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

Journals

Article Types

Countries / Regions

Search Results (173)

Search Parameters:
Keywords = human mesenchymal stromal cells (hMSC)

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 4170 KB  
Article
The Influence of Cisplatin on Functionality and Surface Characteristics of Mesenchymal Stromal Cells In Vitro
by Armin von Fournier, Totta Ehret Kasemo, Miguel Goncalves, Stephan Hackenberg, Marietta Herrmann, Marianne Schmidt, Manuel Stöth, Till Meyer, Thomas Gehrke and Agmal Scherzad
Int. J. Mol. Sci. 2026, 27(1), 76; https://doi.org/10.3390/ijms27010076 - 21 Dec 2025
Viewed by 75
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and play an important role in regenerative processes such as wound healing. Data on possible changes and functional restrictions of MSCs due to cisplatin chemotherapy, for example, in the treatment of head and neck cancer, diverge. The [...] Read more.
Mesenchymal stromal cells (MSCs) are multipotent and play an important role in regenerative processes such as wound healing. Data on possible changes and functional restrictions of MSCs due to cisplatin chemotherapy, for example, in the treatment of head and neck cancer, diverge. The aim of this study was to evaluate the influence of cisplatin on MSCs with regard to their defining characteristics and their ability to differentiate and to migrate. MSCs from four human donors (a 59-year-old man, a 63-year-old woman, a 70-year-old man, and a 61-year-old man) were cultured in vitro with and without cisplatin for 24 h, and toxic and subcytotoxic concentrations were determined using an MTT. We then examined the surface phenotype markers (flow cytometry), migration (scratch assay), histological differentiation markers (adipo-, chondro-, osteogenic), and the expression of selected line-associated genes in real-time quantitative PCR (RT-qPCR) (LEP, SOX9, RUNX2). These characteristics were evaluated after treatment with different subcytotoxic, clinically relevant doses of cisplatin. Flow cytometry confirmed the presence of MSCs-characteristic surface markers, which remained stable under treatment with subcytotoxic doses of cisplatin. Cisplatin exposure reduced the mRNA abundance of leptin (a marker for adipogenic differentiation) but increased SOX9 mRNA abundance (chondrogenic differentiation). RUNX (osteogenic differentiation) did not change post cisplatin exposure. Histological analysis showed no difference with regard to osteogenic, chondrogenic, and adipogenic differentiation at doses up to 10 μM cisplatin. Cell migration was not restricted by cisplatin exposure under the conditions used here. The characteristics of MSCs were not different to controls post cisplatin exposure. mRNA analysis suggested induced changes by cisplatin, although this effect was not histologically detectable even at high doses. Based on the single-molecule markers used here, indications for an inhibitory effect of cisplatin on adipogenic differentiation and a rather enhancing effect on chondrogenic and osteogenic differentiation may be hypothesized. The process observed here could further aggravate the already serious problem of malnutrition in head and neck cancer patients, for example. Taken together though, our study confirms overall MSCs tolerance towards cisplatin. Full article
(This article belongs to the Special Issue Toxicity of Metals, Metal-Based Drugs, and Microplastics)
Show Figures

Figure 1

17 pages, 3354 KB  
Article
Eco-Friendly Fabrication of Secretome-Loaded, Glutathione-Extended Waterborne Polyurethane Nanofibers
by Paolo Accardo, Francesco Cancilla, Annalisa Martorana, Filippo Calascibetta, Giandomenico Amico, Giovanna Pitarresi, Calogero Fiorica, Cinzia Maria Chinnici and Fabio Salvatore Palumbo
Int. J. Mol. Sci. 2025, 26(23), 11556; https://doi.org/10.3390/ijms262311556 - 28 Nov 2025
Viewed by 177
Abstract
The development of advanced delivery systems for bioactive factors is a critical focus in regenerative medicine and tissue engineering. In this study, we present a waterborne polyurethane (WPU)-based scaffold fabricated through a fully aqueous electrospinning process, providing a solvent-free and green method for [...] Read more.
The development of advanced delivery systems for bioactive factors is a critical focus in regenerative medicine and tissue engineering. In this study, we present a waterborne polyurethane (WPU)-based scaffold fabricated through a fully aqueous electrospinning process, providing a solvent-free and green method for delivering secretome derived from human mesenchymal stromal cells (MSCs). We optimized the electrospinning parameters to enable efficient secretome incorporation while preserving fiber morphology, sterility, and biocompatibility. The resulting membranes exhibited a uniform nanofibrous architecture, supported high cell viability, and demonstrated effective secretome loading and release, detected following release of vascular endothelial growth factor (VEGF)-A over 24 h. Overall, our findings highlight the potential of WPU nanofibrous scaffolds as sustainable and functional platforms for the delivery of MSC-derived bioactive factors in biomedical applications. Full article
Show Figures

Figure 1

19 pages, 6135 KB  
Communication
Transient Early Mechanical Loading Induces Hypertrophic Chondrocyte Differentiation of Human Mesenchymal Stromal Cells
by Sina Enzmann, Aline N. Klaus, Romano Matthys, Esther Wehrle, Martin J. Stoddart and Sophie Verrier
Cells 2025, 14(22), 1773; https://doi.org/10.3390/cells14221773 - 12 Nov 2025
Viewed by 398
Abstract
Optimal mechanical parameters for successful bone-healing remain unclear despite their critical influence on fracture outcomes, and the timing of post-surgery mobilization is still controversial despite many clinical observations and pre-clinical studies. In this bioreactor in vitro work, we investigate the effect of fundamental [...] Read more.
Optimal mechanical parameters for successful bone-healing remain unclear despite their critical influence on fracture outcomes, and the timing of post-surgery mobilization is still controversial despite many clinical observations and pre-clinical studies. In this bioreactor in vitro work, we investigate the effect of fundamental parameters such as timing, duration, and frequency of mechanical stimulation on the endochondral bone-healing paths, specifically on the hypertrophic chondrocyte differentiation of naïve human mesenchymal stromal cells (hMSCs). Human MSCs encapsulated in Gelatin-Methacryloyl hydrogels (GelMa) were subjected to three different 10% strain protocols: P1 (168 long-break cycles spread over 14 days), P2ce (cycle equivalent: 168 short-break cycles condensed in 42-min stimulation followed by 14 days free swelling), and P2te (time equivalent—14 days continuous stimulation, 80′640 short-break cycles). In the free-swelling control group, samples were cultured for 14 days without any mechanical stimulation. Our results confirmed that 10% strain induces a robust hypertrophic chondrocyte differentiation of naïve MSCs in all three tested protocols, as demonstrated by enlarged cell size, rounded morphology, robust upregulation of hypertrophic markers (COL10A1, MMP13, RUNX2, ALP), and reduced glycosaminoglycan production. Of particular interest, we show that P2ce (early short stimulation) was as effective as the two extended stimulation protocols, suggesting that initial mechanical signals are sufficient to trigger cell differentiation toward a hypertrophic chondrocyte phenotype that continues even after stimulation ceases. These in vitro findings provide crucial insights into the cellular basis of endochondral ossification during the early phase of loading and show a beneficial long-term effect of early mechanical stimulation. By demonstrating that the cellular mechanobiology of hypertrophic differentiation responds to brief early stimulation, our findings provide a scientific foundation to guide future in vivo investigations on how rehabilitation protocols could influence fracture healing. Full article
(This article belongs to the Section Tissues and Organs)
Show Figures

Graphical abstract

31 pages, 12238 KB  
Article
Micropatterning and Nanodropletting of Titanium by Shifted Surface Laser Texturing Significantly Enhances In Vitro Osteogenesis of Healthy and Osteoporotic Mesenchymal Stromal Cells
by Theresia Stich, Francisca Alagboso, Girish Pattappa, Jin Chu, Denys Moskal, Michal Povolný, Maximilian Saller, Veronika Schönitzer, Konstantin J. Scholz, Fabian Cieplik, Volker Alt, Maximilian Rudert, Tomáš Kovářík, Tomáš Křenek and Denitsa Docheva
J. Funct. Biomater. 2025, 16(11), 401; https://doi.org/10.3390/jfb16110401 - 27 Oct 2025
Viewed by 1087
Abstract
The key to proper implant integration in bone replacement is to orchestrate the complex interactions between materials and tissues. Moreover, due to the rapid demographic shift towards aging societies and the increase in elderly and osteoporotic patients, it is of great importance that [...] Read more.
The key to proper implant integration in bone replacement is to orchestrate the complex interactions between materials and tissues. Moreover, due to the rapid demographic shift towards aging societies and the increase in elderly and osteoporotic patients, it is of great importance that implant materials are osteointegrative in not only healthy but also compromised bone tissues. Here, titanium (Ti) scaffolds were subjected to shifted laser surface texturing (sLST) using a nanosecond pulsed laser to create an open pore macrotopography with micro-and nano-Ti droplets. In contrast to conventional laser texturing, which leads to high heat accumulation; in sLST, the frequency of laser pulses is low, allowing for resolidification, thereby creating a surface with abundant coverage micro-/nanodroplets. The main objective was to compare the cellular responses of human mesenchymal stromal cells (hMSCs) on sLST-textured Ti surfaces (LT-Ti) for the first time with standard sand-blasted, acid-etched surfaces (SLA-Ti). In-depth analyses of cell survival, proliferation, shape, mineralization, and gene expression were performed. Cell survival/proliferation was found to be similar on both surfaces; however, SEM imaging revealed differences in hMSC morphology. On LT-Ti, cells adopted well-rounded shapes, whereas on SLA-Ti they assumed more planar shapes. Bulk RNA sequencing performed after short-term culture on both surfaces disclosed expression changes in genes such as DUSP6, TNFSF12-TNFSF13 and SULT1A4. Remarkably, the osteogenic differentiation capacity of hMSCs was significantly enhanced on LT-Ti compared to SLA-Ti. Furthermore, aged/osteoporotic donor cohorts showed significantly enhanced matrix mineralization on LT-Ti. In conclusion, our novel results demonstrate that sLST-Ti surfaces are safe, highly biocompatible, can rescue patient-cohort-specific mineralization behavior, and therefore hold great potential for the development into next-generation implants, which are suitable for both the elderly and bone-compromised populations. Full article
(This article belongs to the Section Bone Biomaterials)
Show Figures

Graphical abstract

19 pages, 4195 KB  
Article
When Fat Talks: How Adipose-Derived Extracellular Vesicles Fuel Breast Cancer
by Maria Pia Cavaleri, Tommaso Pusceddu, Lucia Sileo, Luna Ardondi, Ilaria Vitali, Ilenia Pia Cappucci, Laura Basile, Giuseppe Pezzotti, Francesco Fiorica, Letizia Ferroni and Barbara Zavan
Int. J. Mol. Sci. 2025, 26(19), 9666; https://doi.org/10.3390/ijms26199666 - 3 Oct 2025
Cited by 1 | Viewed by 1108
Abstract
Adipose tissue plays a crucial role in the tumor microenvironment (TME), where its secreted extracellular vesicles (EVs) are involved in the complex signaling between tumor cells and surrounding stromal components. This study aims to unravel the mechanisms through which adipocyte-derived EVs influence breast [...] Read more.
Adipose tissue plays a crucial role in the tumor microenvironment (TME), where its secreted extracellular vesicles (EVs) are involved in the complex signaling between tumor cells and surrounding stromal components. This study aims to unravel the mechanisms through which adipocyte-derived EVs influence breast cancer (BC) progression. Human mesenchymal stem cells (hMSCs) were differentiated into adipocytes following a 21-day induction protocol that led to significant accumulation of lipid droplets within the cells. EVs were isolated from the conditioned medium of both hMSC-derived adipocytes and BC cells. Particle size distribution, morphology, and uptake into the recipient cell were investigated via nanoparticle tracking analysis, transmission electron microscopy, and fluorescence microscopy, respectively. Our results show that BC-derived EVs notably impaired cell viability and modulated the expression of key genes involved in apoptosis resistance within stromal cells. On the other hand, stromal-derived EVs significantly altered tumor cell behavior, indicating a dynamic, bidirectional exchange of bioactive signals. These findings underscore the pivotal role of EV-mediated communication in the tumor-stroma interplay, suggesting that adipocyte-cancer cell EV crosstalk contributes to the remodeling of the TME, potentially facilitating tumor progression. Full article
Show Figures

Figure 1

23 pages, 3798 KB  
Article
The Impact of IFN-γ Licensing on Mesenchymal Stromal Cells’ Mediated Immunoregulation and HLA Class II Expression: Emerging Evidence from In Vitro Results
by Panagiotis Mallis, Theofanis Chatzistamatiou, Evangelia Gkatzoflia, Hava Zdrava, Eirini-Faidra Sarri, Efstathios Michalopoulos, Alexandros Spyridonidis and Catherine Stavropoulos-Giokas
Int. J. Mol. Sci. 2025, 26(19), 9436; https://doi.org/10.3390/ijms26199436 - 26 Sep 2025
Viewed by 954
Abstract
Mesenchymal stromal cells (MSCs) exert their immunoregulatory properties after licensing by inflammatory signaling cues, e.g., interferon (IFN)-γ. However, MSCs licensing by IFN-γ may result in increased expression of human leukocyte antigen (HLA) class II, which is related to rapid cell elimination, impairment of [...] Read more.
Mesenchymal stromal cells (MSCs) exert their immunoregulatory properties after licensing by inflammatory signaling cues, e.g., interferon (IFN)-γ. However, MSCs licensing by IFN-γ may result in increased expression of human leukocyte antigen (HLA) class II, which is related to rapid cell elimination, impairment of their immunosuppressive properties, and patient sensitization. The aim of this study was to evaluate the impact of IFN-γ on mediated immunoregulation and HLA class II expression. In this study, Wharton’s jelly (WJ) MSCs were isolated from human umbilical cords. Well-defined WJ-MSCs were submitted to IFN-γ exposure, and after 96 h, evaluation of biomolecule secretion and HLA class II expression was performed. Typing of HLA alleles using a next-generation sequencing (NGS) platform was performed. IFN-γ-primed WJ-MSCs secreted a high amount of immunoregulatory biomolecules, while elevated expression of HLA-DRB1 was observed. Analyses the NGS results showed the possibility of WJ-MSCs cluster formation based on their frequency of detected HLA alleles and immunoregulatory potential. Taking into consideration that IFN-γ-primed WJ-MSCs express HLA class II alleles, it is suggested that the HLA histocompatibility between allogeneic donor and recipient should be strongly considered to acquire the most beneficial outcome for the MSCs therapeutic strategy. Full article
Show Figures

Figure 1

17 pages, 3152 KB  
Article
Optimizing the Infusion Route of Human Bone Marrow Mesenchymal Stromal Cells to Mitigate Liver Ischemia–Reperfusion Injury in a Porcine Model
by Stefan H. Luijmes, Job P. van Kooten, Henk P. Roest, Jubi de Haan, Michail Doukas, Cornelia J. Verhoeven, Kairong Wang, Jorke Willemse, Luc J. W. van der Laan, Monique M. A. Verstegen and Jeroen de Jonge
Cells 2025, 14(19), 1496; https://doi.org/10.3390/cells14191496 - 24 Sep 2025
Viewed by 1072
Abstract
Mesenchymal stromal cells (MSC) have been shown to mitigate IRI through their anti-inflammatory and immune-modulating capacities. This study aims to demonstrate the feasibility, safety, and effectiveness of hepatic administration of bone marrow-derived (BM)-MSCs in a large pig model relevant to human anatomy. After [...] Read more.
Mesenchymal stromal cells (MSC) have been shown to mitigate IRI through their anti-inflammatory and immune-modulating capacities. This study aims to demonstrate the feasibility, safety, and effectiveness of hepatic administration of bone marrow-derived (BM)-MSCs in a large pig model relevant to human anatomy. After complete vascular exclusion for 45 min, 3 × 106 human BM-MSCs/kg body weight were infused via the portal vein or hepatic artery. BM-MSC infusion did not cause obstruction of hepatic or pulmonary blood flow within 6 h after infusion. Cells were effectively retained in the liver, being undetectable in peripheral blood, lung, and spleen samples. Human B2M expression, as a marker for BM-MSC presence, was significantly higher for the left liver lobe in arterial infusion compared to portal infusion. In liver samples with high BM-MSC levels, we identified the prevention of up- or downregulation of some genes related to inflammation and energy metabolism that was present in non-treated control samples, indicating biological effects within 6 h of infusion. We conclude that hepatic BM-MSC infusion is feasible and safe, with the hepatic artery serving as the optimal administration route for homogenous distribution. These findings pave the way for clinical studies on MSC infusion in IRI, either in situ in liver conditions or ex situ during machine perfusion. Full article
Show Figures

Figure 1

19 pages, 3181 KB  
Article
Overexpression of BDNF and uPA Combined with the Suppression of Von Hippel–Lindau Tumor Suppressor Enhances the Neuroprotective Activity of the Secretome of Human Mesenchymal Stromal Cells in the Model of Intracerebral Hemorrhage
by Stalik S. Dzhauari, Alexandra L. Primak, Nataliya A. Basalova, Natalia I. Kalinina, Anna O. Monakova, Kirill D. Bozov, Arkadiy Ya. Velichko, Maria E. Illarionova, Olga A. Grigorieva, Zhanna A. Akopyan, Vladimir S. Popov, Pavel G. Malkov, Anastasia Yu. Efimenko, Vsevolod A. Tkachuk and Maxim N. Karagyaur
Int. J. Mol. Sci. 2025, 26(14), 6697; https://doi.org/10.3390/ijms26146697 - 12 Jul 2025
Cited by 1 | Viewed by 1102
Abstract
Nerve tissue damage is an unsolved problem in modern neurology and neurosurgery, which prompts the need to search for approaches to stimulate neuroprotection and regeneration of neural tissue. Earlier we have shown that the secretome of human mesenchymal stromal cells (MSCs) stimulates rat [...] Read more.
Nerve tissue damage is an unsolved problem in modern neurology and neurosurgery, which prompts the need to search for approaches to stimulate neuroprotection and regeneration of neural tissue. Earlier we have shown that the secretome of human mesenchymal stromal cells (MSCs) stimulates rat survival, reduces the severity of neurological deficits, and decreases the volume of brain damage in a hemorrhagic stroke model. A significant disadvantage of using the MSC secretome is the need to concentrate it (at least 5–10 fold) to achieve appreciable pharmacological activity. This increases the cost of obtaining clinically applicable amounts of secretome and slows down the clinical translation of this technology. Here, we created a number of genetically modified human MSC cultures, including immortalized MSCs and those with hyperexpression of brain-derived neurotrophic factor (BDNF) and urokinase-type plasminogen activator (uPA) and with suppressed expression of Von Hippel–Lindau tumor suppressor (VHL), and we evaluated the pharmacological activity of their secretomes in a model of intracerebral hemorrhage (ICH) in rats. The secretome of MSCs immortalized by hyperexpression of the catalytic subunit of human telomerase (hTERT) revealed neuroprotective activity indistinguishable from that of primary MSC cultures, yet it still required 10-fold concentration to achieve neuroprotective efficacy. The secretome of MSC culture with combined hyperexpression of BDNF and uPA and suppressed expression of Von Hippel–Lindau tumor suppressor even without additional concentration reduced the severity of neurological disorders and decreased brain lesion volume in the ICH model. The secretomes of MSCs with separate overexpression of BDNF and uPA or suppression of VHL had no such effect or, on the contrary, revealed a toxic effect in the ICH model. Presumably, this may be due to an imbalance in the representation of individual growth factors in the secretome of genetically modified MSCs, which individually may lead to undesirable effects in damaged nervous tissue, such as increased permeability of the blood–brain barrier (under the influence of pro-angiogenic factors) or neural cell apoptosis (due to an excess of neurotrophic factors). The obtained data show that genetic modification of MSC cultures can enhance or alter the therapeutic activity of their secretomes, which can be used in the creation of promising sources of biopharmaceutical substances. Full article
Show Figures

Figure 1

19 pages, 3292 KB  
Article
Phenothiazine-Based Nanoaggregates: Dual Role in Bioimaging and Stem Cell-Driven Photodynamic Therapy
by Eleonora Calzoni, Alessio Cesaretti, Nicolò Montegiove, Maria Luisa Valicenti, Francesco Morena, Rajneesh Misra, Benedetta Carlotti and Sabata Martino
Nanomaterials 2025, 15(12), 894; https://doi.org/10.3390/nano15120894 - 10 Jun 2025
Cited by 3 | Viewed by 848
Abstract
Nanotechnology is transforming contemporary medicine by providing cutting-edge tools for the treatment and diagnosis of complex disorders. Advanced techniques such as bioimaging and photodynamic therapy (PDT) combine early diagnosis and targeted therapy, offering a more precise approach than conventional treatments. However, a significant [...] Read more.
Nanotechnology is transforming contemporary medicine by providing cutting-edge tools for the treatment and diagnosis of complex disorders. Advanced techniques such as bioimaging and photodynamic therapy (PDT) combine early diagnosis and targeted therapy, offering a more precise approach than conventional treatments. However, a significant obstacle for PDT is the need to selectively deliver photosensitizers to disease sites while minimizing systemic side effects. In this context, mesenchymal stem cells have emerged as promising biological carriers due to their natural tropism towards tumors, low immunogenicity, and their ability to overcome biological barriers. In this study, two push–pull compounds, NPI-PTZ and BTZ-PTZ, phenothiazine derivatives featuring aggregation-induced emission (AIE) abilities, were analyzed. These molecules proved to be excellent fluorescent probes and photosensitizing agents. When administered to human bone marrow-derived multipotent stromal cells (hBM-MSCs) and human adipose multipotent stem cells (hASCs), the compounds were efficiently internalized, maintained a stable fluorescent emission for several days, and showed phototoxicity after irradiation, without inducing major cytotoxic effects under normal conditions. These results highlight the potential of NPI-PTZ and BTZ-PTZ combined with mesenchymal stem cells as theranostic tools, bridging bioimaging and PDT, and suggest new possibilities for advanced therapeutic approaches in clinical applications. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Graphical abstract

29 pages, 4161 KB  
Article
Three-Dimensional Bioprinted Gelatin—Genipin Hydrogels Enriched with hUCMSC-Derived Small Extracellular Vesicles for Regenerative Wound Dressings
by Manal Hussein Taghdi, Maimonah Eissa Al-Masawa, Barathan Muttiah, Mh Busra Fauzi, Jia Xian Law, Ani Amelia Zainuddin and Yogeswaran Lokanathan
Polymers 2025, 17(9), 1163; https://doi.org/10.3390/polym17091163 - 24 Apr 2025
Cited by 4 | Viewed by 1862
Abstract
Mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) have shown great promise in promoting tissue repair, including skin wound healing, but challenges like rapid degradation and short retention have limited their clinical application. Hydrogels have emerged as effective carriers for sustained EV release. Three-dimensional [...] Read more.
Mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) have shown great promise in promoting tissue repair, including skin wound healing, but challenges like rapid degradation and short retention have limited their clinical application. Hydrogels have emerged as effective carriers for sustained EV release. Three-dimensional printing enables the development of personalized skin substitutes tailored to the wound size and shape. This study aimed to develop 3D bioprinted gelatin–genipin hydrogels incorporating human umbilical cord MSC-sEVs (hUCMSC-sEVs) for future skin wound healing applications. Gelatin hydrogels (8% and 10% w/v) were crosslinked with 0.3% genipin (GECL) to improve stability. The hydrogels were evaluated for their suitability for extrusion-based 3D bioprinting and physicochemical properties, such as the swelling ratio, hydrophilicity, enzymatic degradation, and water vapor transmission rate (WVTR). Chemical characterization was performed using EDX, XRD, and FTIR. The hUCMSC-sEVs were isolated via centrifugation and tangential flow filtration (TFF) and characterized. The crosslinked hydrogels were successfully 3D bioprinted and demonstrated superior properties, including high hydrophilicity, a swelling ratio of ~500%, slower degradation, and optimal WVTR. hUCMSC-sEVs, ranging from 50 to 200 nm, were positive for surface and cytosolic markers. Adding 75 μg/mL of hUCMSC-EVs into 10% GECL hydrogels significantly improved the biocompatibility. These hydrogels offer ideal properties for 3D bioprinting and wound healing, demonstrating their potential as biomaterial scaffolds for skin tissue regeneration applications. Full article
(This article belongs to the Special Issue Smart and Bio-Medical Polymers: 2nd Edition)
Show Figures

Figure 1

18 pages, 6069 KB  
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
Cited by 2 | Viewed by 1061
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

21 pages, 6805 KB  
Article
Evaluation of the Anti-Cancer Potential of Extracellular Vesicles Derived from Human Amniotic Fluid Stem Cells: Focus on Effective miRNAs in the Treatment of Melanoma Progression
by Martina Gatti, Francesca Beretti, Gloria Ravegnini, Francesca Gorini, Eleonora Ceneri, Emma Bertucci, Matilde Y. Follo and Tullia Maraldi
Int. J. Mol. Sci. 2024, 25(23), 12502; https://doi.org/10.3390/ijms252312502 - 21 Nov 2024
Cited by 2 | Viewed by 1949
Abstract
Mesenchymal stromal cells (MSCs) and their secretome show intrinsic antitumor properties, however, the anti-cancer effects of MSCs remain debated and depend on the cancer type or model. MSCs derived from discarded samples, such as human amniotic fluid (hAFSC), have been introduced as an [...] Read more.
Mesenchymal stromal cells (MSCs) and their secretome show intrinsic antitumor properties, however, the anti-cancer effects of MSCs remain debated and depend on the cancer type or model. MSCs derived from discarded samples, such as human amniotic fluid (hAFSC), have been introduced as an attractive and potent stem cell source for clinical applications due to their collection procedures, which minimize ethical issues. Until now, various studies have obtained controversial results and poor understanding of the mechanisms behind the effects of perinatal cells on cancer cells. To better clarify this aspect, protein and miRNA expression profiling isolated from Extracellular vesicles (EVs) secreted by hAFSCs, obtained in the II or III trimester, were evaluated. Bioinformatic analysis was performed aiming at evaluating differential expression, pathway enrichment, and miRNA-mRNA networks. We highlighted that most of the highest expressed proteins and miRNAs are mainly involved in antioxidant and anti-cancer effects. Indeed, in the presence of hAFSC-EVs, a reduction of the G2/M phase was observed on melanoma cell lines, an activation of the apoptotic pathway occurred and the migration and invasion ability reduced. Our data demonstrated that II or III trimester hAFSCs can release bioactive factors into EVs, causing an efficient anti-cancer effect inhibiting melanoma progression. Full article
Show Figures

Figure 1

26 pages, 6323 KB  
Article
Design and Characterization of Biomimetic Hybrid Construct Based on Hyaluronic Acid and Alginate Bioink for Regeneration of Articular Cartilage
by Cristina Galocha-León, Cristina Antich, Beatriz Clares-Naveros, Ana Voltes-Martínez, Juan Antonio Marchal and Patricia Gálvez-Martín
Pharmaceutics 2024, 16(11), 1422; https://doi.org/10.3390/pharmaceutics16111422 - 7 Nov 2024
Cited by 4 | Viewed by 2872
Abstract
Background/Objectives: Three-dimensional bioprinting technology has enabled great advances in the treatment of articular cartilage (AC) defects by the biofabrication of biomimetic constructs that restore and/or regenerate damaged tissue. In this sense, the selection of suitable cells and biomaterials to bioprint constructs that mimic [...] Read more.
Background/Objectives: Three-dimensional bioprinting technology has enabled great advances in the treatment of articular cartilage (AC) defects by the biofabrication of biomimetic constructs that restore and/or regenerate damaged tissue. In this sense, the selection of suitable cells and biomaterials to bioprint constructs that mimic the architecture, composition, and functionality of the natural extracellular matrix (ECM) of the native tissue is crucial. In the present study, a novel cartilage-like biomimetic hybrid construct (CBC) was developed by 3D bioprinting to facilitate and promote AC regeneration. Methods: The CBC was biofabricated by the co-bioprinting of a bioink based on hyaluronic acid (HA) and alginate (AL) loaded with human mesenchymal stromal cells (hMSCs), with polylactic acid supporting the biomaterial, in order to mimic the microenvironment and structural properties of native AC, respectively. The CBC was biologically in vitro characterized. In addition, its physiochemical characteristics were evaluated in order to determine if the presence of hMSCs modified its properties. Results: Results from biological analysis demonstrated that CBC supported the high viability and proliferation of hMSCs, facilitating chondrogenesis after 5 weeks in vitro. The evaluation of physicochemical properties in the CBCs confirmed that the CBC developed could be suitable for use in cartilage tissue engineering. Conclusions: The results demonstrated that the use of bioprinted CBCs based on hMSC-AL/HA-bioink for AC repair could enhance the regeneration and/or formation of hyaline cartilaginous tissue. Full article
(This article belongs to the Special Issue 3D Bioprinted Scaffolds for Tissue Engineering)
Show Figures

Figure 1

18 pages, 15413 KB  
Article
Insights into Osteogenesis Induced by Crude Brassicaceae Seeds Extracts: A Role for Glucosinolates
by Laura Gambari, Eleonora Pagnotta, Luisa Ugolini, Laura Righetti, Emanuela Amore, Brunella Grigolo, Giuseppe Filardo and Francesco Grassi
Nutrients 2024, 16(20), 3457; https://doi.org/10.3390/nu16203457 - 12 Oct 2024
Cited by 1 | Viewed by 1877
Abstract
Background/Objectives: Crude extracts from the Brassica genus have recently emerged as promising phytochemicals for preventing bone loss. While the most documented evidence suggests that their general biological activity is due to glucosinolates’ (GLSs’) hydrolysis products, the direct activity of GLSs is beginning [...] Read more.
Background/Objectives: Crude extracts from the Brassica genus have recently emerged as promising phytochemicals for preventing bone loss. While the most documented evidence suggests that their general biological activity is due to glucosinolates’ (GLSs’) hydrolysis products, the direct activity of GLSs is beginning to be uncovered. However, the contribution of GLSs to the bone-sparing activity of crude Brassicaceae extracts has seldom been addressed. Here, we aimed to gain insights into this gap by studying in the same in vitro model of human osteogenesis the effect of two Brassica seed extracts (Eruca sativa and Lepidium sativum) obtained from defatted seed meals, comparing them to the isolated GLSs most represented in their composition, glucoerucin (GER) and glucotropaeolin (GTL), for Eruca sativa and Lepidium sativum, respectively. Methods: Osteogenic differentiation of human mesenchymal stromal cells (hMSCs) was assessed by alizarin red staining assay and real-time PCR, respectively, evaluating mineral apposition and mRNA expression of specific osteogenic genes. Results: Both Brassica extracts and GLSs increased the osteogenic differentiation, indicating that the stimulating effect of Brassica extracts can be at least partially attributed to GLSs. Moreover, these data extend previous evidence of the effect of unhydrolyzed glucoraphanin (GRA) on osteogenesis to other types of GLSs: GER and GTL. Notably, E. sativa extract and GTL induced higher osteogenic stimulation than Lepidium sativum extract and GER, respectively. Conclusions: Overall, this study expands the knowledge on the possible application of Brassica-derived bioactive molecules as natural alternatives for the prevention and treatment of bone-loss pathologies. Full article
(This article belongs to the Special Issue Bioactive Ingredients in Plants Related to Human Health)
Show Figures

Figure 1

20 pages, 3440 KB  
Article
The Comparison of Immunomodulatory Properties of Canine and Human Wharton Jelly-Derived Mesenchymal Stromal Cells
by Anna Burdzinska, Iwona Monika Szopa, Kinga Majchrzak-Kuligowska, Aleksander Roszczyk, Katarzyna Zielniok, Paweł Zep, Filip Andrzej Dąbrowski, Tanushree Bhale, Marek Galanty and Leszek Paczek
Int. J. Mol. Sci. 2024, 25(16), 8926; https://doi.org/10.3390/ijms25168926 - 16 Aug 2024
Cited by 3 | Viewed by 2556
Abstract
Although therapies based on mesenchymal stromal cells (MSCs) are being implemented in clinical settings, many aspects regarding these procedures require further optimization. Domestic dogs suffer from numerous immune-mediated diseases similar to those found in humans. This study aimed to assess the immunomodulatory activity [...] Read more.
Although therapies based on mesenchymal stromal cells (MSCs) are being implemented in clinical settings, many aspects regarding these procedures require further optimization. Domestic dogs suffer from numerous immune-mediated diseases similar to those found in humans. This study aimed to assess the immunomodulatory activity of canine (c) Wharton jelly (WJ)-derived MSCs and refer them to human (h) MSCs isolated from the same tissue. Canine MSC(WJ)s appeared to be more prone to in vitro aging than their human counterparts. Both canine and human MSC(WJ)s significantly inhibited the activation as well as proliferation of CD4+ and CD8+ T cells. The treatment with IFNγ significantly upregulated indoleamine-2,3-dioxygenase 1 (IDO1) synthesis in human and canine MSC(WJ)s, and the addition of poly(I:C), TLR3 ligand, synergized this effect in cells from both species. Unstimulated human and canine MSC(WJ)s released TGFβ at the same level (p > 0.05). IFNγ significantly increased the secretion of TGFβ in cells from both species (p < 0.05); however, this response was significantly stronger in human cells than in canine cells. Although the properties of canine and human MSC(WJ)s differ in detail, cells from both species inhibit the proliferation of activated T cells to a very similar degree and respond to pro-inflammatory stimulation by enhancing their anti-inflammatory activity. These results suggest that testing MSC transplantation in naturally occurring immune-mediated diseases in dogs may have high translational value for human clinical trials. Full article
(This article belongs to the Special Issue Biomedical Applications of Mesenchymal Stem Cells)
Show Figures

Figure 1

Back to TopTop