Soo Kim’s research while affiliated with Asan Medical Center and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (18)


Extracellular Vesicles from Induced Mesenchymal Stem Cells Inhibit Acute Kidney Injury to Chronic Kidney Disease Transition
  • Article

March 2025

·

8 Reads

International Journal of Stem Cells

·

Sungok Hong

·

Soo Kim

·

Compared with conventional mesenchymal stem cells (MSCs), induced mesenchymal stem cells (iMSCs) from induced pluripotent stem cells are unique cell sources for tissue regeneration. The effect of extracellular vesicles (EVs) secreted from iMSCs on inhibiting acute kidney injury (AKI) to chronic kidney disease (CKD) transition was not reported. In this study, we investigated whether EVs from iMSCs (iMSC-EVs) could inhibit AKI-to-CKD transition. iMSC-EVs exhibited the general characteristics of EVs, such as protein marker expression, morphology, and size. Additionally, iMSC-EVs were detected in renal tissues after intravenous injection. In human renal tubular epithelial cells, the increase in pro-fibrotic gene expression in response to transforming growth factor β1 treatment was decreased by iMSC-EVs. In a mouse model of the AKI-to-CKD transtion induced by folic acid, repeated administration of iMSC-EVs restored renal function at day 14. Specifically, iMSC-EVs reduced interstitial fibrosis, sustained inflammation, various types of cell death, and the number of immune cells infiltrating kidneys. Capillary rarefaction in renal tissue was also reversed by iMSC-EVs. Our results demonstrate that iMSC-EVs reduced interstitial fibrosis, inflammation, and cell death occurring during the CKD transition after AKI. Thus, iMSC-EVs have the potential to block AKI-to-CKD transition.




Characterization of iMSC–EVs and HA–iMSC–EVs. A HA–iMSC–EVs were measured the average size is 135.3 nm via NTA. (inset) Representative image of HA–iMSC–EVs observed using Cryo-TEM. Scale bar = 100 nm. B Immunoblotting analysis of HA-iMSCs and HA–iMSC–EVs for markers of extracellular vesicles. Uncropped western blot images are shown in Additional file 1: Fig. S1. C Flow cytometric analysis of iMSC–EVs and HA–iMSC–EVs. D Representative heatmap for analysis of differentially expressed proteins of HA–iMSC–EVs by LC–MS/MS analysis. The differentially expressed proteins in HA–iMSC–EVs against those from iMSC–EVs were marked in red and green, respectively. Enriched pathways, p-values, and fold change are shown in Additional file 1: Table S4. Cryo-TEM, Cryo-transmission electron microscopy; EV, extracellular vesicle; HA, hyaluronic acid; iMSC, induced pluripotent stem cell-derived mesenchymal stem cell; NTA, nanoparticle tracking analysis
HA–iMSC–EVs modulated androgenic alopecia related-mRNA expression in HFDPC. A–E The mRNA expression was measured after being treated with 50 μM testosterone and each testing articles including finasteride (100 nM), HA–iMSC–EVs or iMSC–EVs (25 μg/mL each) for 24 h. Cells not treated with testosterone was used as the normal control. Results were from three replicates. *p < 0.05, **p < 0.01 versus control. EV, extracellular vesicle; HA, hyaluronic acid; HFDPC, hair follicle dermal papilla cell; iMSC, induced pluripotent stem cell-derived mesenchymal stem cell
HA–iMSC–EVs regulated AR-related Wnt/β-catenin signaling and proliferation in HFDPC. A–E The protein expression was detected with 50 μM testosterone in the presence of 100 nM finasteride, 25 μg/mL HA–iMSC–EVs or 25 μg/mL iMSC–EVs for 24 h in HFDPC. A A typical western blotting result showing that HA–iMSC–EVs reduced AR expression and alleviated testosterone-induced alopecia. B–E The expression of each protein was normalized against that of GAPDH (N = 4). Uncropped western blot images are shown in Additional file 1: Fig. S3. F–I Inhibition of anti-AGA effects of HA–iMSC–EVs by rhDKK-1. HDFPCs were treated with 50 μM testosterone in the presence of 5 nM rhDKK-1 or 25 μg/mL HA–iMSC–EVs for 24 h. The expression of AR, β-catenin, and phospho-GSK3βser9 was normalized against that of GAPDH (N = 4). Uncropped western blot images are shown in Additional file 1: Fig. S4. The protein bands were measured by the ImageJ software and plotted as mean ± SEM. *p < 0.05, **p < 0.01 versus control group. #p < 0.05, ##p < 0.01 versus testosterone-only group. EV, extracellular vesicle; HA, hyaluronic acid; HFDPC, hair follicle dermal papilla cell; iMSC, induced pluripotent stem cell-derived mesenchymal stem cell
HA–iMSC–EVs restored hair regrowth and hair follicle cycles in testosterone-induced AGA mice. A Experimental design. B Images of the change of hair growth. Vehicle control denotes mice received topical application of 50% ethanol followed by repeated SC injection of D-PBS. AGA mice received topical application of testosterone followed by repeated SC injection of designated articles. C, D Quantification of hair regrowth area. ImageJ software was used and the relative area of hair growth against those at D1 was obtained. *p < 0.05, **p < 0.01, ****p < 0.0001 versus testosterone-only group E Body weight change. AGA, androgenetic alopecia; D-PBS, Dulbecco’s Phosphate Buffered Saline; EV, extracellular vesicle; HA, hyaluronic acid; iMSC, induced pluripotent stem cell-derived mesenchymal stem cell; SC, subcutaneous
Histological investigation of the enhancement of telogen-to-anagen transition by HA–iMSC–EVs or iMSC–EVs. A Representative hematoxylin and eosin (H&E) stained images of the skin section on day 27. B Anagen:non-anagen (catagen/telogen) ratio in the skin sections. Data was collected from at least 92 hair follicles randomly selected from five mice per group. Progression of the hair follicle cycle on day 27 was quantitatively evaluated. ****p < 0.0001 versus vehicle control group. Scale bars = 250 μm. EV, extracellular vesicle; HA, hyaluronic acid; iMSC, induced pluripotent stem cell-derived mesenchymal stem cell

+2

Improvement of androgenic alopecia by extracellular vesicles secreted from hyaluronic acid-stimulated induced mesenchymal stem cells
  • Article
  • Full-text available

September 2024

·

58 Reads

·

4 Citations

Stem Cell Research & Therapy

Background Androgenetic alopecia (AGA) is a common form of hair loss. Androgens, such as testosterone and dihydrotestosterone, are the main causes of AGA. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) can reduce AGA. However, preparing therapeutic doses of MSCs for clinical use is challenging. Induced pluripotent stem cell-derived MSCs (iMSCs) are homogenous and easily expandable, enabling scalable production of EVs. Hyaluronic acid (HA) can exert various functions including free radical scavenging, immune regulation, and cell migration. Herein, we examined whether hyaluronic acid (HA) stimulation of iMSCs could produce EVs with enhanced therapeutic outcomes for AGA. Methods EVs were collected from iMSCs primed with HA (HA–iMSC–EVs) or without HA (iMSC–EVs). The characteristics of EVs were examined using dynamic light scattering, cryo-transmission electron microscopy, immunoblotting, flow cytometry, and proteomic analysis. In vitro, we compared the potential of EVs in stimulating the survival of hair follicle dermal papilla cells undergoing testosterone-mediated AGA. Additionally, the expression of androgen receptor (AR) and relevant growth factors as well as key proteins of Wnt/β-catenin signaling pathway (β-catenin and phosphorylated GSK3β) was analyzed. Subsequently, AGA was induced in male C57/BL6 mice by testosterone administration, followed by repeated injections of iMSC–EVs, HA–iMSC–EVs, finasteride, or vehicle. Several parameters including hair growth, anagen phase ratio, reactivation of Wnt/β-catenin pathway, and AR expression was examined using qPCR, immunoblotting, and immunofluorescence analysis. Results Both types of EVs showed typical characteristics for EVs, such as size distribution, markers, and surface protein expression. In hair follicle dermal papilla cells, the mRNA levels of AR, TGF-β, and IL-6 increased by testosterone was blocked by HA–iMSC–EVs, which also contributed to the augmented expression of trophic genes related to hair regrowth. However, no notable changes were observed in the iMSC–EVs. Re-activation of Wnt/β-catenin was observed in HA–iMSC–EVs but not in iMSC–EVs, as shown by β-catenin stabilization and an increase in phosphorylated GSK3β. Restoration of hair growth was more significant in HA–iMSC–EVs than in iMSC–EVs, and was comparable to that in mice treated with finasteride. Consistently, the decreased anagen ratio induced by testosterone was reversed by HA–iMSC–EVs, but not by iMSC–EVs. An increased expression of hair follicular β-catenin protein, as well as the reduction of AR was observed in the skin tissue of AGA mice receiving HA–iMSC–EVs, but not in those treated with iMSC–EVs. Conclusions Our results suggest that HA–iMSC–EVs have potential to improve AGA by regulating growth factors/cytokines and stimulating AR-related Wnt/β-catenin signaling.

Download

Figure 5
Figure 6
Figure 7
Figures
Hyaluronic acid stimulation of induced MSCs produces extracellular vesicles with enhanced healing for skin burn wounds

July 2024

·

9 Reads

Background Skin injuries occur for various reasons during whole life. Some chronic wounds could cause an impaired wound healing process characterized by wound hypoxia, high levels of oxygen radicals, elevated levels of matrix metalloproteases, delayed cellular infiltration and granulation tissue formation, reduced angiogenesis, decreased collagen synthesis and organization. In this study, we report the EVs from hyaluronic acid-primed iMSCs (HA-iMSC-EVs) accelerating wound healing and regenerating damaged tissues by inducing the various growth factors in the thermal injury of mice. Methods EVs were collected from iMSCs primed with HA (HA-iMSC-EVs) or without HA (iMSC-EVs) and were isolated using TFF systems. Both EVs analyzed the characteristics. We investigated the proteome of HA-iMSC-EVs using the protein set ontology analysis and protein-protein interaction network. To evaluate the effect of HA-iMSC-EVs on the oxidative stress-induced wound healing delayed model, we assessed the effect of EVs on cell viability, cell migration rate, and the mRNA expression of growth factors using a hydrogen peroxide-exposed HDF model. In addition, we observed elastin and collagen expressions using an ICC staining in the HDF model. In thermal burn wound mice (BALB/c), we compared the effect of EVs in wound closure rate and histological analysis, including expression of elastin, collagen, α-SMA, and CD31. Results HA-iMSC-EVs exhibited typical EV characteristics, including size distribution, markers, and surface protein expression. In GO term analysis, HA-iMSC-EVs increased the proteins associated with ECM, including collagen biosynthesis and elastin fiber formation. In hydrogen peroxide exposed HDF models, HA-iMSC-EVs notably increased cell viability and migration activity. Furthermore, HA-iMSC-EVs increased RNA expression of VEGF, IGF1, and HGF and decreased IL-6 mRNA expression compared to the PBS group. Elastin and collagen expression in the HA-iMSC-EVs group were also significantly increased. In burn-injured mice, HA-iMSC-EVs accelerated wound closure and enhanced histological recovery. HA-iMSC-EVs increased collagen and elastin density on the upper dermis and decreased α-SMA expression. Additionally, HA-iMSC-EVs promoted the capillary density in the dermis. Conclusions Our results suggest that HA-iMSC-EVs accelerated the recovery from burn wound by providing ECM composition signal and regulating growth factors. Our strategy may contribute to the development of alternative treatment option for burn wounds. Trial registration : Not applicable.


Hyaluronic acid stimulation of stem cells for cardiac repair: a cell-free strategy for myocardial infarct

April 2024

·

87 Reads

·

6 Citations

Background Myocardial infarction (MI), a representative form of ischemic heart disease, remains a huge burden worldwide. This study aimed to explore whether extracellular vesicles (EVs) secreted from hyaluronic acid (HA)-primed induced mesenchymal stem cells (HA-iMSC-EVs) could enhance the cardiac repair after MI. Results HA-iMSC-EVs showed typical characteristics for EVs such as morphology, size, and marker proteins expression. Compared with iMSC-EVs, HA-iMSC-EVs showed enhanced tube formation and survival against oxidative stress in endothelial cells, while reduced reactive oxygen species (ROS) generation in cardiomyocytes. In THP-1 macrophages, both types of EVs markedly reduced the expression of pro-inflammatory signaling players, whereas HA-iMSC-EVs were more potent in augmenting anti-inflammatory markers. A significant decrease of inflammasome proteins was observed in HA-iMSC-EV-treated THP-1. Further, phospho-SMAD2 as well as fibrosis markers in TGF-β1-stimulated cardiomyocytes were reduced in HA-iMSC-EVs treatment. Proteomic data showed that HA-iMSC-EVs were enriched with multiple pathways including immunity, extracellular matrix organization, angiogenesis, and cell cycle. The localization of HA-iMSC-EVs in myocardium was confirmed after delivery by either intravenous or intramyocardial route, with the latter increased intensity. Echocardiography revealed that intramyocardial HA-iMSC-EVs injections improved cardiac function and reduced adverse cardiac remodeling and necrotic size in MI heart. Histologically, MI hearts receiving HA-iMSC-EVs had increased capillary density and viable myocardium, while showed reduced fibrosis. Conclusions Our results suggest that HA-iMSC-EVs improve cardiac function by augmenting vessel growth, while reducing ROS generation, inflammation, and fibrosis in MI heart. Graphical Abstract


Characterization of iMSC-EVs and pan PPAR-iMSC-EVs. A Immunoblot detection of markers of extracellular vesicles in iMSC-EVs and pan PPAR-iMSC-EVs. The expression of markers for EVs (CD9, CD81, and TSG101) or organelles (calnexin) analyzed in iMSC-EVs and pan PPAR-iMSC-EVs. Full-length blots are presented in Additional file 2: Fig. S1. B The morphology of iMSC-EVs and pan PPAR-iMSC-EVs. Cryo-TEM was used for imaging. Scale bar: 100 nm. C Size distribution of iMSC-EVs and pan PPAR-iMSC-EVs measured using a nanotracking particle analyzer. D Detection of iMSC-EVs and pan PPAR-iMSC-EVs in the AKI kidney. iMSC-EVs or pan PPAR-iMSC-EVs were stained with CMTMR and subsequently intravascularly administered into mice 8 h after cisplatin treatment. After 72 h of cisplatin administration, the kidneys were harvested, and the presence of iMSC-EVs or pan PPAR-iMSCs was detected under laser confocal microscopy. E The effect of iMSC-EVs or pan PPAR-iMSC-EVs on the growth of HK2 cells. HK2 cells were cultured with iMSC-EVs or pan PPAR-iMSC-EVs under serum-free conditions for 24 (left) and 48 (right) h. The relative number of viable cells was determined by measuring the optical density (OD450) using a CCK-8 assay. F The effect of iMSC-EVs or pan PPAR-iMSC-EVs on the survival of HK2 cells undergoing cisplatin-mediated cell death. HK2 were treated with 15 μM of cisplatin for 24 h in the presence of iMSC-EVs or pan PPAR-iMSC-EVs. The relative number of viable cells was determined by measuring the optical density (OD450) using the CCK-8 assay
Assessment of therapeutic efficacy of EVs and pan PPAR-iMSC-EVs. iMSC-EVs or pan PPAR-iMSC-EVs were administered intravenously 8 and 24 h after cisplatin treatment. After 96 h of cisplatin treatment, mice were sacrificed. A The concentration of blood urea nitrogen, serum creatinine, and body weight. B Light microscopy images of kidney sections stained with hematoxylin and eosin. Magnification: 200x. C Assessment of renal injury (N = 4); Data are presented as mean ± sd. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. D Immunohistochemical staining of proliferating cells with anti-PCNA antibody. Magnification: 400x. Scale bar: 50 μm. *p < 0.05; ***p < 0.001
Alleviation of inflammation by pan PPAR-iMSC-EVs in AKI kidney. Immunohistochemical staining of NGAL (A) and TNF-α (B). (C) The protein expression of NGAL, TNF-α, phosphorylated P38 and ERK1/2 was compared among AKI animals that received the vehicle, iMSC-EV, or pan PPAR-iMSC-EVs. Each lane represents a sample from one animal. After antibodies corresponding to each protein were used for immunoblotting, the relative expression of the proteins was normalized against that of β-actin and quantified using ImageJ software; N = 4; Data are presented as mean ± sd. *p < 0.05; ***p < 0.001; ****p < 0.0001. N represents normal animals that had not undergone cisplatin-mediated AKI. Full-length blots are presented in Additional file 2: Figs. S2, S3
Immunohistochemical detection of inflammatory cells and measurement of capillary density in the AKI mice. A The expression of immune cell markers (CD45, F4/80, and Ly6G) and blood vessels (CD31) are shown; Scale bar: 100 μm. For CD31 staining, enlarged images of each figure are also provided. B Comparison of the number of infiltrated immune cells and capillary structures in AKI kidneys. Data are presented as mean ± sd; *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001
Effect of pan PPAR-iMSC-EVs on apoptotic injury in AKI. A Immunohistochemical detection of BAX protein expression in the kidney from AKI mice. Scale bars are 100 μm. B Detection of cleaved caspase-3-expressing cells in the kidney of AKI mice that received pan PPAR-iMSC-EVs; N = 4. Scale bar: 100 μm. Data are presented as mean ± sd. *p < 0.05; ****p < 0.0001. C Immunoblot analysis of apoptosis markers in AKI kidney. Single lane represents lysate from a single animal. Markers of ER stress (CHOP), apoptosis (BAX, caspase caspase 3) and necroptosis (RIP3, MLKL) were detected using corresponding antibodies. Their relative expression was normalized against that of β-actin and then quantified using ImageJ software; N = 4. Data are presented as mean ± sd. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. Full-length blots are presented in Additional file 2: Fig. S5
Pan PPAR agonist stimulation of induced MSCs produces extracellular vesicles with enhanced renoprotective effect for acute kidney injury

January 2024

·

32 Reads

·

5 Citations

Stem Cell Research & Therapy

Background Acute kidney injury (AKI) has a complex pathophysiology and imposes serious health concerns worldwide. Extracellular vesicles (EVs) derived from induced mesenchymal stem cells (iMSCs) have been recognized as novel cell-free therapeutics for various inflammatory and degenerative disorders. In this study, we investigated whether iMSCs stimulated with a pan-peroxisome proliferator-activated receptor (PPAR) agonist could enhance the therapeutic efficacy of EVs against AKI. Methods Human iMSCs were primed with or without lanifibranor, a PPAR agonist for 24 h, and EVs were collected after an additional 24 h. The basic characteristics of EVs were evaluated using cryo-transmission electron microscopy imaging, immunoblot detection of EV markers, nanoparticle tracking analysis, and localization in AKI kidneys. In vitro, the potential of the EVs to promote the growth and survival of HK-2 cells undergoing cisplatin-induced apoptosis and anti-inflammatory effects in M1-polarized THP-1 was compared. Subsequently, AKI was induced in BALB/c mice using cisplatin. After 8 and 24 h of cisplatin treatment, iMSC-EVs or pan-PPAR-iMSC-EVs were injected intravascularly. At 96 h after cisplatin administration, the renoprotective effects of iMSC-EVs or pan-PPAR-iMSC-EVs in inhibiting inflammation and apoptosis were compared using serum biochemistry, histology, immunohistochemistry, and gene expression analysis by qPCR. Results Both EV types expressed EV markers and had typical EV morphology, and their localization in the renal tissue was confirmed. The proliferation and survival of HK-2 cells were higher in pan-PPAR-iMSC-EVs than those in iMSC-EVs. In M1-polarized THP-1 cells, the reduction in the mRNA expression of inflammatory cytokines was more significant in pan-PPAR-iMSC-EVs than that in iMSC-EVs. In the mouse model of cisplatin-induced AKI, pan-PPAR-iMSC-EVs markedly enhanced renoprotective effects compared to iMSC-EVs. Specifically, pan-PPAR-iMSC-EVs reduced tissue inflammation, immune cell infiltration, and apoptosis. Pan-PPAR-iMSC-EVs also increased renal capillary density. Conclusion Priming iMSCs with a PPAR agonist significantly improved the therapeutic potential of EVs by reducing inflammation and apoptosis. The reported strategy may contribute to the development of a novel cell-free option for AKI treatment. Trial registration: Not applicable.



Figure 3
Figure 4
Figure 5
Figure 6
Hyaluronic acid stimulation of stem cells for cardiac repair: a cell-free strategy for myocardial infarct

October 2023

·

91 Reads

Background Myocardial infarction (MI), a representative form of ischemic heart disease, remains a huge burden worldwide. This study aimed to explore whether extracellular vesicles (EVs) secreted from hyaluronic acid (HA)-primed induced mesenchymal stem cells (HA-iMSC-EVs) could enhance the cardiac repair after MI. Results HA-iMSC-EVs showed typical characteristics for EVs such as morphology, size, and marker proteins expression. Compared with iMSC-EVs, HA-iMSC-EVs showed enhanced tube formation and survival against oxidative stress in endothelial cells, while reduced reactive oxygen species (ROS) generation in cardiomyocytes. In THP-1 macrophages, both types of EVs markedly reduced the expression of pro-inflammatory signaling players, whereas HA-iMSC-EVs were more potent in augmenting anti-inflammatory markers. A significant decrease of inflammasome proteins was observed in HA-iMSC-EV-treated THP-1. Further, phospho-SMAD2 as well as fibrosis markers in TGF-b1-stimulated cardiomyocytes were reduced in HA-iMSC-EVs treatment. Proteomic data showed that HA-iMSC-EVs were enriched with multiple pathways including immunity, extracellular matrix organization, angiogenesis, and cell cycle. The localization of HA-iMSC-EVs in myocardium was confirmed after delivery by either intravenous or intramyocardial route, with the latter increased intensity. Echocardiography revealed that intramyocardial HA-iMSC-EVs injections improved cardiac function and reduced adverse cardiac remodeling and necrotic size in MI heart. Histologically, MI hearts receiving HA-iMSC-EVs had increased capillary density and viable myocardium, while showed reduced fibrosis. Conclusions Our results suggest that HA-iMSC-EVs improve cardiac function by augmenting vessel growth, while reducing ROS generation, inflammation, and fibrosis in MI heart.



Citations (12)


... Specifically, the release of EVs from cells is a slow procedure and cell culture is significant expenses, leading to a relatively low yield and high cost. 33 Actually, platelets are recognized as a prominent source of EVs. PEVs constitute the largest proportion of blood-derived EVs and possess significant potential as an alternative cell-free therapeutic approach in the field of regenerative medicine. ...

Reference:

Platelet-derived extracellular vesicles promote hair follicle growth through β-catenin signaling pathway
Improvement of androgenic alopecia by extracellular vesicles secreted from hyaluronic acid-stimulated induced mesenchymal stem cells

Stem Cell Research & Therapy

... One of the important functions of HA is its antioxidant activity. It can specifically interact with CD44 receptors on myocardial cells, stimulating cell proliferation, maintaining the integrity of myocardial cells during ROS damage, and preventing the activation of death receptors, thereby preserving cardiomyocyte survival and function [34][35][36][37]. ...

Hyaluronic acid stimulation of stem cells for cardiac repair: a cell-free strategy for myocardial infarct

... Kim et al found that a pan-peroxisome proliferator-activated receptor (PPAR) agonist can enhance the protective effect of induced mesenchymal stem cells derived EVs (iMSC-EVs) on kidney in CDDP-AKI mice. 77 Beyond reagents, the researchers also explored physical methods such as pulsed focused ultrasound (pFUS) processing of tissue to increase the therapeutic effect of MSCs-EVs on CDDP-AKI. 78 The possible reason is that pFUS enhances MSCs homing to save renal function safely and effectively. ...

Pan PPAR agonist stimulation of induced MSCs produces extracellular vesicles with enhanced renoprotective effect for acute kidney injury

Stem Cell Research & Therapy

... Furthermore, the therapeutic effects of iMSC-derived EVs have also been explored in many diseases such as cardiovascular disease, musculoskeletal pathology and in the modulation of allogeneic immune responses, where different experimental settings, such as depleted-FBS or human platelet lysate, were employed for the cultivation of iMSCs (Levy et al., 2023;Hong et al., 2024;Meng and Guo, 2023;Tertel et al., 2023). ...

Extracellular vesicles from induced pluripotent stem cell-derived mesenchymal stem cells enhance the recovery of acute kidney injury
  • Citing Article
  • October 2023

Cytotherapy

... Identifying specific regulatory factors, such as let-7c shuttled by adipose-derived MSC exosomes to modulate macrophage polarization, further exemplifies the intricate molecular mechanisms through which MSC exosomes exert their therapeutic effects [165]. These insights pave the way for developing tailored exosome-based therapeutics for various conditions, including skin inflammation and barrier dysfunction [166,167]. In addition, exosomes have also been instrumental in diagnostic applications, with the potential for detecting low-abundant mutant copies of epidermal growth factor receptor (EGFR) in non-small cell lung cancer (NSCLC) using combined exosomal nucleic acid analysis in plasma and pleural fluid [168]. ...

Exosome from IFN-γ-Primed Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Improved Skin Inflammation and Barrier Function

... Clinical trials have already reported the safety and efficacy of intravenous iPSC-MSC administration for steroid-resistant acute graft-versus-host disease [18], with two-year follow-up results further confirming the safety of iMSCs in therapeutic applications [19]. Additionally, iMSCs exhibit superior proliferative capacity compared to traditional MSCs [20,21]. Unlike MSCs derived directly from bone marrow, adipose tissue, or other conventional sources, iMSCs provide a stable cell source that can be obtained through non-invasive methods, avoiding the traumatic procedures required for conventional MSC collection. ...

iPSC-Derived MSCs Are a Distinct Entity of MSCs with Higher Therapeutic Potential than Their Donor-Matched Parental MSCs

... In summary, both cEXO and iEXO harbor a rich repertoire of immunomodulatory proteins, reinforcing their potential to modulate immune responses, which aligns with previous studies [28,29]. Furthermore, GO and KEGG enrichment analyses of differentially expressed proteins indicate a stronger association between IFN-γ-licensed exosomes and macrophage-mediated immune functions, with broad implications for both innate and adaptive immunity. ...

Extracellular vesicles from IFN-γ-primed mesenchymal stem cells repress atopic dermatitis in mice

... In the present study, we observed that infiltration of both M1 and M2 macrophages increased and that all were decreased by iMSC-EVs. The result of M1 macrophage is consistent with that of our previous study, which showed that iMSC-EVs reduced TNF-α, IL-1β, and phosphorylated p65 levels in M1-induced THP1 macrophages (36). Similarly, Lu et al. (37) observed that the polarization of both M1 and M2 macrophages were inhibited by exosomes from bone marrow-derived MSCs through activation of prostaglandin receptor 2 using a mice model of UUO-induced interstitial fibrosis. ...

Cargo proteins in extracellular vesicles: potential for novel therapeutics in non-alcoholic steatohepatitis

... We show that MSC-CM promotes epithelial cell migration and proliferation, resulting in corneal wound-healing effects. Previous studies have identified that MSC-CM or MSC-EV/Exo can both enhance wound healing in vitro and in vivo [3,36,[64][65][66]. We have also previously demonstrated that human corneal MSC-derived EV/exosomes promote the repair of ocular surface injuries [62,67,68]. ...

Mesenchymal Stem Cell-derived Extracellular Vesicles for Skin Wound Healing
  • Citing Chapter
  • April 2021

Advances in Experimental Medicine and Biology

... Instead, the tumorigenic potential, low engraftment, and cell retention restricts the therapeutic application of iPSCs [72]. Further, differentiation of iPSCs into MSCs needs caution and optimized protocols to confirm that not even a single iPSC has been left undifferentiated, else the cell product can become tumorigenic [73]. Moreover, the development of iPSC-based therapies requires higher production costs and is labor-intensive. ...

Generation of mesenchymal stem-like cells for producing extracellular vesicles

World Journal of Stem Cells