Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Bioeng Biotechnol ; 12: 1363241, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38567084

RESUMEN

Background: The cartilage tissue lacks blood vessels, which is composed of chondrocytes and ECM. Due to this vessel-less structure, it is difficult to repair cartilage tissue damages. One of the new methods to repair cartilage damage is to use tissue engineering. In the present study, it was attempted to simulate a three-dimensional environment similar to the natural ECM of cartilage tissue by using hydrogels made of natural materials, including Chitosan and different ratios of Alginate. Material and methods: Chitosan, alginate and Chitosan/Alginate hydrogels were fabricated. Fourier Transform Infrared, XRD, swelling ratio, porosity measurement and degradation tests were applied to scaffolds characterization. After that, human adipose derived-mesenchymal stem cells (hADMSCs) were cultured on the hydrogels and then their viability and chondrogenic differentiation capacity were studied. Safranin O and Alcian blue staining, immunofluorescence staining and real time RT-PCR were used as analytical methods for chondrogenic differentiation potential evaluation of hADMSCs when cultured on the hydrogels. Results: The highest degradation rate was detected in Chitosan/Alginate (1:0.5) group The scaffold biocompatibility results revealed that the viability of the cells cultured on the hydrogels groups was not significantly different with the cells cultured in the control group. Safranin O staining, Alcian blue staining, immunofluorescence staining and real time PCR results revealed that the chondrogenic differentiation potential of the hADMSCs when grown on the Chitosan/Alginate hydrogel (1:0.5) was significantly higher than those cell grown on the other groups. Conclusion: Taken together, these results suggest that Chitosan/Alginate hydrogel (1:0.5) could be a promising candidate for cartilage tissue engineering applications.

2.
In Vitro Cell Dev Biol Anim ; 60(1): 106-114, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38123755

RESUMEN

Using stem cells is one of the most important determining factors in repairing lesions using regenerative medicine. Obtaining adult stem cells from patients is a perfect choice, but it is worth noting that their differentiation and proliferation potential decreases as the patient ages. For this reason, the use of amniotic fluid stem cells can be one of the excellent alternatives. This research aimed to investigate the osteogenic differentiation potential of the amniotic fluid stem cells while cultured on the polycaprolactone/poly L-lactic acid nanofibrous scaffold. Scaffolds were qualitatively evaluated by a scanning electron microscope, and their hydrophilicity and mechanical properties were studied using contact angle and tensile test, respectively. The biocompatibility and non-toxicity of the nanofibers were also evaluated using viability assay. The osteo-supportive capacity of the nanofibers was examined using alizarin red staining, alkaline phosphatase activity, and calcium release measurement. Finally, the expression level of four important bone-related genes was determined quantitatively. The results demonstrated that the mineralization rate, alkaline phosphatase activity, intracellular calcium, and bone-related genes increased significantly in the cells cultured on the polycaprolactone/poly L-lactic acid scaffold compared to the cells cultured on the tissue culture plate as a control. According to the results, it can be concluded that the polycaprolactone/poly L-lactic acid nanofibrous scaffold surprisingly improved the osteogenic differentiation potential of the amniotic fluid stem cells and, in combination with polycaprolactone/poly L-lactic acid nanofibers could be a promising candidate as bone implants.


Asunto(s)
Nanofibras , Animales , Andamios del Tejido , Osteogénesis , Ingeniería de Tejidos/métodos , Calcio , Fosfatasa Alcalina , Líquido Amniótico , Células Cultivadas , Poliésteres/farmacología , Diferenciación Celular , Células Madre , Ácido Láctico/farmacología
3.
In Vitro Cell Dev Biol Anim ; 59(6): 401-409, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37405626

RESUMEN

Regulation of mesenchymal stem cell (MSC) fate for targeted cell therapy applications has been a subject of interest, particularly for tissues such as tendons that possess a marginal regenerative capacity. Control of MSCs' fate into the tendon-specific lineage has mainly been achieved by implementation of chemical growth factors. Mechanical stimuli or 3-dimensional (D) scaffolds have been used as an additional tool for the differentiation of MSCs into tenocytes, but oftentimes, they require a sophisticated bioreactor or a complex scaffold fabrication technique which reduces the feasibility of the proposed method to be used in practice. Here, we used nanovibration to induce the differentiation of MSCs toward the tenogenic fate solely by the use of nanovibration and without the need for growth factors or complex scaffolds. MSCs were cultured on 2D cell culture dishes that were connected to piezo ceramic arrays to apply nanovibration (30-80 nm and 1 kHz frequency) over 7 and 14 d. We observed that nanovibration resulted in significant overexpression of tendon-related markers in both gene expression and protein expression levels, while there was no significant differentiation into adipose and cartilage lineages. These findings could be of assistance in the mechanoregulation of MSCs for stem cell engineering and regenerative medicine applications.


Asunto(s)
Células Madre Mesenquimatosas , Ingeniería de Tejidos , Animales , Ingeniería de Tejidos/métodos , Vibración , Diferenciación Celular , Cordón Umbilical
4.
Sci Rep ; 13(1): 12145, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37500679

RESUMEN

Self-healing and autologous bone graft of calvaraial defects can be challenging. Therefore, the fabrication of scaffolds for its rapid and effective repair is a promising field of research. This paper provided a comparative study on the ability of Three-dimensional (3D) printed polycaprolactone (PCL) scaffolds and PCL-modified with the hydroxyapatite (HA) and bioglasses (BG) bioceramics scaffolds in newly bone formed in calvaria defect area. The studied 3D-printed PCL scaffolds were fabricated by fused deposition layer-by-layer modeling. After the evaluation of cell adhesion on the surface of the scaffolds, they were implanted into a rat calvarial defect model. The rats were divided into four groups with scaffold graft including PCL, PCL/HA, PCL/BG, and PCL/HA/BG and a non-explant control group. The capacity of the 3D-printed scaffolds in calvarial bone regeneration was investigated using micro computed tomography scan, histological and immunohistochemistry analyses. Lastly, the expression levels of several bone related genes as well as the expression of miR-20a and miR-17-5p as positive regulators and miR-125a as a negative regulator in osteogenesis pathways were also investigated. The results of this comparative study have showed that PCL scaffolds with HA and BG bioceramics have a great range of potential applications in the field of calvaria defect treatment.


Asunto(s)
MicroARNs , Andamios del Tejido , Ratas , Animales , Microtomografía por Rayos X , Osteogénesis , Regeneración Ósea , Durapatita/farmacología , Cráneo/diagnóstico por imagen , Impresión Tridimensional , MicroARNs/farmacología , Poliésteres/farmacología , Ingeniería de Tejidos/métodos
5.
Adv Pharm Bull ; 12(4): 658-672, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36415646

RESUMEN

Unique mechanical properties, miscibility potency, and biodegradability are the three prominent features of polycaprolactone (PCL), making it an attractive biomaterial which commonly applied in regenerative medicine and biomedical engineering. Different strategies developed for fabricating nanofibrous construct, electrospinning is a practical, simple, and efficient technique based on electro-hydrodynamic systems that use an electrified viscous fluid jet drawn by the air toward a collector at a changing electric potential. PCL electrospun-based nanofibrous composites as proper scaffolds are employed in stem cell-related research, particularly in tissue engineering, wound dressing, and systems designed for sending drugs. A compilation of mechanochemical properties and most common biological performance on PCL-based electrospun fibrous structures in biomedical application are included in this study. Therefore, electrospun PCL nanofiber applying has been presented, and after that, current progress and prospects have been discussed. Literature reviews revealed that electrospun PCL nanofibrous composites had gained significant attention in regenerative medicine, and these structures have shown notable development in mechanobiological properties. This evidence is a crucial success for biomedical strategies, especially in regenerative medicine.

6.
Sci Rep ; 12(1): 12359, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35859093

RESUMEN

Bone tissue engineering uses various methods and materials to find suitable scaffolds that regenerate lost bone due to disease or injury. Poly(ε-caprolactone) (PCL) can be used in 3D printing for producing biodegradable scaffolds by fused deposition modeling (FDM). However, the hydrophobic surfaces of PCL and its non-osteogenic nature reduces adhesion and cell bioactivity at the time of implantation. This work aims to enhance bone formation, osteogenic differentiation, and in vitro biocompatibility via PCL scaffolds modification with Hydroxyapatite (HA) and Collagen type I (COL). This study evaluated the osteosupportive capacity, biological behavior, and physicochemical properties of 3D-printed PCL, PCL/HA, PCL/COL, and PCL/HA/COL scaffolds. Biocompatibility and cells proliferation were investigated by seeding human adipose tissue-derived mesenchymal stem cells (hADSCs) onto the scaffolds, which were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, and 6-diamidino-2-phenylindole (DAPI) staining. In addition, the bone differentiation potential of the hADSCs was assessed using calcium deposition, alkaline phosphatase (ALP) activity, and bone-related protein and genes. Although all constructed scaffolds support hADSCs proliferation and differentiation, the results showed that scaffold coating with HA and COL can boost these capacities in a synergistic manner. According to the findings, the tricomponent 3D-printed scaffold can be considered as a promising choice for bone tissue regeneration and rebuilding.


Asunto(s)
Durapatita , Osteogénesis , Colágeno/química , Durapatita/química , Durapatita/farmacología , Humanos , Poliésteres/química , Impresión Tridimensional , Células Madre , Andamios del Tejido/química
7.
Stem Cell Rev Rep ; 18(6): 1892-1911, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35080745

RESUMEN

Wound healing is a serious obstacle due to the complexity of evaluation and management. While novel approaches to promoting chronic wound healing are of critical interest at the moment, several studies have demonstrated that combination therapy is critical for the treatment of a variety of diseases, particularly chronic wounds. Among the various approaches that have been proposed for wound care, regenerative medicine-based methods have garnered the most attention. As is well known, regenerative medicine's three primary tools are gene/cell therapy, biomaterials, and tissue engineering. Multifunctional biomaterials composed of synthetic and natural components are highly advantageous for exosome carriers, which utilizing them is an exciting wound healing method. Recently, stem cell-secreted exosomes and certain biomaterials have been identified as critical components of the wound healing process, and their combination therapy appears to produce significant results. This paper presents a review of literature and perspectives on the use of stem cell-derived exosomes and biomaterials in wound healing, particularly chronic wounds, and discusses the possibility of future clinical applications.


Asunto(s)
Exosomas , Materiales Biocompatibles/farmacología , Células Madre , Ingeniería de Tejidos , Cicatrización de Heridas
8.
Curr Stem Cell Res Ther ; 17(8): 772-792, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34615453

RESUMEN

INTRODUCTION: It has been shown that mechanical forces can induce or promote osteogenic differentiation as well as remodeling of the new created bone tissues. To apply this characteristic in bone tissue engineering, it is important to know which mechanical stimuli through which signaling pathway has a more significant impact on osteogenesis. METHODS: In this systematic study, an electronic search was conducted using PubMed and Google Scholar databases. This study has been prepared and organized according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Included studies were first categorized according to the in vivo and in vitro studies. RESULTS: Six types of mechanical stresses were used in these articles and the most commonly used mechanical force and cell source were tension and bone marrow-derived mesenchymal stem cells (BMMSCs), respectively. These forces were able to trigger twelve signaling pathways in which Wnt pathway was so prominent. CONCLUSION: 1) Although specific signaling pathways are induced through specific mechanical forces, Wnt signaling pathways are predominantly activated by almost all types of force/stimulation, 2) All signaling pathways regulate expression of RUNX2, which is known as a master regulator of osteogenesis, 3) In Tension force, the mode of force administration, i.e, continuous or noncontinuous tension is more important than the percentage of elongation.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Vía de Señalización Wnt/fisiología
9.
Mol Biol Rep ; 49(2): 1389-1412, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34716502

RESUMEN

Currently, nanoscale materials and scaffolds carrying antitumor agents to the tumor target site are practical approaches for cancer treatment. Immunotherapy is a modern approach to cancer treatment in which the body's immune system adjusts to deal with cancer cells. Immuno-engineering is a new branch of regenerative medicine-based therapies that uses engineering principles by using biological tools to stimulate the immune system. Therefore, this branch's final aim is to regulate distribution, release, and simultaneous placement of several immune factors at the tumor site, so then upgrade the current treatment methods and subsequently improve the immune system's handling. In this paper, recent research and prospects of nanotechnology-based cancer immunotherapy have been presented and discussed. Furthermore, different encouraging nanotechnology-based plans for targeting various innate and adaptive immune systems will also be discussed. Due to novel views in nanotechnology strategies, this field can address some biological obstacles, although studies are ongoing.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Humanos , Sistema Inmunológico , Factores Inmunológicos/uso terapéutico , Nanopartículas/administración & dosificación , Nanotecnología/métodos , Neoplasias/inmunología
10.
Cell Tissue Res ; 384(2): 403-421, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33433691

RESUMEN

Production of a 3D bone construct with high-yield differentiated cells using an appropriate cell source provides a reliable strategy for different purposes such as therapeutic screening of the drugs. Although adult stem cells can be a good source, their application is limited due to invasive procedure of their isolation and low yield of differentiation. Patient-specific human-induced pluripotent stem cells (hiPSCs) can be an alternative due to their long-term self-renewal capacity and pluripotency after several passages, resolving the requirement of a large number of progenitor cells. In this study, a new biphasic 3D-printed collagen-coated HA/ß-TCP scaffold was fabricated to provide a 3D environment for the cells. The fabricated scaffolds were characterized by the 3D laser scanning digital microscopy, X-ray diffraction, Fourier transform infrared spectroscopy, and mechanical test. Then, the osteogenesis potential of the hiPSC-seeded scaffolds was investigated compared to the buccal fat pad stem cell (BFPSC)-seeded scaffolds through in vitro and in vivo studies. In vitro results demonstrated up-regulated expressions of osteogenesis-related genes of RUNX2, ALP, BMP2, and COL1 compared to the BFPSC-seeded scaffolds. In vivo results on calvarial defects in the rats confirmed a higher bone formation in the hiPSC-seeded scaffolds compared to the BFPSC-seeded groups. The immunofluorescence assay also showed higher expression levels of collagen I and osteocalcin proteins in the hiPSC-seeded scaffolds. It can be concluded that using the hiPSC-seeded scaffolds can lead to a high yield of osteogenesis, and the hiPSCs can be used as a superior stem cell source compared to BFPSCs for bone-like construct bioengineering.


Asunto(s)
Tejido Adiposo/diagnóstico por imagen , Células Madre Pluripotentes Inducidas/metabolismo , Osteogénesis/fisiología , Impresión Tridimensional/normas , Andamios del Tejido/normas , Tejido Adiposo/fisiopatología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Masculino , Ratas , Ratas Wistar
11.
ACS Omega ; 6(51): 35284-35296, 2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-34984260

RESUMEN

With advances in bone tissue engineering, various materials and methods have been explored to find a better scaffold that can help in improving bone growth and regeneration. Three-dimensional (3D) printing by fused deposition modeling can produce customized scaffolds from biodegradable polyesters such as polycaprolactone (PCL). Although the fabricated PCL scaffolds exhibited a lack of bioactivity and poor cell attachment on their surfaces, herein, using a simple postfabrication modification method with hydroxyapatite (HA) and bioglasses (BGs), we obtained better cell proliferation and attachment. Biological behavior and osteosupportive capacity of the 3D-printed scaffolds including PCL, PCL/HA, PCL/BG, and PCL/HA/BG were evaluated in this study, while human adipose tissue-derived mesenchymal stem cells (hADSCs) were cultured on the scaffolds. The cell morphology, attachment, and proliferation were investigated using scanning electron microscopy (SEM), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, and 4',6-diamidino-2-phenylindole (DAPI) staining. In the next step, the ability of stem cells to differentiate into osteoblasts was evaluated by measuring alkaline phosphatase (ALP) activity, calcium deposition, and bone-related gene and protein expression. In the end, the expression levels of miR-20a, miR-125a, and their target genes were also investigated as positive and negative regulators in osteogenesis pathways. The results showed that the coated scaffolds with bioceramics present a more appropriate surface for cell adhesion and proliferation, as well as efficient potential in inducing osteoconduction and osteointegration compared to PCL alone and control. The PCL/HA/BG scaffold exhibited higher in vitro cell viability and bone formation compared to the other groups, which can be due to the synergistic effect of HA and BG. On the whole, this tricomponent 3D-printing scaffold has a promising prospect for bone tissue engineering applications.

