Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Drug Deliv Transl Res ; 14(2): 342-359, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37587289

RESUMEN

Glioblastoma multiforme (GBM) is regarded as a highly aggressive brain cancer with a poor prognosis. There is an increase in the expression of P-glycoprotein (P-gp), responsible for multidrug resistance (MDR), making it a potential target for improving drug responses. Additionally, glioblastoma stem cells (GSCs) increase resistance to chemo- and radiotherapy and play a major role in cancer relapse. In this study, we targeted P-gp using a small molecule inhibitor, reversan (RV), to inhibit MDR that prolonged the retention of drugs in the cytosolic milieu. To eliminate GBM and GSCs, we have used two well-established anti-cancer drugs, regorafenib (RF) and curcumin (CMN). To improve the pharmacokinetics and decrease systemic delivery of drugs, we developed nanostructure hybrid lipid capsules (nHLCs), where hydrophobic drugs can be loaded in the core, and their physicochemical properties were determined by dynamic light scattering (DLS) and cryo-scanning electron microscopy (SEM). Inhibition of MDR by RV has also shown enhanced retention of nHLC in GBM cells. Co-delivery of drug-loaded nHLCs, pre-treated with RV, exhibited superior cytotoxicity in both GBM and GSCs than their individual doses and effectively reduced the size and stemness of tumor spheres and accelerated the rate of apoptosis, suggesting a promising treatment for glioblastoma.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Células Madre Neoplásicas , Resistencia a Múltiples Medicamentos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Lípidos , Línea Celular Tumoral
2.
ACS Appl Mater Interfaces ; 15(50): 58151-58165, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38063494

RESUMEN

Cancer stem cells (CSCs) present a formidable challenge in cancer treatment due to their inherent resistance to chemotherapy, primarily driven by the overexpression of ABC transporters and multidrug resistance (MDR). Despite extensive research on pharmacological small-molecule inhibitors, effectively managing MDR and improving chemotherapeutic outcomes remain elusive. On the other hand, magnetic hyperthermia (MHT) holds great promise as a cancer therapeutic, but there is limited research on its potential to reverse MDR in breast CSCs and effectively eliminate CSCs through combined chemo-hyperthermia. To address these gaps, we developed tumor microenvironment-sensitive, drug-loaded poly(propylene sulfide) (PPS)-coated magnetic nanoparticles (PPS-MnFe). These nanoparticles were employed to investigate hyperthermia sensitivity and MDR reversion in breast CSCs, comparing their performance to that of small-molecule inhibitors. Additionally, we explored the efficacy of combined chemo-hyperthermia in killing CSCs. CSC-enriched breast cancer cells were subjected to low-dose MHT at 42 °C for 30 min and then treated with the chemical MDR inhibitor salinomycin (SAL). The effectiveness of each treatment in inhibiting MDR was assessed by measuring the efflux of the MDR substrate, rhodamine 123 (R123) dye. Notably, MHT induced a prolonged reversal of MDR activity compared with SAL treatment alone. After successfully inhibiting MDR, the breast CSCs were exposed to chemotherapy using paclitaxel to trigger synergistic cell death. The combination of MHT and chemotherapy demonstrated remarkable reductions in stemness properties, MDR reversal, and the effective eradication of breast CSCs in this innovative dual-modality approach.


Asunto(s)
Neoplasias de la Mama , Hipertermia Inducida , Humanos , Femenino , Polipropilenos/farmacología , Resistencia a Antineoplásicos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Resistencia a Múltiples Medicamentos , Células Madre Neoplásicas/patología , Concentración de Iones de Hidrógeno , Fenómenos Magnéticos , Línea Celular Tumoral , Microambiente Tumoral
3.
Biomater Sci ; 11(11): 3938-3951, 2023 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-37093244

RESUMEN

With the increasing dependence on fluorescence bioimaging, luminogens with aggregation-induced emission (AIE) properties have gained significant attention due to their excellent photostabilization, minimal photobleaching, high reliability, and superior biocompatibility. Since mitochondria are crucial subcellular organelles in eukaryotic cells with important biological functions, organelle-specific AIE emitters with distinct functions have been highly sought after, but with limited success using simple synthetic methods. Here, we describe a strategy for synthesizing two triphenylamine (TPA) based acrylonitriles, tethered to different donor groups, TPA and phenothiazine (PTZ), respectively, with superior AIE properties using Suzuki coupling. We conducted a systematic and detailed experimental analysis of the structural characteristics of both AIE luminogens, which exhibited excellent photostability, a large Stokes shift, and bright solid-state emission. A cell viability study carried out with F1 and F2 dyes revealed that both luminogens exhibited excellent biocompatibility. Based on fluorescence experiments, F2 displayed excellent AIE characteristics, permeability, biocompatibility, and photostability compared to rhodamine 123, allowing it to selectively stain and track mitochondria in cancer cells over an extended period of time. The Pearson correlation coefficient of F2 and rhodamine 123 was estimated to have an r-value of 0.99. Our findings are expected to provide insight into the synthesis of an extensive archive of AIE-based acrylonitriles with fascinating properties for mitochondrial staining.


Asunto(s)
Colorantes Fluorescentes , Mitocondrias , Humanos , Rodamina 123 , Reproducibilidad de los Resultados , Colorantes Fluorescentes/química , Células HeLa
4.
Acta Biomater ; 161: 1-36, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36907233

RESUMEN

Immunotherapy involves the therapeutic alteration of the patient's immune system to identify, target, and eliminate cancer cells. Dendritic cells, macrophages, myeloid-derived suppressor cells, and regulatory T cells make up the tumor microenvironment. In cancer, these immune components (in association with some non-immune cell populations like cancer-associated fibroblasts) are directly altered at a cellular level. By dominating immune cells with molecular cross-talk, cancer cells can proliferate unchecked. Current clinical immunotherapy strategies are limited to conventional adoptive cell therapy or immune checkpoint blockade. Targeting and modulating key immune components presents an effective opportunity. Immunostimulatory drugs are a research hotspot, but their poor pharmacokinetics, low tumor accumulation, and non-specific systemic toxicity limit their use. This review describes the cutting-edge research undertaken in the field of nanotechnology and material science to develop biomaterials-based platforms as effective immunotherapeutics. Various biomaterial types (polymer-based, lipid-based, carbon-based, cell-derived, etc.) and functionalization methodologies for modulating tumor-associated immune/non-immune cells are explored. Additionally, emphasis has been laid on discussing how these platforms can be used against cancer stem cells, a fundamental contributor to chemoresistance, tumor relapse/metastasis, and failure of immunotherapy. Overall, this comprehensive review strives to provide up-to-date information to an audience working at the juncture of biomaterials and cancer immunotherapy. STATEMENT OF SIGNIFICANCE: Cancer immunotherapy possesses incredible potential and has successfully transitioned into a clinically lucrative alternative to conventional anti-cancer therapies. With new immunotherapeutics getting rapid clinical approval, fundamental problems associated with the dynamic nature of the immune system (like limited clinical response rates and autoimmunity-related adverse effects) have remained unanswered. In this context, treatment approaches that focus on modulating the compromised immune components within the tumor microenvironment have garnered significant attention amongst the scientific community. This review aims to provide a critical discussion on how various biomaterials (polymer-based, lipid-based, carbon-based, cell-derived, etc.) can be employed along with immunostimulatory agents to design innovative platforms for selective immunotherapy directed against cancer and cancer stem cells.


Asunto(s)
Materiales Biocompatibles , Neoplasias , Humanos , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Neoplasias/patología , Inmunoterapia/métodos , Células Madre Neoplásicas/patología , Lípidos , Microambiente Tumoral
5.
Nanoscale ; 15(16): 7384-7402, 2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-36751724

RESUMEN

Magnetic nanoparticle (MNP) delivery systems are promising for targeted drug delivery, imaging, and chemo-hyperthermia of cancer; however, their uses remain limited primarily due to their toxicity associated with reactive oxygen species (ROS) generation, targeted delivery, and biodegradation. Attempts employing polymer coatings to minimize the toxicity, along with other challenges, have had limited success. We designed a novel yet generic 'one-for-all' polypropylene sulphide (PPS) coated magnetic nano-delivery system (80 ± 15 nm) as a multi-faceted approach for significant biocompatibility improvement, loading of multiple drugs, ROS-responsive delivery, and combined chemo-hyperthermia therapy for biomedical applications. Three distinct MNP systems (15 ± 1 nm) were fabricated, coated with PPS polymer, and investigated to validate our hypothesis and design. Simultaneous degradation of MNPs and PPS coatings with ROS-scavenging characteristics boosted the biocompatibility of MNPs 2-3 times towards non-cancerous fibroblasts (NIH3T3) and human epithelial cells (HEK293). In an alternating magnetic field, PPS-MNPs (MnFe) had the strongest heating characteristics (SAR value of 240 W g-1). PPS-MNP drug-loaded NPs were efficiently internalised into cells and released 80% of the drugs under tumor microenvironment-mimicking (pH 5-7, ROS) conditions, and demonstrated effective chemo-hyperthermia (45 °C) application for breast cancer cells with 95% cell death in combined treatment vs. 55% and 30% cell death in only hyperthermia and chemotherapy respectively.


Asunto(s)
Hipertermia Inducida , Nanopartículas de Magnetita , Nanopartículas , Neoplasias , Animales , Ratones , Humanos , Polipropilenos/farmacología , Nanopartículas de Magnetita/uso terapéutico , Especies Reactivas de Oxígeno , Células HEK293 , Células 3T3 NIH , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Hipertermia Inducida/métodos , Fenómenos Magnéticos , Microambiente Tumoral
6.
Macromol Biosci ; 23(1): e2200347, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36353916

RESUMEN

Nanofibrous microspheres (NFM) are emerging as prominent next-generation biomimetic injectable scaffold system for stem cell delivery and different tissue regeneration where nanofibrous topography facilitates ECM-like stem cells niches. Addition of osteogenic bioactive nanosilicate platelets within NFM can provide osteoconductive cues to facilitate matrix mediated osteogenic differentiation of stem cells and enhance the efficiency of bone tissue regeneration. In this study, gelatin nanofibrous microspheres are prepared containing fluoride-doped laponite XL21 (LP) using the emulsion mediated thermal induce phase separation (TIPS) technique. Systematic studies are performed to understand the effect of physicochemical properties of biomimicking NFM alone and with different concentrations of LP on human dental follicle stem cells (hDFSCs), their cellular attachment, proliferation, and osteogenic differentiation. The study highlights the effect of LP nanosilicate with biomimicking nanofibrous injectable scaffold system aiding in enhancing stem cell differentiation under normal physiological conditions compared to NFM without LP. The laponite-NFM shows suitability as excellent injectable biomaterials system for stem cell attachment, proliferation and osteogenic differentiation for stem cell transplantation and bone tissue regeneration.


Asunto(s)
Nanofibras , Osteogénesis , Humanos , Gelatina/farmacología , Gelatina/química , Microesferas , Nanofibras/química , Saco Dental , Diferenciación Celular , Trasplante de Células Madre , Andamios del Tejido/química , Ingeniería de Tejidos/métodos
7.
Biomater Adv ; 139: 212981, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35882137

RESUMEN

Stem cells based novel treatment modality for degenerative and immune dysfunction diseases created a huge demand of suitable carriers to support ex-vivo production of quality stem cells, and effective in-vivo transplantation of stem cells and their fate. In spite of promising candidature of nanofibrous microspheres (NFM) to recreate native stem cell niches to be used for possible scaling-up for ex-vivo stem cells expansion, it remains fairly unexplored. A systematic study on the stem cell-NFM interaction comparative with commercial microspheres (CM) has been performed for the first time. Gelatin NFM with variable physicochemical properties such as size, surface properties, surface chemistry, and variable degradability were prepared using microemulsion coupled with thermally induced phase separation (TIPS) method. Effect of physicochemical properties of NFM and their cellular interaction such as binding, morphology, metabolic activity and proliferation studies were performed using human bone marrow-derived mesenchymal stem cells (hBMSCs), human dental follicle stem cells (hDFSCs) and human gingival fibroblast (HGF) cells and compared with the commercial and solid microspheres. Gelatin NFM supports excellent cell binding, proliferation, metabolic activities and chemical cues specific differentiation. All out-turns indicate that NFM stand to be an outstanding candidate for ex-vivo cells' expansion and injectable carriers for stem cell transplantation.


Asunto(s)
Gelatina , Nanofibras , Gelatina/química , Humanos , Microesferas , Nanofibras/química , Nicho de Células Madre , Trasplante de Células Madre
8.
Int J Pharm ; 613: 121358, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34896560

RESUMEN

Ideal dressing materials for complex and large asymmetric burns should have the dual properties of anti-bacterial and regenerative with advanced applicability of direct deposit on the wound at the patient bedside. In this study, core-shell nanofibers (polycaprolactone; PCL and polyethylene oxide; PEO) with different percent of silver sulfadiazine (SSD) loading (2-10%) were prepared by the airbrushing method using a custom build device. Results indicate a sustained release profile of silver sulfadiazine (SSD) up to 28 days and concentration-dependent anti-bacterial activity. The morphology and proliferation of human dermal fibroblast (HDF) cells and human dental follicle stem cells (HDFSC) on the silver sulfadiazine loaded nanofibers confirm the biocompatibility of airbrushed nanofibers. Moreover, upregulation of extracellular matrix (ECM) proteins (Col I, Col III, and elastin) support the differentiation and regenerative properties of silver sulfadiazine nanofiber mats. This was further confirmed by the complete recovery of rabbit burn wound models within 7 days of silver sulfadiazine loaded nanofiber dressing. Histopathology data show silver sulfadiazine loaded core-shell nanofibers' anti-inflammatory and proliferative activity without any adverse response on the tissue. Overall data display that the airbrushed silver sulfadiazine-loaded core-shell nanofibers are effective dressing material with the possibility of direct fiber deposition on the wound to cover, heal, and regenerate large asymmetric burn wounds.


Asunto(s)
Quemaduras , Nanofibras , Animales , Vendajes , Quemaduras/tratamiento farmacológico , Humanos , Conejos , Sulfadiazina de Plata , Cicatrización de Heridas
9.
Colloids Surf B Biointerfaces ; 204: 111775, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33940518

RESUMEN

Cancer stem cells (CSCs) comprise a diminutive population of the tumor but pose major obstacles in cancer treatment, often their presence being correlated with poor prognosis, therapeutic resistance and relapse. Nanocarriers of combined drugs regimes demonstrate improved pharmacokinetics and decreased systemic toxicity by targeting the bulk tumor cells along with CSCs, holding the key to future successful chemotherapy. Herein, we developed lipid nanocapsules (LNCs) with co-encapsulated paclitaxel (PTX) and salinomycin (SAL) to eliminate breast cancer cells (MCF-7; non-bCSCs) and cancer stem cells (bCSCs) respectively. LNCs loaded with either PTX or SAL alone or in combination were fabricated by the phase inversion temperature (PIT) method. Physicochemical properties such as nano-size (90 ±â€¯5 nm) and spherical morphology of LNCs were confirmed by dynamic light scattering (DLS) and scanning electron microscopy (SEM) respectively. More than 98 % encapsulation efficiency of drug, alone or in combination, and their controlled drug release was obtained. Drug loaded LNCs were efficiently internalized and exhibited cytotoxicity in non-bCSCs and bCSCs, with dual drug loaded LNCs offering superior cytotoxicity and anti-bCSCs property. Drug loaded nanocapsules induced apoptosis in bCSCs, potentiated with the co-delivery of paclitaxel and salinomycin. Synergistic cytotoxic effect on both cells, non-bCSCs and bCSCs and effective reduction of the tumor mammospheres growth by co-encapsulated paclitaxel and salinomycin suggest LNCs to be promising for treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama , Nanocápsulas , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Lípidos , Células Madre Neoplásicas , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Piranos
10.
Colloids Surf B Biointerfaces ; 202: 111702, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33780906

RESUMEN

C1, a synthetic analog of curcumin, has been reported to show potent antiproliferative effects against a variety of cancer cells. Here, we report a strong anticancer activity of the folate receptor-targeted lipid nanoparticle formulation of C1 against cancer cells and cancer stem cells both in 2D culture and 3D spheroids. The size of the C1 encapsulated folic acid functionalized nanoliposomes (Lipos-C1) was determined to be 83 ± 17 nm. Lipos-C1 nanoparticles displayed sustained C1 release kinetics at both pH 7.4 and 5.5. The folate receptor (FR) targeted nanoliposomes were internalized into FR-positive KB cells via the folate receptor-mediated endocytosis process. Lipos-C1 killed KB cells much more efficiently than C1. Lipos-C1 depolymerized microtubules, generated ROS, caused DNA damage, and induced apoptosis in KB cells. Importantly, Lipos-C1 strongly inhibited the growth of the 3D KB spheroids than C1. Interestingly, Lipos-C1 also suppressed cancer stem cells (CSCs) enriched MCF-7 mammosphere growth by impeding breast cancer stem cells (BCSCs) enrichment, growth, and proliferation. The results suggested that Lipos-C1 could be a promising nanoformulation for cancer chemotherapy.


Asunto(s)
Neoplasias de la Mama , Curcumina , Nanopartículas , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Curcumina/farmacología , Femenino , Ácido Fólico , Humanos , Células MCF-7 , Células Madre Neoplásicas
11.
Colloids Surf B Biointerfaces ; 190: 110927, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32169777

RESUMEN

Lipid Nanocapsules (LNCs) have been used for drug delivery in cells and animal models for several years. LNCs with unique physicochemical properties for favorable biorecognition, biocompatibility and stimuli responsive (pH/temperature etc.) properties i.e., smart-LNCs, are most promising for future nanomedicine applications. However, conventional phase inversion temperature (PIT) method of LNCs preparation may not be suitable for the fabrication of thermally labile drug loaded LNCs and smart-LNCs. Herein, we report for the first time, a novel low temperature (LT) method for the preparation of LNCs (including smart-LNCs of size 25-150 nm), hereafter, named as nanostructure hybrid lipid capsules (nHLCs), comprising safe excipients such as oil (Labrafac™ PG), surfactant (Kolliphor® HS 15, Brij® S100), and lipid (Lipoid S-75, Lipoid S PC-3, Lipoid PE 18:1/18:1, Lipoid PC 16:0/16:0 etc.). Effects of process parameters on the physicochemical properties of nHLCs were probed to optimize the process. Ternary phase diagram shows that our method allows for great flexibility in the formation of nHLCs with tailored size and composition. This method resulted in drug loaded (regorafenib used as model drug) nHLCs with 95 % encapsulation efficiency and sustained release profile at 37 °C. The drug loaded nHLCs (as prepared or in lyophilized form) has excellent storage stability at 4 °C (for more than one month) as well as biocompatibility similar to that of LNCs prepared by PIT method. Our novel LT method of LNCs (i.e. nHLCs) preparation is a generic method for the development of drug loaded (including thermally labile) and smart-LNCs for future nanomedicine applications.


Asunto(s)
Materiales Biocompatibles/química , Sistemas de Liberación de Medicamentos , Lípidos/química , Nanoestructuras/química , Compuestos de Fenilurea/química , Piridinas/química , Temperatura , Cápsulas/química , Tamaño de la Partícula , Propiedades de Superficie
12.
ACS Appl Bio Mater ; 3(10): 6811-6822, 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-35019344

RESUMEN

Co-eradication of cancer stem cells (CSCs) along with cancer cells have emerged as an immediate necessity to combat the rapid progression, therapeutic resistance, and relapse of cancer. Curcumin (CMN) has been well established for anticancer activity against a variety of cancers with an ability to eliminate CSCs. In spite of its extensive therapeutic potential, clinical applicability is impeded due to its highly hydrophobic nature. In this study, we developed CMN-loaded nanostructure hybrid lipid capsules (CMN-nHLCs) of three sizes (25, 75, and 150 nm) with 4% (w/w) loading capacity using our low-temperature (LT) method. Molecular interaction between different ingredients using fourier transform infrared (FTIR) analysis shows self-arrangement of ingredients into CMN-loaded nHLCs without any chemical bonding. CMN-nHLCs show a controlled release of CMN from nHLCs at 37 °C and long-term storage stability at 4 °C. CMN-nHLCs show ∼2.5-fold enhanced anticancer efficacy compared to free CMN in breast cancer cells (non-bCSCs) and breast cancer stem-like cells (bCSCs). CMN-nHLCs are effectively internalized into MCF-7 cells (non-bCSCs and bCSCs) and cause significant reduction in their mammosphere size/number and stemness. nHLCs provide improved physicochemical properties of CMN and superior anticancer efficacy by co-eradiating both non-bCSCs and bCSCs, suggesting a promising candidature of CMN-nHLCs for breast cancer treatment.

13.
Mol Pharm ; 17(1): 284-300, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31794223

RESUMEN

Therapeutic protein depots have limited clinical success because of the presence of critical preparation barriers such as low encapsulation, uncontrolled release, and activity loss during processing and storage. In the present study, we used our novel protein-nanoencapsulation (into sugar-glass nanoparticle; SGnP) platform to prepare a protein depot to overcome the abovementioned formidable challenges. The SGnP-mediated microparticle protein depot has been validated using four model proteins (bovine serum albumin, horseradish peroxidase, fibroblastic growth factor, and epidermal growth factor) and model biodegradable poly(lactic-co-glycolic acid) polymer system. The results show that our protein-nanoencapsulation-mediated platform provides a new generic platform to prepare a protein depot through the conventional emulsion method of any polymer and single/multiple protein systems. This protein depot has the required pharmaceutical properties such as high encapsulation efficiency, burst-free sustained release, and protein preservation during processing and storage, making it suitable for off-the-shelf use in therapeutic protein delivery and tissue engineering applications.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Proteínas/administración & dosificación , Preparaciones de Acción Retardada/química , Composición de Medicamentos/métodos , Emulsiones , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/metabolismo , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/metabolismo , Vidrio/química , Peroxidasa de Rábano Silvestre/química , Peroxidasa de Rábano Silvestre/metabolismo , Humanos , Células MCF-7 , Microscopía Electrónica de Rastreo , Nanopartículas/ultraestructura , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Proteínas/química , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/metabolismo , Esferoides Celulares/efectos de los fármacos , Azúcares
14.
ACS Appl Bio Mater ; 2(12): 5340-5348, 2019 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-35021534

RESUMEN

Gelatin nanofibers have gained significant attention for different biomedical applications, as they provide a suitable environment for cell attachment, growth, and proliferation compared to the other biopolymers and synthetic polymers. Airbrushing/solution-blow-spinning could overcome the limitation of the conventional electrospinning method of nanofiber preparation. The present study reports the fabrication of nano/microfibers from commercially available low-molecular-weight gelatin of animal origin as a first-time study. The effect of various airbrushing parameters, namely, the concentration of gelatin solution, air pressure, and polymer solution flow rate on the fiber quality, morphology, and diameters, was studied. Finally, the biological evaluation of the airbrushed gelatin nanofibers was performed using human bone marrow-derived mesenchymal stem cells (hBMSCs). Gelatin nanofibers exhibit excellent biocompatibility and support the growth of hBMSCs similar to electrospun gelatin nanofibers. Our airbrushing technique is an easy, low-cost, and scalable method to fabricate the gelatin nanofibers for different biomedical applications such as tissue engineering, wound healing, and substrate for delivery of bioactive molecules.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...