Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
J Control Release ; 371: 386-405, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38844177

RESUMO

Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.


Assuntos
Proteína Morfogenética Óssea 4 , Diferenciação Celular , Gelatina , Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes Induzidas , Esferoides Celulares , Fator de Células-Tronco , Proteína Morfogenética Óssea 4/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator de Células-Tronco/metabolismo , Gelatina/química , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Animais , Humanos , Camundongos
2.
Bioeng Transl Med ; 9(2): e10629, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38435815

RESUMO

Human induced pluripotent stem cells (iPSCs) hold great promise for personalized medicine, as they can be differentiated into specific cell types, especially mesenchymal stem cells (MSCs). Therefore, our study sought to assess the feasibility of deriving MSCs from teratomas generated from human iPSCs. Teratomas serve as a model to mimic multilineage human development, thus enriching specific somatic progenitors and stem cells. Here, we discovered a small, condensed mass of MSCs within iPSC-generated teratomas. Afterward, we successfully isolated MSCs from this condensed mass, which was a byproduct of teratoma development. To evaluate the characteristics and cell behaviors of iPSC-derived MSCs (iPSC-MSCs), we conducted comprehensive assessments using qPCR, immunophenotype analysis, and cell proliferation-related assays. Remarkably, iPSC-MSCs exhibited an immunophenotype resembling that of conventional MSCs, and they displayed robust proliferative capabilities, similar to those of higher pluripotent stem cell-derived MSCs. Furthermore, iPSC-MSCs demonstrated the ability to differentiate into multiple lineages in vitro. Finally, we evaluated the therapeutic potential of iPSC-MSCs using an osteochondral defect model. Our findings demonstrated that teratomas are a promising source for the isolation of condensed MSCs. More importantly, our results suggest that iPSC-MSCs derived from teratomas possess the capacity for tissue regeneration, highlighting their promise for future therapeutic applications.

3.
Int J Biol Macromol ; 253(Pt 5): 127154, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37793524

RESUMO

The rising demand for green and clean energy urges the enlargement of economical and proficient electrode materials for supercapacitors. Herein, we designed a novel electrode material by porous cellulose graphitic carbon (CC) derived from bio-waste cornhusk via the pyrolysis route, and α-Fe2O3 decorated nanostructure with CC (CCIO) was achieved in situ pyrolysis of corn-husk and Fe(NO3)3·9H2O metal salt followed by a coating of polypyrrole (CCIOP). The CC, CCIO, and CCIOP nanocomposite electrodes were characterized by XRD, Raman, FTIR, FE-SEM/EDX, FE-TEM, XPS, and BET analysis. The CCIOP nanocomposite electrode exhibits an enhanced specific capacitance (Csp) of 290.9 F/g, which is substantial to its pristine CC (128.3 F/g), PPy (140.3 F/g), and CCIO (190.7 F/g). The Csp of CCIOP in a three-electrode system, using 1 M Na2SO4 electrolyte exhibits excellent capacity retention of 79.1 % even at a high current density of 10 A/g. The as-fabricated asymmetric supercapacitor (ASC) delivered a remarkable capacity retention of 88.7 % with a coulombic efficiency of 98.8 % even after 3000 cycles. The study shows successful utilization of cellulose from bio-waste cornhusk into a substantial template applicable in future alternative energy storage devices.


Assuntos
Grafite , Nanocompostos , Polímeros , Celulose , Carbono , Pirróis , Eletrodos
4.
Tissue Eng Regen Med ; 20(3): 461-471, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37041434

RESUMO

BACKGROUND: Matrix metalloproteinases (MMPs) are proteins involved in the repair and remodeling the extracellular matrix (ECM). MMP13 is essential for bone development and healing through the remodeling of type I collagen (COL1), the main component of the ECM in bone tissue. Mesenchymal stem cells (MSCs)-based cell therapy has been considered a promising approach for bone regeneration because of their osteogenic properties. However, the approaches using MSC to completely regenerate bone tissue have been limited. To overcome the limitation, genetic engineering of MSC could be a strategy for promoting regeneration efficacy. METHODS: We performed in vitro and in vivo experiments using MMP13-overexpressing MSCs in the presence of COL1. To examine MMP13-overexpressing MSCs in vivo, we prepared a fibrin/COL1-based hydrogel to encapsulate MSCs and subcutaneously implanted gel-encapsulated MSCs in nude mice. We found that the osteogenic marker genes, ALP and RUNX2, were upregulated in MMP13-overexpressing MSCs through p38 phosphorylation. In addition, MMP13 overexpression in MSCs stimulated the expression of integrin α3, which is up-stream receptor of p38, and substantially increased osteogenic differentiation potential of MSCs. Bone tissue formation in MMP13-overexpressing MSCs was significantly higher than that in control MSCs. Taken together, our findings demonstrate that MMP13 is not only an essential factor for bone development and bone healing but also has a pivotal role in promoting osteogenic differentiation of MSCs to induce bone formation. CONCLUSION: MSCs Genetically engineered to overexpress MMP13, which have a powerful potential to differentiate into the osteogenic cells, might be beneficial in bone disease therapy.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Camundongos , Animais , Hidrogéis , Camundongos Nus , Metaloproteinase 13 da Matriz/genética , Osso e Ossos , Células-Tronco Mesenquimais/metabolismo , Colágeno/metabolismo
5.
J Control Release ; 354: 45-56, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36586671

