Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
J Korean Med Sci ; 37(31): e244, 2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35942557

ABSTRACT

BACKGROUND: To deliver therapeutics into the brain, it is imperative to overcome the issue of the blood-brain-barrier (BBB). One of the ways to circumvent the BBB is to administer therapeutics directly into the brain parenchyma. To enhance the treatment efficacy for chronic neurodegenerative disorders, repeated administration to the target location is required. However, this increases the number of operations that must be performed. In this study, we developed the IntraBrain Injector (IBI), a new implantable device to repeatedly deliver therapeutics into the brain parenchyma. METHODS: We designed and fabricated IBI with medical grade materials, and evaluated the efficacy and safety of IBI in 9 beagles. The trajectory of IBI to the hippocampus was simulated prior to surgery and the device was implanted using 3D-printed adaptor and surgical guides. Ferumoxytol-labeled mesenchymal stem cells (MSCs) were injected into the hippocampus via IBI, and magnetic resonance images were taken before and after the administration to analyze the accuracy of repeated injection. RESULTS: We compared the planned vs. insertion trajectory of IBI to the hippocampus. With a similarity of 0.990 ± 0.001 (mean ± standard deviation), precise targeting of IBI was confirmed by comparing planned vs. insertion trajectories of IBI. Multiple administrations of ferumoxytol-labeled MSCs into the hippocampus using IBI were both feasible and successful (success rate of 76.7%). Safety of initial IBI implantation, repeated administration of therapeutics, and long-term implantation have all been evaluated in this study. CONCLUSION: Precise and repeated delivery of therapeutics into the brain parenchyma can be done without performing additional surgeries via IBI implantation.


Subject(s)
Ferrosoferric Oxide , Mesenchymal Stem Cells , Animals , Brain/diagnostic imaging , Brain/surgery , Dogs , Humans , Imaging, Three-Dimensional , Magnetic Resonance Imaging/methods
2.
Bioengineering (Basel) ; 9(4)2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35447737

ABSTRACT

Mesenchymal stem cells (MSCs) are one of the most extensively studied stem cell types owing to their capacity for differentiation into multiple lineages as well as their ability to secrete regenerative factors and modulate immune functions. However, issues remain regarding their further application for cell therapy. Here, to demonstrate the superiority of the improvement of MSCs, we divided umbilical cord blood-derived MSCs (UCB-MSCs) from 15 donors into two groups based on efficacy and revealed donor-dependent variations in the anti-inflammatory effect of MSCs on macrophages as well as their immunoregulatory effect on T cells. Through surface marker analyses (242 antibodies), we found that HLA-A2 was positively related to the anti-inflammatory and immunoregulatory function of MSCs. Additionally, HLA-A2 mRNA silencing in MSCs attenuated their therapeutic effects in vitro; namely, the suppression of LPS-stimulated macrophages and phytohemagglutinin-stimulated T cells. Moreover, HLA-A2 silencing in MSCs significantly decreased their therapeutic effects in a rat model of hyperoxic lung damage. The present study provides novel insights into the quality control of donor-derived MSCs for the treatment of inflammatory conditions and diseases.

3.
Cells ; 10(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34572070

ABSTRACT

Mesenchymal stem cells (MSCs) are accessible, abundantly available, and capable of regenerating; they have the potential to be developed as therapeutic agents for diseases. However, concerns remain in their further application. In this study, we developed a SMall cell+Ultra Potent+Scale UP cell (SMUP-Cell) platform to improve whole-cell processing, including manufacturing bioreactors and xeno-free solutions for commercialization. To confirm the superiority of SMUP-Cell improvements, we demonstrated that a molecule secreted by SMUP-Cells is capable of polarizing inflammatory macrophages (M1) into their anti-inflammatory phenotype (M2) at the site of injury in a pain-associated osteoarthritis (OA) model. Lipopolysaccharide-stimulated macrophages co-cultured with SMUP-Cells expressed low levels of M1-phenotype markers (CD11b, tumor necrosis factor-α, interleukin-1α, and interleukin-6), but high levels of M2 markers (CD163 and arginase-1). To identify the paracrine action underlying the anti-inflammatory effect of SMUP-Cells, we employed a cytokine array and detected increased levels of pentraxin-related protein-3 (PTX-3). Additionally, PTX-3 mRNA silencing was applied to confirm PTX-3 function. PTX-3 silencing in SMUP-Cells significantly decreased their therapeutic effects against monosodium iodoacetate (MIA)-induced OA. Thus, PTX-3 expression in injected SMUP-Cells, applied as a therapeutic strategy, reduced pain in an OA model.


Subject(s)
C-Reactive Protein/metabolism , Macrophages/immunology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Osteoarthritis/therapy , Osteocytes/cytology , Pain/prevention & control , Serum Amyloid P-Component/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Injections, Intra-Articular , Iodoacetic Acid/toxicity , Macrophage Activation/immunology , Male , Osteoarthritis/chemically induced , Osteoarthritis/metabolism , Osteoarthritis/pathology , Pain/etiology , Pain/metabolism , Pain/pathology , Rats , Rats, Sprague-Dawley
4.
Stem Cells Int ; 2021: 5582792, 2021.
Article in English | MEDLINE | ID: mdl-33936211

ABSTRACT

Autophagy plays a critical role in stem cell maintenance and is related to cell growth and cellular senescence. It is important to find a quality-control marker for predicting senescent cells. This study verified that CD47 could be a candidate to select efficient mesenchymal stem cells (MSCs) to enhance the therapeutic effects of stem cell therapy by analyzing the antibody surface array. CD47 expression was significantly decreased during the expansion of MSCs in vitro (p < 0.01), with decreased CD47 expression correlated with accelerated senescence phenotype, which affected cell growth. UCB-MSCs transfected with CD47 siRNA significantly triggered the downregulation of pRB and upregulation of pp38, which are senescence-related markers. Additionally, autophagy-related markers, ATG5, ATG12, Beclin1, and LC3B, revealed significant downregulation with CD47 siRNA transfection. Furthermore, autophagy flux following treatment with an autophagy inducer, rapamycin, has shown that CD47 is a key player in autophagy and senescence to maintain and regulate the growth of MSCs, suggesting that CD47 may be a critical key marker for the selection of effective stem cells in cell therapy.

5.
J Clin Med ; 10(2)2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33466868

ABSTRACT

Umbilical cord blood (UCB) is used as a source of donor cells for hematopoietic stem cell (HSC) transplantation. The success of transplantation is dependent on the quality of cord blood (CB) units for maximizing the chance of engraftment. Improved outcomes following transplantation are associated with certain factors of cryopreserved CB units: total volume and total nucleated cell (TNC) count, mononuclear cell (MNC) count, and CD34+ cell count. The role of the storage period of CB units in determining the viability and counts of cells is less clear and is related to the quality of cryopreserved CB units. Herein, we demonstrate the recovery of viable TNCs and CD34+ cells, as well as the MNC viability in 20-year-old cryopreserved CB units in a CB bank (MEDIPOST Co., Ltd., Seongnam-si, Gyeonggi-do, Korea). In addition, cell populations in CB units were evaluated for future clinical applications. The stable recovery rate of the viability of cryopreserved CB that had been stored for up to 20 years suggested the possibility of uses of the long-term cryopreservation of CB units. Similar relationships were observed in the recovery of TNCs and CD34+ cells in units of cryopreserved and fresh CB. The high-viability recovery of long-term cryopreserved CB suggests that successful hematopoietic stem cell (HSC) transplantation and other clinical applications, which are suitable for treating incurable diseases, may be performed regardless of long-term storage.

6.
Cells ; 10(1)2021 01 03.
Article in English | MEDLINE | ID: mdl-33401590

ABSTRACT

In order to provide a sufficient number of cells for clinical use, mesenchymal stem cells (MSCs) must be cultured for long-term expansion, which inevitably triggers cellular senescence. Although the small size of MSCs is known as a critical determinant of their fate, the main regulators of stem cell senescence and the underlying signaling have not been addressed. Umbilical cord blood-derived MSCs (UCB-MSCs) were obtained using size-isolation methods and then cultured with control or small cells to investigate the major factors that modulate MSC senescence. Cytokine array data suggested that the secretion of interukin-8 (IL-8) or growth-regulated oncogene-alpha (GROa) by senescent cells was markedly inhibited during incubation of small cells along with suppression of cognate receptor (C-X-C motif chemokine receptor2, CXCR2) via blockade of the autocrine/paracrine positive loop. Moreover, signaling via toll-like receptor 2 (TLR2) and TLR5, both pattern recognition receptors, drove cellular senescence of MSCs, but was inhibited in small cells. The activation of TLRs (2 and 5) through ligand treatment induced a senescent phenotype in small cells. Collectively, our data suggest that small cell from UCB-MSCs exhibit delayed cellular senescence by inhibiting the process of TLR signaling-mediated senescence-associated secretory phenotype (SASP) activation.


Subject(s)
Cell Size , Cellular Senescence , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 5/metabolism , Autocrine Communication , Chemokine CXCL1/metabolism , Fetal Blood/cytology , Humans , Infant, Newborn , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Phenotype , Receptors, Interleukin-8B/metabolism
7.
Stem Cells Int ; 2020: 8878412, 2020.
Article in English | MEDLINE | ID: mdl-32733573

ABSTRACT

The formation of neurofibrillary tangles has been implicated as an important pathological marker for Alzheimer's disease (AD). Studies have revealed that the inhibition of abnormal hyperphosphorylation and aggregation of tau in the AD brain might serve as an important drug target. Using in vitro and in vivo experimental models, such as the AD mouse model (5xFAD mice), we investigated the inhibition of hyperphosphorylation of tau using the human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs). Administration of hUCB-MSCs not only ameliorated the spatial learning and memory impairments but also mitigated the hyperphosphorylation of tau in 5xFAD mice. Furthermore, in vivo experiments in mice and in vitro ThT fluorescence assay validated galectin-3 (GAL-3) as an essential factor of hUCB-MSC. Moreover, GAL-3 was observed to be involved in the removal of aberrant forms of tau, by reducing hyperphosphorylation through decrements in the glycogen synthase kinase 3 beta (GSK-3ß). Our results confirm that GAL-3, secreted by hUCB-MSC, regulates the abnormal accumulation of tau by protein-protein interactions. This study suggests that hUCB-MSCs mitigate hyperphosphorylation of tau through GAL-3 secretion. These findings highlight the potential role of hUCB-MSCs as a therapeutic agent for aberrant tau in AD.

8.
Cells ; 9(6)2020 05 28.
Article in English | MEDLINE | ID: mdl-32481584

ABSTRACT

Conventional therapeutic applications of mesenchymal stromal cells (MSCs) focus on cell replacement and differentiation; however, increasing evidence suggests that most of their therapeutic effects are carried out by their various secretions. This study investigated the application of conditioned medium (CM) from human umbilical cord blood-derived MSCs (hUCB-MSCs) to improve hair growth and developed a method to reliably produce this optimized CM. Primed MSC-derived CM (P-CM) with combinations of TGF-ß1 and LiCl was optimized by comparing its effects on the cell viability of dermal papilla cells (DPCs). P-CM significantly increased the viability of DPCs compared to CM. The secretion of vascular endothelial growth factor (VEGF) in DPCs was regulated by the macrophage migration inhibitory factor (MIF) in the P-CM secreted by MSCs. These findings suggest that P-CM can improve the efficacy in hair growth via a paracrine mechanism and that MIF in P-CM exerts hair growth-promoting effects via a VEGF-related ß-catenin and p-GSK-3ß [SER9] signaling pathway. Furthermore, clinical trials have shown that 5% P-CM improved androgenetic alopecia through producing an increased hair density, thickness, and growth rate, suggesting that this topical agent may be a novel and effective treatment option for patients with androgenetic alopecia.


Subject(s)
Culture Media, Conditioned/chemistry , Fetal Blood/cytology , Hair/growth & development , Macrophage Migration-Inhibitory Factors/pharmacology , Mesenchymal Stem Cells/cytology , Adult , Alopecia/pathology , Cell Survival/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , Glycogen Synthase Kinase 3 beta/metabolism , Hair/cytology , Hair/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Lithium Chloride/pharmacology , Mesenchymal Stem Cells/drug effects , Middle Aged , Models, Biological , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Young Adult , beta Catenin/metabolism
9.
Stem Cells Int ; 2020: 1802976, 2020.
Article in English | MEDLINE | ID: mdl-32399038

ABSTRACT

Therapeutic treatment of various inflammation-related diseases using mesenchymal stem cells (MSCs) has increased in recent years because of the paracrine action of these cells but shows several limitations. First, MSC-based therapies exhibit varying efficacies; thus, biomarkers should be determined to identify who may benefit from these candidate therapeutic agents. Second, the mechanism underlying the therapeutic effects is poorly understood. To evaluate the effects of human umbilical cord blood-derived MSCs (UCB-MSCs) on macrophages, the macrophage cell line NR8383 stimulated with lipopolysaccharide (LPS) was cocultured by UCB-MSCs. We found that UCB-MSCs mediated changes in macrophage polarization towards M2 from M1 macrophages. To identify the paracrine action underlying the anti-inflammation effect of UCB-MSCs, the secretion of UCB-MSCs exposed to LPS-stimulated NR8383 cells was tested using a biotin label-based 507 antibody array. Among the secreted proteins, we selected pentraxin-related protein PTX3/tumor necrosis factor-inducible gene 14 protein (PTX3) to investigate its association with UCB-MSCs in macrophage polarization. We found that human PTX3 was secreted from UCB-MSCs under inflammation condition and reinforced the M2 macrophage marker via the Dectin-1 receptor by activating MSK1/2 phosphorylation signaling in NR8383 cells. Accordingly, knockdown of PTX3 in UCB-MSCs significantly attenuated their therapeutic effects in a neonatal hyperoxic lung injury resulting in reduced survival, lung alveolarization, M2 marker expression, Dectin-1 levels, anti-inflammatory cytokines, and improved M1 marker expression and inflammatory cytokines compared to control MSC-injected rats. UCB-MSCs show therapeutic potential by controlling macrophage polarization. Interestingly, higher PTX3 levels in UCB-MSCs induced greater improvement in the therapeutic effects than lower PTX3 levels. Collectively, PTX3 is a potential marker with critical paracrine effects for predicting the therapeutic potential of MSC therapy in inflammatory diseases; quality control assessments using PTX3 may be useful for improving the therapeutic effects of UCB-MSCs.

10.
Stem Cells Int ; 2020: 5924983, 2020.
Article in English | MEDLINE | ID: mdl-32399043

ABSTRACT

Mesenchymal stem cells (MSCs) represent a promising means to promote tissue regeneration. However, the heterogeneity of MSCs impedes their use for regenerative medicine. Further investigation of this phenotype is required to develop cell therapies with improved clinical efficacy. Here, a small-sized population of human umbilical cord blood-derived MSCs (UCB-MSCs) was isolated using a filter and centrifuge system to analyze its stem cell characteristics. Consequently, this population showed higher cell growth and lower senescence. Additionally, it exhibited diverse stem cell properties including differentiation, stemness, and adhesion, as compared to those of the population before isolation. Using cell surface protein array or sorting analysis, both EGFR and CD49f were identified as markers associated with the small-sized population. Accordingly, suppression of these surface proteins abolished the superior characteristics of this population. Moreover, compared to that with large or nonisolated populations, the small-sized population showed greater therapeutic efficacy by promoting the engraftment potential of infused cells and reducing lung damage in an emphysema mouse model. Therefore, the isolation of this small-sized population of UCB-MSCs could be a simple and effective way to enhance the efficacy of cell therapy.

11.
Tissue Eng Regen Med ; 17(2): 193-202, 2020 04.
Article in English | MEDLINE | ID: mdl-32008170

ABSTRACT

BACKGROUND: Regeneration of soft tissue defects is essential for adipose tissue pathologies and disease, trauma, or injury-induced damage. Here, we show that umbilical cord blood-derived mesenchymal stem cells could potentially be tailored and used for the reconstruction of specific damaged sites. Adipogenesis can be exploited in soft tissue reconstruction. Also, primary cilia play a role in the control of adipogenesis. METHODS: The adipogenic differentiation capacity of mesenchymal stem cells (MSCs) was shown to influence ciliogenesis. MSCs transfected with intraflagellar transport 88 (IFT88) small interfering RNA (siRNA), which blocks the assembly and maintenance of cilia, were examined to confirm the relationship between adipogenesis and ciliogenesis. Also, 1,2-Bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA-AM), calcium chelator, inhibited the ciliogenesis of MSCs in adipogenic differentiation. RESULTS: IFT88-knockdown led to decreased cilia formation and limitation of cilia elongation in adipogenesis. Additionally, intracellular calcium triggered cilia formation in MSCs adipogenesis. Interestingly, intracellular calcium cannot overcome the inhibition of adipogenesis caused by low numbers of cilia in MSCs. CONCLUSION: Our data suggested that ciliogenesis was negatively regulated by Wnt5a/ß-catenin signaling during adipogenesis. Thus, we suggest that calcium induction triggers adipogenesis and ciliogenesis.


Subject(s)
Adipogenesis/physiology , Calcium/metabolism , Cell Differentiation/physiology , Cilia/metabolism , Fetal Blood/metabolism , Wnt-5a Protein/metabolism , beta Catenin/metabolism , Adipose Tissue , Cell Differentiation/genetics , Humans , Mesenchymal Stem Cells , RNA, Small Interfering
12.
Int J Mol Med ; 45(2): 556-568, 2020 02.
Article in English | MEDLINE | ID: mdl-31894311

ABSTRACT

Alopecia is a common and distressing condition, and developing new therapeutic agents to prevent hair loss is important. Human umbilical cord blood­derived mesenchymal stem cells (hUCB­MSCs) have been studied intensively in regenerative medicine. However, the therapeutic potential of these cells against hair loss and hair organ damage remains unclear, and the effects of hUCB­MSC transplantation on hair loss require evaluation. The current study aimed to investigate the effects of hUCB­MSCs on hair regression in vivo and restoration of anagen conduction on hair growth in vitro. The effects of hUCB­MSCs were explored in mouse catagen induction models using a topical treatment of 0.1% dexamethasone to induce hair regression. Dexamethasone was also used to simulate a stress environment in vitro. The results demonstrated that hUCB­MSCs significantly prevented hair regression induced by dexamethasone topical stimulation in vivo. Additionally, hUCB­MSCs significantly increased the proliferation of human dermal papilla cells (hDPCs) and HaCaT cells, which are key constituent cells of the hair follicle. Stimulation of vascular endothelial growth factor secretion and decreased expression of DKK­1 by hUCB­MSCs were also observed in hDPCs. Restoration of cell viability by hUCB­MSCs suggested that these cells exerted a protective effect on glucocorticoid stress­associated hair loss. In addition, anti­apoptotic effects and regulation of the autophagic flux recovery were observed in HaCaT cells. The results of the present study indicated that hUCB­MSCs may have the capacity to protect hair follicular dermal papilla cells and keratinocytes, thus preventing hair loss. Additionally, the protective effects of hUCB­MSCs may be resistant to dysregulation of autophagy under harmful stress.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Dexamethasone/adverse effects , Hair Follicle/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Apoptosis/drug effects , Cell Line , Cells, Cultured , Female , Fetal Blood/cytology , Hair/cytology , Hair/drug effects , Hair/growth & development , Hair/ultrastructure , Hair Follicle/drug effects , Hair Follicle/ultrastructure , Humans , Mice, Inbred C57BL
13.
Antioxidants (Basel) ; 9(1)2020 Jan 11.
Article in English | MEDLINE | ID: mdl-31940867

ABSTRACT

Umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) are accessible, available in abundance, and have been shown to be a promising source that can regenerate cartilage in patients with osteoarthritis or other orthopedic diseases. Recently, a three-dimensional (3D) cell culture system was developed to mimic the naive tissue microenvironment. However, the efficacy of cells generated from the 3D spheroid culture system has not yet been elucidated. In the present study, we demonstrate the changes in superoxide dismutase 2 (SOD2) gene expression, an indicator of oxidative stress, on 3D spheroid MSCs. Moreover, siRNA transfection and neutralizing antibody investigations were performed to confirm the function of SOD2 and E-cadherin. Overall, we found that SOD2 siRNA transfection in the spheroid form of MSCs increases the expression of apoptotic genes and decreases the clearance of mitochondrial reactive oxygen species (ROS). As a result, we confirm that 3D spheroid formation increases E-cadherin and SOD2 expression, ultimately regulating the phosphoinositide 3-kinase (PI3K/pAkt/pNrf2 and pERK/pNrf2 signaling pathway. Additionally, we show that SOD2 expression on 3D spheroid MSCs affects the regeneration rates of destructive cartilage in an osteoarthritic model. We postulate that the impact of SOD2 expression on 3D spheroid MSCs reduces oxidative stress and apoptosis, and also promotes cartilage regeneration.

14.
World J Stem Cells ; 12(12): 1511-1528, 2020 Dec 26.
Article in English | MEDLINE | ID: mdl-33505598

ABSTRACT

Umbilical cord blood (UCB) is a primitive and abundant source of mesenchymal stem cells (MSCs). UCB-derived MSCs have a broad and efficient therapeutic capacity to treat various diseases and disorders. Despite the high latent self-renewal and differentiation capacity of these cells, the safety, efficacy, and yield of MSCs expanded for ex vivo clinical applications remains a concern. However, immunomodulatory effects have emerged in various disease models, exhibiting specific mechanisms of action, such as cell migration and homing, angiogenesis, anti-apoptosis, proliferation, anti-cancer, anti-fibrosis, anti-inflammation and tissue regeneration. Herein, we review the current literature pertaining to the UCB-derived MSC application as potential treatment strategies, and discuss the concerns regarding the safety and mass production issues in future applications.

15.
Cartilage ; 11(3): 338-347, 2020 07.
Article in English | MEDLINE | ID: mdl-30079757

ABSTRACT

OBJECTIVE: The objective of this study was to describe in life methods by which osteoarthritis can be staged in order to time therapeutic interventions that are relevant to osteoarthritis (OA) clinical trials. METHODS: Twenty-two sheep underwent arthroscopic meniscal destabilization to induce OA. Serial computed tomography (CT) imaging and arthroscopy were used to monitor osteoarthritis progression at 3-month intervals over 9 months. Eleven sheep received 1 intra-articular injection of hyaluronate 3 months after OA induction and another group of 11 received saline. A linear mixed model was used to define the trajectory of shape change in the medial joint compartment. Ordinal logistic regression was used to investigate the association between morphological changes and sclerosis. RESULTS: Three months after meniscal destabilization there were early bipolar chondral lesions in the medial compartment of the knee, as well as osteophytes and bone remodeling. Superficial fissures and cartilage cracks progressed to discrete areas of cartilage thinning and fibrillation on the medial tibial plateau by 6 months that became cartilage erosions by nine months. A linear mixed effect model demonstrated significant change in medial compartment length and width with over time (P < 0.05) for both groups. A significant association between severity of sclerosis and medial compartment morphology was also observed. CONCLUSIONS: The induction of osteoarthritic lesions with meniscal release model can be followed using noninvasive and minimally invasive procedures allowing for real-time decisions about redosing therapies, or other changes such as extending trial timelines without sacrificing animals to conduct assessments.


Subject(s)
Arthroscopy/methods , Disease Models, Animal , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/pathology , Tomography, X-Ray Computed/methods , Animals , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Disease Progression , Knee Joint/diagnostic imaging , Knee Joint/pathology , Menisci, Tibial , Osteoarthritis, Knee/etiology , Sheep , Tibia/diagnostic imaging , Tibia/pathology
16.
Int J Mol Sci ; 20(19)2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31569732

ABSTRACT

Bronchopulmonary dysplasia (BPD), caused by hyperoxia in newborns and infants, results in lung damage and abnormal pulmonary function. However, the current treatments for BPD are steroidal and pharmacological therapies, which cause neurodevelopmental impairment. Treatment with umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) is an efficient alternative approach. To prevent pulmonary inflammation in BPD, this study investigated the hypothesis that a key regulator was secreted by MSCs to polarize inflammatory macrophages into anti-inflammatory macrophages at inflammation sites. Lipopolysaccharide-induced macrophages co-cultured with MSCs secreted low levels of the inflammatory cytokines, IL-8 and IL-6, but high levels of the anti-inflammatory cytokine, IL-10. Silencing decorin in MSCs suppressed the expression of CD44, which mediates anti-inflammatory activity in macrophages. The effects of MSCs were examined in a rat model of hyperoxic lung damage. Macrophage polarization differed depending on the levels of decorin secreted by MSCs. Moreover, intratracheal injection of decorin-silenced MSCs or MSCs secreting low levels of decorin confirmed impaired alveolarization of damaged lung tissues by down-regulation of decorin. In tissues, a decrease in the anti-inflammatory macrophage marker, CD163, was observed via CD44. Thus, we identified decorin as a key paracrine factor, inducing macrophage polarization via CD44, a master immunoregulator in mesenchymal stem cells.


Subject(s)
Decorin/biosynthesis , Fetal Blood/cytology , Hyaluronan Receptors/blood , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Biomarkers , Disease Models, Animal , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Hyperoxia/complications , Lung Injury/diagnosis , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/therapy , Rats
17.
Exp Mol Med ; 51(7): 1-14, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31285429

ABSTRACT

The administration of mesenchymal stem cells (MSCs) was shown to attenuate overt as well as early diabetic nephropathy in rodents, but the underlying mechanism of this beneficial effect is largely unknown. Inflammation and mitochondrial dysfunction are major pathogenic factors in diabetic nephropathy. In this study, we found that the repeated administration of MSCs prevents albuminuria and injury to tubular epithelial cells (TECs), an important element in the progression of diabetic nephropathy, by improving mitochondrial function. The expression of M1 macrophage markers was significantly increased in diabetic kidneys compared with that in control kidneys. Interestingly, the expression of arginase-1 (Arg1), an important M2 macrophage marker, was reduced in diabetic kidneys and increased by MSC treatment. In cultured TECs, conditioned media from lipopolysaccharide-activated macrophages reduced peroxisomal proliferator-activated receptor gamma coactivator 1α (Pgc1a) expression and impaired mitochondrial function. The coculture of macrophages with MSCs increased and decreased the expression of Arg1 and M1 markers, respectively. Treatment with conditioned media from cocultured macrophages prevented activated macrophage-induced mitochondrial dysfunction in TECs. In the absence of MSC coculture, Arg1 overexpression in macrophages reversed Pgc1a suppression in TECs. These observations suggest that MSCs prevent the progression of diabetic nephropathy by reversing mitochondrial dysfunction in TECs via the induction of Arg1 in macrophages.


Subject(s)
Albuminuria/prevention & control , Arginase/metabolism , Diabetes Complications/prevention & control , Diabetic Nephropathies/prevention & control , Mesenchymal Stem Cells/metabolism , Animals , Arginase/genetics , Cell Line , Cord Blood Stem Cell Transplantation , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney/metabolism , Kidney/pathology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mice , Mitochondria/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
18.
Stem Cells Int ; 2018: 4978763, 2018.
Article in English | MEDLINE | ID: mdl-30254683

ABSTRACT

Human mesenchymal stem cells (hMSCs), including human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), which have high proliferation capacity and immunomodulatory properties, are considered to be a good candidate for cell-based therapies. hMSCs show enhanced therapeutic effects via paracrine secretion or cell-to-cell contact that modulates inflammatory or immune reactions. Here, treatment with cobalt chloride (CoCl2) was more effective than naïve hUCB-MSCs in suppressing inflammatory responses in a coculture system with phytohemagglutinin- (PHA-) activated human peripheral blood mononuclear cells (hPBMCs). Furthermore, the effect of CoCl2 is exerted by promoting the expression of anti-inflammatory mediators (e.g., PGE2) and inhibiting that of inflammatory cytokines (e.g., TNF-α and IFN-γ). Treatment of hUCB-MSCs with CoCl2 leads to increased expression of microRNA- (miR-) 146a, which was reported to modulate anti-inflammatory responses. Hypoxia-inducible factor- (HIF-) 1α silencing and ERK inhibition abolished CoCl2-induced miR-146a expression, suggesting that ERK and HIF-1α signals are required for CoCl2-induced miR-146a expression in hUCB-MSCs. These data suggest that treatment with CoCl2 enhances the immunosuppressive capacity of hUCB-MSCs through the ERK-HIF-1α-miR-146a-mediated signaling pathway. Furthermore, pretreatment of transplanted MSCs with CoCl2 can suppress lung inflammation more than naïve MSCs can in a mouse model of asthma. These findings suggest that CoCl2 may improve the therapeutic effects of hUCB-MSCs for the treatment of inflammatory diseases.

19.
Biochem Biophys Res Commun ; 504(4): 933-940, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30224067

ABSTRACT

Alzheimer's disease (AD), which is the most common progressive neurodegenerative disease, causes learning and memory impairment. The pathological progress of AD can derive from imbalanced homeostasis of amyloid beta (Aß) in the brain. In such cases, microglia play important roles in regulating the brain Aß levels. In the present study, we found that human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) can increase, through paracrine action, the ability of microglial cells to clear Aß. In order to identify the associated paracrine factors, a secretome of hUCB-MSCs co-cultured with Aß-treated BV2 microglial cells was analyzed using a human cytokine protein array. As a result, growth differentiation factor-15 (GDF-15) was identified as a predominant candidate, and its association with Aß clearance by microglial cells was investigated in vitro and in a 5XFAD mouse model. When Aß-treated BV2 cells were treated with exogenous recombinant GDF-15, the Aß levels in the culture medium decreased. Moreover, GDF-15 injection in the brain parenchyma of 5XFAD mice also led to decrease in Aß plaques. In contrast, co-culture of BV2 cells and hUCB-MSCs treated with GDF-15-specific siRNA did not influence the Aß levels in the culture medium. To elucidate how these phenomena are related, we confirmed that GDF-15 specifically increases insulin-degrading enzyme (IDE) expression in microglial cells through TGFß receptor type II (TGFßRII), both in vitro and in vivo. These findings suggest that hUCB-MSCs promote the Aß clearance ability of microglial cells through regulation of GDF-15 secretion, thus elucidating a therapeutic mechanism for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Growth Differentiation Factor 15/metabolism , Mesenchymal Stem Cells/metabolism , Alzheimer Disease/pathology , Animals , Coculture Techniques , Disease Models, Animal , Fetal Blood/cytology , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/pharmacology , Humans , Insulysin/metabolism , Mesenchymal Stem Cells/cytology , Mice, Mutant Strains , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Paracrine Communication , Peptide Fragments/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
20.
Korean J Physiol Pharmacol ; 22(5): 555-566, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30181702

ABSTRACT

Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) are used in tissue repair and regeneration; however, the mechanisms involved are not well understood. We investigated the hair growth-promoting effects of hUCB-MSCs treatment to determine whether hUCB-MSCs enhance the promotion of hair growth. Furthermore, we attempted to identify the factors responsible for hair growth. The effects of hUCB-MSCs on hair growth were investigated in vivo, and hUCB-MSCs advanced anagen onset and hair follicle neogeneration. We found that hUCB-MSCs co-culture increased the viability and up-regulated hair induction-related proteins of human dermal papilla cells (hDPCs) in vitro. A growth factor antibody array revealed that secretory factors from hUCB-MSCs are related to hair growth. Insulin-like growth factor binding protein-1 (IGFBP-1) and vascular endothelial growth factor (VEGF) were increased in co-culture medium. Finally, we found that IGFBP-1, through the co-localization of an IGF-1 and IGFBP-1, had positive effects on cell viability; VEGF secretion; expression of alkaline phosphatase (ALP), CD133, and ß-catenin; and formation of hDPCs 3D spheroids. Taken together, these data suggest that hUCB-MSCs promote hair growth via a paracrine mechanism.

SELECTION OF CITATIONS
SEARCH DETAIL
...