Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Biotechnol Bioeng ; 120(12): 3638-3654, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37668186

RESUMEN

Mesenchymal stem cells and macrophages (MQ) are two very important cells involved in the normal wound healing process. It is well understood that topological cues and mechanical factors can lead to different responses in stem cells and MQ by influencing their shape, cytoskeleton proliferation, migration, and differentiation, which play an essential role in the success or failure of biomaterial implantation and more importantly wound healing. On the other hand, the polarization of MQ from proinflammatory (M1) to prohealing (M2) phenotypes has a critical role in the acceleration of wound healing. In this study, the morphology of different MQ subtypes (M0, M1, and M2) was imprinted on a silicon surface (polydimethylsiloxane [PDMS]) to prepare a nano-topography cell-imprinted substrate with the ability to induce anti-inflammatory effects on the mouse adipose-derived stem cells (ADSCs) and RAW264.7 monocyte cell line (MO). The gene expression profiles and flow cytometry of MQ revealed that the cell shape microstructure promoted the MQ phenotypes according to the specific shape of each pattern. The ELISA results were in agreement with the gene expression profiles. The ADSCs on the patterned PDMS exhibited remarkably different shapes from no-patterned PDMS. The MOs grown on M2 morphological patterns showed a significant increase in expression and section of anti-inflammatory cytokine compared with M0 and M1 patterns. The ADSCs homing in niches heavily deformed the cytoskeletal, which is probably why the gene expression and phenotype unexpectedly changed. In conclusion, wound dressings with M2 cell morphology-induced surfaces are suggested as excellent anti-inflammatory and antiscarring dressings.


Asunto(s)
Macrófagos , Células Madre Mesenquimatosas , Ratones , Animales , Macrófagos/metabolismo , Citocinas/metabolismo , Cicatrización de Heridas , Células Madre Mesenquimatosas/metabolismo , Antiinflamatorios/farmacología
2.
Biomed Mater ; 18(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36693281

RESUMEN

It is well established that surface topography can affect cell functions. However, finding a reproducible and reliable method for regulating stem cell behavior is still under investigation. It has been shown that cell imprinted substrates contain micro- and nanoscale structures of the cell membrane that serve as hierarchical substrates, can successfully alter stem cell fate. This study investigated the effect of the overall cell shape by fabricating silicon wafers containing pit structure in the average size of spherical-like chondrocytes using photolithography technique. We also used chondrocyte cell line (C28/I2) with spindle-like shape to produce cell imprinted substrates. The effect of all substrates on the differentiation of adipose-derived mesenchymal stem cells (ADSCs) has been studied. The AFM and scanning electron microscopy images of the prepared substrates demonstrated that the desired shapes were successfully transferred to the substrates. Differentiation of ADSCs was investigated by immunostaining for mature chondrocyte marker, collagen II, and gene expression of collagen II, Sox9, and aggrecan markers. C28/I2 imprinted substrate could effectively enhanced chondrogenic differentiation compared to regular pit patterns on the wafer. It can be concluded that cell imprinted substrates can induce differentiation signals better than engineered lithographic substrates. The nanostructures on the cell-imprinted patterns play a crucial role in harnessing cell fate. Therefore, the patterns must include the nano-topographies to have reliable and reproducible engineered substrates.


Asunto(s)
Condrocitos , Células Madre Mesenquimatosas , Diferenciación Celular , Células Madre , Colágeno/metabolismo , Condrogénesis , Células Cultivadas
3.
J Cell Mol Med ; 26(23): 5929-5942, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36412036

RESUMEN

Different growth factors can regulate stem cell differentiation. We used keratinocyte growth factor (KGF) to direct adipose-derived stem cells (ASCs) differentiation into keratinocytes. To enhance KGF bioavailability, we targeted KGF for collagen by fusing it to collagen-binding domain from Vibrio mimicus metalloprotease (vibrioCBD-KGF). KGF and vibrioCBD-KGF were expressed in Escherichia coli and purified to homogeneity. Both proteins displayed comparable activities in stimulating proliferation of HEK-293 and MCF-7 cells. vibrioCBD-KGF demonstrated enhanced collagen-binding affinity in immunofluorescence and ELISA. KGF and vibrioCBD-KGF at different concentrations (2, 10, and 20 ng/ml) were applied for 21 days on ASCs cultured on collagen-coated plates. Keratinocyte differentiation was assessed based on morphological changes, the expression of keratinocyte markers (Keratin-10 and Involucrin), and stem cell markers (Collagen-I and Vimentin) by real-time PCR or immunofluorescence. Our results indicated that the expression of keratinocyte markers was substantially increased at all concentrations of vibrioCBD-KGF, while it was observed for KGF only at 20 ng/ml. Immunofluorescence staining approved this finding. Moreover, down-regulation of Collagen-I, an indicator of differentiation commitment, was more significant in samples treated with vibrioCBD-KGF. The present study showed that vibrioCBD-KGF is more potent in inducing the ASCs differentiation into keratinocytes compared to KGF. Our results have important implications for effective skin regeneration using collagen-based biomaterials.