RESUMO

Extracellular vesicles (EVs) are nano-sized carriers that reflect the parent cell's information and are known to mediate cell-cell communication. In order to overcome the disadvantages of mesenchymal stem cells (MSCs) in cell therapy, such as unexpected differentiation leading to tumorization, immune rejection, and other side effects, EVs derived from MSCs (MSC-EVs) with the tissue regenerative function have been studied as new cell-free therapeutics. However, therapeutic applications of EVs require overcoming several challenges. First, the production efficiency of MSC-EVs should be increased at least as much as the quantity of them are required to their clinical application; second, MSC-EVs needs to show various functionality further, thereby increasing tissue regeneration efficiency. In this study, we treated tauroursodeoxycholic acid (TUDCA), a biological derivative known to regulate cholesterol, to MSCs and investigated whether TUDCA treatment would be able to increase EV production efficiency and tissue regenerative capacity of EVs. Indeed, it appears that TUDCA priming to MSC increases the yield of MSC-EVs >2 times by reducing the cellular cholesterol level in MSCs and increasing the exocytosis-related CAV1 expression. Interestingly, it was found that the EVs derived from TUDCA-primed MSCs (T-EV) contained higher amounts of anti-inflammatory cytokines (IL1RN, IL6, IL10, and IL11) and osteogenic proteins (ALP, RUNX2, BMP2, BMPR1, and BMPR2) than those in control MSC-EVs (C-EV). Besides, it was shown that T-EV not only regulated M1/M2 macrophages differentiation of monocytes, also effectively increased the osteogenic differentiation of MSCs as well as bone tissue regeneration in a bone defect rat model. Based on these results, it is concluded that TUDCA treatment to MSC as a new approach endows EV with high-yield production and functionality. Thus, we strongly believe T-EV would be a powerful therapeutic material for bone tissue regeneration and potentially could be expanded to other types of tissue regeneration for clinical applications.


Assuntos
Vesículas Extracelulares , Osteogênese , Ratos , Animais , Citocinas/metabolismo , Regeneração Óssea , Vesículas Extracelulares/metabolismo
6.
Nat Biomed Eng ; 7(1): 72-84, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36564626

RESUMO

The effectivity of cancer immunotherapies is hindered by immunosuppressive tumour microenvironments that are poorly infiltrated by effector T cells and natural killer cells. In infection and autoimmune disease, the recruitment and activation of effector immune cells is coordinated by pro-inflammatory T helper 17 (TH17) cells. Here we show that pathogen-mimicking hollow nanoparticles displaying mannan (a polysaccharide that activates TH17 cells in microbial cell walls) limit the fraction of regulatory T cells and induce TH17-cell-mediated anti-tumour responses. The nanoparticles activate the pattern-recognition receptor Dectin-2 and Toll-like receptor 4 in dendritic cells, and promote the differentiation of CD4+ T cells into the TH17 phenotype. In mice, intra-tumoural administration of the nanoparticles decreased the fraction of regulatory T cells in the tumour while markedly increasing the fractions of TH17 cells (and the levels of TH17-cell-associated cytokines), CD8+ T cells, natural killer cells and M1-like macrophages. The anti-tumoural activity of the effector cells was amplified by an agonistic antibody against the co-stimulatory receptor OX40 in multiple mouse models. Nanomaterials that induce TH17-cell-mediated immune responses may have therapeutic potential.


Assuntos
Linfócitos T CD8-Positivos , Nanopartículas , Animais , Camundongos , Diferenciação Celular , Citocinas , Linfócitos T Reguladores , Células Th17/imunologia
7.
Tissue Eng Regen Med ; 20(1): 143-154, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36482140

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) are used for tissue regeneration due to their wide differentiation capacity and anti-inflammatory effects. Extracellular vesicles (EVs) derived from MSCs are also known for their regenerative effects as they contain nucleic acids, proteins, lipids, and cytokines similar to those of parental cells. There are several studies on the use of MSCs or EVs for tissue regeneration. However, the combinatorial effect of human MSCs (hMSCs) and EVs is not clear. In this study, we investigated the combinatorial effect of hMSCs and EVs on cartilage regeneration via co-encapsulation in a hyaluronic-acid (HA)-based hydrogel. METHODS: A methacrylic-acid-based HA hydrogel was prepared to encapsulate hMSCs and EVs in hydrogels. Through in vitro and in vivo analyses, we investigated the chondrogenic potential of the HA hydrogel-encapsulated with hMSCs and EVs. RESULTS: Co-encapsulation of hMSCs with EVs in the HA hydrogel increased the chondrogenic differentiation of hMSCs and regeneration of damaged cartilage tissue compared with that of the HA hydrogel loaded with hMSCs only. CONCLUSION: Co-encapsulation of hMSCs and EVs in the HA hydrogel effectively enhances cartilage tissue regeneration due to the combinatorial therapeutic effect of hMSCs and EVs. Thus, in addition to cartilage tissue regeneration for the treatment of osteoarthritis, this approach would be a useful strategy to improve other types of tissue regeneration.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Humanos , Hidrogéis/farmacologia , Cartilagem/metabolismo , Ácido Hialurônico/farmacologia , Células-Tronco Mesenquimais/metabolismo , Vesículas Extracelulares/metabolismo
8.
Stem Cells Transl Med ; 11(10): 1072-1088, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36180050

RESUMO

Spinal fusion surgery is a surgical technique that connects one or more vertebrae at the same time to prevent movement between the vertebrae. Although synthetic bone substitutes or osteogenesis-inducing recombinant proteins were introduced to promote bone union, the rate of revision surgery is still high due to pseudarthrosis. To promote successful fusion after surgery, stem cells with or without biomaterials were introduced; however, conventional 2D-culture environments have resulted in a considerable loss of the innate therapeutic properties of stem cells. Therefore, we conducted a preclinical study applying 3D-spheroids of human bone marrow-dewrived mesenchymal stem cells (MSCs) to a mouse spinal fusion model. First, we built a large-scale manufacturing platform for MSC spheroids, which is applicable to good manufacturing practice (GMP). Comprehensive biomolecular examinations, which include liquid chromatography-mass spectrometry and bioinformatics could suggest a framework of quality control (QC) standards for the MSC spheroid product regarding the identity, purity, viability, and potency. In our animal study, the mass-produced and quality-controlled MSC spheroids, either undifferentiated or osteogenically differentiated were well-integrated into decorticated bone of the lumbar spine, and efficiently improved angiogenesis, bone regeneration, and mechanical stability with statistical significance compared to 2D-cultured MSCs. This study proposes a GMP-applicable bioprocessing platform and QC directions of MSC spheroids aiming for their clinical application in spinal fusion surgery as a new bone graft substitute.


Assuntos
Substitutos Ósseos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Fusão Vertebral , Animais , Camundongos , Humanos , Fusão Vertebral/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Medula Óssea , Osteogênese , Materiais Biocompatíveis , Proteínas Recombinantes
9.
Sci Rep ; 12(1): 2456, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35165342

RESUMO

Determining the exact positional relationship between mandibular third molar (M3) and inferior alveolar nerve (IAN) is important for surgical extractions. Panoramic radiography is the most common dental imaging test. The purposes of this study were to develop an artificial intelligence (AI) model to determine two positional relationships (true contact and bucco-lingual position) between M3 and IAN when they were overlapped in panoramic radiographs and compare its performance with that of oral and maxillofacial surgery (OMFS) specialists. A total of 571 panoramic images of M3 from 394 patients was used for this study. Among the images, 202 were classified as true contact, 246 as intimate, 61 as IAN buccal position, and 62 as IAN lingual position. A deep convolutional neural network model with ResNet-50 architecture was trained for each task. We randomly split the dataset into 75% for training and validation and 25% for testing. Model performance was superior in bucco-lingual position determination (accuracy 0.76, precision 0.83, recall 0.67, and F1 score 0.73) to true contact position determination (accuracy 0.63, precision 0.62, recall 0.63, and F1 score 0.61). AI exhibited much higher accuracy in both position determinations compared to OMFS specialists. In determining true contact position, OMFS specialists demonstrated an accuracy of 52.68% to 69.64%, while the AI showed an accuracy of 72.32%. In determining bucco-lingual position, OMFS specialists showed an accuracy of 32.26% to 48.39%, and the AI showed an accuracy of 80.65%. Moreover, Cohen's kappa exhibited a substantial level of agreement for the AI (0.61) and poor agreements for OMFS specialists in bucco-lingual position determination. Determining the position relationship between M3 and IAN is possible using AI, especially in bucco-lingual positioning. The model could be used to support clinicians in the decision-making process for M3 treatment.