12.
Adv Exp Med Biol ; 1312: 19-37, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33159303

RESUMEN

The first isolation of human embryonic stem cells (hESC) reported in the late 90s opened a new window to promising possibilities in the fields of human developmental biology and regenerative medicine. Subsequently, the differentiation of hESC lines into different precursor cells showed their potential in treating different incurable diseases. However, this promising field has consistently had remarkable ethical and experimental limitations. This paper is a review of clinical trial studies dealing with hESC and their advantages, limitations, and other specific concerns. Some of the hESC limitations have been solved, and several clinical trial studies are ongoing so that recent clinical trials have strived to improve the clinical applications of hESC, especially in macular degeneration and neurodegenerative diseases. However, regarding hESC-based therapy, several important issues need more research and discussion. Despite considerable studies to Date, hESC-based therapy is not available for conventional clinical applications, and more studies and data are needed to overcome current clinical and ethical limitations. When all the limitations of Embryonic stem cells (ESC) are wholly resolved, perhaps hESC can become superior to the existing stem cell sources. This overview will be beneficial for understanding the standard and promising applications of cell and tissue-based therapeutic approaches and for developing novel therapeutic applications of hESC.


Asunto(s)
Células Madre Embrionarias , Células Madre Embrionarias Humanas , Diferenciación Celular , Línea Celular , Ensayos Clínicos como Asunto , Humanos , Medicina Regenerativa
13.
Cell Tissue Bank ; 21(3): 495-505, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32388594

RESUMEN

The use of bioactive scaffolds in tissue engineering has a significant effect on the damaged tissue healing by an increase in speed and quality of the process. Herein, electrospinning was applied to fabricate composite nanofibrous scaffolds by Poly lactic-co-glycolic acid (PLGA) and Polyurethane (PU) with and without poly-phosphate (poly-P). Scaffolds were characterized morphologically by scanning electron microscope (SEM), and their biocompatibility was also investigated by SEM, protein adsorption, cell attachment and survival assays. The applicability of the scaffolds for bladder tissue engineering was also evaluated by culturing mesenchymal stem cells (MSCs) on the scaffolds and their differentiation into smooth muscle cell (SMC) was studied at the gene and protein levels. The results demonstrated that scaffold biocompatibility was increased significantly by loading poly-P. SMC related gene and protein expression level in MSCs cultured on poly-P-loaded scaffold was also increased significantly compared to those cells cultured on empty scaffold. It can be concluded that poly-P hasn't also increased scaffold biocompatibility, but also SMC differentiation potential of MSCs was also increased while cultured on the poly-P containing scaffold compared to the empty scaffold. Taken together, our study showed that PLGA-PU-poly-P alone and in combination with MSCs has a promising potential for support urinary bladder smooth muscle tissue engineering.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Miocitos del Músculo Liso/citología , Nanofibras/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Polifosfatos/farmacología , Poliuretanos/química , Andamios del Tejido/química , Adsorción , Separación Celular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Nanofibras/ultraestructura
14.
Stem Cell Rev Rep ; 16(3): 427-433, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32281052

RESUMEN

"COVID-19" is the word that certainly isn't forgotten by everybody who lives in the first half of the twenty-first century. COVID-19, as a pandemic, has led many researchers from different biomedical fields to find solutions or treatments to manage the pandemic. However, no standard treatment for this disease has been discovered to date. Probably, preventing the severe acute respiratory infection form of COVID-19 as the most dangerous phase of this disease can be helpful for the treatment and reduction of the death rate. In this regard, mesenchymal stem cells (MSCs)-based immunomodulation treatment has been proposed as a suitable therapeutic approach and several clinical trials have begun. Recently, MSCs according to their immunomodulatory and regenerative properties attract attention in clinical trials. After the intravenous transplantation of MSCs, a significant population of cells accumulates in the lung, which they alongside immunomodulatory effect could protect alveolar epithelial cells, reclaim the pulmonary microenvironment, prevent pulmonary fibrosis, and cure lung dysfunction. Given the uncertainties in this area, we reviewed reported clinical trials and hypotheses to provide useful information to researchers and those interested in stem cell therapy. In this study, we considered this new approach to improve patient's immunological responses to COVID-19 using MSCs and discussed the aspects of this proposed treatment. However, currently, there are no approved MSC-based approaches for the prevention and/or treatment of COVID-19 patients but clinical trials ongoing.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/terapia , Inmunomodulación , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Pandemias , Neumonía Viral/terapia , Linfocitos B/inmunología , Linfocitos B/virología , Betacoronavirus/inmunología , COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/inmunología , Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Pulmón/inmunología , Pulmón/virología , Trasplante de Células Madre Mesenquimatosas/tendencias , Células Madre Mesenquimatosas/citología , Neumonía Viral/epidemiología , Neumonía Viral/inmunología , Neumonía Viral/virología , Respiración Artificial , SARS-CoV-2 , Linfocitos T/inmunología , Linfocitos T/virología
15.
In Vitro Cell Dev Biol Anim ; 56(4): 313-321, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32307668

RESUMEN

Bladder dysfunction is one of the most common diseases that occur for a number of reasons and the current treatment modalities do not improve much in its recovery process. Tissue engineering in the last two decades has given great hope for the treatment of these disorders. In this study, a composite nanofibrous scaffold was fabricated from chitosan, collagen, and polyvinyl-alcohol polymer blend while curcumin incorporated in scaffold fibers. The scaffold supportive functions from smooth muscle cell differentiation were studied when human-induced pluripotent stem cells were cultured on the scaffolds under differentiation medium. Biocompatibility of the fabricated scaffold increased significantly by incorporating curcumin in the scaffold fibers, where protein adsorption, cell attachment, and viability were increased in the nanofiber/curcumin group compared with the other groups. In addition, the expression level of smooth muscle cell-related genes, including alpha-smooth muscle actin (αSMA), smooth muscle 22 alpha (SM-22a), Caldesmon1, and Calponin1in the stem cells upregulated while cultured in the presence of curcumin, but this increase was significantly improved while cells cultured on the nanofibers/curcumin. In addition, αSMA protein in the cells cultured on the nanofibers/curcumin expressed significantly higher than those cells cultured on the nanofibers without curcumin. It can be concluded that smooth muscle cell differentiation of the induced pluripotent stem cells promoted by curcumin and this promotion was synergistically improved while curcumin incorporated in the nanofibers. Graphical abstract.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Quitosano/química , Colágeno/química , Curcumina/farmacología , Células Madre Pluripotentes Inducidas/citología , Miocitos del Músculo Liso/citología , Nanofibras/química , Alcohol Polivinílico/química , Animales , Diferenciación Celular/genética , Línea Celular , Células Nutrientes/citología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Nanofibras/ultraestructura , Ratas
16.
Gene ; 740: 144534, 2020 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-32145328

RESUMEN

The function of tissue cells is strongly depends on the extracellular matrix (ECM) that can guide and support cell structure. This support plays a crucial role in the process of cell proliferation and differentiation. Herein, three different nanofibrous scaffolds that are highly attractive for tissue engineering were selected and then osteogenic related genes and protein expression patterns of human adipose-derived mesenchymal stem cells (AT-MSCs) were investigated when grown on substrates. Polycaprolactone, Poly (L-lactic acid) and Polyvinylidene-fluoride nanofibrous scaffolds were fabricated using Electrospinning method and then AT-MSCs viability and osteogenic differentiation were evaluated while cultured on them. The highest AT-MSCs survival rate when grown on the scaffolds was detected when grown on Polyvinylidene-fluoride. In addition, the highest ALP activity and mineralization were also observed in differentiated AT-MSCs has grown on Polyvinylidene-fluoride. The expression levels of Runx2, osteonectin and osteocalcin genes and osteocalcin protein in the AT-MSCs has grown on the Polyvinylidene-fluoride were also significantly higher than the rest of the scaffolds. Based on the results, it seems that since the studied substrate have a similar structural characteristics, their nature may have an important role in the stem cell's osteogenesis process, where the Polyvinylidene-fluoride piezoelectricity was a most distinguished characteristic.


Asunto(s)
Células Madre Mesenquimatosas , Nanofibras , Osteogénesis , Ingeniería de Tejidos/métodos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Nanofibras/química , Osteocalcina/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Osteonectina/metabolismo , Poliésteres , Polivinilos
17.
J Cell Physiol ; 235(5): 4239-4246, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31613005

RESUMEN

Diabetes is one of the most common diseases in the world that is chronic, progressive, and costly, and causes many complications. Common drug therapies are not able to cure it, and pancreas transplantation is not responsive to the high number of patients. The production of the insulin producing cells (IPCs) from the stem cells in the laboratory and their transplantation to the patient's body is one of the most promising new approaches. In this study, the differentiation potential of the induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) into IPCs was compared to each other while cultured on poly(lactic-co-glycolic) acid (PLGA)/polyethylene glycol (PEG) nanofibrous scaffold as a 3D substrate and tissue culture polystyrene (TCPS) as a 2D substrate. Although the expression level of the insulin, Glut2 and pdx-1 genes in stem cells cultured on 3D substrate was significantly higher than the stem cells cultured on 2D substrate, the highest expression level of these genes was detected in the iPSCs cultured on PLGA-PEG. Insulin and C-peptide secretions from differentiated cells were also investigated and the results showed that secretions in cultured iPSCs on the PLGA-PEG were significantly higher than cultured iPSCs on the TCPS and cultured MSCs on both PLGA-PEG and TCPS. In addition, insulin protein was also expressed in the cultured iPSCs on the PLGA-PEG significantly higher than cultured MSCs on the PLGA-PEG. It can be concluded that differentiation potential of iPSCs into IPCs is significantly higher than human MSCs at both 2D and 3D culture systems.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Insulina/metabolismo , Células Madre Mesenquimatosas/fisiología , Péptido C/metabolismo , Técnicas de Cultivo de Célula/métodos , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
18.
J Biomed Mater Res A ; 108(2): 377-386, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31654461

RESUMEN

Smart scaffolds have a great role in the damaged tissue reconstruction. The aim of this study was developing a scaffold that in addition to its fiber's topography has also content of micro-RNAs (miRNAs), which play a regulatory role during osteogenesis. In this study, we inserted two important miRNAs, including miR-22 and miR-126 in the electrospun polycaprolactone (PCL) nanofibers and after scaffold characterization, osteoinductivity of the fabricated nanofibers was investigated by evaluating of the osteogenic differentiation potential of induced pluripotent stem cells (iPSCs) when grown on miRNAs-incorporated PCL nanofibers (PCL-miR) and empty PCL. MiRNAs incorporation had no effect on the fibers size and morphology, cell attachment, and protein adsorption, although viability and proliferation rate of the human iPSCs were increased after a week in PCL-miR compared to the empty PCL. The results obtained from alkaline phosphatase activity, calcium content, bone-related genes, and proteins expression assays demonstrated that the highest osteogenic markers were observed in iPSCs grown on the PCL-miR compared to the cells cultured on PCL and culture plate. According to the results, miR-incorporated PCL nanofibers could be considered as a promising potential tissue-engineered construct for the treatment of patients with bone lesions and defects.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , MicroARNs/administración & dosificación , Nanofibras/química , Osteogénesis , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Materiales Biocompatibles/química , Diferenciación Celular , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/genética , Poliésteres/química
19.
J Cell Biochem ; 121(5-6): 3185-3196, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31886565

RESUMEN

Tissue engineering is fast becoming a key approach in bone medicine studies. Designing the ideally desirable combination of stem cells and scaffolds are at the hurt of efforts for producing implantable bone substitutes. Clinical application of stem cells could be associated with serious limitations, and engineering scaffolds that are able to imitate the important features of extracellular matrix is a major area of challenges within the field. In this study, electrospun scaffolds of polyvinylidene fluoride (PVDF), PVDF-graphene oxide (GO), PVDF-polyvinyl alcohol (PVA) and PVDF-PVA-GO were fabricated to study the osteogenic differentiation potential of human induced pluripotent stem cells (iPSCs) while cultured on fabricated scaffolds. Scanning electron microscopy study, viability assay, relative gene expression analysis, immunocytochemistry, alkaline phosphates activity, and calcium content assays confirmed that the osteogenesis rate of hiPSCs cultured on PVDF-PVA-Go is significantly higher than other scaffolds. Here, we showed that the biocompatible, nontoxic, flexible, piezoelectric, highly porous and interconnected three-dimensional structure of electrospun PVDF-PVA-Go scaffold in combination with hiPSCs (as the stem cells with significant advantageous in comparison to other types) makes them a highly promising scaffold-stem cell system for bone remodeling medicine. There was no evidence for the superiority of PVDF-GO or PVDF-PVA scaffold for osteogenesis, compared to each other; however both of them showed better potentials as to PVDF scaffold.


Asunto(s)
Grafito/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Alcohol Polivinílico/farmacología , Polivinilos/farmacología , Adsorción , Materiales Biocompatibles/química , Remodelación Ósea , Sustitutos de Huesos , Calcio/metabolismo , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Electricidad , Perfilación de la Expresión Génica , Humanos , Microscopía Electrónica de Rastreo , Estrés Mecánico , Ingeniería de Tejidos/métodos , Andamios del Tejido
20.
J Cancer Res Ther ; 15(5): 1114-1119, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31603120

RESUMEN

AIM OF STUDY: One of the new methods that have promising results is the use of cell-derived microvesicles (MVs) to kill tumor cells. Given that MVs contain apoptotic materials, genes, and proteins, they can interfere with the fate of adjacent cells. MATERIALS AND METHODS: In the present study, after adipose tissue-derived mesenchymal stem cells (AT-MSCs) isolation and characterization, MVs were derived from AT-MSCs and then characterized morphologically by standard error of the mean and size determination by DLS, and after that, the influence of MVs on human breast cancer cells (MCF-7) was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay and apoptosis-related gene expression. The raw data were analyzed in SPSS.17 software. RESULTS: The results indicated that MVs have a size range of 500-1500 nm, and the viability of MCF-7 was significantly decreased when treated by different concentrations of MVs and it was confirmed when apoptosis-related genes' expression level was measured by real-time reverse transcription polymerase chain reaction whereas demonstrated that apoptosis genes including Bax, P53, P21, and EP300 (2- ΔΔ CT) and ΔCT values were expressed significantly in MCF-7 treated by MVs higher than those nontreated, and decrease of Bcl-2 expression level in MVs-treated MCF-7 was also significant as an antiapoptosis-related gene. CONCLUSIONS: Taking together, AT-MSC-derived MVs demonstrated anticancer or antitumoral properties on MCF-7 cells, and it could also be effective for other types of cancer cells.


Asunto(s)
Neoplasias de la Mama/terapia , Células Madre Mesenquimatosas/citología , Microvasos/citología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Micropartículas Derivadas de Células/genética , Femenino , Expresión Génica/genética , Humanos , Células MCF-7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...