Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
J Control Release ; 371: 258-272, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38815704

ABSTRACT

Currently, exosomes showed appropriate potential in the repair of skin injury. However, the functions of the exosomes could be compromised rapidly due to their short half-life and high clearance rate in vivo. In addition, the controlled release of effective concentrations of exosomes could increase the utilization efficiency of exosomes in wound healing. Accordingly, the design of an effective system for the controlled delivery of exosomes during the wound treatment period was necessary. In this contribution, we designed a novel exosome-based multifunctional nanocomposite platform with photothermal-controlled release performance for the repair of skin injury. Based on the agarose hydrogel, two-dimensional Ti3C2 (Ti3C2 MXene) and human umbilical cord mesenchymal stem cell (hucMSC)-derived exosomes, the as-prepared platform (i.e., hucMSC-derived exosome/Ti3C2 MXene hydrogel) was synthesized for the first time. Apart from possessing injectability, the hucMSC-derived exosome/Ti3C2 MXene hydrogel utilized the excellent photothermal effect of Ti3C2 MXene and proper phase transition performance of agarose hydrogel to provide a photothermal-controlled release system for the hucMSC-derived exosomes, which was beneficial for the personalized on-demand drug delivery. Importantly, the hucMSC-derived exosomes maintained their inherent structure and activity after being released from the Ti3C2 MXene hydrogel. Additionally, the as-prepared hydrogel with multifunctional performance also presented remarkable biocompatibility and photothermal-antibacterial property, and could efficiently accelerate wound healing by promoting cell proliferation, angiogenesis, collagen deposition, and reducing the level of inflammation at the wound site. The results suggested that the exosome-based multifunctional nanocomposite platform with great potential for wound healing would make significant advances in the revolution of traditional treatment methods in skin injury.

2.
Food Funct ; 15(7): 3446-3462, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38450419

ABSTRACT

Endothelial dysfunction (ED) is an initiating trigger and key factor in vascular complications, leading to disability and mortality in individuals with diabetes. The research concerning therapeutic interventions for ED has gained considerable interest. Fenugreek, a commonly used edible plant in dietary consumption, has attracted significant attention due to its management of diabetes and its associated complications. The research presented in this study examines the potential therapeutic benefits of fenugreek in treating ED and investigates the underlying mechanism associated with its effects. The analysis on fenugreek was performed using 70% ethanol extract, and its chemical composition was analyzed using ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). In total, we identified 49 compounds present in the fenugreek extract. These compounds encompass flavonoids, saponins, and phospholipids. Then, the models of ED in streptozotocin-induced diabetic mice and high glucose-induced isolated rat aortas were established for research. Through vascular function testing, it was observed that fenugreek extract effectively improved ED induced by diabetes or high glucose. By analyzing the protein expression of arginase 1 (Arg1), Arg activity, Arg1 immunohistochemistry, nitric oxide (NO) level, and the protein expression of endothelial nitric oxide synthase (eNOS), p38 mitogen-activated protein kinase (p38 MAPK), and p-p38 MAPK in aortas, this study revealed that the potential mechanism of fenugreek extract in anti-ED involves the downregulation of Arg1, leading to enhanced NO production. Furthermore, analysis of serum exosomes carrying Arg activity indicates that fenugreek may decrease the activity of Arg transported by serum exosomes, potentially preventing the increase in Arg levels triggered by the uptake of serum exosomes by vascular endothelial cells. In general, this investigation offers valuable observations regarding the curative impact of fenugreek extract on anti-ED in diabetes, revealing the involvement of the Arg1 pathway in its mechanism.


Subject(s)
Diabetes Mellitus, Experimental , Endothelial Cells , Plant Extracts , Trigonella , Rats , Mice , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Arginase , p38 Mitogen-Activated Protein Kinases/metabolism , Glucose/metabolism , Nitric Oxide Synthase Type III/metabolism
3.
Aging (Albany NY) ; 15(18): 9544-9560, 2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37724890

ABSTRACT

Inflammation of chondrocytes plays a critical role in the occurrence and development of osteoarthritis (OA). Recent evidence indicated exosomes derived from mesenchymal stem cells (MSCs-Exos) exhibit excellent anti-inflammatory ability in many troublesome inflammatory diseases including OA. In the present study, we aimed to explore the role of human umbilical cord-derived MSCs-Exos (hUC-MSCs-Exos) in treating the inflammation of chondrocytes and its related mechanisms. Ultracentrifugation was applied to isolate hUC-MSCs-Exos from the culture supernatant of hUC-MSCs. Two OA-like in vitro inflammation models of human articular chondrocytes induced with interleukin 1ß (IL-1ß) and co-incubation with macrophage utilizing transwell cell culture inserts were both used to evaluate the anti-inflammatory effects of hUC-MSCs-Exos. The mRNA sequencing of chondrocytes after treatment and microRNA (miRNA) sequencing of hUC-MSCs-Exos were detected and analyzed for possible mechanism analysis. The results of the study confirmed that hUC-MSCs-Exos had a reversed effect of IL-1ß on chondrocytes in the expression of collagen type II alpha 1 (COL2A1) and matrix metalloproteinase 13 (MMP13). The addition of hUC-MSCs-Exos to M1 macrophages in the upper chamber showed down-regulation of IL-1ß and tumor necrosis factor α (TNF-α), up-regulation of IL-10 and arginase1 (ARG1), and reversed the gene and protein expression of COL2A1 and MMP13 of the chondrocytes seeded in the lower chamber. The results of this study confirmed the anti-inflammatory effects of hUC-MSCs-Exos in the human articular chondrocytes inflammation model. hUC-MSCs-Exos may be used as a potential cell-free treatment strategy for chondrocyte inflammation in OA.

4.
Ann Transl Med ; 10(18): 976, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36267713

ABSTRACT

Background: Osteoarthritis (OA) is one of the most common joint diseases and a major global public health concern. Mesenchymal stem cells (MSCs) have been widely used for the treatment of OA owing to their paracrine secretion of trophic factors, a phenomenon in which exosomes may play a major role. Here, we investigate the potential of exosomes from human umbilical cord-derived MSCs (hUC-MSCs-Exos) in alleviating OA. Methods: The hUC-MSCs-Exos were harvested from hUC-MSC-conditioned medium using ultracentrifugation. Rats with surgically-induced OA were intra-articularly injected with hUC-MSCs-Exos. The effect of hUC-MSCs-Exos in repairing osteoarticular cartilage was assessed using hematoxylin and eosin (HE) staining, safranin-O and fast green staining and immunohistochemistry. The in vitro experiments were further carried out to verify the therapeutic effect. The effects of hUC-MSCs-Exos on the proliferation and migration of human chondrocytes were evaluated using the cell counting kit-8, EdU-555 cell proliferation kit, and transwell assays. Annexin V-FITC/PI staining were used to evaluate the effect of exosomes on chondrocyte apoptosis. An in vitro model of human articular chondrocytes treated with interleukin 1 beta (IL-1ß) was used to evaluate the effects of exosomes, analyses involved using quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence, and western blotting. The role of hUC-MSCs-Exos in macrophage polarization was examined in the monocyte cell line, Tohoku Hospital Pediatrics-1 (THP-1) by qRT-PCR and immunofluorescence. Results: The results showed that hUC-MSCs-Exos prevented severe damage to the knee articular cartilage in the rat OA model. We confirmed the high efficacy of hUC-MSCs-Exos in promoting chondrocyte proliferation and migration and inhibiting chondrocyte apoptosis. Additionally, hUC-MSCs-Exos could reverse IL-1ß-induced injury of chondrocytes and regulate the polarization of macrophages in vitro. Conclusions: There is potential for hUC-MSCs-Exos to be used as a treatment strategy for OA.

5.
Acta Biomater ; 152: 255-272, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36041647

ABSTRACT

Fibroblasts can be directly reprogrammed via a combination of small molecules to generate induced neurons (iNs), bypassing intermediate stages. This method holds great promise for regenerative medicine; however, it remains inefficient. Recently, studies have suggested that physical cues may improve the direct reprogramming of fibroblasts into neurons, but the underlying mechanisms remain to be further explored, and the physical factors reported to date do not exhibit the full properties of the extracellular matrix (ECM). Previous in vitro studies mainly used rigid polystyrene dishes, while one of the characteristics of the native in-vivo environment of neurons is the soft nature of brain ECM. The reported stiffness of brain tissue is very soft ranging between 100 Pa and 3 kPa, and the effect of substrate stiffness on direct neuronal reprogramming has not been explored. Here, we show for the first time that soft substrates substantially improved the production efficiency and quality of iNs, without needing to co-culture with glial cells during reprogramming, producing more glutamatergic neurons with electrophysiological functions in a shorter time. Transcriptome sequencing indicated that soft substrates might promote glutamatergic neuron reprogramming through integrins, actin cytoskeleton, Hippo signalling pathway, and regulation of mesenchymal-to-epithelial transition, and competing endogenous RNA network analysis provided new targets for neuronal reprogramming. We demonstrated that soft substrates may promote neuronal reprogramming by inhibiting microRNA-615-3p-targeting integrin subunit beta 4. Our findings can aid the development of regenerative therapies and help improve our understanding of neuronal reprogramming. STATEMENT OF SIGNIFICANCE: First, we have shown that low stiffness promotes direct reprogramming on the basis of small molecule combinations. To the best of our knowledge, this is the first report on this type of method, which may greatly promote the progress of neural reprogramming. Second, we found that microRNA (miR)-615-3p may interact with integrin subunit beta 4 (ITGB4), and the soft substrates may promote neural reprogramming by inhibiting miR-615-3p targeting ITGB4. We are the first to report on this mechanism. Our findings will provide more functional neurons for subsequent basic and clinical research in neurological regenerative medicine, and will help to improve the overall understanding of neural reprogramming. This work also provides new ideas for the design of medical biomaterials for nerve regeneration.


Subject(s)
MicroRNAs , Polystyrenes , Biocompatible Materials/pharmacology , Fibroblasts , Integrins/metabolism , MicroRNAs/pharmacology , Neurons/metabolism , Polystyrenes/pharmacology
6.
Stem Cell Res Ther ; 13(1): 394, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35922863

ABSTRACT

BACKGROUND: Retinitis pigmentosa is a rod-cone degenerative disease that induces irreversible vision loss. This study probed the protective capacity of mesenchymal stem cell-derived small EVs (MSC-EVs) on the retinas of rd10 mice and the underlying mechanism. METHODS: MSC-EVs were injected into the vitreous of rd10 mice at postnatal day 14 and P21; morphology and function were examined at P28. The mechanism of action was explored by using co-culture of photoreceptor cell line 661 W and microglia cell line BV2. RESULTS: Treatment with MSC-EVs increased the survival of photoreceptors and preserved their structure. Visual function, as reflected by optomotor and electroretinogram responses, was significantly enhanced in MSC-EVs-treated rd10 mice. Mechanistically, staining for Iba1, GFAP, F4/80, CD68 and CD206 showed that MSC-EVs suppressed the activation of microglial, Müller glial and macrophages. Furthermore, western blotting showed that the treatment inhibited the NF-κB pathway. RNA-seq and qPCR showed that MSC-EVs upregulated anti-inflammatory cytokines while downregulating pro-inflammatory cytokines. MSC-EVs application in vitro decreased the number of TUNEL-positive 661 W cells co-cultured with LPS-stimulated BV2, with similar impact on the cytokine expression as in vivo study. Genetic screening predicted miR-146a to be the downstream target of MSC-EVs, which was detected in MSC-EVs and upregulated in co-cultured 661 W cells and BV2 cells after MSC-EVs treatment. Upregulation of miR-146a by using its mimic decreased the expression of the transcription factor Nr4a3, and its downregulation inhibition promoted Nr4a3 expression in both 661 W and BV2 cells. Nr4a3 was further identified as the target gene of miR-146a by dual-luciferase assay. Furthermore, overexpressing miR-146a significantly decreased the expression of LPS-induced pro-inflammatory cytokines in BV2 cells. CONCLUSIONS: MSC-EVs delays retinal degeneration in rd10 mice mainly by its anti-inflammatory effect via the miR-146a-Nr4a3axis. Hence, MSC-EVs may be used in the treatment of neurodegenerative diseases.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Receptors, Steroid , Retinitis Pigmentosa , Animals , Anti-Inflammatory Agents , Cytokines/metabolism , DNA-Binding Proteins , Disease Models, Animal , Extracellular Vesicles/metabolism , Lipopolysaccharides , Mesenchymal Stem Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins , Receptors, Thyroid Hormone , Retina/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/therapy
7.
Biomed Res Int ; 2021: 8834795, 2021.
Article in English | MEDLINE | ID: mdl-33575346

ABSTRACT

The positive effect of low-intensity pulsed ultrasound (LIPUS) on bone fracture healing has been proved. However, during the period of LIPUS therapy, it is undetermined whether LIPUS promotes the formation of heterotopic ossification (HO), which usually occurs in muscle tissues after trauma such as bone fracture and spinal cord injury. Here, we used 6-week LIPUS therapy in a 42-year-old Chinese male patient with a fracture nonunion in combination with ultrasonography for monitoring fracture healing and HO formation. After the LIPUS therapy, the mineralized bone formation in the area of defect of the distal tibia was presented in an ultrasound image, which was consistent with the outcome of plain radiography showing callus formation and the blurred fracture line in the area exposed to LIPUS. In addition, ultrasound images revealed no evidence of HO development within soft tissues during the period of LIPUS therapy. This study suggests that ultrasonography is a potential tool to guarantee the performance of LIPUS therapy with monitoring HO formation. Easy to use, the integration of the handheld ultrasound scanner and the ultrasonic therapeutic apparatus is entirely dedicated to help orthopedists make high-quality care and diagnosis.


Subject(s)
Fractures, Ununited , Ultrasonic Therapy , Ultrasonography , Adult , Feasibility Studies , Fractures, Ununited/diagnostic imaging , Fractures, Ununited/therapy , Humans , Male , Tibial Fractures/diagnostic imaging , Tibial Fractures/therapy , Ultrasonic Waves
8.
Stem Cell Res Ther ; 11(1): 392, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32917285

ABSTRACT

BACKGROUND: Human bone marrow-derived mesenchymal stem cells (hBMSCs) have chondrocyte differentiation potential and are considered to be a cell source for cell-transplantation-mediated repair of cartilage defects, including those associated with osteoarthritis (OA). However, chondrocyte hypertrophic differentiation is a major obstacle for the application of hBMSCs in articular cartilage defect treatment. We have previously shown that microRNA-27b (miR-27b) inhibits hypertrophy of chondrocytes from rat knee cartilage. In this study, we investigated the role of miR-27b in chondrocyte hypertrophic differentiation of hBMSCs. METHODS: Chondrogenic marker and microRNA expression in hBMSC chondrogenic pellets were evaluated using RT-qPCR and immunohistochemistry. The hBMSCs were transfected with miR-27b before inducing differentiation. Gene and protein expression levels were analyzed using RT-qPCR and western blot. Coimmunoprecipitation was used to confirm interaction between CBFB and RUNX2. Luciferase reporter assays were used to demonstrate that CBFB is a miR-27b target. Chondrogenic differentiation was evaluated in hBMSCs treated with shRNA targeting CBFB. Chondrogenic hBMSC pellets overexpressing miR-27b were implanted into cartilage lesions in model rats; therapeutic effects were assessed based on histology and immunohistochemistry. RESULTS: The hBMSCs showed typical MSC differentiation potentials. During chondrogenic differentiation, collagen 2 and 10 (COL2 and COL10), SOX9, and RUNX2 expression was upregulated. Expression of miR-140, miR-143, and miR-181a increased over time, whereas miR-27b and miR-221 were downregulated. Cartilage derived from hBMSC and overexpressing miR-27b exhibited higher expression of COL2 and SOX9, but lower expression of COL10, RUNX2, and CBFB than did the control cartilage. CBFB and RUNX2 formed a complex, and CBFB was identified as a novel miR-27b target. CBFB knockdown by shRNA during hBMSC chondrogenic differentiation led to significantly increased COL2 and SOX9 expression and decreased COL10 expression. Finally, miR-27b-overexpressing hBMSC chondrogenic pellets had better hyaline cartilage morphology and reduced expression of hypertrophic markers and tend to increase repair efficacy in vivo. CONCLUSION: MiR-27b plays an important role in preventing hypertrophic chondrogenesis of hBMSCs by targeting CBFB and is essential for maintaining a hyaline cartilage state. This study provides new insights into the mechanism of hBMSC chondrocyte differentiation and will aid in the development of strategies for treating cartilage injury based on hBMSC transplantation.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Animals , Cell Differentiation , Cells, Cultured , Chondrocytes , Chondrogenesis/genetics , Core Binding Factor beta Subunit , Humans , Hypertrophy/genetics , MicroRNAs/genetics , Rats
9.
Stem Cell Res Ther ; 11(1): 174, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393338

ABSTRACT

BACKGROUND: Skin wounding is very common and may be slow to heal. Increasing evidence shows that exosomes derived from mesenchymal stem cells (MSCs) dramatically enhance skin wound healing in a paracrine manner. However, the mechanism underlying this phenomenon has not yet been elucidated. Thus, the objective of the present study was to identify the signaling pathways and paracrine factors by which MSC-derived exosomes promote de novo skin tissue regeneration in response to wound healing. METHODS: In vitro and in vivo skin wound healing models were created by treating immortalized human keratinocytes (HaCaT) with hydrogen peroxide (H2O2) and excising full-thickness mouse skin, respectively. Exosomes were extracted from human umbilical cord Wharton's jelly MSCs (hucMSC-Ex) by ultracentrifugation of cell culture supernatant. RESULTS: The hucMSC-Ex treatment significantly increased HaCaT cell proliferation and migration in a time- and dose-dependent manner, suppressed HaCaT apoptosis induced with H2O2 by inhibiting nuclear translocation of apoptosis-inducing factor (AIF) and upregulating poly ADP ribose polymerase 1 (PARP-1) and poly (ADP-ribose) (PAR). The animal experiments showed that relative to hucMSCs, hucMSC-Ex attenuated full-thickness skin wounding by enhancing epidermal re-epithelialization and dermal angiogenesis. CONCLUSIONS: These findings indicated that direct administration of hucMSC-Ex may effectively treat cutaneous wounding and could be of great value in clinical settings.


Subject(s)
Exosomes , Animals , Apoptosis , Apoptosis Inducing Factor/genetics , Cell Proliferation , Hydrogen Peroxide/pharmacology , Wound Healing
10.
Front Neurol ; 9: 771, 2018.
Article in English | MEDLINE | ID: mdl-30271377

ABSTRACT

Traumatic injury is one of varying causes of heterotopic ossification (HO). After HO occurrence, rehabilitation training need alterations to avoid the aggravation of HO. Therefore, monitoring of HO development plays an important role in the rehabilitation procedure. The aims of this study are to evaluate the post-traumatic HO occurring at various joints, to describe the features of HO development in ultrasound images, and to provide a guidance for the orthopedist to make individualized rehabilitation therapy. Eight subjects with the post-traumatic HO were recruited in this study. The joints on the injured side was examined by plain radiography. The joints on the injured side and the corresponding sites on the uninjured sides were scanned by ultrsonography. The HO tissues were segmented automatically using a semi-supervised segmentation algorithm. Then the HO tissues were evaluated in comparison with the corresponding region of the uninjured side. During the development stage of immature HO, ultrasonography was sensitive to observe the involved soft tissue and the calcification of HO. The characteristics of HO tissues in ultrasound image included the hyperechoic mass occasionally accompanied with acoustic shadow and the irregular muscular architecture. It was found that the mean grayscale value of HO was significantly higher (p < 0.001) than that of the uninjured side at the middle and late stages. During the development period of HO, the HO grayscale value gradually increased and the mean grayscale of value of mature HO was significantly higher (p < 0.05) than that of immature HO. According to the information of HO provided by ultrasound, the orthopedist properly adjusted the rehabilitation treatment. The results demonstrated that the visualization of HO using ultrasonography revealed the development of HO in the muscle tissues around the injured joints and thus provide a guidance for the orthopedist to make individualized rehabilitation therapy. Ultrasound could be a useful imaging modality for quantitative evaluation of HO during the rehabilitation of traumatic injury.

11.
Stem Cell Res Ther ; 9(1): 211, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30092828

ABSTRACT

BACKGROUND: Differentiation of human induced pluripotent stem cells (hiPSCs) into hepatocytes has important clinical significance in providing a new stem cell source for cell therapy of terminal liver disease. The differential gene expression analysis of hiPSCs, induced hepatocyte-like cells (HLCs), and primary human hepatocytes (PHHs) provides valuable information for optimization of an induction scheme and exploration of differentiation mechanisms. METHODS: Human hair follicle-derived iPSCs (hHF-iPSCs) were induced in vitro by mimicking the environment of a developing liver for 19 days. Expression of specific proteins was determined by immunofluorescence staining; the function of HLCs in storage and metabolism was identified by detecting periodic acid-Schiff, indocyanine green, and low-density lipoprotein. Based on the transcriptomics data, the differential gene expression profiles of hHF-iPSCs, HLCs, and PHHs were analyzed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, FunRich, and network analysis methods. RESULTS: HLCs were able to express albumin (ALB), alpha-fetoprotein, CYP3A4, and CYP7A1, and exhibited matured liver cell functions such as glycogen synthesis and storage. Complement and coagulation cascades and metabolic pathways ranked top in the downregulated list of HLCs/PHHs, while the cell cycle ranked top in the upregulated list of HLCs/PHHs. In the protein-protein interaction network, according to the degree rankings, TOP2A, CDK1, etc. were the important upregulated differentially expressed genes (DEGs), while ALB, ACACB, etc. were the major downregulated DEGs in HLCs/PHHs; the module analysis indicated that CDCA8, AURKB, and AURKA were the top upregulated DEGs in HLCs/PHHs. CONCLUSIONS: We presented the differences in gene expression among hHF-iPSCs, HLCs, and PHHs through transcriptome array data and provided new ideas for the optimization of induction.


Subject(s)
Hair Follicle/cytology , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Gene Expression/physiology , Hair Follicle/metabolism , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
12.
Article in English | MEDLINE | ID: mdl-30008302

ABSTRACT

Fenugreek is a well-known medicinal plant used for treatment of diabetes. In this study, the antidiabetic effect of fenugreek flavonoids was investigated by metabonomics based on ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). Fenugreek flavonoids were purified using polyamide resin and D101 macroporous adsorption resin, characterized by UPLC-Q-TOF-MS, and administered to streptozotocin (STZ)-induced diabetic rats for 28 days. Pharmacological study results indicated that fenugreek flavonoids exerted a strong antidiabetic effect characterized by significant reduction of fasting blood glucose (P < 0.01), increase in serum insulin level (P < 0.01) and liver glycogen content (P < 0.01), attenuation of weight loss, and improvement of pancreatic islet and kidney conditions. The antidiabetic effect of fenugreek flavonoids was further analyzed by metabonomics. Serum samples of health and diabetic rats treated or not with fenugreek flavonoids were evaluated by UPLC-Q-TOF-MS, followed by principal component analysis (PCA) and orthogonal projection to latent structures squares-discriminant analysis (OPLS-DA). The PCA model revealed significant differences among the animal groups, and OPLS-DA identified fenugreek flavonoids-induced changes of 11 potential biomarkers involved in lipid metabolism (docosahexaenoic acid, arachidonic acid, sphinganine, sphingosine­1­phosphate, and lysophosphatidylcholines 20:4, 18:2, 16:0, and 20:2), amino acid metabolism (hippuric acid and tryptophan), and kidney function-related metabolism (2­phenylethanol glucuronide). Our study demonstrates that flavonoids are bioactive components of fenugreek with potent antidiabetic activity, which exert their therapeutic effects by multiple mechanisms, including reducing insulin resistance, improving gluconeogenesis, and protecting islet cells and kidneys from damage.


Subject(s)
Biomarkers/blood , Diabetes Mellitus, Experimental/metabolism , Flavonoids/pharmacology , Hypoglycemic Agents/pharmacology , Metabolomics/methods , Plant Extracts/pharmacology , Trigonella , Animals , Blood Glucose/drug effects , Chromatography, High Pressure Liquid/methods , Kidney/drug effects , Male , Mass Spectrometry , Pancreas/drug effects , Principal Component Analysis , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Streptozocin
13.
Int J Med Sci ; 15(3): 257-268, 2018.
Article in English | MEDLINE | ID: mdl-29483817

ABSTRACT

BMMSCs have drawn great interest in tissue engineering and regenerative medicine attributable to their multi-lineage differentiation capacity. Increasing evidence has shown that the mechanical stiffness of extracellular matrix is a critical determinant for stem cell behaviors. However, it remains unknown how matrix stiffness influences MSCs commitment with changes in cell morphology, adhesion, proliferation, self-renewal and differentiation. We employed fibronectin coated polyacrylamide hydrogels with variable stiffnesses ranging from 13 to 68 kPa to modulate the mechanical environment of BMMSCs and found that the morphology and adhesion of BMMSCs were highly dependent on mechanical stiffness. Cells became more spread and more adhesive on substrates of higher stiffness. Similarly, the proliferation of BMMSCs increased as stiffness increased. Sox2 expression was lower during 4h to 1 week on the 13-16 kPa and 62-68 kPa, in contrast, it was higher during 4h to 1 week on the 48-53 kPa. Oct4 expression on 13-16 kPa was higher than 48-53 kPa at 4h, and it has no significant differences at other time point among three different stiffness groups. On 62-68 kPa, BMMSCs were able to be induced toward osteogenic phenotype and generated a markedly high level of RUNX2, ALP, and Osteopontin. The cells exhibited a polygonal morphology and larger spreading area. These results suggest that matrix stiffness modulates commitment of BMMSCs. Our findings may eventually aid in the development of novel, effective biomaterials for the applications in tissue engineering.


Subject(s)
Cell Differentiation/genetics , Mesenchymal Stem Cells/cytology , Osteogenesis/genetics , Tissue Engineering , Cell Adhesion/genetics , Cell Proliferation/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Humans , Mesenchymal Stem Cell Transplantation , Osteopontin/genetics , SOXB1 Transcription Factors/genetics , Tissue Scaffolds
14.
Sci Rep ; 7(1): 12266, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28947775

ABSTRACT

Despite recent progress in the preparation of feeder cells for human induced pluripotent stem cells (hiPSCs), there remain issues which limit the acquisition of feeder cells in large scale. Approaches for obtaining feeder cells quickly on a large scale are in immediate need. To reach this goal, we established suspension-adhesion method (SAM) and three-dimensional (3D) suspension method (3DSM). In SAM, mouse embryonic fibroblast (MEF) growth were fully inhibited by 10 µg/ml mitomycin-C (MMC) in 0.5 hours, and the feeder cells generated display higher adherent and recovery rates as well as longer survival time compared to conventional method (CM). 3DSM, an optimized method of SAM in which MEFs were cultured and MMC treated in suspension, was developed to lower the costs and workload using CELLSPIN System. The yield of feeder cells is several times the yield of SAM while the adherent and recovery rates and the capacity of supporting hiPSCs growth were not sacrificed. Hence, 3DSM is an economical and easy way to generate large-scale feeder cells for hiPSCs cultures.


Subject(s)
Cell Culture Techniques/methods , Cell Proliferation , Feeder Cells/physiology , Animals , Cell Survival , Cells, Cultured , Fibroblasts/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Mice , Time Factors
15.
Differentiation ; 96: 30-39, 2017.
Article in English | MEDLINE | ID: mdl-28753444

ABSTRACT

Mesenchymal stem cells (MSCs) are a compatible cellular alternative for regenerative medicine and tissue engineering because of their powerful multipotency. Matrix stiffness plays a profound role on stem cell behavior. Nevertheless, the effect of matrix stiffness on umbilical cordmesenchymal stem cells (UC-MSCs) remains unexplored. To conduct an in-depth exploration, we cultured UC-MSCs on different stiffness (Young's modulus: 13-16, 35-38, 48-53, and 62-68 kPa) polyacrylamide gels coated with fibronectin. We found that the proliferation and adhesion of UC-MSCs varied when cultured on the different matrices, and the spreading capacity was stronger as the stiffness increased (*P<0.05). Real-time quantitative PCR results showed that the soft matrix promoted adipogenic differentiation, with higher expression levels of adipocytic markers like PPARγ and C/EBPα (*P<0.05). In contrast, cells tended to differentiate into muscle when cultured on the 48-53 kPa matrix, which was validated by increased expression of myogenic makers like desminand MOYG (*P<0.05). Moreover, increased expression of osteoblastic makers (*P<0.05), such as ALP, collagen type I, osteocalcin, and Runx2, confirmed that cells differentiated into bone on the high-stiffness matrix.


Subject(s)
Cell Differentiation , Cell Proliferation , Elastic Modulus , Mesenchymal Stem Cells/cytology , Acrylic Resins/chemistry , Adipocytes/cytology , Adipocytes/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Adhesion , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Fibronectins/chemistry , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Umbilical Cord/cytology
16.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1044-1045: 8-16, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28063302

ABSTRACT

Fenugreek is a traditional plant for the treatment of diabetes. Galactomannan, an active major component in fenugreek seeds, has shown hypoglycemic activity. The present study was performed to investigate the therapeutic mechanism underlying fenugreek galactomannan (F-GAL) in treating diabetes, using a metabonomics approach based on ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-QTOF/MS). The F-GAL used for study was highly purified, and its yield, purity, and galactose/mannose ratio were characterized by capillary zone electrophoresis (CZE) and a modified phenol-sulfuric acid method. After treatment of streptozotocin (STZ)-induced diabetic rats with F-GAL for 28days, urine and serum samples were analyzed by UPLC-QTOF/MS. Multivariate statistical approaches such as principal component analysis (PCA) and orthogonal projection to latent structures squares-discriminant analysis (OPLS-DA) were applied to distinguish the non-diabetic/untreated, diabetic/untreated, and diabetic/F-GAL-treated groups. Then, potential biomarkers were identified that may help elucidate the underlying therapeutic mechanism of F-GAL in diabetes. The results demonstrated that there was a clear separation among the three groups in the PCA model. Fourteen potential biomarkers were identified by OPLS-DA, and they were determined to be produced in response to the therapeutic effects of F-GAL. These biomarkers were involved in histidine metabolism, tryptophan metabolism, energy metabolism, phenylalanine metabolism, sphingolipid metabolism, glycerophospholipid metabolism, and arachidonic acid metabolism. In conclusion, our study demonstrates that a metabonomics approach is a powerful, novel tool that can be used to evaluate the underlying therapeutic mechanisms of herb extracts.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents , Mannans , Metabolomics/methods , Plant Extracts , Trigonella/chemistry , Animals , Biomarkers/blood , Blood Glucose/drug effects , Chromatography, High Pressure Liquid/methods , Galactose/analogs & derivatives , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Mannans/pharmacology , Mannans/therapeutic use , Metabolic Networks and Pathways/drug effects , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Principal Component Analysis , Random Allocation , Rats
17.
Cell Tissue Res ; 362(1): 69-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25948482

ABSTRACT

The use of human mesenchymal stem cells (hMSCs) in cell therapies has increased the demand for strategies that allow efficient cell scale-up. Preliminary data on the three-dimensional (3D) spinner culture describing the potential use of microcarriers for hMSCs culture scale-up have been reported. We exploited a rich source of autologous stem cells (human hair follicle) and demonstrated the robust in vitro long-term expansion of human hair follicle-derived mesenchymal stem cells (hHF-MSCs) by using CultiSpher(®)-G microcarriers. We analyzed the feasibility of 3D culture by using hHF-MSCs/CultiSpher(®)-G microcarrier constructs for its potential applicability in regenerative medicine by comparatively analyzing the performance of hHF-MSCs adhered to the CultiSpher(®)-G microspheres in 3D spinner culture and those grown on the gelatin-coated plastic dishes (2D culture), using various assays. We showed that the hHF-MSCs seeded at various densities quickly adhered to and proliferated well on the microspheres, thus generating at least hundreds of millions of hHF-MSCs on 1 g of CultiSpher(®)-G within 12 days. This resulted in a cumulative cell expansion of greater than 26-fold. Notably, the maximum and average proliferation rates in 3D culture were significantly greater than that of the 2D culture. However, the hHF-MSCs from both the cultures retained surface marker and nestin expression, proliferation capacity and differentiation potentials toward adipocytes, osteoblasts and smooth muscle cells and showed no significant differences as evidenced by Edu incorporation, cell cycle, colony formation, apoptosis, biochemical quantification and qPCR assays.


Subject(s)
Hair Follicle/metabolism , Mesenchymal Stem Cells/metabolism , Adult , Cell Differentiation , Cell Proliferation , Female , Hair Follicle/cytology , Humans , Male , Middle Aged , Regenerative Medicine
18.
Stem Cell Res Ther ; 6: 38, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25889402

ABSTRACT

INTRODUCTION: Successful stem cell therapy relies on large-scale generation of stem cells and their maintenance in a proliferative multipotent state. This study aimed to establish a three-dimension culture system for large-scale generation of hWJ-MSC and investigated the self-renewal activity, genomic stability and multi-lineage differentiation potential of such hWJ-MSC in enhancing skin wound healing. METHODS: hWJ-MSC were seeded on gelatin microbeads and cultured in spinning bottles (3D). Cell proliferation, karyotype analysis, surface marker expression, multipotent differentiation (adipogenic, chondrogenic, and osteogenic potentials), and expression of core transcription factors (OCT4, SOX2, NANOG, and C-MYC), as well as their efficacy in accelerating skin wound healing, were investigated and compared with those of hWJ-MSC derived from plate cultres (2D), using in vivo and in vitro experiments. RESULTS: hWJ-MSC attached to and proliferated on gelatin microbeads in 3D cultures reaching a maximum of 1.1-1.30×10(7) cells on 0.5 g of microbeads by days 8-14; in contrast, hWJ-MSC derived from 2D cultures reached a maximum of 6.5 -11.5×10(5) cells per well in a 24-well plate by days 6-10. hWJ-MSC derived by 3D culture incorporated significantly more EdU (P<0.05) and had a significantly higher proliferation index (P<0.05) than those derived from 2D culture. Immunofluorescence staining, real-time PCR, flow cytometry analysis, and multipotency assays showed that hWJ-MSC derived from 3D culture retained MSC surface markers and multipotency potential similar to 2D culture-derived cells. 3D culture-derived hWJ-MSC also retained the expression of core transcription factors at levels comparable to their 2D culture counterparts. Direct injection of hWJ-MSC derived from 3D or 2D cultures into animals exhibited similar efficacy in enhancing skin wound healing. CONCLUSIONS: Thus, hWJ-MSC can be expanded markedly in gelatin microbeads, while retaining MSC surface marker expression, multipotent differential potential, and expression of core transcription factors. These cells also efficiently enhanced skin wound healing in vivo, in a manner comparable to that of hWJ-MSC obtained from 2D culture.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cells/cytology , Skin/injuries , Wharton Jelly/cytology , Wound Healing/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Gelatin/metabolism , Homeodomain Proteins/biosynthesis , Humans , Mice , Microspheres , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Tissue Culture Techniques
19.
Fundam Clin Pharmacol ; 29(2): 191-203, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25619112

ABSTRACT

Cyclosporine A (CsA) enhances hair growth through caspase-dependent pathways by retarding anagen-to-catagen phase transition in the hair follicle growth cycle. Whether apoptosis-inducing factor (AIF), a protein that induces caspase-independent apoptosis, can regulate the hair follicle cycle in response to CsA is currently unclear. Here, we show that the pro-hair growth properties of CsA are in part due to blockage of AIF nuclear translocation. We first isolate hair follicles from murine dorsal skin. We then used Western blot, immunohistochemistry and immunofluorescence to evaluate the expression and localization of AIF in hair follicles. We also determined whether modulation of AIF was responsible for the effects of CsA at the anagen-to-catagen transition. AIF was expressed in hair follicles during the anagen, catagen and telogen phases. There was significant nuclear translocation of AIF as hair follicles transitioned from anagen to late catagen phase; this was inhibited by CsA, likely due to reduced cyclophilin A expression and attenuated AIF release from mitochondria. However, we note that AIF translocation was not completely eliminated, which likely explains why the transition to catagen phase was severely retarded by CsA, rather than being completely inhibited. We speculate that blockade of the AIF signalling pathway is a critical event required for CsA-dependent promotion of hair growth in mice. The study of AIF-related signalling pathways may provide insight into hair diseases and suggest potential novel therapeutic strategies.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cell Nucleus/metabolism , Cyclosporine/pharmacology , Hair Follicle/metabolism , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Cell Nucleus/drug effects , Hair Follicle/drug effects , Mice , Mice, Inbred C57BL
20.
Cell Transplant ; 24(5): 891-907, 2015.
Article in English | MEDLINE | ID: mdl-24835482

ABSTRACT

Genetically engineered stem cells that overexpress genes encoding therapeutic products can be exploited to correct metabolic disorders by repairing and regenerating diseased organs or restoring their function. Hair follicles are readily accessible and serve as a rich source of autologous stem cells for cell-based gene therapy. Here we isolated mesenchymal stem cells from human hair follicles (HF-MSCs) and engineered them to overexpress the human insulin gene and release human insulin in a time- and dose-dependent manner in response to rapamycin. The engineered HF-MSCs retained their characteristic cell surface markers and retained their potential to differentiate into adipocytes and osteoblasts. When mice with streptozotocin-induced type 1 diabetes were engrafted with these engineered HF-MSCs, these cells expressed and released a dose of human insulin, dramatically reversed hyperglycemia, and significantly reduced death rate. Moreover, the engineered HF-MSCs did not form detectable tumors throughout the 120-day animal tests in our experiment. Our results show that HF-MSCs can be used to safely and efficiently express therapeutic transgenes and therefore show promise for cell-based gene therapy of human disease.


Subject(s)
Cell Engineering , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Hair Follicle/metabolism , Hyperglycemia/therapy , Insulin/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Adult , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Heterografts , Humans , Hyperglycemia/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...