinas/metabolismo , Enfermedades Uterinas/patología , Regeneración , Fibrosis
8.
Small ; 19(17): e2207211, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36651038

RESUMEN

Paracrine is an important mechanism in mesenchymal stem cells (MSCs) that promotes tissue regeneration. However, anoikis is attributed to unsuitable adhesion microenvironment hindered this paracrine effect. In this study, a living and injectable porous hydrogel microsphere with long-term paracrine activity is constructed via the freeze-drying microfluidic technology and the incorporation of platelet-derived growth factor-BB (PDGF-BB) and exogenous MSCs. Benefiting from the porous structure and superior mechanical property of methacrylate gelatin (GelMA) hydrogel microspheres (GMs), exogenous stem cells are able to adhere and proliferate on GMs, thereby facilitating cell-to-extracellular matrix (ECM) and cell-to-cell interactions and enhancing paracrine effect. Furthermore, the sustained release of PDGF-BB can recruit endogenous MSCs to prolong the paracrine activity of the living GMs. In vitro and in vivo experiments validated that the living GMs exhibit superior secretion properties and anti-inflammatory efficacy and can attenuate osteoarthritis (OA) progression by favoring the adherent microenvironment and utilizing the synergistic effect of exogenous and endogenous MSCs. Overall, a living injectable porous hydrogel microsphere that can enhance the paracrine activity of stem cells is fabricated and anticipated to hold the potential of future clinical translation in OA and other diseases.


Asunto(s)
Materiales Biocompatibles , Cartílago , Microesferas , Becaplermina , Porosidad , Materiales Biocompatibles/química , Hidrogeles/química
9.
Small ; 19(45): e2303456, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37438648

RESUMEN

The pathogenesis of postmenopausal osteoporosis (PMOP) is mainly determined by the adhesion of osteoclasts to the bone matrix and the involvement of various molecules in bone resorption. The dual regulation strategy of the physical barriers of bone matrix and intracellular gene regulation generated by advanced biomaterials is a decent alternative for the treatment of PMOP. Herein, for the first time, it is identified that hsa-miR-378i/mmu-miR-378a-3p are closely associated with PMOP. Then, an osteophilic and dual-regulated alendronate-gene lipoplex (antagomir@Aln-Lipo), composed of medicative alendronate-functionalized liposomal vehicle and encapsulated specific microRNAs is engineered, for bone-targeting delivery of genes to achieve combined mitigation of bone loss. Alendronate targets hydroxyapatite in the bone matrix and occupies the adhesion site of osteoclasts, thus providing the "physical barriers". Antagomir is coupled precisely to specific endogenous microRNAs, thus providing the "genetic signals". These functionalized lipoplexes exhibited long-term stability and good transfection efficiency. It is proven that antagomir@Aln-Lipo could synergistically regulate osteoclastogenesis and bone resorption in vitro and in vivo. Furthermore, intravenous injection of antagomir@Aln-Lipo efficiently reverses bone loss through a dual mechanism driven by alendronate and antagomir-378a-3p. In conclusion, the osteophilic and dual-regulated antagomir@Aln-Lipo offers a brand-new bifunctional strategy for the precise treatment of PMOP.


Asunto(s)
Resorción Ósea , MicroARNs , Humanos , Alendronato , Antagomirs , Huesos/patología , MicroARNs/genética
10.
Small ; 19(15): e2207030, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36604983

RESUMEN

The "double-edged sword" effect of macrophages under the influence of different microenvironments determines the outcome and prognosis of tissue injury. Accurate and stable reprogramming macrophages (Mφ) are the key to rapid wound healing. In this study, an immunized microsphere-engineered GelMA hydrogel membrane is constructed for oral mucosa treatment. The nanoporous poly(lactide-co-glycolide) (PLGA) microsphere drug delivery system combined with the photo-cross-linkable hydrogel is used to release the soybean lecithin (SL)and IL-4 complexes (SL/IL-4) sustainedly. In this way, it is realized effective wound fit, improvement of drug encapsulation, and stable triphasic release of interleukin-4 (IL-4). In both in vivo and in vitro experiments, it is demonstrated that the hydrogel membrane can reprogram macrophages in the microenvironment into M2Mφ anti-inflammatory types, thereby inhibiting the local excessive inflammatory response. Meanwhile, high levels of platelet-derived growth factor (PDGF) secreted by M2Mφ macrophages enhanced neovascular maturation by 5.7-fold, which assisted in achieving rapid healing of oral mucosa. These findings suggest that the immuno-engineered hydrogel membrane system can re-modulating the biological effects of Mφ, and potentiating the maturation of neovascularization, ultimately achieving the rapid repair of mucosal tissue. This new strategy is expected to be a safe and promising immunomodulatory biomimetic material for clinical translation.


Asunto(s)
Hidrogeles , Interleucina-4 , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Microesferas , Macrófagos , Membrana Mucosa
11.
Small ; : e2309060, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38063818

RESUMEN

The cellular functions, such as tissue-rebuilding ability, can be directly affected by the metabolism of cells. Moreover, the glucose metabolism is one of the most important processes of the metabolism. However, glucose cannot be efficiently converted into energy in cells under ischemia hypoxia conditions. In this study, a high-energy intermediate fructose hydrogel (HIFH) is developed by the dynamic coordination between sulfhydryl-functionalized bovine serum albumin (BSA-SH), the high-energy intermediate in glucose metabolism (fructose-1,6-bisphosphate, FBP), and copper ion (Cu2+ ). This hydrogel system is injectable, self-healing, and biocompatible, which can intracellularly convert energy with high efficacy by regulating the glucose metabolism in situ. Additionally, the HIFH can greatly boost cell antioxidant capacity and increase adenosine triphosphate (ATP) in the ischemia anoxic milieu by roughly 1.3 times, improving cell survival, proliferation and physiological functions in vitro. Furthermore, the ischemic skin tissue model is established in rats. The HIFH can speed up the healing of damaged tissue by promoting angiogenesis, lowering reactive oxygen species (ROS), and eventually expanding the healing area of the damaged tissue by roughly 1.4 times in vivo. Therefore, the HIFH can provide an impressive perspective on efficient in situ cell energy supply of damaged tissue.

12.
Small ; 18(39): e2203265, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36031400

RESUMEN

Overexpression of inflammatory cytokines and chemokines occurs at deep soft tissue injury sites impeding the inflammation self-limiting and impairing the tissue remodeling process. Inspired by the electrostatically extracellular matrix (ECM) binding property of the inflammatory signals, an inflammation self-limiting fibrous tape is designed by covalently modifying the thermosensitive methacrylated gelatin (GelMA) and negatively charged methacrylated heparin (HepMA) hydrogel mixture with proper ratio onto the electrospun fibrous membrane by mild alkali hydrolysis and carboxyl-amino condensation reaction to restore inflammation self-limiting and promote tissue repair via regional immunity regulation. While the GelMA guarantees cell compatibility, the negatively charged HepMA successfully adsorbs the inflammatory cytokines and chemokines by electrostatic interactions and inhibits immune cell migration in vitro. Furthermore, in vivo inflammation self-limiting and regional immunity regulation efficacy is evaluated in a rat abdominal hernia model. Reduced local inflammatory cytokines and chemokines in the early stage and increased angiogenesis and ECM remodeling in the later phase confirm that the tape is an approach to maintain an optimal regional immune activation level after soft tissue injury. Overall, the reported electrospun fibrous tape will find its way into clinical transformation and solve the challenges of deep soft tissue injury.


Asunto(s)
Traumatismos de los Tejidos Blandos , Andamios del Tejido , Álcalis , Animales , Citocinas/metabolismo , Matriz Extracelular/química , Gelatina/química , Heparina , Hidrogeles/química , Inflamación/metabolismo , Ratas , Traumatismos de los Tejidos Blandos/metabolismo , Ingeniería de Tejidos , Andamios del Tejido/química
13.
Small ; 18(40): e2202156, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36056898

RESUMEN

The localization and accumulation of drugs in the body determine their therapeutic effects; however, the specific microstructure of damaged tissues hinders drug delivery. Currently, there is a shortage of effective drug carriers to breach these barriers and achieve efficient tissue and cellular delivery of drugs. In this study, an injectable double positively charged functional hydrogel microsphere with "targeting cartilage extracellular matrix", "cartilage penetration", and "cellular phagocytosis" is designed for matching the structural characteristics of joints, addressing the difficulties of drug delivery in joints. The microspheres could be adsorbed on the negatively charged cartilage surface because of their positively charged poly-lysine surface. Furthermore, the internally loaded positively charged polyamidoamine contained kartogenin, which helped further the penetration of the cartilage under the guidance of electrical charge. The microspheres could release kartogenin for more than 21 days. In in vivo experiments, the microspheres effectively improve the efficiency of drug delivery, inhibit the degradation of cartilage matrix and subchondral bone, and delay the development of osteoarthritis. As a double positively charged drug delivery system, the versatile microsphere has great potential for treating osteoarthritis and other diseases.


Asunto(s)
Hidrogeles , Osteoartritis , Anilidas , Portadores de Fármacos/química , Humanos , Hidrogeles/química , Lisina , Microesferas , Osteoartritis/tratamiento farmacológico , Fagocitosis , Ácidos Ftálicos
14.
Small ; 18(36): e2106591, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35106912

RESUMEN

The least damaging and most economical method to deliver drugs or carriers into the inner ear for treatment of disease is through the middle ear. However, the retention of drug in the middle ear is an obstacle. Here, inspired by the adhesion of mussels, a methacrylate gelatin microspheres (GM) coupling polydopamine (PDA) layer (GM@PDA) with excellent adhesive ability is constructed, and Ebselen liposomes are further loaded into the GM@PDA (GM@PDA@Lipo-Ebselen). The loading capacity of GM@PDA for Ebselen liposomes is 25 ± 1 µg mg-1 microspheres. GM@PDA@Lipo-Ebselen could be injected on round windows membrane (RWM) and tightly adheres to the surface of RWM by PDA, and the microspheres are even still attached to the RWM after 360° rotation and inverted shaking. The in vivo imaging system shows that the adhesive microspheres can prolong the retention of the middle ear cavity for more than 7 days. The hearing of mice in the GM@PDA@Lipo-Ebselen group is significantly recovered, especially on day 14 after noise exposure, and the hearing of each frequency is restored to baseline level. At 32 kHz frequency, the survival of outer hair cells recovers from 48 0± 6% to 93 ± 2%. Therefore, the adhesive and injectable hydrogel microspheres provide a promising strategy for the treatment of hearing loss.


Asunto(s)
Oído Interno , Hidrogeles , Adhesivos , Animales , Gelatina , Liposomas , Ratones , Microesferas
15.
Small ; 18(36): e2200799, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35266631

RESUMEN

Exudate management is critical to improve chronic wound healing. Herein, inspired by a Janus-structured lotus leaf with asymmetric wettability, a Janus electrospun short fiber scaffold is fabricated via electrospinning technologies and short fiber modeling. This scaffold is composed of hydrophilic 2D curcumin-loaded electrospun fiber and hydrophobic 3D short fiber via layer-by-layer assembly and electrostatic interactions which can aggregate the wound exudate by pumping from the hydrophobic layer to the hydrophilic via multiple contact points between hydrophilic and hydrophobic fibers, and simultaneously trigger the cascade release of curcumin in the upper 2D electrospun fiber. The 3D short fiber with high porosity and hydrophobicity can quickly aggregate exudate within 30 s after compounding with hydrophilic 2D electrospun fiber via a spontaneous pump. In vitro experiments show that Janus electrospun short fiber has good biocompatibility, and the cascade release of curcumin can significantly promote the proliferation and migration of fibroblasts. In vivo experiments show that it can trigger cascade release of curcumin by aggregating wound exudate, so as to accelerate wound healing process and promote collagen deposition and vascularization. Hence, this unique biometric Janus scaffold provides an alternative for chronic wound healing.


Asunto(s)
Curcumina , Nanofibras , Colágeno , Curcumina/farmacología , Fibroblastos , Nanofibras/química , Porosidad , Cicatrización de Heridas
16.
Small ; 18(46): e2204316, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36192165

RESUMEN

Tissue-specific natural anisotropic microstructures play an important role in the normal functioning of tissues, yet they remain difficult to construct by current printing techniques. Herein, a stepwise algorithm-assisted bioprinting technology for the construction of biomimetic tissues with a customizable anisotropic microstructure by combining the Adaptive Mesh Generation algorithm and the Greedy Search algorithm is developed. Based on the mechanical topology optimization design mechanism, the Adaptive Mesh Generation algorithm can generate controllable anisotropic mesh patterns with the minimum free energy in plane models according to tissue-specific requirements. Subsequently, the Greedy Search algorithm can program the generated pattern data into optimized printing paths, effectively avoiding structural deformations caused by the multiple stacking of materials and reducing the printing time. The developed bioprinting technique is suitable for various types of bioinks including polymers, hydrogels, and organic/inorganic complexes. After combining with a calcium phosphorus bioink, the compound algorithm-assisted bioprinting technique successfully customizes femurs with biomimetic chemical compositions, anisotropic microstructures, and biological properties, demonstrating its effectiveness. Additionally, algorithm-assisted bioprinting is generally suitable for most commercial extrusion bioprinters that function in the geometric code (G-code) drive mode. Therefore, the algorithm-assisted extrusion bioprinting technology offers an intelligent manufacturing strategy for the customization of anisotropic microstructures in biomimetic tissues.


Asunto(s)
Bioimpresión , Bioimpresión/métodos , Biomimética , Impresión Tridimensional , Hidrogeles/química , Algoritmos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
17.
Small ; 18(36): e2201656, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35419952

RESUMEN

Bone defect regeneration depends on the population and lifespan of M2 macrophages, which are regulated by dual signals generated by the "physical" spatial configuration of biological tissues and "molecular" chemokines. Herein, inspired by the reprogramming of macrophages, immunoengineered porous microspheres are constructed to accelerate bone repair through the regulation of both "physical" and "molecular" signals. The porous structure of injectable poly (l-lactic acid) (PLLA) microspheres prepared by the microfluidic technique provides a "physical signal" for osteogenic differentiation. Additionally, interleukin (IL)-4-loaded liposomes (Ls) are modified on PLLA microspheres through amide bonds to produce IL-4/Ls/PLLA microspheres, providing a "molecular signal" in stimulating the differentiation of macrophages to M2 type. It is confirmed that IL-4/Ls/PLLA microspheres could induce M2-macrophages polarization and potentiate osteoblast proliferation and differentiation while coculturing with macrophages and osteoblasts in vitro. Besides, IL-4/Ls/PLLA microspheres are proved to promote bone defect regeneration by inducing the conversion of M1 macrophages to M2 through dual biosignal-functional regulation in both the calvaria defect and maxillary sinus defect models. Overall, the immuno-reprogrammed IL-4/Ls/PLLA microspheres achieve the precise immuno-reprogramming of macrophages by dual biosignal-functional regulation. This immune reengineering strategy paves a way for clinical bone defect treatment.


Asunto(s)
Interleucina-4 , Osteogénesis , Regeneración Ósea/fisiología , Microesferas , Osteoblastos , Poliésteres/química
18.
J Nanobiotechnology ; 20(1): 294, 2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729570

RESUMEN

Diabetic wound is the leading cause of non-traumatic amputations in which oxidative stress and chronic inflammation are main factors affecting wound healing. Although mesenchymal stem cells (MSCs) as living materials can promote skin regeneration, they are still vulnerable to oxidative stress which limits their clinical applications. Herein, we have prepared (polylactic-co-glycolic acid) (PLGA) nanofibers electrospun with LPS/IFN-γ activated macrophage cell membrane. After defining physicochemical properties of the nanofibers modified by LPS/IFN-γ activated mouse RAW264.7 cell derived membrane (RCM-fibers), we demonstrated that the RCM-fibers improved BMMSC proliferation and keratinocyte migration upon oxidative stress in vitro. Moreover, bone marrow derived MSCs (BMMSCs)-loaded RCM-fibers (RCM-fiber-BMMSCs) accelerated wound closure accompanied by rapid re-epithelialization, collagen remodeling, antioxidant stress and angiogenesis in experimental diabetic wound healing in vivo. Transcriptome analysis revealed the upregulation of genes related to wound healing in BMMSCs when co-cultured with the RCM-fibers. Enhanced healing capacity of RCM-fiber-BMMSCs living material was partially mediated through CD200-CD200R interaction. Similarly, LPS/IFN-γ activated THP-1 cell membrane coated nanofibers (TCM-fibers) exhibited similar improvement of human BMMSCs (hBMMSCs) on diabetic wound healing in vivo. Our results thus demonstrate that LPS/IFN-γ activated macrophage cell membrane-modified nanofibers can in situ immunostimulate the biofunctions of BMMSCs, making this novel living material promising in wound repair of human diabetes.