Asunto(s)
Diferenciación Celular , Factor 7 de Crecimiento de Fibroblastos , Queratinocitos , Células Madre , Humanos , Colágeno , Colágeno Tipo I/genética , Factor 7 de Crecimiento de Fibroblastos/farmacología , Células HEK293 , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos
4.
Sci Rep ; 11(1): 12130, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34108580

RESUMEN

It has been proved that cell-imprinted substrates molded from template cells can be used for the re-culture of that cell while preserving its normal behavior or to differentiate the cultured stem cells into the template cell. In this study, a microfluidic device was presented to modify the previous irregular cell-imprinted substrate and increase imprinting efficiency by regular and objective cell culture. First, a cell-imprinted substrate from template cells was prepared using a microfluidic chip in a regular pattern. Another microfluidic chip with the same pattern was then aligned on the cell-imprinted substrate to create a chondrocyte-imprinted-based integrated microfluidic device. Computational fluid dynamics (CFD) simulations were used to obtain suitable conditions for injecting cells into the microfluidic chip before performing experimental evaluations. In this simulation, the effect of input flow rate, number per unit volume, and size of injected cells in two different chip sizes were examined on exerted shear stress and cell trajectories. This numerical simulation was first validated with experiments with cell lines. Finally, chondrocyte was used as template cell to evaluate the chondrogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs) in the chondrocyte-imprinted-based integrated microfluidic device. ADSCs were positioned precisely on the chondrocyte patterns, and without using any chemical growth factor, their fibroblast-like morphology was modified to the spherical morphology of chondrocytes after 14 days of culture. Both immunostaining and gene expression analysis showed improvement in chondrogenic differentiation compared to traditional imprinting methods. This study demonstrated the effectiveness of cell-imprinted-based integrated microfluidic devices for biomedical applications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Condrocitos/citología , Condrogénesis , Dispositivos Laboratorio en un Chip/estadística & datos numéricos , Células Madre Mesenquimatosas/citología , Técnicas Analíticas Microfluídicas/métodos , Animales , Bioimpresión , Células Cultivadas , Humanos , Conejos
5.
Vet Med Sci ; 7(4): 1417-1425, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33939304

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is a systemic chronic disease with synovial membrane, tendon and articular tissue inflammation. Current treatments of RA have many side effects and are quite expensive. Today, new treatments procedures and inexpensive herbal drugs are developed. Marham-Mafasel is mainly made out of two traditional herbs (Arnebia euchroma and Martricaria chamomilla). OBJECTIVE: In this study, for the first time, the impact of Marham-Mafasel on joint inflammation, histopathological changes and IL-1ß gene expression was evaluated in RA animal model. METHODS: The RA was induced by a single s.c. injection of 0.1 ml Freund's complete adjuvant into the left hind footpad. In continuous, 15 RA male Wistar rats were used in three groups: I: Control; II: Treatment I (Piroxicam) and III: Treatment II (Marham-Mafasel). The volume of the hind paw was measured every day from 0 to 19 using water changed volume approach. The inflammation in the joint was evaluated using histopathology assay and gene expression of IL-1ß was evaluated with use of Real-Time PCR. RESULTS: Hind paw swelling of Marham-Mafasel at days 10th and 19th was reduced compared with the control group (p < 0.05). There was no statistically difference in histological degrading and changes index in three groups (p ≥ 0.05). Relative expression of IL-1ß in Marham-Mafasel group was significantly decreased compared with other groups. CONCLUSION: The co-administration of M. Chamomile and A. euchroma, called Marham-Mafasel, decreases IL-1ß gene expression that leads to a reduction in inflammation in rheumatoid arthritis (RA) animal model.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Reumatoide/tratamiento farmacológico , Boraginaceae/química , Inflamación/tratamiento farmacológico , Interleucina-1beta/metabolismo , Matricaria/química , Animales , Masculino , Medicina Tradicional , Extractos Vegetales/química , Extractos Vegetales/farmacología , Ratas , Ratas Wistar
6.
Mater Sci Eng C Mater Biol Appl ; 121: 111794, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33579444

RESUMEN

Separating cells from the body and cultivating them in vitro will alter the function of cells. Therefore, for optimal cell culture in the laboratory, conditions similar to those of their natural growth should be provided. In previous studies, it has been shown that the use of cellular shape at the culture surface can regulate cellular function. In this work, the efficiency of the imprinting method increased by using microfluidic chip design and fabrication. In this method, first, a cell-imprinted substrate of chondrocytes was made using a microfluidic chip. Afterwards, stem cells were cultured on a cell-imprinted substrate using a second microfluidic chip aligned with the substrate. Therefore, stem cells were precisely placed on the chondrocyte patterns on the substrate and their fibroblast-like morphology was changed to chondrocyte's spherical morphology after 14-days culture in the chip without using any chemical growth factor. After chondrogenic differentiation and in vitro assessments (real-time PCR and immunocytotoxicity), differentiated stem cells were transferred on a collagen-hyaluronic acid scaffold and transplanted in articular cartilage defect of the rabbit. After 6 months, the post-transplantation analysis showed that the articular cartilage defect had been successfully regenerated in differentiated stem cell groups in comparison with the controls. In conclusion, this study showed the potency of the imprinting method for inducing chondrogenicity in stem cells, which can be used in clinical trials due to the safety of the procedure.


Asunto(s)
Cartílago Articular , Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Células Cultivadas , Condrocitos , Condrogénesis , Dispositivos Laboratorio en un Chip , Conejos , Regeneración , Ingeniería de Tejidos
7.
Mater Sci Eng C Mater Biol Appl ; 121: 111836, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33579474

RESUMEN

To some extent, cell therapy for myocardial infarction (MI) has supported the idea of cardiac repair; however, further optimizations are inevitable. Combined approaches that comprise suitable cell sources and supporting molecules considerably improved its effect. Here, we devised a strategy of simultaneous transplantation of human cardiac progenitor cells (CPCs) and an optimized oxygen generating microparticles (MPs) embedded in fibrin hydrogel, which was injected into a left anterior descending artery (LAD) ligating-based rat model of acute myocardial infarction (AMI). Functional parameters of the heart, particularly left ventricular systolic function, markedly improved and reached pre-AMI levels. This functional restoration was well correlated with substantially lower fibrotic tissue formation and greater vascular density in the infarct area. Our novel approach promoted CPCs retention and differentiation into cardiovascular lineages. We propose this novel co-transplantation strategy for more efficient cell therapy of AMI which may function by providing an oxygen-rich microenvironment, and thus regulate cell survival and differentiation.


Asunto(s)
Infarto del Miocardio , Oxígeno , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Infarto del Miocardio/terapia , Ratas , Células Madre , Función Ventricular Izquierda
8.
J Gene Med ; 22(12): e3288, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33047833

RESUMEN

BACKGROUND: Dentin matrix protein 1 (DMP1) is highly expressed in mineralized tooth and bone, playing a critical role in mineralization and phosphate metabolism. One important role for the expression of DMP1 in the nucleus of preosteoblasts is the up-regulation of osteoblast-specific genes such as osteocalcin and alkaline phosphatase1 . The present study aimed to investigate the potential application of human DMP1 promoter as an indicator marker of osteoblastic differentiation. METHODS: In the present study, we developed DMP1 promoter-DsRed-GFP knock-in mesenchymal stem cell (MSCs) via the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system that enabled automatic detection of osteoblast differentiation. With the application of a homology-directed knock-in strategy, a 2-kb fragment of DMP1 promoter, which was inserted upstream of the GFP and DsRed reporter cassette, was integrated into the human ROSA locus to generate double fluorescent cells. We further differentiated MSCs under osteogenic media to monitor the fate of MSCs. First, cells were transfected using CRISPR/Cas9 plasmids, which culminated in MSCs with a green fluorescence intensity, then GFP-positive cells were selected using puromycin. Second, the GFP-positive MSCs were differentiated toward osteoblasts, which demonstrated an increased red fluorescence intensity. The osteoblast differentiation of MSCs was also verified by performing alkaline phosphatase and Alizarin Red assays. RESULTS: We have exploited the DMP1 promoter as a predictive marker of MSC differentiation toward osteoblasts. Using the CRISPR/Cas9 technology, we have identified a distinctive change in the fluorescence intensities of GFP knock-in (green) and osteoblast differentiated MSCs 2 . CONCLUSIONS: The data show that DMP1-DsRed-GFP knock-in MSCs through CRISPR/Cas9 technology provide a valuable indicator for osteoblast differentiation. Moreover, The DMP1 promoter might be used as a predictive marker of MSCs differentiated toward osteoblasts.


Asunto(s)
Sistemas CRISPR-Cas , Diferenciación Celular , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Técnicas de Sustitución del Gen/métodos , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteogénesis , Fosfoproteínas/antagonistas & inhibidores , Proliferación Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas
9.
Anticancer Agents Med Chem ; 20(17): 2041-2052, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32532197

RESUMEN

BACKGROUND AND PURPOSE: This study subjected a rat model to the extracts of muscle and shell tissues from Portunus segnis to assess their therapeutic effects on the HT-29 colon cancer cells as well as on colonic Aberrant Crypt Foci (ACF) induced by Azoxymethane (AOM). METHODS: The cell line was exposed to the extracts to compare the cytotoxicity of hexane, butanol, ethyl acetate, and water extract of muscle and ethanolic extract of the shell. Male rats (n=40) were assigned into control, positive, negative, and treatment groups. The animals were injected with AOM, except the control group, and then exposed to 250 and 500mg/kg of the crude extracts. Immunohistochemical localization of Bax and Bcl-2, as well as ACF and antioxidant enzymes, were evaluated in the rat colon. RESULTS: The butanolic muscle extract and ethanolic shell one demonstrated an IC50 of 9.02±0.19µg/ml and 20.23±0.27µg/ml towards the cell line, respectively. Dietary exposure inhibited the ACF formation and crypt multiplicity in the colon compared to the cancer control group. The activity of SOD and CAT increased, while that of MDA decreased. The expression of Bax and Bcl-2 increased and decreased, respectively. CONCLUSION: Taken together, the results show that both extractions were suggested to be suppressive to AOMinduced colon cancer.


Asunto(s)
Focos de Criptas Aberrantes/tratamiento farmacológico , Antineoplásicos/farmacología , Antioxidantes/farmacología , Braquiuros/química , Neoplasias Colorrectales/tratamiento farmacológico , Músculos/química , Focos de Criptas Aberrantes/inducido químicamente , Focos de Criptas Aberrantes/patología , Animales , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Antioxidantes/química , Antioxidantes/aislamiento & purificación , Azoximetano/administración & dosificación , Compuestos de Bifenilo/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inyecciones Intradérmicas , Masculino , Estructura Molecular , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Picratos/antagonistas & inhibidores , Ratas , Ratas Wistar , Relación Estructura-Actividad , Células Tumorales Cultivadas
10.
Cell Tissue Res ; 381(2): 255-272, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32405685

RESUMEN

Mesenchymal stem cells (MSCs) are promising cell candidates for cartilage regeneration. Furthermore, it is important to control the cell-matrix interactions that have a direct influence on cell functions. Providing an appropriate microenvironment for cell differentiation in response to exogenous stimuli is a critical step towards the clinical utilization of MSCs. In this study, hydrogels consisted of different proportions of alginates that were modified using gelatin, collagen type I and arginine-glycine-aspartic acid (RGD) and were evaluated regarding their effects on mesenchymal stem cells. The effect of applying hydrostatic pressure on MSCs encapsulated in collagen-modified alginate with and without chondrogenic medium was evaluated 7, 14 and 21 days after culture, which is a comprehensive evaluation of chondrogenesis in 3D hydrogels with mechanical and chemical stimulants. Alcian blue, safranin O and dimethyl methylene blue (DMMB) staining showed the chondrogenic phenotype of cells seeded in the collagen- and RGD-modified alginate hydrogels with the highest intensity after 21 days of culture. The results of real-time PCR for cartilage-specific extracellular matrix genes indicated the chondrogenic differentiation of MSCs in all hydrogels. Also, the synergic effects of chemical and mechanical stimuli are indicated. The highest expression levels of the studied genes were observed in the cells embedded in collagen-modified alginate by loading after 14 days of exposure to the chondrogenic medium. The effect of using IHP on encapsulated MSCs in modified alginate with collagen type I is equal or even higher than using TGF-beta on encapsulated cells. The results of immunohistochemical assessments also confirmed the real-time PCR data.


Asunto(s)
Condrogénesis , Matriz Extracelular/metabolismo , Hidrogeles/química , Mecanotransducción Celular , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos , Alginatos/química , Animales , Cartílago Articular , Células Cultivadas , Condrocitos , Colágeno Tipo I/química , Gelatina/química , Masculino , Péptidos/química , Conejos , Andamios del Tejido
11.
J Biomater Appl ; 35(1): 72-82, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32192388

RESUMEN

Oxygen is a vital molecule for cell and tissue processes. Electrospun fibers have been extensively used as drug loading carriers due to possibility of well control over drug release with modulating fiber properties. However, they have not been used as depots for oxygen release. In the present study, an oxygen-releasing nanofibrous scaffold has been developed by electrospinning of polylactic acid/nano-calcium peroxide suspension with different polylactic acid concentrations (6.5 and 13% w/v). The electrospun fibers with calcium peroxide cargo provided oxygen content of 30-94 mmHg in a period of 14 days which lies well within the oxygen level of osseous tissue. The release profile of 13% polylactic acid fibers was different with that of 6.5% fibers with respects to the initial content of released oxygen and the release rate. Not only did 13% fibers supply oxygen with a slower rate, but also they resulted in a lower burst release of oxygen. Cell culture studies in hypoxia corroborated that 13% polylactic acid fibers better preserve cell viability comparing 6.5% counterparts as perceived by MTT assay. Moreover, they endowed more favored milieu for adherence, arrangement and migration of mesenchymal stem cells as confirmed by microscopy images. The oxygen-releasing fibers equally affected alkaline phosphatase, osteocalcin, and calcium deposition by mesenchymal stem cells most likely due to interplay between topographical and metabolic cues offered by 6.5 and 13% formulations.


Asunto(s)
Nanofibras/química , Oxígeno/administración & dosificación , Peróxidos/química , Poliésteres/química , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Liberación de Fármacos , Células Madre Mesenquimatosas/citología , Osteogénesis , Oxígeno/química , Conejos , Ingeniería de Tejidos
12.
Biomed Mater ; 15(4): 045010, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32120352

RESUMEN

Endothelial cell migration is a crucial step in the process of new blood vessel formation-a necessary process to maintain cell viability inside thick tissue constructs. Here, we report a new method for maintaining cell viability and inducing cell migration using a perfused microfluidic platform based on collagen gel and a gradient hydrogel sheet. Due to the helpful role of the extracellular matrix components in cell viability, we developed a hydrogel sheet from decellularized tissue (DT) of the bovine heart and chitosan (CS). The results showed that hydrogel sheets with an optimum weight ratio of CS/DT = 2 possess a porosity of around 75%, a mechanical strength of 23 kPa, and display cell viability up to 78%. Then, we immobilized a radial gradient of vascular endothelial growth factor (VEGF) on the hydrogel sheet to promote human umbilical vein endothelial cell migration. Finally, we incorporated the whole system as an entirety on the top of the microfluidic platform and studied cell migration through the hydrogel sheet in the presence of soluble and immobilized VEGF. The results demonstrated that immobilized VEGF stimulated cell migration in the hydrogel sheet at all depths compared with soluble VEGF. The results also showed that applying a VEGF gradient in both soluble and immobilized states had a significant effect on cell migration at limited depths (<100 µm). The main finding of this study is a significant improvement in cell migration using an in vivo imitating, cost-efficient and highly reproducible platform, which may open up a new perspective for tissue engineering applications.


Asunto(s)
Movimiento Celular , Hidrogeles/química , Microfluídica , Miocardio/metabolismo , Animales , Bovinos , Supervivencia Celular , Colágeno/metabolismo , ADN/química , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microscopía Electrónica de Rastreo , Porosidad , Reproducibilidad de los Resultados , Espectroscopía Infrarroja por Transformada de Fourier , Estrés Mecánico , Ingeniería de Tejidos/métodos , Andamios del Tejido , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Biomed Mater ; 15(3): 035014, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-31896091

RESUMEN

Tendon tissue engineering based on stem cell differentiation has attracted a great deal of attention in recent years. Previous studies have examined the effect of cell-imprinted polydimethylsiloxane (PDMS) substrate on induction differentiation in stem cells. In this study, we used tenocyte morphology as a positive mold to create a tenocyte-imprinted substrate on PDMS. The morphology and topography of this tenocyte replica on PDMS was evaluated with scanning electron microscopy (SEM) and atomic force microscopy. The tenogenic differentiation induction capacity of the tenocyte replica in adipose tissue-derived mesenchymal stem cells (ADSCs) was then investigated and compared with other groups, including tissue replica (which was produced similarly to the tenocyte replica and was evaluated by SEM), decellularized tendon, and bone morphogenic protein (BMP)-12, as other potential inducers. This comparison gives us an estimate of the ability of tenocyte-imprinted PDMS (called cell replica in the present study) to induce differentiation compared to other inducers. For this reason, ADSCs were divided into five groups, including control, cell replica, tissue replica, decellularized tendon and BMP-12. ADSCs were seeded on each group separately and investigated by the real-time reverse transcription polymerase chain reaction (RT-PCR) technique after seven and 14 days. Our results showed that in spite of the higher effect of the growth factor on tenogenic differentiation, the cell replica can also induce tenocyte marker expression (scleraxis and tenomodulin) in ADSCs. Moreover, the tenogenic differentiation induction capacity of the cell replica was greater than tissue replica. Immunocytochemistry analysis revealed that ADSCs seeding on the cell replica for 14 days led to scleraxis and tenomodulin expression at the protein level. In addition, immunohistochemistry indicated that contrary to the promising results in vitro, there was little difference between ADSCs cultured on tenocyte-imprinted PDMS and untreated ADSCs. The results of such studies could lead to the production of inexpensive cell culture plates or biomaterials that can induce differentiation in stem cells without growth factors or other supplements.


Asunto(s)
Tejido Adiposo/metabolismo , Células Madre Mesenquimatosas/citología , Tenocitos/citología , Ingeniería de Tejidos/métodos , Adulto , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Materiales Biocompatibles , Proteínas Morfogenéticas Óseas/química , Diferenciación Celular , Dimetilpolisiloxanos/química , Factores de Diferenciación de Crecimiento/química , Humanos , Inmunohistoquímica , Masculino , Proteínas de la Membrana/química , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Impresión Molecular , Ratas , Tendones/citología
14.
J Biomed Mater Res A ; 108(1): 136-147, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31515881

RESUMEN

Oxygen is an important signaling molecule which affects many behaviors of bone progenitor cells. Oxygen releasing biomaterials depend on their material and design are able to provide and modulate the desired oxygen for cells. To date, many oxygen releasing vehicles have been developed by incorporating microsized calcium peroxide (CPO) into polymeric matrixes. However, an oxygen releasing system based on nano CPO is still lacking. Not only can nanosized CPO provide more controllable oxygen release, but also can be loaded in vehicles of different shapes and sizes. Current research was conducted to take the advantages of nanomaterials as oxygen releasing components. To this end, CPO nanoparticles were synthesized using a hydrolysis-precipitation procedure and then loaded into the poly (lactide-co-glycolide) (PLGA) matrix via an electrospray process. The surface of PLGA/CaO2 particles was decorated with amine functionalities to render them more bioactive through a controlled aminolysis reaction. The studies on PLGA/CaO2 microparticles revealed that biconcave disk-like morphology with a mean diameter of 5.3 µm was formed. The particles persistently provide oxygen content of 35-67.5 mmHg up to 14 days which lies within the acceptable range for bone tissue engineering applications. PLGA/CaO2 microparticles induced 208 and 76% increase in number of viable mesenchymal cells on 6th and 14th days of cell seeding comparing PLGA counterparts. Furthermore, the expression of two bone biomarkers, that is, alkaline phosphatase and osteocalcin, at protein level as well as the extent of calcium deposition was increased in the presence of PLGA/CaO2 microparticles compared to PLGA ones.


Asunto(s)
Aminas/química , Microesferas , Oxígeno/farmacología , Peróxidos/química , Fosfatasa Alcalina/metabolismo , Animales , Calcio/análisis , Células Cultivadas , Preparaciones de Acción Retardada , Peróxido de Hidrógeno/análisis , Concentración de Iones de Hidrógeno , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Nanopartículas/ultraestructura , Osteocalcina/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Conejos , Espectrometría por Rayos X , Espectroscopía Infrarroja por Transformada de Fourier
15.
Eur J Pharm Biopharm ; 146: 133-142, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31698041

RESUMEN

Doxorubicin is an anti-cancer drug that is important for breast cancer therapy. In this study, the effects of the membrane potential of breast cancer cells (-30 mV) and normal breast epithelial cells (-60 mV) on doxorubicin (DOX) permeability was studied. To achieve this goal, black lipid membranes (BLMs) as a model cell membrane were formed with DPhPC phospholipids in a single aperture of a Teflon sheet by the Montal and Mueller method. The presence of the BLM was characterized by capacitive measurements. The measured specific capacitance of 0.6 µF/cm2 after applying the Montal and Mueller method, confirming the presence of a BLM in the aperture. In addition, the very low current leakage of the BLM (9-24 pA) and ClyA-protein channel insertion in the BLM indicate the compactness, high quality, and thickness of 3-5 nm of the BLM. Afterwards, the permeability of doxorubicin through the BLM was studied at defined cell conditions (37 °C and pH 7.4), as well as cancerous and healthy epithelial-cell membrane potentials (-30 mV and -60 mV, respectively). The results show a slow DOX penetration within the first few hours, which increases rapidly with time. The initial slow penetration can be attributed to an electrostatic interaction between doxorubicin and DPhPC molecules in the model cell membrane. Furthermore, a MTT assay on MCF-10A and MCF-7 under different concentrations of doxorubicin confirmed that the cancerous MCF-7 cell line is more resistant to doxorubicin in comparison with the non-cancerous MCF-10A. Such studies highlight important strategies for designing and tuning the interaction efficacy of novel pharmaceuticals at molecular level.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Permeabilidad de la Membrana Celular/fisiología , Doxorrubicina/farmacocinética , Potenciales de la Membrana , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Humanos , Membrana Dobles de Lípidos/metabolismo , Células MCF-7 , Fosfolípidos/metabolismo
16.
Artif Cells Nanomed Biotechnol ; 47(1): 1022-1035, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30942113

RESUMEN

Differentiation of stem cells to Schwann is considered efficient way for nerve regeneration since the sources of human Schwann cells are limited for clinical application. It is demonstrated that mimicking micromechanical forces or micro/nanotopographical environments that stem cells are experienced in vivo could control their fate. Here, the potency of substrates with imprinted cell-like topographies for direct differentiation of adipose-derived mesenchymal stem cells (ADSCs) into Schwann cells (SCs) is reported. For the preparation of substrates with imprinted SC-Like topographies, SCs are isolated from the sciatic nerve, grown, fixed, and then SC morphologies are transferred to polydimethylsiloxane (PDMS) substrates by mold casting. Subsequently, mesenchymal stem cells (MSCs) are seeded on the SC-imprinted substrates and their differentiation to SCs is evaluated by immunocytochemistry, real-time PCR, and western blotting. Analysis of morphology and expression of SC-specific markers show that MSCs cultured on the imprinted substrates have the typical SC-like morphology and express SC-specific markers including S100b, p75NTR, and Sox10. It is believed that specific cell-like topographies and related micromechanical cues can be sufficient for direct differentiation of ADSCs into Schwann cells by cell-imprinting method as a physical technique.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Ingeniería , Células Madre Mesenquimatosas/citología , Células de Schwann/citología , Animales , Regeneración Nerviosa , Ratas
17.
Int J Pharm ; 559: 1-12, 2019 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-30664992

RESUMEN

Passing the Blood-Brain-Barrier (BBB) is a challenging aspect in nanomedicine. Utilizing surfactant particles is reported to be a potent strategy for easier BBB penetration. On the other hand, loading different functional molecules on a single particle is therapeutically and economically beneficial. In this study, multifunctional amphiphilic Janus nanoparticles have been prepared on the basis of superparamagnetic iron oxide nanoparticles. This Janus platform is armed with folic acid targeting agent and Doxorubicin (DOX) drug that have been conjugated on different sides of the nanoparticles. DOX has been conjugated via imine bond that makes these particles pH sensitive. Chemo-physical characters, in-vitro drug release pattern and toxicity of nanoparticles on rat C6 glioma cell line were studied that confirmed the preparation and pH-dependent behavior of nanoparticles. Microscopy observations showed the Janus morphology of nanoparticles and their cell penetration behavior. Prepared Janus nanoparticle can be utilized as a multifunctional nanomedicine platform for brain cancer treatment.


Asunto(s)
Compuestos Férricos/química , Nanopartículas/química , Animales , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Ácido Fólico/química , Glioma/tratamiento farmacológico , Glioma/metabolismo , Nanopartículas de Magnetita , Nanomedicina/métodos , Nanopartículas/metabolismo , Ratas , Distribución Tisular
18.
J Biomed Mater Res A ; 107(2): 403-413, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30485631

RESUMEN

Providing affinity sites on alginate (ALG) matrix enables specific binding of growth factors to the polymer backbone and allows their release in a controlled fashion. In this study, we used a blend of alginate sulfate (ALG-S) and polyvinyl alcohol (PVA) to fabricate electrospun scaffolds capable of delivering a heparin-like growth factor, transforming growth factor-beta1 (TGF-ß1). The alginate was sulfated with different degrees of sulfation (DS, from 0.8, 3.4 to 12.4%) by a simple process. The success of sulfation was determined by Fourier-transform infrared spectroscopy (FTIR), nuclear magnetic resonance spectroscopy (NMR), elemental analysis, ultraviolet (UV) spectroscopy and staining with dimethylmethylene blue. The physical-mechanical properties of nanofibrous mats were characterized by scanning electron microscopy (SEM), FTIR, energy-dispersive X-ray spectroscopy (EDX), tensile strength and mass loss analysis. Additionally, the release kinetics of transforming growth factor-ß1 (TGF-ß1) from PVA/ALG-S and PVA/ALG scaffolds were compared. The results showed that the binding and entrapment of TGF-ß1 to the nanofibrous scaffolds are improved by the addition of sulfate group to alginate. In conclusion, our results support that nanofibrous scaffold based on PVA/ALG-S can deliver growth factors in tissue engineering application. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 403-413, 2019.


Asunto(s)
Alginatos/química , Portadores de Fármacos/química , Nanofibras/química , Alcohol Polivinílico/química , Factor de Crecimiento Transformador beta1/administración & dosificación , Línea Celular , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Células Madre Mesenquimatosas/citología , Nanofibras/ultraestructura , Sulfatos/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Factor de Crecimiento Transformador beta1/farmacocinética
19.
J Mater Sci Mater Med ; 30(1): 7, 2018 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-30594964

RESUMEN

Strategies based on growth factor (GF) delivery have attracted considerable attention in tissue engineering applications. Among different GFs, transforming growth factor beta 1 (TGF-ß1) is considered to be a potent factor for inducing chondrogenesis. In the present study, an expression cassette encoding the TGF-ß1 protein was prepared and transfected into the SP2/0-Ag14 cell line. The confocal microscopy of the transfected cells was performed to confirm the correct transfection process. The expression and in vitro release kinetics of the recombinant TGF-ß1 were assessed by western blot analysis and ELISA, respectively. Moreover, the biological activity of the expressed protein was compared with that of a commercially available product. The chondrogenic effects of the sustained release of the recombinant TGF-ß1 in an in vitro co-culture system were evaluated using a migration assay and real-time PCR. Results of confocal microscopy confirmed the successful transfection of the vector-encoding TGF-ß1 protein into the SP2/0-Ag14 cells. The bioactivity of the produced protein was in the range of the commercial product. The sustained release of the TGF-ß1 protein via SP2/0-Ag14 cells encapsulated in hydrogels encouraged the migration of adipose-derived MSCs. In addition, the expression analysis of chondrogenesis-related genes revealed that the pretreatment of encapsulated Ad-MSCs cells in alginate sulfate hydrogels through their exposure to the sustained release of TGF-ß1 is an efficient approach before transplantation of cells into the body.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Condrogénesis/fisiología , Células Madre Mesenquimatosas/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Alginatos/química , Animales , Línea Celular , Células Madre Mesenquimatosas/fisiología , Ratones , Factor de Crecimiento Transformador beta1/genética
20.
Biosens Bioelectron ; 118: 51-57, 2018 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-30055420

RESUMEN

Determining the migratory and invasive capacity of cancer cells as well as clarifying the underlying mechanisms are most relevant for developing biosensors in cancer diagnosis, prognosis, drug development and treatment. Intravasation of metastatic cells into blood stream initiated by their invasion to vascular layer would be a significant characteristic of metastasis. Many types of biochemical and bioelectrical sensors were developed for early detection of metastasis. The simplicity of the setup, the ease of the readout, detection of the trace of rare metastatic cells and the feasibility to perform the assay with standard laboratory equipment are some of the challenges limiting the usability of the sensors in tracing the metastasis. Here we describe a biosensor based on recently reported metastatic diagnosis assay; Metas-Chip, with the assistance of nanoroughened Poly-methyl methacrylate (PMMA) layer to diagnose populated metastatic breast cells from primary cancerous ones. Retraction and detachment of Human Umbilical Vein Endothelial Cells (HUVECs) invaded by metastatic cells as a recently found phenomena is the mechanism of the action. A population of HUVECs would be detached from the gold microelectrodes, patterned on nanoroughened surface, which would lead to large changes in impedance. Here, applying biocompatible and patternable nanoroughened surface instead of using adhesive layers which might produce electrical noises resulted in great sensitivity and detectivity of the sensor. Apart from the tight interaction between endothelial cells and nanocontacts of the electrodes, using low concentration (10%) of tumor cells in this invasion assay, might enhance its application in clinical trials.


Asunto(s)
Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Polimetil Metacrilato/química , Línea Celular Tumoral , Impedancia Eléctrica , Electrodos , Oro/química , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA