Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
J Control Release ; 370: 583-599, 2024 May 13.
Article En | MEDLINE | ID: mdl-38729435

Stem cells are recognized as an important target and tool in regenerative engineering. In this study, we explored the feasibility of engineering amniotic fluid-derived mesenchymal stem cell-secreted molecules (afMSC-SMs) as a versatile bioactive material for skin regenerative medicine applications in a time- and cost-efficient and straightforward manner. afMSC-SMs, obtained in powder form through ethanol precipitation, effectively contributed to preserving the self-renewal capacity and differentiation potential of primary human keratinocytes (pKCs) in a xeno-free environment, offering a potential alternative to traditional culture methods for their long-term in vitro expansion, and allowed them to reconstitute a fully stratified epithelium sheet on human dermal fibroblasts. Furthermore, we demonstrated the flexibility of afMSC-SMs in wound healing and hair regrowth through injectable hydrogel and nanogel-mediated transdermal delivery systems, respectively, expanding the pool of regenerative applications. This cell-free approach may offer several potential advantages, including streamlined manufacturing processes, scalability, controlled formulation, longer shelf lives, and mitigation of risks associated with living cell transplantation. Accordingly, afMSC-SMs could serve as a promising therapeutic toolbox for advancing cell-free regenerative medicine, simplifying their broad applicability in various clinical settings.

2.
Exp Mol Med ; 55(6): 1182-1192, 2023 06.
Article En | MEDLINE | ID: mdl-37258581

Spinal cord injury (SCI) is a clinical condition that leads to permanent and/or progressive disabilities of sensory, motor, and autonomic functions. Unfortunately, no medical standard of care for SCI exists to reverse the damage. Here, we assessed the effects of induced neural stem cells (iNSCs) directly converted from human urine cells (UCs) in SCI rat models. We successfully generated iNSCs from human UCs, commercial fibroblasts, and patient-derived fibroblasts. These iNSCs expressed various neural stem cell markers and differentiated into diverse neuronal and glial cell types. When transplanted into injured spinal cords, UC-derived iNSCs survived, engrafted, and expressed neuronal and glial markers. Large numbers of axons extended from grafts over long distances, leading to connections between host and graft neurons at 8 weeks post-transplantation with significant improvement of locomotor function. This study suggests that iNSCs have biomedical applications for disease modeling and constitute an alternative transplantation strategy as a personalized cell source for neural regeneration in several spinal cord diseases.


Neural Stem Cells , Spinal Cord Injuries , Humans , Rats , Animals , Neural Stem Cells/metabolism , Spinal Cord Injuries/therapy , Spinal Cord Injuries/metabolism , Neurons/metabolism , Axons , Spinal Cord , Cell Differentiation/physiology
3.
Int J Mol Sci ; 24(10)2023 May 12.
Article En | MEDLINE | ID: mdl-37239986

Let-7 miRNAs have pleiotropic cellular functions in cell proliferation, migration, and regenerative processes. Here, we investigate whether the inhibition of let-7 miRNAs with antisense oligonucleotides (ASOs) can be a transient and safe strategy enhancing the therapeutic potential of mesenchymal stromal cells (MSCs) to overcome their limitations in cell therapeutic trials. We first identified major subfamilies of let-7 miRNAs preferentially expressed in MSCs, and efficient ASO combinations against these selected subfamilies that mimic the effects of LIN28 activation. When let-7 miRNAs were inhibited with an ASO combination (anti-let7-ASOs), MSCs exhibited higher proliferation with delayed senescence during the passaging into a culture. They also exhibited increased migration and enhanced osteogenic differentiation potential. However, these changes in MSCs were not accompanied by cell-fate changes into pericytes or the additional acquisition of stemness, but instead occurred as functional changes accompanied by changes in proteomics. Interestingly, MSCs with let-7 inhibition exhibited metabolic reprogramming characterized by an enhanced glycolytic pathway, decreased reactive oxygen species, and lower transmembrane potential in mitochondria. Moreover, let-7-inhibited MSCs promoted the self-renewal of neighboring hematopoietic progenitor cells, and enhanced capillary formation in endothelial cells. These findings together show that our optimized ASO combination efficiently reprograms the MSC functional state, allowing for more efficient MSC cell therapy.


Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/metabolism , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Cell Differentiation/genetics , MicroRNAs/metabolism
4.
Biochem Biophys Res Commun ; 629: 128-134, 2022 11 12.
Article En | MEDLINE | ID: mdl-36116375

Chronic Kidney Disease (CKD) is increasingly recognized as a global public health issue. Diabetic nephropathy (DN), also known as diabetic kidney disease, is a leading cause of CKD. Regenerative medicine strategy employing nephron progenitor cells (NPCs) is worthy of consideration as an alternative to shortage of donor organs for kidney transplantation. In previous study, we successfully generated induced NPCs (iNPCs) from human urine-derived cells that resembled human embryonic stem cell-derived NPCs. Here, we aimed to investigate the therapeutic potential of iNPCs in DN animal model. The results revealed the therapeutic effect of iNPCs as follows: (1) diminished glomerular hypertrophy, (2) reduced tubulointerstitial fibrosis, (3) low blood urea nitrogen, serum creatinine and albuminuria value, (4) decreased inflammation/fibrosis, (5) enhanced renal regeneration and (6) confirmed safety. This study demonstrates that human iNPCs have a therapeutic potential as a cell source for transplantation in patients with kidney diseases.


Diabetes Mellitus , Diabetic Nephropathies , Renal Insufficiency, Chronic , Animals , Creatinine , Diabetes Mellitus/pathology , Diabetic Nephropathies/drug therapy , Fibrosis , Humans , Kidney/pathology , Mice , Nephrons , Renal Insufficiency, Chronic/pathology , Stem Cells
5.
Tissue Eng Regen Med ; 19(3): 643-658, 2022 06.
Article En | MEDLINE | ID: mdl-35325404

BACKGROUND: Immunoglobulin A (IgA) nephropathy (IgAN) is one of an important cause of progressive kidney disease and occurs when IgA settles in the kidney resulted in disrupts kidney's ability to filter waste and excess water. Hydrogels are promising material for medical applications owing to their excellent adaptability and filling ability. Herein, we proposed a hyaluronic acid/gelatin (CHO-HA/Gel-NH2) bioactive hydrogel as a cell carrier for therapeutic kidney regeneration in IgAN. METHODS: CHO-HA/Gel-NH2 hydrogel was fabricated by Schiff-base reaction without any additional crosslinking agents. The hydrogel concentrations and ratios were evaluated to enhance adequate mechanical properties and biocompatibility for further in vivo study. High serum IgA ddY mice kidneys were treated with human urine-derived renal progenitor cells encapsulated in the hydrogel to investigate the improvement of IgA nephropathy and kidney regeneration. RESULTS: The stiffness of the hydrogel was significantly enhanced and could be modulated by altering the concentrations and ratios of hydrogel. CHO-HA/Gel-NH2 at a ratio of 3/7 provided a promising milieu for cells viability and cells proliferation. From week four onwards, there was a significant reduction in blood urea nitrogen and serum creatinine level in Cell/Gel group, as well as well-organized glomeruli and tubules. Moreover, the expression of pro-inflammatory and pro-fibrotic molecules significantly decreased in the Gel/Cell group, whereas anti-inflammatory gene expression was elevated compared to the Cell group. CONCLUSION: Based on in vivo studies, the renal regenerative ability of the progenitor cells could be further increased by this hydrogel system.


Glomerulonephritis, IGA , Hydrogels , Animals , Gelatin , Glomerulonephritis, IGA/drug therapy , Hyaluronic Acid , Immunoglobulin A , Kidney , Mice , Regeneration
6.
NPJ Regen Med ; 7(1): 4, 2022 Jan 13.
Article En | MEDLINE | ID: mdl-35027563

The generation of human oligodendrocyte progenitor cells (OPCs) may be therapeutically valuable for human demyelinating diseases such as multiple sclerosis. Here, we report the direct reprogramming of human somatic cells into expandable induced OPCs (iOPCs) using a combination of OCT4 and a small molecule cocktail. This method enables generation of A2B5+ (an early marker for OPCs) iOPCs within 2 weeks retaining the ability to differentiate into MBP-positive mature oligodendrocytes. RNA-seq analysis revealed that the transcriptome of O4+ iOPCs was similar to that of O4+ OPCs and ChIP-seq analysis revealed that putative OCT4-binding regions were detected in the regulatory elements of CNS development-related genes. Notably, engrafted iOPCs remyelinated the brains of adult shiverer mice and experimental autoimmune encephalomyelitis mice with MOG-induced 14 weeks after transplantation. In conclusion, our study may contribute to the development of therapeutic approaches for neurological disorders, as well as facilitate the understanding of the molecular mechanisms underlying glial development.

7.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article En | MEDLINE | ID: mdl-34948246

BACKGROUND: Regenerative medicine strategies employing nephron progenitor cells (NPCs) are a viable approach that is worthy of substantial consideration as a promising cell source for kidney diseases. However, the generation of induced nephron progenitor-like cells (iNPCs) from human somatic cells remains a major challenge. Here, we describe a novel method for generating NPCs from human urine-derived cells (UCs) that can undergo long-term expansion in a serum-free condition. RESULTS: Here, we generated iNPCs from human urine-derived cells by forced expression of the transcription factors OCT4, SOX2, KLF4, c-MYC, and SLUG, followed by exposure to a cocktail of defined small molecules. These iNPCs resembled human embryonic stem cell-derived NPCs in terms of their morphology, biological characteristics, differentiation potential, and global gene expression and underwent a long-term expansion in serum-free conditions. CONCLUSION: This study demonstrates that human iNPCs can be readily generated and expanded, which will facilitate their broad applicability in a rapid, efficient, and patient-specific manner, particularly holding the potential as a transplantable cell source for patients with kidney disease.


Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/metabolism , Nephrons/metabolism , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Humans , Induced Pluripotent Stem Cells/cytology , Nephrons/growth & development , Nephrons/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Urine/cytology
8.
Stem Cell Res ; 54: 102392, 2021 07.
Article En | MEDLINE | ID: mdl-34091428

Leigh syndrome is a progressive neurodegenerative disease due to defects in the mitochondrial genes, including mitochondrial DNA cytochrome b (MTCYB) mutation, that typically begins in infancy or early childhood. Exercise intolerance and fatigue are common symptoms of mitochondrial disorders. Here, we generated induced pluripotent stem cell (iPSC) line from a 1-year-old patient with Leigh syndrome with MTCYB through temporal expression of exogenes, synthetic self-replicative mRNAs which were regulated by B18R protein. The established iPSCs showed expression of various pluripotency markers, a normal karyotype and differentiation potential to three germ layers in vitro while retaining MTCYB mutation.


Induced Pluripotent Stem Cells , Leigh Disease , Neurodegenerative Diseases , Cell Differentiation , Child, Preschool , Cytochromes b/genetics , DNA, Mitochondrial/genetics , Humans , Infant , Leigh Disease/genetics , Mutation/genetics , RNA, Messenger/genetics
9.
Stem Cells Transl Med ; 9(12): 1643-1650, 2020 12.
Article En | MEDLINE | ID: mdl-32716131

Glial cells are crucial for the development of the central nervous system and the maintenance of chemical homeostasis. The process of gliogenesis has been well studied in the rodent brain, but it remains less well studied in the human brain. In addition, rodent glial cells differ from human counterparts in terms of morphologies, functions, and anatomical locations. Cerebral organoids (also referred to as spheroids) derived from human pluripotent stem cells (hPSCs) have been developed and are suitable cell-based models for researching developmental and neurodegenerative diseases. The in vitro generation of glia, including astrocytes and oligodendrocytes, from such organoids represents a promising tool to model neuronal diseases. Here, we showed that three-dimensional (3D) culture of OLIG2- and NKX2.2-expressing neurospheres produced efficiently mature astrocytes and oligodendrocytes in terms of morphologies and expression pattern recapitulating native 3D environment. Our findings provide important insights for developmental research of the human brain and glial specification that may facilitate patient-specific disease modeling.


Homeodomain Proteins/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/metabolism , Zebrafish Proteins/metabolism , Homeobox Protein Nkx-2.2 , Humans , Nuclear Proteins , Transcription Factors
10.
Obstet Gynecol Sci ; 63(5): 594-604, 2020 Sep.
Article En | MEDLINE | ID: mdl-32698560

OBJECTIVE: Exploiting their ability to differentiate into mesenchymal lineages like cartilage, bone, fat, and muscle, and to elicit paracrine effects, mesenchymal stem cells (MSCs) are widely used in clinical settings to treat tissue injuries and autoimmune disorders. One of accessible sources of MSC is the samples used for Papanicolaou (Pap) test, which is a cervical screening method for detecting potentially pre-cancerous and cancerous alterations in the cervical cells and to diagnose genetic abnormalities in fetuses. This study aimed to identify and isolate the stem cells from Pap smear samples collected from pregnant women, and to trace the origin of these cells to maternal or fetal tissue, and characterize their stem cell properties. METHODS: To investigate the possibility and efficiency of establishing MSC lines from the Pap smear samples, we were able to establish 6 cell lines from Pap smear samples from 60 pregnant women at different stages of gestation. RESULTS: The 3 cell lines randomly selected among the 6 established in this study, displayed high proliferation rates, several characteristics of MSCs, and the capacity to differentiate into adipocytes, osteocytes, and chondrocytes. Our study identified that the stem cell lines obtainable from Pap smear sampling were uterine cervical stromal cells (UCSCs) and had 10% efficiency of establishment. CONCLUSION: Despite their low efficiency of establishment, human UCSCs from Pap smear samples can become a simple, safe, low-cost, and donor-specific source of MSCs for stem cell therapy and regenerative medicine.

11.
J Biomed Sci ; 27(1): 56, 2020 Apr 20.
Article En | MEDLINE | ID: mdl-32312260

BACKGROUND: Human keratinocytes and derived products are crucial for skin repair and regeneration. Despite substantial advances in engineered skin equivalents, their poor availability and immunorejection remain major challenges in skin grafting. METHODS: Induced keratinocyte-like cells (iKCs) were directly reprogrammed from human urine cells by retroviral transduction of two lineage-specific transcription factors BMI1 and △NP63α (BN). Expression of keratinocyte stem cell or their differentiation markers were assessed by PCR, immunofluorescence and RNA-Sequencing. Regeneration capacity of iKCs were assessed by reconstitution of a human skin equivalent under air-interface condition. RESULTS: BN-driven iKCs were similar to primary keratinocytes (pKCs) in terms of their morphology, protein expression, differentiation potential, and global gene expression. Moreover, BN-iKCs self-assembled to form stratified skin equivalents in vitro. CONCLUSIONS: This study demonstrated an approach to generate human iKCs that could be directly reprogrammed from human somatic cells and extensively expanded in serum- and feeder cell-free systems, which will facilitate their broad applicability in an efficient and patient-specific manner.


Cellular Reprogramming/physiology , Keratinocytes/physiology , Cells, Cultured/physiology , Cellular Reprogramming Techniques , Female , Humans , In Vitro Techniques , Male , Skin Physiological Phenomena
12.
Cells ; 8(9)2019 09 06.
Article En | MEDLINE | ID: mdl-31489945

Human neural stem cells (NSCs) hold enormous promise for neurological disorders, typically requiring their expandable and differentiable properties for regeneration of damaged neural tissues. Despite the therapeutic potential of induced NSCs (iNSCs), a major challenge for clinical feasibility is the presence of integrated transgenes in the host genome, contributing to the risk for undesired genotoxicity and tumorigenesis. Here, we describe the advanced transgene-free generation of iNSCs from human urine-derived cells (HUCs) by combining a cocktail of defined small molecules with self-replicable mRNA delivery. The established iNSCs were completely transgene-free in their cytosol and genome and further resembled human embryonic stem cell-derived NSCs in the morphology, biological characteristics, global gene expression, and potential to differentiate into functional neurons, astrocytes, and oligodendrocytes. Moreover, iNSC colonies were observed within eight days under optimized conditions, and no teratomas formed in vivo, implying the absence of pluripotent cells. This study proposes an approach to generate transplantable iNSCs that can be broadly applied for neurological disorders in a safe, efficient, and patient-specific manner.


Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Neural Stem Cells/cytology , RNA, Messenger/metabolism , Urine/cytology , Adult , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neural Stem Cells/metabolism , RNA, Messenger/genetics , Transgenes
13.
Exp Mol Med ; 51(7): 1-15, 2019 07 04.
Article En | MEDLINE | ID: mdl-31273189

Alopecia, one of the most common chronic diseases, can seriously affect a patient's psychosocial life. Dermal papilla (DP) cells serve as essential signaling centers in the regulation of hair growth and regeneration and are associated with crosstalk between autocrine/paracrine factors and the surrounding environment. We previously demonstrated that amniotic fluid-derived mesenchymal stem cell-conditioned medium (AF-MSC-CM) accelerates hair regeneration and growth. The present study describes the effects of overexpression of a reprogramming factor, Nanog, on MSC properties, the paracrine effects on DP cells, and in vivo hair regrowth. First, we examined the in vitro proliferation and lifespan of AF-MSCs overexpressing reprogramming factors, including Oct4, Nanog, and Lin28, alone or in combination. Among these factors, Nanog was identified as a key factor in maintaining the self-renewal capability of AF-MSCs by delaying cellular senescence, increasing the endogenous expression of Oct4 and Sox2, and preserving stemness. Next, we evaluated the paracrine effects of AF-MSCs overexpressing Nanog (AF-N-MSCs) by monitoring secretory molecules related to hair regeneration and growth (IGF, PDGF, bFGF, and Wnt7a) and proliferation of DP cells. In vivo studies revealed that CM derived from AF-N-MSCs (AF-N-CM) accelerated the telogen-to-anagen transition in hair follicles (HFs) and increased HF density. The expression of DP and HF stem cell markers and genes related to hair induction were higher in AF-N-CM than in CM from AF-MSCs (AF-CM). This study suggests that the secretome from autologous MSCs overexpressing Nanog could be an excellent candidate as a powerful anagen inducer and hair growth stimulator for the treatment of alopecia.


Alopecia/therapy , Hair Follicle/physiology , Nanog Homeobox Protein/metabolism , Regeneration , Alopecia/pathology , Amniotic Fluid/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cellular Senescence , Dermis/metabolism , Female , Gene Expression , Hair/physiology , Humans , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
14.
Stem Cells Dev ; 28(10): 633-648, 2019 05 15.
Article En | MEDLINE | ID: mdl-30880587

Engraftment of oligodendrocyte progenitor cells (OPCs), which form myelinating oligodendrocytes, has the potential to treat demyelinating diseases such as multiple sclerosis. However, conventional strategies for generating oligodendrocytes have mainly focused on direct differentiation into forebrain- or spinal cord-restricted oligodendrocytes without establishing or amplifying stem/progenitor cells. Taking advantage of a recently established culture system, we generated expandable EN1- and GBX2-positive glial-restricted progenitor-like cells (GPLCs) near the anterior hindbrain. These cells expressed PDGFRα, CD9, S100ß, and SOX10 and mostly differentiated into GFAP-positive astrocytes and MBP-positive oligodendrocytes. RNA-seq analysis revealed that the transcriptome of GPLCs was similar to that of O4-positive OPCs, but distinct from that of rosette-type neural stem cells. Notably, engrafted GPLCs not only differentiated into GFAP-positive astrocytes but also myelinated the brains of adult shiverer mice 8 weeks after transplantation. Our strategy for establishing anterior hindbrain-specific GPLCs with gliogenic potency will facilitate their use in the treatment of demyelinating diseases and studies of the molecular mechanisms underlying glial development in the hindbrain.


Astrocytes/cytology , Demyelinating Diseases/therapy , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/transplantation , Oligodendroglia/cytology , Pluripotent Stem Cells/cytology , Animals , Astrocytes/metabolism , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Transgenic , Oligodendroglia/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Rhombencephalon/cytology , S100 Calcium Binding Protein beta Subunit/metabolism , SOXE Transcription Factors/metabolism , Tetraspanin 29/metabolism
15.
Metab Eng ; 53: 35-47, 2019 05.
Article En | MEDLINE | ID: mdl-30779965

Reprogramming of 'adult' differentiated somatic cells to 'embryonic' pluripotent stem cells accompanied by increased rate of glycolysis. Conversely, glycolysis triggers accumulation of advanced glycation end products (AGEs), a potential causative factor in aging, by promoting methylglyoxal production. Therefore, it is reasonable that pluripotent stem cells (PSCs) would specifically regulate glycolysis to maintain their embryonic features. In this study, we focused on glycine decarboxylase (GLDC), a key enzyme in the glycine cleavage system that regulates glycolysis and methylglyoxal production in cancer. GLDC was exclusively expressed in PSCs, and inhibition of this enzyme induced alterations of metabolome and AGE accumulation, thereby suppressing the embryonic pluripotent state. Surprisingly, the level of accumulated AGEs in somatic cells gradually decreased during reprogramming, ultimately disappearing in iPSCs. In addition, ectopic expression of GLDC or treatment with the AGE inhibitor LR-90 promoted reprogramming. Together, these findings suggest that GLDC-mediated regulation of glycolysis and controlling AGE accumulation is related to maintenance and induction of pluripotency.


Cellular Reprogramming , Gene Expression Regulation, Enzymologic , Glycation End Products, Advanced/metabolism , Glycine Dehydrogenase (Decarboxylating)/biosynthesis , Glycolysis , Induced Pluripotent Stem Cells/enzymology , Butyrates/pharmacology , Cell Line , Glycation End Products, Advanced/genetics , Glycine/genetics , Glycine/metabolism , Glycine Dehydrogenase (Decarboxylating)/genetics , Humans , Induced Pluripotent Stem Cells/cytology
16.
Biochem Biophys Res Commun ; 507(1-4): 236-241, 2018 12 09.
Article En | MEDLINE | ID: mdl-30414673

Control of adipogenesis in mesenchymal stem cells (MSCs) offers enormous potential for management of obesity- and aging-related diseases. Celastrol, the traditional Chinese medicine extracted from Tripterygium wilfordi, exhibits anti-obesity effects in in vitro and in vivo murine models. This study describes how celastrol affects multilineage differentiation potential of human adipose-derived stem cells (hADSCs). We performed in vitro adipogenic differentiation of hADSCs and investigated how celastrol-induced lipid accumulation and expression of adipocyte differentiation markers varied with dose, duration, and donor age. In addition, we assessed the effect of celastrol on osteogenic and chondrogenic differentiation of hADSCs. During adipogenic induction of hADSCs, the inhibitory effect of celastrol on lipid accumulation and adipogenesis depended on dose, duration, time of administration, and individual donor. Inhibition was mediated by proliferator-activated receptor-γ (PPARG) and CCAAT/enhancer-binding protein alpha (CEBPA). Celastrol also suppressed differentiation of hADSCs into the osteogenic and chondrogenic lineages. Celastrol plays a regulatory role in multilineage differentiation of human MSCs. Our findings provide important insights regarding management of obesity and stem cell therapy.


Adipogenesis/drug effects , Adipose Tissue/cytology , Stem Cells/cytology , Triterpenes/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Aged , Aged, 80 and over , Aging/genetics , Biomarkers/metabolism , Gene Expression Regulation/drug effects , Humans , Middle Aged , Pentacyclic Triterpenes , Stem Cells/drug effects , Stem Cells/metabolism , Triterpenes/administration & dosage , Young Adult
17.
Stem Cell Res Ther ; 9(1): 293, 2018 11 08.
Article En | MEDLINE | ID: mdl-30409167

BACKGROUND: Mesenchymal stem cell-derived conditioned medium (MSC-CM) has emerged as a promising cell-free tool for restoring degenerative diseases and treating traumatic injuries. The present study describes the effect of selenium as a reactive oxygen species (ROS) scavenger and its additive effect with basic fibroblast growth factor (bFGF) on in vitro expansion of amniotic fluid (AF)-MSCs and the paracrine actions of AF-MSC-CM as well as the associated cellular and molecular mechanisms. METHODS: In this study, we obtained CM from human AF-MSCs cultured with selenium. The stemness of selenium-treated AF-MSCs was evaluated by cell growth and differentiation potential. Human fibroblasts were treated with AF-MSC-CM and analyzed for cell signaling changes. For in vivo wound healing assay, ICR mice with a full-thickness skin wound were used. RESULTS: Selenium played a critical role in in vitro expansion of AF-MSCs through activation of the AKT-ERK1/2, Smad2, and Stat3 signaling pathways along with inactivation of GSK3ß. When administered together with bFGF, it showed remarkable effect in inhibiting ROS accumulation and preserving their multipotency. Proliferation and migration of human dermal fibroblasts and in vivo wound healing were improved in the CMs derived from AF-MSCs exposed to selenium and bFGF, which was caused by the Smad2, AKT-MEK1/2-ERK, and NFκB signaling triggered by the paracrine factors of AF-MSCs, such as TGF-ß, VEGF, and IL-6. Our results suggest the following: (a) supplementation of selenium in AF-MSC culture contributes to in vitro expansion and preservation of multipotency, (b) ROS accumulation causes progressive losses in proliferative and differentiation potential, (c) the separate activities of bFGF and selenium in MSCs exert an additive effect when used together, and (d) the additive combination improves the therapeutic effects of AF-MSC-derived CMs on tissue repair and regeneration. CONCLUSION: Antioxidants, such as selenium, should be considered as an essential supplement for eliciting the paracrine effects of MSC-CMs.


Amniotic Fluid/cytology , Fibroblast Growth Factor 2/pharmacology , Mesenchymal Stem Cells/cytology , Paracrine Communication , Selenium/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Intracellular Space/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred ICR , Models, Biological , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Paracrine Communication/drug effects , Phenotype , Reactive Oxygen Species/metabolism
18.
Drug Deliv Transl Res ; 8(3): 857-862, 2018 06.
Article En | MEDLINE | ID: mdl-29372538

The ability of glucagon-like peptide-1 analogs to enhance glucose-dependent insulin secretion and to inhibit ß cell apoptosis could be of potential benefit for islet transplantation. In this study, we investigated the effect of sustained local delivery of exenatide, a synthetic exendin-4, on the in vitro viability and function of encapsulated porcine islets. Prior to encapsulation, we fabricated exenatide-loaded poly(latic-co-glycolic acid) microspheres, and investigated their release behavior with different initial drug-loading amounts. Exenatide-loaded microspheres, exhibiting a sustained release over 21 days, were subsequently chosen and co-encapsulated with porcine islets in alginate microcapsules. During the 21-day period, the islets co-encapsulated with the exenatide-loaded microspheres exhibited improved survival and glucose-stimulated insulin secretion, compared to those without. This suggested that the intracapsular sustained delivery of exenatide via microspheres could be a promising strategy for improving survival and function of microencapsulated porcine islets for islet xenotransplantation.


Alginates/administration & dosage , Hypoglycemic Agents/administration & dosage , Islets of Langerhans Transplantation , Islets of Langerhans/chemistry , Microspheres , Peptides/administration & dosage , Venoms/administration & dosage , Alginates/chemistry , Animals , Cell Survival/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Drug Liberation , Exenatide , Glucuronic Acid/administration & dosage , Glucuronic Acid/chemistry , Hexuronic Acids/administration & dosage , Hexuronic Acids/chemistry , Hypoglycemic Agents/chemistry , Islets of Langerhans/drug effects , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Peptides/chemistry , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Swine , Venoms/chemistry
19.
J Microencapsul ; 34(1): 57-62, 2017 Feb.
Article En | MEDLINE | ID: mdl-28097929

In this study, we investigated the effect of intracapsular environment on the survival of anchorage-dependent cells (ADCs) encapsulated in alginate microcapsules with three different core structures, i.e. liquid, semi-liquid and microsphere-encapsulating semi-liquid core, using NIH 3T3 fibroblasts as an ADC model. For the latter, we fabricated poly (ɛ-caprolactone) microspheres and co-encapsulated them with the cells, to establish cell-substrate interactions in the capsule. The fibroblast cells co-encapsulated with the microspheres exhibited higher survival and growth than those without. This study provides a "proof of concept" for employing microspheres as a cell-friendly surface to establish intracapsular cell-substrate interactions thus prolonging the survival of encapsulated therapeutic ADCs.


Alginates/chemistry , Fibroblasts/cytology , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Polyesters/chemistry , Animals , Capsules , Cell Survival , Cells, Immobilized/cytology , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Mice , Microspheres , NIH 3T3 Cells
20.
Nanomedicine ; 11(6): 1407-16, 2015 Aug.
Article En | MEDLINE | ID: mdl-25819884

Monodisperse spherical silica nanoparticles (SNPs) with diameters of 20-200 nm were employed to study size, dose, and cell-type dependent cytotoxicity in A549 and HepG2 epithelial cells and NIH/3T3 fibroblasts. These uniform SNPs of precisely controlled sizes eliminated uncertainties arising from mixed sizes, and uniquely allowed the probing of effects entirely size-dependent. Cell viability, membrane disruption, oxidative stress, and cellular uptake were studied. The extent and mechanism of SNP cytotoxicity were found to be not only size and dose dependent, but also highly cell type dependent. Furthermore, the 60 nm SNPs exhibited highly unusual behavior in comparison to particles of other sizes tested, implying interesting possibilities for controlling cellular activities using nanoparticles. Specifically, the 60 nm SNPs were preferentially endocytosed by cells and, at high doses, caused a disproportionate decrease in cell viability. The present work may help elucidate certain contradictions among existing results on nanoparticle-induced cytotoxicity. FROM THE CLINICAL EDITOR: Silica nanoparticles are being investigated in many research areas for their use in clinical applications. Nonetheless, the relationship between particle size and potential toxicity remains to be elucidated. In this article, the authors studied the biological effects of spherical SNPs with precise diameters between 20 and 200 nm on three different cell types and their results should provide more data on safety for better drug design.


Nanoparticles/toxicity , Silicon Dioxide/toxicity , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Mice , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Particle Size , Silicon Dioxide/chemistry
...