Assuntos
Tomada de Decisão Clínica/métodos , Aprendizado Profundo , Mandíbula/diagnóstico por imagem , Traumatismos do Nervo Mandibular/prevenção & controle , Nervo Mandibular/diagnóstico por imagem , Dente Serotino/diagnóstico por imagem , Radiografia Panorâmica/métodos , Adulto , Idoso , Tomografia Computadorizada de Feixe Cônico/métodos , Confiabilidade dos Dados , Feminino , Humanos , Masculino , Traumatismos do Nervo Mandibular/etiologia , Pessoa de Meia-Idade , Extração Dentária/efeitos adversos , Adulto Jovem
10.
Acta Biomater ; 125: 219-230, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33677160

RESUMO

Extracellular matrix (ECM) remodeling is necessary for the development and self-healing of tissue, and the process is tissue specific. Matrix metalloproteinases (MMPs) play a role in ECM remodeling by unwinding and cleaving ECM. We hypothesized that ECM remodeling by MMPs is involved in the differentiation of stem cells into specific lineages during self-healing. To prove the hypothesis, we investigated which MMPs are involved in the osteogenic differentiation of human mesenchymal stem cells (hMSCs) grown on a type I collagen (Col I) matrix, and we found that specifically high expression of MMP13 in hMSCs grown on a Col I matirx during osteogenic differentiation. Moreover, knocking down of MMP13 decreased the osteogenic differentiation of hMSCs grown on a Col I matrix. In addition, pre-treatment of recombinant human MMP13 lead to remodeling of Col I matrix and increased the osteogenic differentiation of hMSCs and in vivo bone formation following the upregulation of the expression of runt-related transcription factor 2 (RUNX2), integrin α3 (ITGA3), and focal adhesion kinase. Furthermore, the transcription factor RUNX2 bound to the MMP13 promoter. These results suggest that growth on a remodeled Col I matrix by MMP13 stimulates osteogenic differentiation of hMSCs and self-healing of bone tissue via an MMP13/ITGA3/RUNX2 positive feedback loop. STATEMENT OF SIGNIFICANCE: Self-healing of tissue could be the key to treating diseases that cannot be overcome by present technology. We investigated the mechanism underlying the self-healing of tissue and we found that the osteogenic differentiation was increased in hMSCs grown on a remodeled Col I matrix by the optimized concentration of MMP13 not in hMSCs grown on a Col I fragments cleaved by a high concentration of MMP13. In addition, we found the remodeled Col I matrix by MMP13 increased the osteogenic capacity through a MMP13/integrin α3/RUNX2 positive feedback loop. This result would be able to not only provide a strategy for bone tissue-specific functional materials following strong evidence about the self-healing mechanism of bone through the interaction between stem cells and the ECM matrix. As such, we strongly believe our finding will be of interest to researchers studying biomaterials, stem cell biology and matrix interaction for regenerative medicine and therapy.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Regeneração Óssea , Osso e Ossos , Diferenciação Celular , Células Cultivadas , Colágeno , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Retroalimentação , Humanos , Integrina alfa3 , Ligantes , Metaloproteinase 13 da Matriz/genética
11.
Int J Biol Macromol ; 178: 464-476, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33662416

RESUMO

Polymer-clay nanocomposite hydrogel films (PCNCHFs) were prepared from caboxymethyl cellulose, polyvinylpyrrolidone, agar and nanosepiolite clay (0, 0.3, 0.5, 0.7, 0.9 and 1.5% reinforcement) by treating thermally in a simple, rapid, and inexpensive route. The PCNCHFs and its 5-fluorouracil (FU)-loaded composites (PCNCHFs@FU) were tested for FU release and characterized by FTIR, XRD, FE-SEM, EDX, DSC, and TGA analyses to investigate their structural, morphological, and thermal properties. The nanosepiolite-loaded polymer composites (PCNCHF1 to PCNCHF5) exhibited higher tensile strength than the pristine polymer hydrogel (PCNCHF0); consequently, the thermal properties (glass- and melting-transition) were improved. The PCNCHFs@FU demonstrated prolonged FU release at pH 7.4 for 32 h. The biocompatibility of PCNCHFs was tested against human skin fibroblast (CCDK) cells. The viability of cells exposed to all PCNCHFs was >95% after 72 h of culture. The live/dead assay show the proliferation of fibroblast cells, confirming the biocompatibility of the hydrogels. The pH-sensitive PCNCHFs@FU release could be suitable for drug release in cancer therapy, and the developed PCNCHFs may also be useful for tissue engineering, food packaging, and other biological applications.


Assuntos
Carboximetilcelulose Sódica/química , Portadores de Fármacos/química , Fluoruracila , Hidrogéis/química , Silicatos de Magnésio/química , Nanocompostos/química , Fluoruracila/química , Fluoruracila/farmacocinética
12.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255398

RESUMO

Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for articular cartilage repair and regeneration. However, the terminal hypertrophic differentiation of Ad-MSC-derived cartilage is a critical barrier during hyaline cartilage regeneration. In this study, we investigated the role of matrilin-3 in preventing Ad-MSC-derived chondrocyte hypertrophy in vitro and in an osteoarthritis (OA) destabilization of the medial meniscus (DMM) model. Methacrylated hyaluron (MAHA) (1%) was used to encapsulate and make scaffolds containing Ad-MSCs and matrilin-3. Subsequently, the encapsulated cells in the scaffolds were differentiated in chondrogenic medium (TGF-ß, 1-14 days) and thyroid hormone hypertrophic medium (T3, 15-28 days). The presence of matrilin-3 with Ad-MSCs in the MAHA scaffold significantly increased the chondrogenic marker and decreased the hypertrophy marker mRNA and protein expression. Furthermore, matrilin-3 significantly modified the expression of TGF-ß2, BMP-2, and BMP-4. Next, we prepared the OA model and transplanted Ad-MSCs primed with matrilin-3, either as a single-cell suspension or in spheroid form. Safranin-O staining and the OA score suggested that the regenerated cartilage morphology in the matrilin-3-primed Ad-MSC spheroids was similar to the positive control. Furthermore, matrilin-3-primed Ad-MSC spheroids prevented subchondral bone sclerosis in the mouse model. Here, we show that matrilin-3 plays a major role in modulating Ad-MSCs' therapeutic effect on cartilage regeneration and hypertrophy suppression.


Assuntos
Cartilagem Hialina/crescimento & desenvolvimento , Hipertrofia/genética , Células-Tronco Mesenquimais/citologia , Osteoartrite/genética , Animais , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrogênese/genética , Humanos , Ácido Hialurônico/farmacologia , Hipertrofia/patologia , Hipertrofia/prevenção & controle , Hipertrofia/terapia , Proteínas Matrilinas/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Osteoartrite/terapia , Regeneração/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Alicerces Teciduais , Fator de Crescimento Transformador beta/genética
13.
Int J Nanomedicine ; 15: 8437-8449, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162754

RESUMO

BACKGROUND: Lipid polymer hybrid nanoparticles (LPHNPs) have been widely investigated in drug and gene delivery as well as in medical imaging. A knowledge of lipid-based surface engineering and its effects on how the physicochemical properties of LPHNPs affect the cell-nanoparticle interactions, and consequently how it influences the cytological response, is in high demand. METHODS: Herein, we have engineered antibiotic-loaded (doxycycline or vancomycin) LPHNPs with cationic and zwitterionic lipids and examined the effects on their physicochemical characteristics (size and charge), antibiotic entrapment efficiency, and the in vitro intracellular bacterial killing efficiency against Mycobacterium smegmatis or Staphylococcus aureus infected macrophages. RESULTS: The incorporation of cationic or zwitterionic lipids in the LPHNP formulation resulted in a size reduction in LPHNPs formulations and shifted the surface charge of bare NPs towards positive or neutral values. Also observed were influences on the drug incorporation efficiency and modulation of the drug release from the biodegradable polymeric core. The therapeutic efficacy of LPHNPs loaded with vancomycin was improved as its minimum inhibitory concentration (MIC) (2 µg/mL) versus free vancomycin (4 µg/mL). Importantly, our results show a direct relationship between the cationic surface nature of LPHNPs and its intracellular bacterial killing efficiency as the cationic doxycycline or vancomycin loaded LPHNPs reduced 4 or 3 log CFU respectively versus the untreated controls. CONCLUSION: In our study, modulation of surface charge in the nanomaterial formulation increased macrophage uptake and intracellular bacterial killing efficiency of LPHNPs loaded with antibiotics, suggesting alternate way for optimizing their use in biomedical applications.


Assuntos
Antibacterianos/farmacologia , Sistemas de Liberação de Medicamentos , Espaço Intracelular/microbiologia , Macrófagos/microbiologia , Nanopartículas/química , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Lipídeos/química , Macrófagos/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Mycobacterium smegmatis/efeitos dos fármacos , Tamanho da Partícula , Polímeros/química , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia
14.
J Control Release ; 328: 596-607, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32946872

RESUMO

A high level of reactive oxygen species (ROS) such as hydrogen peroxide (H2O2) upregulates pro-inflammatory cytokines and inhibits the osteogenic differentiation of mesenchymal stem cells (MSCs), which are key factors in bone regeneration. Ursodeoxycholic acid (UDCA), a hydrophilic bile acid, has antioxidant and anti-inflammatory activities and also plays beneficial roles in bone regeneration by stimulating the osteogenic differentiation of MSCs while suppressing their adipogenic differentiation. Despite its remarkable capacity for bone regeneration, multiple injections of UDCA induce adverse side effects such as mechanical stress and contamination in bone defects. To fully exploit the beneficial roles of UDCA, a concept polymeric prodrug was developed based on the hypothesis that removal of overproduced H2O2 will potentiate the osteogenic functions of UDCA. In this work, we report bone regenerative nanoparticles (NPs) formulated from a polymeric prodrug of UDCA (PUDCA) with UDCA incorporated in its backbone through H2O2-responsive peroxalate linkages. The PUDCA NPs displayed potent antioxidant and anti-inflammatory activities in MSCs and induced osteogenic rather than adipogenic differentiation of the MSCs. In rat models of bone defect, the PUDCA NPs exhibited significantly better bone regeneration capacity and anti-inflammatory effects than equivalent amounts of UDCA. We anticipate that PUDCA NPs have tremendous translational potential as bone regenerative agents.


Assuntos
Células-Tronco Mesenquimais , Nanopartículas , Pró-Fármacos , Animais , Ácidos e Sais Biliares , Regeneração Óssea , Diferenciação Celular , Peróxido de Hidrogênio , Osteogênese , Ratos
15.
Stem Cell Res Ther ; 11(1): 363, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32831130

RESUMO

BACKGROUND: Chronic low back pain is a prevalent disability, often caused by intervertebral disc (IVD) degeneration. Mesenchymal stem cell (MSC) therapy could be a safe and feasible option for repairing the degenerated disc. However, for successful translation to the clinic, various challenges need to be overcome including unwanted adverse effects due to acidic pH, hypoxia, and limited nutrition. Matrilin-3 is an essential extracellular matrix (ECM) component during cartilage development and ossification and exerts chondrocyte protective effects. METHODS: This study evaluated the effects of matrilin-3-primed adipose-derived MSCs (Ad-MSCs) on the repair of the degenerated disc in vitro and in vivo. We determined the optimal priming concentration and duration and developed an optimal protocol for Ad-MSC spheroid generation. RESULTS: Priming with 10 ng/ml matrilin-3 for 5 days resulted in the highest mRNA expression of type 2 collagen and aggrecan in vitro. Furthermore, Ad-MSC spheroids with a density of 250 cells/microwell showed the increased secretion of favorable growth factors such as transforming growth factor beta (TGF-ß1), TGF-ß2, interleukin-10 (IL-10), granulocyte colony-stimulating factor (G-CSF), and matrix metalloproteinase 1 (MMP1) and decreased secretion of hypertrophic ECM components. In addition, matrilin-3-primed Ad-MSC spheroid implantation was associated with optimal repair in a rabbit model. CONCLUSION: Our results suggest that priming MSCs with matrilin-3 and spheroid formation could be an effective strategy to overcome the challenges associated with the use of MSCs for the treatment of IVD degeneration.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Degeneração do Disco Intervertebral/terapia , Proteínas Matrilinas/genética , Coelhos
16.
Int J Mol Sci ; 21(12)2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560070

RESUMO

Tauroursodeoxycholic acid (TUDCA) is a US FDA-approved hydrophilic bile acid for the treatment of chronic cholestatic liver disease. In the present study, we investigate the effects of TUDCA on the proliferation and differentiation of osteoblasts and its therapeutic effect on a mice model of osteoporosis. Following treatment with different concentrations of TUDCA, cell viability, differentiation, and mineralization were measured. Three-month-old female C57BL/6 mice were randomly divided into three groups (n = 8 mice per group): (i) normal mice as the control group, (ii) ovariectomy (OVX) group (receiving phosphate-buffered saline (PBS) treatment every other day for 4 weeks), and (iii) OVX group with TUDCA (receiving TUDCA treatment every other day for 4 weeks starting 6 weeks after OVX). At 11 weeks post-surgery, serum levels of procollagen type I N-terminal propeptides (PINP) and type I collagen crosslinked C-telopeptides (CTX) were measured, and all mice were sacrificed to examine the distal femur by micro-computed tomography (CT) scans and histology. TUDCA (100 nM, 1 µM) significantly increased the proliferation and viability of osteoblasts and osteoblast differentiation and mineralization when used in vitro. Furthermore, TUDCA neutralized the detrimental effects of methylprednisolone (methylprednisolone-induced osteoblast apoptosis). In the TUDCA treatment group the PINP level was higher and the CTX level was lower, but these levels were not significantly different compared to the PBS treatment group. Micro-CT and histology showed that the TUDCA treatment group preserved more trabecular structures in the distal femur compared to the PBS treatment group. In addition, the TUDCA treatment group increased the percentage bone volume with respect to the total bone volume, bone mineral density, and mice distal femur trabeculae compared with the PBS treatment group. Taken together, our findings suggest that TUDCA may provide a favorable effect on bones and could be used for the prevention and treatment of osteoporosis.


Assuntos
Osteoporose/tratamento farmacológico , Ovariectomia/efeitos adversos , Fragmentos de Peptídeos/metabolismo , Pró-Colágeno/metabolismo , Ácido Tauroquenodesoxicólico/administração & dosagem , Animais , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metilprednisolona/efeitos adversos , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoporose/etiologia , Osteoporose/metabolismo , Distribuição Aleatória , Ácido Tauroquenodesoxicólico/farmacologia , Resultado do Tratamento
17.
Int J Biol Macromol ; 159: 474-486, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32437816

RESUMO

Poly(acrylamide-co-acrylamidoglycolic acid)/guar gum@ Ag-nanocomposite (AgNC@PAAG) hydrogels has been fabricated by a green protocol utilizing rhubarb stem-extract as bioreductant. The prepared nanocomposites (NCs) are formulated by varying guar gum (GG) polymer and cross-linker content, and used remarkably to study the release of an anticancer drug, 5-fluorouracil (FU). The AgNC@PAAG has demonstrated its potential in bacterial inactivation and p-nitrophenol (PNP) reduction. The AgNC@PAAG hydrogels showed extended FU release time, which was up to 23 h in pH 7.4. The higher zone of inhibition was documented for AgNC@PAAG against B. subtilis and E. coli. It was noticed that, the inhibition activity of AgNC@PAAG, was directly proportional to cross-linker content than the GG polymer. The efficiency of AgNC@PAAG as a nanocatalyst was evaluated for a model reduction reaction of p-nitrophenol (PNP) reduction by aqueous sodium borohydride (NaBH4), with an apparent rate constant of 121.8 × 10-3 min-1 at ambient temperature. The proposed nanocatalysts are reliable and recyclable, demonstrated its catalytic recycle efficacy of 85% after the third successive run. These NCs robust its biological and catalytic activity after embedding silver nanoparticles (AgNPs) by the bioreduction process; these optimized nanocatalysts can be remarkably used in biomedical healthcare sectors and industrial catalysis.


Assuntos
Tecnologia Biomédica , Ciência Ambiental , Galactanos/química , Mananas/química , Nanopartículas Metálicas/química , Nanogéis/química , Gomas Vegetais/química , Prata/química , Antibacterianos/administração & dosagem , Antibacterianos/farmacologia , Biodegradação Ambiental , Técnicas de Química Sintética , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Ciência Ambiental/métodos , Hidrogéis/síntese química , Hidrogéis/química , Cinética , Nanopartículas Metálicas/ultraestrutura , Testes de Sensibilidade Microbiana , Estrutura Molecular , Porosidade , Espectroscopia de Infravermelho com Transformada de Fourier , Termodinâmica , Difração de Raios X
18.
Cells ; 8(12)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31842346

RESUMO

Owing to the importance of stem cell culture systems in clinical applications, researchers have extensively studied them to optimize the culture conditions and increase efficiency of cell culture. A spheroid culture system provides a similar physicochemical environment in vivo by facilitating cell-cell and cell-matrix interaction to overcome the limitations of traditional monolayer cell culture. In suspension culture, aggregates of adjacent cells form a spheroid shape having wide utility in tumor and cancer research, therapeutic transplantation, drug screening, and clinical study, as well as organic culture. There are various spheroid culture methods such as hanging drop, gel embedding, magnetic levitation, and spinner culture. Lately, efforts are being made to apply the spheroid culture system to the study of drug delivery platforms and co-cultures, and to regulate differentiation and pluripotency. To study spheroid cell culture, various kinds of biomaterials are used as building forms of hydrogel, film, particle, and bead, depending upon the requirement. However, spheroid cell culture system has limitations such as hypoxia and necrosis in the spheroid core. In addition, studies should focus on methods to dissociate cells from spheroid into single cells.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Esferoides Celulares/citologia , Animais , Materiais Biocompatíveis/química , Diferenciação Celular , Hipóxia Celular , Células Cultivadas , Humanos
19.
Tissue Eng Part A ; 25(23-24): 1646-1657, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30982407

RESUMO

The directed differentiation of human adipose-derived stem cells (hASCs) into different cell types has shown great therapeutic potential in treating various diseases. To maximize the therapeutic potentials, researchers have tried manipulating master transcriptional genes that promote efficient differentiation of mesenchymal stem cells (MSCs) such as the MAPK/ERK signaling pathway. Sprouty (SPRY) is a family of proteins that are known to inhibit the MAPK/ERK signaling pathway. Although the role of some SPRY isoforms in MSC differentiation is known, the function of SPRY4 isoform has not been fully elucidated. In the present study, the role of SPRY4 in the multilineage differentiation of hASCs has been elucidated. To investigate the role of SPRY4 in hASC differentiation and tissue regeneration, we performed a transient knockdown of SPRY expression via a small interfering RNA (siSPRY4). Western blot and quantitative polymerase chain reaction results revealed that the treatment of siSPRY4 before induction of differentiation had no significant effect on adipogenic, but reduced chondrogenic, differentiation of hASCs. Interestingly, SPRY4 transient knockdown had a significant effect on the osteogenic differentiation as indicated by the increased messenger RNA (mRNA) and protein expression of osteogenic markers such as alkaline phosphatase (ALP; 2.3-fold) and osteopontin (OPN; 3.5-fold) and increased calcium deposition measured via Alizarin red staining (3.3-fold). Moreover, in vivo tissue regeneration of siSPRY4-treated hASCs in ectopic bone formation and calvarial defect mouse models showed higher bone volume (5.24-fold) and trabecular number (4.59-fold) assessed via histological and microcomputed tomography analyses. We also determined that the enhanced osteogenic differentiation in SPRY4-treated hASCs was due to the induction of ERK1/2 phosphorylation. Taken together, our results suggest that the regulation of SPRY4 through MAPK signaling is a potentially critical aspect on the osteogenic differentiation of hASCs and for bone tissue regeneration, and thus, may be utilized as a potent technique in the development of effective bone therapeutics. Impact Statement This study tried to expand our current understanding of the osteogenic differentiation of mesenchymal stem cells. The transient downregulation of the SPRY4 expression via small interfering RNA (siRNA) showed significant enhancement of the osteogenic differentiation of adipose-derived stem cells via the induction of ERK 1/2 phosphorylation. This suggests the possible mechanism to maximize the potential of stem cell as therapeutics and has a great potential in treating various bone-related diseases.


Assuntos
Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Osteogênese , Adipogenia , Animais , Proliferação de Células , Condrogênese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/enzimologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação
20.
Stem Cell Res Ther ; 10(1): 32, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30654843

RESUMO

BACKGROUND: Ell3 is a RNA polymerase II elongation factor that has various cell type-dependent functions, such as regulating the differentiation efficiency of embryonic stem cells and sensitizing cancer cells to anticancer drugs. However, there has been little research on the role of Ell3 on the regulation of senescence and apoptosis of stem cells. METHODS: We analyzed the senescence of Ell3-suppressed stem cells by mitochondrial activity, ß-gal (+) cells, and lineage differentiation efficiency. The apoptosis of Ell3-overexpressing stem cells was analyzed by Annexin V staining, Immunoblot, and Live&dead assay. In addition, chromatin immunoprecipitation and luciferase assays were used to demonstrate p53 functions as a direct transcriptional activator of Ell3. RESULTS: Suppression of Ell3 expression induced senescence in stem cells by increasing Bcl-2 expression. Unlike the effect of Ell3 suppression, the ectopic expression of Ell3 induces apoptosis of stem cells and induces apoptosis of adjacent cells. In addition, p53 functions as a direct transcriptional activator of Ell3 during the stem cell apoptosis. CONCLUSIONS: We suggest that the function of Ell3 is associated with the p53-Bcl2 axis in both senescent and apoptotic ADSCs.


Assuntos
Células-Tronco Adultas/citologia , Fatores de Elongação da Transcrição/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Células-Tronco Adultas/metabolismo , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Senescência Celular/fisiologia , Humanos , Células MCF-7 , Osteócitos/citologia , Osteócitos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ativação Transcricional , Fatores de Elongação da Transcrição/antagonistas & inibidores , Fatores de Elongação da Transcrição/biossíntese , Fatores de Elongação da Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA