Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (350)

Search Parameters:
Keywords = MSC migration

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 2087 KiB  
Article
Liposome-Loaded Mesenchymal Stem Cells Enhance Tumor Accumulation and Anti-Tumor Efficacy of Doxorubicin in Mouse Tumor Models of Melanoma
by Yusuke Kono, Himi Kanbara, Saki Danjo, Aiga Yoshikawa, Yoshihiro Iwayama and Ken-ichi Ogawara
Pharmaceutics 2025, 17(8), 947; https://doi.org/10.3390/pharmaceutics17080947 - 22 Jul 2025
Abstract
Background: Mesenchymal stem cells (MSCs) possess an intrinsic tumor-tropic ability, and therefore, MSCs may potentially be used as biomimetic carriers for active drug delivery systems targeting tumors. We previously developed a method to efficiently load liposomes onto the surface of MSCs via [...] Read more.
Background: Mesenchymal stem cells (MSCs) possess an intrinsic tumor-tropic ability, and therefore, MSCs may potentially be used as biomimetic carriers for active drug delivery systems targeting tumors. We previously developed a method to efficiently load liposomes onto the surface of MSCs via electrostatic interactions. The prepared liposome-loaded MSCs (Lip-MSCs) spontaneously accumulated in solid melanoma tumors with low vascular permeability while stably carrying liposomes. Methods: To explore Lip-MSC applications in cancer chemotherapy, doxorubicin (DOX)-encapsulated liposomes (DOX-Lip) were prepared and loaded onto MSCs. The cell viability, DOX-releasing properties, tumor-homing capacity, and anti-tumor efficacy of DOX-Lip-MSCs were analyzed. Results: Small liposomes (100 nm) retained DOX, whereas significant leakage of DOX was observed from 600 nm-sized liposomes. Based on this result, we used 100 nm DOX-Lip for the preparation of DOX-Lip-MSCs. Compared with MSCs loaded with DOX by incubation with DOX solution, DOX-Lip-MSCs could load a larger amount of DOX with minimal cytotoxicity. DOX-Lip-MSCs also showed sustained DOX release. DOX-Lip-MSCs efficiently migrated toward the conditioned medium of B16/BL6 melanoma cells in vitro and accumulated in B16/BL6 tumors in vivo, leading to a significant inhibitory effect on tumor growth. Conclusions: Lip-MSCs can serve as an efficient carrier to deliver anti-cancer drugs into solid tumors. Full article
(This article belongs to the Special Issue Cell-Mediated Delivery Systems)
Show Figures

Figure 1

22 pages, 1940 KiB  
Article
Equine Colostrum-Derived Mesenchymal Stromal Cells: A Potential Resource for Veterinary Regenerative Medicine
by Angelita Capone, Barbara Merlo, Fabiana Begni and Eleonora Iacono
Vet. Sci. 2025, 12(7), 681; https://doi.org/10.3390/vetsci12070681 - 19 Jul 2025
Viewed by 213
Abstract
Beyond its immunological role, colostrum has emerged as a promising, non-invasive source of bioactive factors, including mesenchymal stem/stromal cells (MSCs). This study represents the first attempt to isolate and characterize MSCs from equine colostrum (C-MSCs) to assess their potential use in veterinary regenerative [...] Read more.
Beyond its immunological role, colostrum has emerged as a promising, non-invasive source of bioactive factors, including mesenchymal stem/stromal cells (MSCs). This study represents the first attempt to isolate and characterize MSCs from equine colostrum (C-MSCs) to assess their potential use in veterinary regenerative medicine. Colostrum (n = 6) was collected from mares immediately after their delivery and centrifuged, and the recovered cells were cultured under standard conditions. The C-MSCs displayed plastic adherence and a heterogeneous morphology, including spindle-shaped and epithelial-like cells. The population doubling time (PDT) values varied among the samples, and four out of six showed rapid proliferation (<2 days). Colony-forming unit (CFU) assays confirmed their clonogenic potential, though significant inter-sample variability was observed (p < 0.05). Spheroid formation assays revealed differences in cell–cell adhesion: four out of six samples formed stable spheroids within four days. A migration assay showed significant variability (p < 0.05): one out of six achieved complete wound closure within 72 h, whereas five out of six reached ~30% at 96 h. All samples were positive for adipogenic, chondrogenic, and osteogenic differentiation as shown via staining. RT-PCR confirmed MSC marker expression, while hematopoietic markers were absent. MHC-I expression was weak in five out of six samples, whereas MHC-II was consistently negative. These findings support equine colostrum as a viable MSC source, though its variability requires further validation with larger samples. Additional research is needed to investigate C-MSCs’ immunomodulatory properties and therapeutic potential. Full article
Show Figures

Figure 1

18 pages, 2381 KiB  
Article
Influence of Low-Level Red Laser Irradiation on the Proliferation, Viability, and Differentiation of Human Embryonic Stem Cell-Derived Mesenchymal Stem Cells
by Khalid M. AlGhamdi, Ashok Kumar, Musaad Alfayez and Amer Mahmood
Life 2025, 15(7), 1125; https://doi.org/10.3390/life15071125 - 17 Jul 2025
Viewed by 351
Abstract
The present investigation was conducted to observe the effects of different energy densities of a low-level red laser (LLRL) on human embryonic stem cell-derived mesenchymal stem cells (hESC-MSCs). hESC-MSCs were cultured and irradiated with a LLRL from 0.5 to 5.0 J/cm2 at [...] Read more.
The present investigation was conducted to observe the effects of different energy densities of a low-level red laser (LLRL) on human embryonic stem cell-derived mesenchymal stem cells (hESC-MSCs). hESC-MSCs were cultured and irradiated with a LLRL from 0.5 to 5.0 J/cm2 at a wavelength of 635 nm. Biological parameters such as proliferation, viability, and migration were observed after 72 h of LLRL irradiation. Compared with the control, LLRL irradiation significantly increased the proliferation and viability of hESC-MSCs from 0.5 to 2.5 J/cm2 (p < 0.001, p < 0.05). LLRL irradiation from 0.5 to 3.0 J/cm2 significantly increased the migration of hESC-MSCs (p < 0.01). These results revealed that LLRL irradiation at lower energy densities significantly increased the proliferation, viability, and migration of hESC-MSCs. However, higher energy densities were ineffective; this was also true when we examined osteogenic differentiation, as low energy densities of LLRL had a positive effect on differentiation, whereas higher energy densities had a negative effect on alkaline phosphatase activity, Alizarin Red staining and gene expression analysis. In addition, not all stem cell markers were affected by the laser, and a slight decrease in the expression of CD146, which is a stemness marker, was detected, indicating improved differentiation. These findings indicate that low energy densities of LLRL irradiation have positive effects on the proliferation, migration, and differentiation of hESC-MSCs. However, higher energy densities showed inhibitory effects. Full article
Show Figures

Figure 1

12 pages, 5543 KiB  
Article
Hypoxic Conditions Promote Cartilage Repair in a Rat Knee Osteochondral Defect Model via Hypoxia-Inducible Factor-1α
by Kei Nakamura, Atsuo Inoue, Yuji Arai, Shuji Nakagawa, Yuta Fujii, Ryota Cha, Keisuke Sugie, Kentaro Hayashi, Tsunao Kishida, Osam Mazda and Kenji Takahashi
Int. J. Mol. Sci. 2025, 26(13), 6370; https://doi.org/10.3390/ijms26136370 - 2 Jul 2025
Viewed by 275
Abstract
Bone marrow stimulation is a treatment for articular cartilage injuries that promotes cartilage repair by inducing the migration and accumulation of mesenchymal stem cells (MSCs), but often results in fibrocartilage with limited durability. This study aimed to investigate the effect of hypoxic conditions [...] Read more.
Bone marrow stimulation is a treatment for articular cartilage injuries that promotes cartilage repair by inducing the migration and accumulation of mesenchymal stem cells (MSCs), but often results in fibrocartilage with limited durability. This study aimed to investigate the effect of hypoxic conditions on cartilage repair using a rat osteochondral defect model. Osteochondral defects (1.0 mm in diameter) were created in the femoral trochlear groove, and rats were exposed to hypoxic conditions (12% O2) for 4 weeks postoperatively. Histological analysis was performed, and protein expression of hypoxia-inducible factor-1α (HIF-1α) and SRY-box transcription factor 9 (SOX9) in the repair tissue was evaluated after 1 week. As a result, after 1 week, protein expression of HIF-1α and SOX9 in the Hypoxia group was significantly increased compared to the Normoxia group. After 4 weeks, the Hypoxia group exhibited a hyaline cartilage-like tissue structure with a significantly lower Modified Wakitani score compared to the Normoxia group. Furthermore, after 4 weeks, the inhibition of HIF-1α suppressed cartilage repair. These findings suggest that hypoxic conditions promote SOX9 expression via HIF-1α during the early phase of MSC chondrogenic differentiation and promote the formation of hyaline cartilage-like repair tissue. In conclusion, bone marrow stimulation under hypoxic conditions may enhance the repair effect on articular cartilage injuries. Full article
(This article belongs to the Special Issue Molecular Biology of Hypoxia)
Show Figures

Figure 1

18 pages, 1711 KiB  
Article
Exosomes Derived from Induced and Wharton’s Jelly-Derived Mesenchymal Stem Cells Promote Senescence-like Features and Migration in Cancer Cells
by Nidaa A. Ababneh, Razan AlDiqs, Sura Nashwan, Mohammad A. Ismail, Raghda Barham, Renata M. Alatoom, Fairouz Nairat, Mohammad H. Gharandouq, Talal Al-Qaisi, Abdalla Awidi and Tareq Saleh
Int. J. Mol. Sci. 2025, 26(13), 6178; https://doi.org/10.3390/ijms26136178 - 26 Jun 2025
Viewed by 513
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) play a key role in tissue repair, immune regulation, and cancer biology. Due to limitations in MSC expansion and source variability, interest has shifted to induced pluripotent stem cell-derived MSCs (iMSCs) as a promising alternative. This study compares [...] Read more.
Mesenchymal stem cell-derived exosomes (MSC-Exos) play a key role in tissue repair, immune regulation, and cancer biology. Due to limitations in MSC expansion and source variability, interest has shifted to induced pluripotent stem cell-derived MSCs (iMSCs) as a promising alternative. This study compares effects of exosomes derived from iMSCs (iMSC-Exos) and Wharton’s jelly MSCs (WJMSC-Exos) on MCF7 and A549 cancer cells. Both types of exosomes reduced MCF7 proliferation and induced a senescence-like state, rather than apoptosis, although the antiproliferative effect was transient in A549 cells. Notably, WJMSC-Exos promoted migration in both MCF7 and A549, whereas iMSC-Exos did not exhibit this effect. Overall, WJMSC-Exos had a more robust impact on cancer cell proliferation and migration. These findings highlight the diverse effects of exosomes on cancer and the development of a senescence-like state as an important response to Exos exposure. Moreover, these findings invite for more careful evaluation of the therapeutic role of iMSC-derived Exos. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

16 pages, 76646 KiB  
Article
Cytokinesis in Suspension: A Distinctive Trait of Mesenchymal Stem Cells
by Bhavna Rani, Hong Qian and Staffan Johansson
Cells 2025, 14(12), 932; https://doi.org/10.3390/cells14120932 - 19 Jun 2025
Viewed by 468
Abstract
Mesenchymal stem cells (MSCs) have a broad clinical potential, but their selection and expansion on plastic cause unknown purity and phenotypic alterations, reducing therapy efficiency. Furthermore, their behavior in non-adherent conditions during systemic transplantation remains poorly understood. The sphere formation from single cells [...] Read more.
Mesenchymal stem cells (MSCs) have a broad clinical potential, but their selection and expansion on plastic cause unknown purity and phenotypic alterations, reducing therapy efficiency. Furthermore, their behavior in non-adherent conditions during systemic transplantation remains poorly understood. The sphere formation from single cells is commonly used to assess stemness, but MSCs lack this ability, raising questions about their anchorage dependence for proliferation. We investigated whether bone marrow-derived MSCs can complete cytokinesis in non-adherent environments. Primary human and mouse bone marrow-derived MSCs were synchronized in early mitosis using nocodazole and were cultured on soft, rigid, or non-adherent surfaces. Both human and mouse MSCs displayed an ALIX (abscission licensor) recruitment to the midbody 40–90 min post-nocodazole release, regardless of the substrate adherence. Cells maintained for 4hr in the suspension remained viable, and daughter cells rapidly migrated apart upon the re-adhesion to fibronectin-coated surfaces, demonstrating cytokinesis completion in suspension. These findings distinguish MSCs from fibroblasts (which require adhesion for division), provide a more general stemness feature, and suggest that adhesion-independent cytokinesis is a trait relevant to the post-transplantation survival and tissue homing. This property may offer strategies to expand MSCs with an improved purity and functionality and to enhance engraftment by leveraging cell cycle manipulation to promote an early extracellular matrix deposition at target sites. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Figure 1

20 pages, 8483 KiB  
Article
Comparative Efficacy of Exosomes Derived from Different Mesenchymal Stem Cell Sources in Osteoarthritis Models: An In Vitro and Ex Vivo Analysis
by Jaishree Sankaranarayanan, Hyung Keun Kim, Ju Yeon Kang, Sree Samanvitha Kuppa, Hong Yeol Yang and Jong Keun Seon
Int. J. Mol. Sci. 2025, 26(12), 5447; https://doi.org/10.3390/ijms26125447 - 6 Jun 2025
Viewed by 706
Abstract
Osteoarthritis (OA) is a prevalent and debilitating joint disorder that affects a substantial proportion of the global population, underscoring the urgent need for therapeutic strategies that extend beyond symptomatic management. Although mesenchymal stem cells (MSCs) have emerged as a promising therapeutic modality, their [...] Read more.
Osteoarthritis (OA) is a prevalent and debilitating joint disorder that affects a substantial proportion of the global population, underscoring the urgent need for therapeutic strategies that extend beyond symptomatic management. Although mesenchymal stem cells (MSCs) have emerged as a promising therapeutic modality, their clinical application remains constrained by several inherent limitations. This study explores a cell-free alternative by investigating the therapeutic potential of exosomes derived from bone marrow (BMSCs), adipose tissue (ADSCs), and umbilical cord (UMSCs) MSCs in mitigating OA pathogenesis, utilizing both in vitro and ex vivo models. Exosomes from each MSC source were isolated and characterized through nanoparticle tracking analysis, transmission electron microscopy, and Western blotting to confirm their identity and purity. Subsequently, their chondroprotective, anti-inflammatory, and regenerative properties were systematically assessed through evaluations of cell viability, expression profiles of inflammatory and chondroprotective markers, and chondrocyte migration assays. The results demonstrate that all three types of MSC-derived exosomes (MSC-Exos) exhibit low cytotoxicity while significantly suppressing proinflammatory markers and enhancing the expression of chondroprotective genes. Notably, BMSC-Exos and UMSC-Exos displayed superior efficacy in attenuating inflammation, promoting cartilage protection, and inhibiting chondrocyte apoptosis. Furthermore, all MSC-Exos markedly enhanced chondrocyte motility, a critical component of cartilage repair. Collectively, these findings support the therapeutic promise of MSC-Exos, particularly those derived from BMSCs and UMSCs, as a targeted, cell-free approach for the treatment of OA compared to ADSCs. By modulating inflammation, promoting cartilage regeneration, and preventing chondrocyte apoptosis, MSC-Exos may serve as a viable and scalable alternative to current MSC-based therapies for this widespread degenerative disease. Full article
(This article belongs to the Special Issue Molecular Advances and Perspectives in Rheumatic Diseases)
Show Figures

Figure 1

15 pages, 4213 KiB  
Article
Dose-Dependent Effects of TGF-β Inhibition on Osteoblast Differentiation and Wound Healing
by Nihal Almuraikhi, Latifa Alkhamees, Sumaiya Tareen, Hessah Alshammari and Manikandan Muthurangan
Curr. Issues Mol. Biol. 2025, 47(5), 360; https://doi.org/10.3390/cimb47050360 - 14 May 2025
Viewed by 533
Abstract
TGF-β is a multifunctional pathway that controls significant cellular and physiological processes and several pathological activities. TGF-β-induced signaling can be triggered upon binding to specific receptors to initiate the transcriptional activation of several genes and cellular processes. However, the detailed role of TGF-β [...] Read more.
TGF-β is a multifunctional pathway that controls significant cellular and physiological processes and several pathological activities. TGF-β-induced signaling can be triggered upon binding to specific receptors to initiate the transcriptional activation of several genes and cellular processes. However, the detailed role of TGF-β signaling in osteoblast differentiation remains to be explicated. SB525334, a selective TGF-βRI inhibitor, was investigated for its effect on the osteoblastic differentiation of human bone marrow MSCs at different concentrations. Alkaline phosphatase (ALP) activity was used to assess osteoblast differentiation marker, while Alizarin red staining was used as a marker for mineralization. Expressions of osteoblast-specific genes were evaluated using real-time PCR. A migration assay was performed to assess the effect of TGF-β on wound healing. Moreover, immunofluorescent staining for SMAD2/3 and SMAD4 was employed to confirm the activation of the TGF-β pathway. The inhibition of TGF-β1 signaling using a high concentration of SB525334 (3 µM) significantly reduced ALP activity and mineralization and downregulated osteoblast-specific genes. However, the opposite effect was reported using a lower concentration (0.03 µM), where osteoblast-associated genes were significantly upregulated, and ALP activity and mineralization were higher. Significant scratch/wound healing was achieved at a lower concentration of SB525334, while a higher concentration of SB525334 resulted in lower healing. Moreover, a low concentration of SB525334 demonstrated nuclear translocation of SMAD 2/3 and 4. Our study confirms that the effect of TGF-β signaling in bone formation and wound healing is dose-dependent, and the use of TGF-β is recommended as a valuable therapeutic approach. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

18 pages, 7599 KiB  
Article
Injectable, Manganese-Labeled Alginate Hydrogels as a Matrix for Longitudinal and Rapidly Retrievable 3D Cell Culture
by Izabela Malysz-Cymborska, Dominika Golubczyk, Piotr Walczak, Luiza Stanaszek and Miroslaw Janowski
Int. J. Mol. Sci. 2025, 26(10), 4574; https://doi.org/10.3390/ijms26104574 - 10 May 2025
Viewed by 518
Abstract
Hydrogels are one of the most attractive biomaterials, used in both three-dimensional (3D) and in vivo cultures. They facilitate the reconstruction of tissue microenvironments by preserving the spatial arrangement of cells, cell–cell interactions, and functional dynamics in the tissue. In this work, the [...] Read more.
Hydrogels are one of the most attractive biomaterials, used in both three-dimensional (3D) and in vivo cultures. They facilitate the reconstruction of tissue microenvironments by preserving the spatial arrangement of cells, cell–cell interactions, and functional dynamics in the tissue. In this work, the long-term effect of alginate hydrogel on cell culture and the possibility of rapid cell recovery by dissolving the hydrogel were investigated. Mouse glial-restricted progenitors (GRPs) and porcine mesenchymal stem cells (MSCs) were suspended in hydrogels; their metabolic activity, viability, and expression of genes, which are involved in oxidative stress, apoptosis, proliferation, migration, and differentiation, were assessed using quantitative polymerase chain reaction (qPCR). The concentration that was able to dissolve the hydrogel and was the least harmful to the cells was 0.005 M ethylenediaminetetraacetic acid (EDTA). The metabolism of both cell types was reduced from the beginning of the experiment to day 3. From day 7 to the end of the experiment, the normalization of the GRP metabolism was observed, in contrast to the MSCs. For the apoptosis-related genes, caspase 3, 7, and B-cell leukemia (Casp3, Casp 7, Bcl2) were increased in GRPs and MSCs on days 0 and 1. After 3 and 7 days, an increase in the expression of oxidative stress genes (nuclear factor of activated T-cells 5—NFAT5 and autophagy-related 14-ATG14) was observed in cells cultured in calcium chloride (CaCl2). GRPs cultured in calcium alginate (CaM) were not affected and, remarkably, showed increased Antigen Kiel 67 (Ki67) levels after 30 days. In conclusion, alginate hydrogels provide an excellent environment for stem cell culture in 3D for a longer period of time, but this is dependent on the cell type. Therefore, an individual approach to cell culture is necessary, taking into account the requirements of the cells to be used. Full article
(This article belongs to the Special Issue Biocomposite Hydrogels for Biomedical Applications)
Show Figures

Graphical abstract

14 pages, 475 KiB  
Review
The Therapeutic Use and Potential of MSCs: Advances in Regenerative Medicine
by Alin Constantin Pînzariu, Roxana Moscalu, Radu Petru Soroceanu, Minela Aida Maranduca, Ilie Cristian Drochioi, Vlad Ionut Vlasceanu, Sergiu Timofeiov, Daniel Vasile Timofte, Bogdan Huzum, Mihaela Moscalu, Dragomir Nicolae Serban and Ionela Lacramioara Serban
Int. J. Mol. Sci. 2025, 26(7), 3084; https://doi.org/10.3390/ijms26073084 - 27 Mar 2025
Viewed by 1312
Abstract
Mesenchymal stem cells (MSCs) have emerged as a relevant strategy in regenerative medicine due to their multipotent differentiation capacity, immunomodulatory properties, and therapeutic applications in various medical fields. This review explores the therapeutic use of MSCs, focusing on their role in treating autoimmune [...] Read more.
Mesenchymal stem cells (MSCs) have emerged as a relevant strategy in regenerative medicine due to their multipotent differentiation capacity, immunomodulatory properties, and therapeutic applications in various medical fields. This review explores the therapeutic use of MSCs, focusing on their role in treating autoimmune disorders and neoplastic diseases and in tissue regeneration. We discuss the mechanisms underlying MSC-mediated tissue repair, including their paracrine activity, migration to injury sites, and interaction with the immune system. Advances in cellular therapies such as genome engineering and MSC-derived exosome treatments further enhance their applicability. Key methodologies analyzed include genomic studies, next-generation sequencing (NGS), and bioinformatics approaches to optimize MSC-based interventions. Additionally, we reviewed preclinical and clinical evidence demonstrating the therapeutic potential of MSCs in conditions such as graft-versus-host disease, osteoarthritis, liver cirrhosis, and neurodegenerative disorders. While promising, challenges remain regarding standardization, long-term safety, and potential tumorigenic risks associated with MSC therapy. Future research should focus on refining MSC-based treatments to enhance efficacy and minimize risks. This review underscores the need for large-scale clinical trials to validate MSC-based interventions and fully harness their therapeutic potential. Full article
(This article belongs to the Special Issue Latest Research on Mesenchymal Stem Cells)
Show Figures

Figure 1

18 pages, 6069 KiB  
Article
Cisplatin-Mediated IL-6 and IDO1 Suppression in Mesenchymal Stromal Cells: Implications for Tumor Microenvironment Modulation In Vitro
by Armin von Fournier, Erik Würflein, Helena Moratin, Manuel Stöth, Totta Ehret Kasemo, Marietta Herrmann, Miguel Goncalves, Rudolf Hagen, Stephan Hackenberg, Thomas Gehrke and Agmal Scherzad
Curr. Issues Mol. Biol. 2025, 47(4), 231; https://doi.org/10.3390/cimb47040231 - 27 Mar 2025
Viewed by 489
Abstract
Mesenchymal stromal cells (MSCs) influence tumor biology and immunology by releasing cytokines, chemokines and growth factors. Currently, cisplatin is an integral part of drug-based tumor therapy, for example, in head and neck squamous cell carcinoma (HNSCC). Cisplatin treatment induces apoptosis as a primary [...] Read more.
Mesenchymal stromal cells (MSCs) influence tumor biology and immunology by releasing cytokines, chemokines and growth factors. Currently, cisplatin is an integral part of drug-based tumor therapy, for example, in head and neck squamous cell carcinoma (HNSCC). Cisplatin treatment induces apoptosis as a primary mechanism of action; however, additional immunomodulatory effects of cisplatin are gaining interest. The aim of this study is to evaluate the possible immunomodulatory effects of cisplatin in human MSCs (hMSCs). The MSCs, obtained from human bone marrow, were characterized by analyzing plastic adherence, typical surface features, and ability to differentiate. Toxicity analysis of cisplatin’s effects on primary MSCs, including the determination of a subtoxic concentration, was performed using the MTT assay. Enzyme-linked immunosorbent assays (ELISA) and a quantitative real-time polymerase chain reaction (qRT-PCR) were used to identify potentially immunomodulatory factors. Additionally, a scratch assay was performed to evaluate cell migration. First, subtoxic cisplatin concentrations were determined. A significantly reduced protein expression of indoleamine 2,3-dioxygenase 1 (IDO1) in MSCs under the influence of subtoxic cisplatin concentrations was demonstrated. Similarly, IL-6 protein expression was qualitatively reduced at subtoxic concentrations, although without statistical significance. At the mRNA level, qRT-PCR showed a non-significant, cisplatin concentration-dependent reduction in the expression of both IL-6 and IDO1. The scratch assay showed no statistically significant influence on migration after cisplatin treatment. In MSCs, there is tendency to a decrease in IL-6 and IDO1 at both protein and mRNA level after cisplatin exposure. These effects are congruent with each other and dose-dependent. This indicates that cisplatin not only acts via the known cytotoxic effect, but may induce a reduction in tumor-supporting proteins, like IL-6 and IDO1, by MSCs in the tumor microenvironment at subtoxic concentrations. Traditional cytostatic compounds, which can favorably modulate the immune system in the tumor microenvironment, may open new avenues to explore treatment strategies specifically targeting immunomodulation. Overall, the results indicate beneficial immunomodulation by cisplatin. Full article
(This article belongs to the Special Issue Targeting Tumor Microenvironment for Cancer Therapy, 3rd Edition)
Show Figures

Figure 1

14 pages, 4619 KiB  
Article
Cobalt-Incorporated Hydroxyapatite Conditioned Media Promotes In Vitro Scratch Wound Healing and Mesenchymal Stem Cell Migration
by Weerapat Leelasangsai, Krongrat Thummachot, Puttita Thammasarnsophon, Autcharaporn Srion, Jintamai Suwanprateeb and Somying Patntirapong
J. Funct. Biomater. 2025, 16(3), 72; https://doi.org/10.3390/jfb16030072 - 20 Feb 2025
Viewed by 1146
Abstract
Cell migration of mesenchymal stem cells (MSCs) is critical for bone healing and remodeling. Cobalt is a well-known hypoxia mimic, which can enhance MSC migration. Therefore, the objective of this study was to investigate the migratory response of MSCs to a developed cobalt-incorporated [...] Read more.
Cell migration of mesenchymal stem cells (MSCs) is critical for bone healing and remodeling. Cobalt is a well-known hypoxia mimic, which can enhance MSC migration. Therefore, the objective of this study was to investigate the migratory response of MSCs to a developed cobalt-incorporated hydroxyapatite (HACo) material. HACo was fabricated by a simple ion exchange procedure at concentrations ranging from 40 to 8000 μM into disc shape. HACo discs were incubated in the media and conditioned media (CM; HACoCM) were collected for MSC culture. HACM served as a control. MSCs were cultured until reaching 90% confluence before the wound was generated by scraping. Time-lapse imaging of wound migration was monitored, recorded, and assessed. Statistical analysis was performed by one-way ANOVA followed by a Dunnett’s test. The wound area gradually declined from 0 to 40 h for all samples. HACoCM at 40 µM (HACo40CM) promoted wound closure at the early period of wound healing. Both HACo40CM and HACo8000CM enhanced the distance and velocity of individual cell migration. However, only HACo40CM affected cell persistence and direction at the early period of cell migration. Exposure to HACoCM accelerated the speed of MSC migration, which is necessary for wound healing. The migratory ability of individual cells could help the rate of wound healing. Therefore, HACo materials may serve as potential biomaterials for enhanced bone healing. Full article
(This article belongs to the Section Bone Biomaterials)
Show Figures

Figure 1

15 pages, 2851 KiB  
Article
Human Placenta MSC-Derived DNA Fragments Exert Therapeutic Effects in a Skin Wound Model via the A2A Receptor
by Hankyu Lee, Hyun-Jung Lee, Hyeon-Jun Jang, Hyeri Park and Gi Jin Kim
Int. J. Mol. Sci. 2025, 26(4), 1769; https://doi.org/10.3390/ijms26041769 - 19 Feb 2025
Cited by 1 | Viewed by 1785
Abstract
PDRN, polydeoxyribonucleotide, which is used as a tissue-regeneration material, is present in human cells under physiological conditions and stimulates regeneration and metabolic activity. PDRN can be used as a biomaterial for several types of regeneration, including wound healing, to promote cell growth and [...] Read more.
PDRN, polydeoxyribonucleotide, which is used as a tissue-regeneration material, is present in human cells under physiological conditions and stimulates regeneration and metabolic activity. PDRN can be used as a biomaterial for several types of regeneration, including wound healing, to promote cell growth and growth-factor production. The aims of this study were to determine the effect of PDRN derived from human placenta-derived mesenchymal stem cells (hPD-MSCs) on cellular regeneration through A2A receptor signaling and to investigate its therapeutic effects in a mouse model of wound healing. Human PDRN (UNIPlax) was extracted from hPD-MSCs fragmented via a sonication system and evaluated for its effect on the migration of HaCaT cells in an in vitro system and in a wound-healing mouse model in vivo. Compared with the sham treatment, UNIPlax treatment significantly increased the migration of injured HaCaT cells (p < 0.05). Additionally, the tube formation of human umbilical vein endothelial cells (HUVECs) was greater than that of the sham group (p < 0.05), and the effects of this treatment were mediated through the A2A receptor. Furthermore, UNIPlax treatment led to a decrease in wound size; in addition, the area of granulation and the rate of collagen formation at the wound site were significantly greater than those in the sham group in the wound-healing mouse model (p < 0.001). We also confirmed that UNIPlax promoted tissue regeneration and the expression of VEGF through the A2A receptor. Taken together, these findings indicate that UNIPlax has potential for regeneration of damaged tissues, including during wound healing. Full article
Show Figures

Figure 1

23 pages, 8162 KiB  
Article
The Role of Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles in Modulating Dermal Fibroblast Activity: A Pathway to Enhanced Tissue Regeneration
by Muttiah Barathan, Kow Jack Ham, Hui Yin Wong and Jia Xian Law
Biology 2025, 14(2), 150; https://doi.org/10.3390/biology14020150 - 1 Feb 2025
Viewed by 1877
Abstract
Extracellular vesicles (EVs) secreted by umbilical cord-derived mesenchymal stem cells (UC-MSCs) hold significant promise as therapeutic agents in regenerative medicine. This study investigates the effects of UC-MSC-derived EVs on dermal fibroblast function, and their potential in wound healing applications. EVs were characterized by [...] Read more.
Extracellular vesicles (EVs) secreted by umbilical cord-derived mesenchymal stem cells (UC-MSCs) hold significant promise as therapeutic agents in regenerative medicine. This study investigates the effects of UC-MSC-derived EVs on dermal fibroblast function, and their potential in wound healing applications. EVs were characterized by nanoparticle tracking analysis and transmission electron microscopy, revealing a mean size of 118.6 nm, consistent with exosomal properties. Dermal fibroblasts were treated with varying concentrations of EVs (25–100 µg/mL), and their impacts on cellular metabolism, mitochondrial activity, reactive oxygen species (ROS) production, wound closure, inflammatory cytokine secretion, growth factor production, and extracellular matrix (ECM) gene expression were evaluated. At lower concentrations (25–50 µg/mL), EVs significantly enhanced fibroblast metabolic and mitochondrial activity. However, higher concentrations (≥75 µg/mL) increased ROS levels, suggesting potential hormetic effects. EVs also modulated inflammation by reducing pro-inflammatory cytokines (IL-6, TNF-α) while promoting pro-regenerative cytokines (IL-33, TGF-β). Treatment with 50 µg/mL of EVs optimally stimulated wound closure and growth factor secretion (VEGF, BDNF, KGF, IGF), and upregulated ECM-related gene expression (type I and III collagen, fibronectin). These findings demonstrate that UC-MSC-derived EVs exert multifaceted effects on dermal fibroblast function, including enhanced cellular energetics, stimulation of cell migration, regulation of inflammation, promotion of growth factor production, and increased ECM synthesis. This study highlights the potential of EVs as a novel therapeutic strategy for wound healing and tissue regeneration, emphasizing the importance of optimizing EV concentration for maximal therapeutic efficacy. Full article
(This article belongs to the Section Cell Biology)
Show Figures

Figure 1

19 pages, 5112 KiB  
Article
Electrical Stimulation Generates Induced Tumor-Suppressing Cells, Offering a Potential Option for Combatting Breast Cancer and Bone Metastasis
by Changpeng Cui, Yinzhi Xu, Xue Xiong, Uma K. Aryal, Andy Chen, Stanley Chien, Lidan You, Baiyan Li and Hiroki Yokota
Int. J. Mol. Sci. 2025, 26(3), 1030; https://doi.org/10.3390/ijms26031030 - 25 Jan 2025
Viewed by 1583
Abstract
Treating advanced metastatic cancer, particularly with bone metastasis, remains a significant challenge. In previous studies, induced tumor-suppressing (iTS) cells were successfully generated through genetic, chemical, and mechanical interventions. This study investigates the potential of electrical stimulation to generate iTS cells. Using a custom [...] Read more.
Treating advanced metastatic cancer, particularly with bone metastasis, remains a significant challenge. In previous studies, induced tumor-suppressing (iTS) cells were successfully generated through genetic, chemical, and mechanical interventions. This study investigates the potential of electrical stimulation to generate iTS cells. Using a custom electrical stimulator with platinum electrodes, mesenchymal stem cells (MSCs) and Jurkat T cells were stimulated under optimized conditions (50 mV/cm, 10–100 Hz, 1 h). Conditioned medium (CM) from electrically stimulated cells demonstrated tumor-suppressing capabilities, inhibiting tumor cell migration, 3D spheroid growth, and cancer tissue fragment viability. Additionally, the CM reduced osteoclast maturation while promoting osteoblast differentiation. Proteomic analysis revealed enrichment of tumor-suppressing proteins, including histone H4, in the CM. Functional studies identified Piezo1 as a key mediator, as its knockdown significantly impaired the tumor-suppressive effects. Mechanistically, the process was distinct from other methods, such as mechanical vibration, with SUN1 inhibition showing no effect on iTS cell generation by electrical stimulation. These findings demonstrate the efficacy of electrical stimulation in enhancing the antitumor capabilities of MSCs and T cells, offering a novel approach to cancer therapy. Further exploration of this strategy could provide valuable insights into developing new treatments for metastatic cancer. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

Back to TopTop