Asunto(s)
Diabetes Mellitus , Células Madre Mesenquimatosas , Nanofibras , Animales , Diabetes Mellitus/metabolismo , Lipopolisacáridos , Células Madre Mesenquimatosas/metabolismo , Ratones , Nanofibras/química , Cicatrización de Heridas
19.
J Nanobiotechnology ; 20(1): 47, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35062972

RESUMEN

BACKGROUND: Hemostasis and repair are two essential processes in wound healing, yet early hemostasis and following vascularization are challenging to address in an integrated manner. RESULTS: In this study, we constructed a hemostatic sponge OBNC-DFO by fermentation of Komagataeibacter xylinus combined with TEMPO oxidation to obtain oxidized bacterial nanocellulose (OBNC). Then angiogenetic drug desferrioxamine (DFO) was grafted through an amide bond, and it promoted clot formation and activated coagulation reaction by rapid blood absorption due to the high total pore area (approximately 42.429 m2/g measured by BET). The further release of DFO stimulated the secretion of HIF-1α and the reconstruction of blood flow, thus achieving rapid hemostasis and vascularization in damaged tissue. This new hemostatic sponge can absorb water at a rate of approximate 1.70 g/s, rapidly enhancing clot formation in the early stage of hemostasis. In vitro and in vivo coagulation experiments (in rat tail amputation model and liver trauma model) demonstrated superior pro-coagulation effects of OBNC and OBNC-DFO to clinically used collagen hemostatic sponges (COL). They promoted aggregation and activation of red blood cells and platelets with shorter whole blood clotting time, more robust activation of endogenous coagulation pathways and less blood loss. In vitro cellular assays showed that OBNC-DFO prevailed over OBNC by promoting the proliferation of human umbilical vein endothelial cells (HUVECs). In addition, the release of DFO enhanced the secretion of HIF-1α, further strengthening vascularization in damaged skin. In the rat skin injury model, 28 days after being treated with OBNC-DFO, skin appendages (e.g., hair follicles) became more intact, indicating the achievement of structural and functional regeneration of the skin. CONCLUSION: This hemostatic and vascularization-promoting oxidized bacterial nanocellulose hemostatic sponge, which rapidly activates coagulation pathways and enables skin regeneration, is a highly promising hemostatic and pro-regenerative repair biomaterial.


Asunto(s)
Bacterias/metabolismo , Vendajes , Materiales Biocompatibles , Hemostáticos , Animales , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Células Cultivadas , Celulosa/química , Deferoxamina , Hemorragia , Hemostasis/efectos de los fármacos , Hemostáticos/metabolismo , Hemostáticos/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Nanoestructuras/química , Neovascularización Patológica/metabolismo , Porosidad , Ratas , Ratas Sprague-Dawley
20.
Proc Natl Acad Sci U S A ; 116(16): 7744-7749, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30926671

RESUMEN

Effective cancer therapies often demand delivery of combinations of drugs to inhibit multidrug resistance through synergism, and the development of multifunctional nanovehicles with enhanced drug loading and delivery efficiency for combination therapy is currently a major challenge in nanotechnology. However, such combinations are more challenging to administer than single drugs and can require multipronged approaches to delivery. In addition to being stable and biodegradable, vehicles for such therapies must be compatible with both hydrophobic and hydrophilic drugs, and release drugs at sustained therapeutic levels. Here, we report synthesis of porous silicon nanoparticles conjugated with gold nanorods [composite nanoparticles (cNPs)] and encapsulate them within a hybrid polymersome using double-emulsion templates on a microfluidic chip to create a versatile nanovehicle. This nanovehicle has high loading capacities for both hydrophobic and hydrophilic drugs, and improves drug delivery efficiency by accumulating at the tumor after i.v. injection in mice. Importantly, a triple-drug combination suppresses breast tumors by 94% and 87% at total dosages of 5 and 2.5 mg/kg, respectively, through synergy. Moreover, the cNPs retain their photothermal properties, which can be used to significantly inhibit multidrug resistance upon near-infrared laser irradiation. Overall, this work shows that our nanovehicle has great potential as a drug codelivery nanoplatform for effective combination therapy that is adaptable to other cancer types and to molecular targets associated with disease progression.


Asunto(s)
Antineoplásicos , Sistemas de Liberación de Medicamentos/métodos , Nanotubos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Antineoplásicos/uso terapéutico , Femenino , Oro , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Ratones Desnudos , Técnicas Analíticas Microfluídicas , Nanomedicina , Nanotubos/química , Nanotubos/efectos de la radiación , Neoplasias Experimentales/tratamiento farmacológico , Procesos Fotoquímicos , Porosidad , Silicio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA