Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (80)

Search Parameters:
Keywords = MSC immune suppression

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 1103 KB  
Review
Therapeutic Potential of Chimeric Antigen Receptor-Expressing Mesenchymal Stem Cells in the Treatment of Inflammatory and Autoimmune Diseases
by Vladislav Volarevic, Carl Randall Harrell, Crissy Fellabaum, Valentin Djonov and Ana Volarevic
Int. J. Mol. Sci. 2025, 26(16), 7795; https://doi.org/10.3390/ijms26167795 - 12 Aug 2025
Viewed by 725
Abstract
Chimeric antigen receptor-engineered mesenchymal stem cells (CAR-MSCs) represent a novel and highly adaptable platform for the targeted treatment of inflammatory and autoimmune diseases. By integrating the inflammation-homing and immunomodulatory properties of mesenchymal stem cells (MSCs) with the antigen-specific recognition and activation potential of [...] Read more.
Chimeric antigen receptor-engineered mesenchymal stem cells (CAR-MSCs) represent a novel and highly adaptable platform for the targeted treatment of inflammatory and autoimmune diseases. By integrating the inflammation-homing and immunomodulatory properties of mesenchymal stem cells (MSCs) with the antigen-specific recognition and activation potential of chimeric antigen receptors (CARs), CAR-MSCs enable site-specific delivery of therapeutic agents directly to inflamed or diseased tissues. This dual functionality enhances therapeutic precision while minimizing off-target effects and systemic toxicity. Recent preclinical studies have demonstrated the efficacy of CAR-MSCs in modulating pathogenic immune responses, reducing local inflammation, and promoting tissue repair in various disease models. CAR-MSCs have been engineered to recognize and interact with disease-specific antigens or inflammatory markers, allowing them to selectively suppress the activation and proliferation of autoreactive immune cells. This targeted immunosuppression offers a promising strategy for restoring immune tolerance without the risks associated with systemic immunosuppression. In this review, we provide a comprehensive overview of recent developments in CAR-MSC design, highlight mechanisms by which CARs enhance MSC functionality, and discuss key challenges, including safety, scalability, and regulatory considerations. Collectively, these emerging approaches hold substantial promise for reshaping future therapies for inflammatory and autoimmune diseases. Full article
(This article belongs to the Special Issue Therapeutic Uses of Adult Stem Cells)
Show Figures

Figure 1

25 pages, 4674 KB  
Review
Research Progress on Icariin Promoting Bone Injury Repair and Regeneration
by Weijian Hu, Yameng Si, Xin Xie and Jiabin Xu
Pharmaceuticals 2025, 18(8), 1174; https://doi.org/10.3390/ph18081174 - 8 Aug 2025
Cited by 1 | Viewed by 1758
Abstract
Icariin (ICA) is a bioactive flavonoid compound extracted from Epimedium plants. In recent years, it has attracted significant research interest in the field of bone tissue repair due to its pharmacological effects via multiple targets and pathways. Studies have shown that ICA promotes [...] Read more.
Icariin (ICA) is a bioactive flavonoid compound extracted from Epimedium plants. In recent years, it has attracted significant research interest in the field of bone tissue repair due to its pharmacological effects via multiple targets and pathways. Studies have shown that ICA promotes the osteogenic differentiation of mesenchymal stem cells (MSCs) and enhances bone matrix formation by regulating signaling pathways such as Akt and Wnt/β-catenin. It concurrently inhibits osteoclast activity to maintain the balance of bone remodeling, thereby simultaneously stimulating new bone regeneration and suppressing bone resorption. At the same time, ICA exerts potent anti-inflammatory and antioxidant effects and promotes angiogenesis, improving the local microenvironment of bone injury and significantly facilitating the regeneration of bone and cartilage tissues. Additionally, ICA exhibits notable protective effects in multiple organ systems including the cardiovascular, hepatic, renal, and nervous systems. Specifically, ICA reduces cardiomyocyte apoptosis and fibrosis to preserve cardiac function, improves hepatic metabolic function and alleviates oxidative stress, attenuates renal inflammation and fibrosis, and—through neuroprotective actions—reduces neuroinflammation and promotes neuronal survival. These multi-organ effects help optimize the systemic environment for bone healing. However, ICA faces significant pharmacokinetic challenges. It has low oral bioavailability (due to poor absorption and extensive first-pass metabolism) as well as a short half-life. Consequently, maintaining effective drug concentrations in vivo is difficult, which limits its therapeutic efficacy and impedes clinical translation. To fully realize its regenerative potential, advanced drug delivery strategies (e.g., nanocarrier-based delivery systems) are being explored to enhance ICA’s bioavailability and prolong its duration of action. Overall, ICA’s multi-modal actions on bone cells, the immune microenvironment, and systemic factors make it a promising multi-target agent for bone regeneration. Addressing its pharmacokinetic limitations through optimized delivery and conducting further clinical studies will be crucial to realize its full therapeutic potential. This review provides a comprehensive overview of recent advances and challenges in translating ICA’s benefits into orthopedic therapy. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

23 pages, 8937 KB  
Article
Neuro-Cells Mitigate Amyloid Plaque Formation and Behavioral Deficits in the APPswe/PS1dE9 Model of Alzheimer Disease While Also Reducing IL-6 Production in Human Monocytes
by Johannes de Munter, Kirill Chaprov, Ekkehard Lang, Kseniia Sitdikova, Erik Ch. Wolters, Evgeniy Svirin, Aliya Kassenova, Andrey Tsoy, Boris W. Kramer, Sholpan Askarova, Careen A. Schroeter, Daniel C. Anthony and Tatyana Strekalova
Cells 2025, 14(15), 1168; https://doi.org/10.3390/cells14151168 - 29 Jul 2025
Viewed by 696
Abstract
Neuroinflammation is a key feature of Alzheimer’s disease (AD), and stem cell therapies have emerged as promising candidates due to their immunomodulatory properties. Neuro-Cells (NC), a combination of unmodified mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), have demonstrated therapeutic potential in [...] Read more.
Neuroinflammation is a key feature of Alzheimer’s disease (AD), and stem cell therapies have emerged as promising candidates due to their immunomodulatory properties. Neuro-Cells (NC), a combination of unmodified mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), have demonstrated therapeutic potential in models of central nervous system (CNS) injury and neurodegeneration. Here, we studied the effects of NC in APPswe/PS1dE9 mice, an AD mouse model. Twelve-month-old APPswe/PS1dE9 mice or their wild-type littermates were injected with NC or vehicle into the cisterna magna. Five to six weeks post-injection, cognitive, locomotor, and emotional behaviors were assessed. The brain was stained for amyloid plaque density using Congo red, and for astrogliosis using DAPI and GFAP staining. Gene expression of immune activation markers (Il-1β, Il-6, Cd45, Tnf) and plasticity markers (Tubβ3, Bace1, Trem2, Stat3) was examined in the prefrontal cortex. IL-6 secretion was measured in cultured human monocytes following endotoxin challenge and NC treatment. Untreated APPswe/PS1dE9 mice displayed impaired learning in the conditioned taste aversion test, reduced object exploration, and anxiety-like behavior, which were improved in the NC-treated mutants. NC treatment normalized the expression of several immune and plasticity markers and reduced the density of GFAP-positive cells in the hippocampus and thalamus. NC treatment decreased amyloid plaque density in the hippocampus and thalamus, targeting plaques of <100 μm2. Additionally, NC treatment suppressed IL-6 secretion by human monocytes. Thus, NC treatment alleviated behavioral deficits and reduced amyloid plaque formation in APPswe/PS1dE9 mice, likely via anti-inflammatory mechanisms. The reduction in IL-6 production in human monocytes further supports the potential of NC therapy for the treatment of AD. Full article
Show Figures

Figure 1

15 pages, 1845 KB  
Article
In Vitro Investigation of Statin Effects on Genes Associated with Severe COVID-19 in Cancerous and Non-Cancerous Cells
by Adriana Kapustová, Patrik Macášek, Bibiána Baďurová, Jana Melegová, Silvie Rimpelová, Jan Kubovčiak, Jana Šáchová, Miluše Hradilová, Michal Kolář, Libor Vítek, Tomáš Ruml and Helena Gbelcová
Biomedicines 2025, 13(7), 1714; https://doi.org/10.3390/biomedicines13071714 - 14 Jul 2025
Viewed by 669
Abstract
Background: The progressive course of coronavirus disease 2019 (COVID-19) is more frequently observed in individuals with obesity, diabetes, pulmonary and cardiovascular disease, or arterial hypertension. Many patients with these conditions are prescribed statins to treat hypercholesterolaemia. However, statins exhibit additional pleiotropic effects. The [...] Read more.
Background: The progressive course of coronavirus disease 2019 (COVID-19) is more frequently observed in individuals with obesity, diabetes, pulmonary and cardiovascular disease, or arterial hypertension. Many patients with these conditions are prescribed statins to treat hypercholesterolaemia. However, statins exhibit additional pleiotropic effects. The present study aims to investigate the effects of all eight currently existing statins on the expression of genes whose products have been reported to be directly associated with complicated COVID-19 disease. Methods: We extended the interpretation of the whole-genome DNA microarray analyses of pancreatic cancer cells MiaPaCa-2 and whole-transcriptome analyses of adipose tissue-derived mesenchymal stem cells AD-MSC that we had performed in the past. From the number of genes with altered expression induced by statins, we focused on those reported to be involved in a complicated course of COVID-19, including APOE and ACE2, genes encoding proteins involved in innate antiviral immunity and respiratory failure genes. Results: Although we did not observe statin-induced changes in the expression of APOE, ACE2 and any of the six genes clustered in the locus associated with respiratory failure in patients with COVID-19, some statins induced changes in the expression of genes encoding their interaction partners. Among genes associated with the immune system, all statins, which are effective in vitro affected the expression of genes encoding IL-6 and IL-8 and interaction partners of NF-kB, which may influence the duration of viral persistence. Conclusions: Statins act on multiple pathways simultaneously, some of which support COVID-19 development, while others suppress it. Full article
Show Figures

Figure 1

15 pages, 719 KB  
Review
Mesenchymal Stem-Cell-Derived Exosomes and MicroRNAs: Advancing Cell-Free Therapy in Systemic Sclerosis
by Cristiano Barbetta, Francesco Bonomi, Gemma Lepri, Daniel E. Furst, Silvia Bellando Randone and Serena Guiducci
Cells 2025, 14(13), 1018; https://doi.org/10.3390/cells14131018 - 3 Jul 2025
Viewed by 1864
Abstract
Mesenchymal stem cell (MSC) transplantation has emerged as a potential therapeutic strategy for systemic sclerosis (SSc), a rare autoimmune disease characterized by inflammation, fibrosis, and vasculopathy. Recent evidence suggests that the therapeutic benefits of MSCs do not depend directly on their ability to [...] Read more.
Mesenchymal stem cell (MSC) transplantation has emerged as a potential therapeutic strategy for systemic sclerosis (SSc), a rare autoimmune disease characterized by inflammation, fibrosis, and vasculopathy. Recent evidence suggests that the therapeutic benefits of MSCs do not depend directly on their ability to proliferate but rather on their capacity to release extracellular nanovesicles known as exosomes (MSC-Exos). MSC-Exos are rich in bioactive molecules such as microRNAs, which can modulate gene expression and trigger significant biological responses, playing a central role in modulating immune responses, inhibiting fibrotic pathways and promoting tissue repair and angiogenesis. Preclinical studies have demonstrated that MSC-Exos can attenuate fibrosis, modulate macrophage polarization, suppress autoreactive lymphocyte activity, and even reverse pulmonary arterial hypertension in animal models of SSc. Compared to cell-based therapies, MSC-Exos offer several advantages, including lower immunogenicity and better safety profile. This review provides an overview of the immunomodulatory, antifibrotic, and angiogenic properties of MSC-Exos and explores their potential as novel cell-free therapy for SSc. Full article
Show Figures

Figure 1

20 pages, 3807 KB  
Review
Effects of Mesenchymal Stem Cells on Functions of Chimeric Antigen Receptor-Expressing T Lymphocytes and Natural Killer Cells
by Vladislav Volarevic, Carl Randall Harrell, Aleksandar Arsenijevic, Valentin Djonov and Ana Volarevic
Cells 2025, 14(13), 978; https://doi.org/10.3390/cells14130978 - 25 Jun 2025
Viewed by 942
Abstract
Chimeric antigen receptor (CAR)-engineered immune cells, particularly CAR T lymphocytes and CAR natural killer (NK) cells, have revolutionized cancer immunotherapy. However, their therapeutic efficacy and safety can be influenced by the tumor microenvironment, particularly the presence of mesenchymal stem cells (MSCs). MSCs are [...] Read more.
Chimeric antigen receptor (CAR)-engineered immune cells, particularly CAR T lymphocytes and CAR natural killer (NK) cells, have revolutionized cancer immunotherapy. However, their therapeutic efficacy and safety can be influenced by the tumor microenvironment, particularly the presence of mesenchymal stem cells (MSCs). MSCs are immunomodulatory cells which can alter the function of tumor-infiltrated immune cells in both supportive and suppressive ways. Results obtained in recently conducted experimental studies demonstrate that MSCs modulate proliferation, cytotoxicity, cytokine production and anti-tumor activity in CAR-expressing immune cells in both a juxtacrine and a paracrine manner. While MSCs can enhance CAR cell viability and persistence through trophic support, they may also impair cytotoxic function and promote an immunosuppressive phenotype under certain conditions. Understanding the dualistic nature of MSCs in CAR-based immunotherapy for malignant diseases is critical for optimizing clinical outcomes. Additionally, MSCs may serve as vehicles for targeted delivery of immunomodulatory agents, and should be considered as active components in the design of next-generation CAR-based immunotherapies. Accordingly, in this review article we emphasize molecular and cellular mechanisms involved in MSC-dependent modulation of CAR-expressing immune cells, paving the way for more efficient CAR-based immunotherapy for malignant diseases. Full article
(This article belongs to the Special Issue Immunoregulatory Functions of Mesenchymal Stem Cells (MSCs))
Show Figures

Figure 1

14 pages, 16089 KB  
Article
Effects of Adipose-Derived Mesenchymal Stem Cell-Secretome on Pyroptosis of Laparoscopic Hepatic Ischemia Reperfusion Injury in a Porcine Model
by Yajun Ma, Lei Cao, Pujun Li, Zhihui Jiao, Xiaoning Liu, Xiangyu Lu, Tao Liu and Hongbin Wang
Cells 2025, 14(10), 722; https://doi.org/10.3390/cells14100722 - 15 May 2025
Cited by 1 | Viewed by 795
Abstract
Extensive research has been conducted on mesenchymal stem cells (MSCs) regarding their ability to modify the immune response and reduce tissue damage. Many researchers have found that the regulatory capacity of MSCs primarily comes from their secretome. As a result, there has been [...] Read more.
Extensive research has been conducted on mesenchymal stem cells (MSCs) regarding their ability to modify the immune response and reduce tissue damage. Many researchers have found that the regulatory capacity of MSCs primarily comes from their secretome. As a result, there has been much interest in utilizing “cell-free” therapies as alternatives to stem cell treatments. In this study, the secretome from adipose mesenchymal stem cells (ADSC-secretome) was extracted and injected into minipigs with established liver injury models. Blood and liver tissue samples were obtained prior to the procedure, as well as on days 1, 3, and 7 after surgery. It was found that ADSC-secretome effectively suppressed the synthesis of the NOD-like receptor protein 3 (NLRP3) inflammasome, leading to a downregulation of gasdermin-D (GSDMD) expression, and demonstrated a more prominent anti-pyroptosis effect compared to ADSCs. Furthermore, ADSC-secretome inhibited the high mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) inflammatory pathway. In summary, both ADSC-secretome and ADSCs inhibited pyroptosis in right hemihepatic ischemia–reperfusion combined with left hemihepatectomy injury, and ADSC-secretome exhibited a stronger therapeutic effect. ADSC-secretome exerted these therapeutic effects through the inhibition of the HMGB1/TLR4/NF-κB inflammatory pathway. In the future, “cell-free” therapy is expected to replace cell-based methods. Full article
Show Figures

Figure 1

19 pages, 889 KB  
Review
Therapeutic Potential of Mesenchymal Stem Cell-Derived Extracellular Vesicles in the Treatment of Parkinson’s Disease
by Ana Volarevic, Carl Randall Harrell, Aleksandar Arsenijevic, Valentin Djonov and Vladislav Volarevic
Cells 2025, 14(8), 600; https://doi.org/10.3390/cells14080600 - 16 Apr 2025
Cited by 1 | Viewed by 1492
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the gradual loss of dopamine-producing neurons. Oxidative stress, mitochondrial dysfunction, detrimental immune response, and neuroinflammation are mainly responsible for the injury and degeneration of dopaminergic neurons in the brains of patients suffering from [...] Read more.
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the gradual loss of dopamine-producing neurons. Oxidative stress, mitochondrial dysfunction, detrimental immune response, and neuroinflammation are mainly responsible for the injury and degeneration of dopaminergic neurons in the brains of patients suffering from PD. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as a promising therapeutic approach for treating PD due to their ability to suppress the activation of inflammatory immune cells and enhance the viability and function of dopamine-producing neurons. MSC-EVs can easily bypass the blood-brain barrier and deliver their cargo (neuroprotective factors, immunosuppressive proteins, and microRNAs) to injured dopamine-producing neurons and brain-infiltrated inflammatory immune cells. A large number of recently published experimental studies demonstrated that MSC-EVs efficiently alleviated PD-related motor and behavioral deficits in animal models, indicating that MSC-EVs should be considered as potentially new therapeutic agents for the treatment of PD. Accordingly, in this review article, we summarized current knowledge about the therapeutic potential of MSCs-EVs in the treatment of PD, paving the way for their future clinical use in the treatment of neurodegenerative and neuroinflammatory disorders. Full article
(This article belongs to the Special Issue Molecular Therapeutic Advances for Neurodegenerative Diseases)
Show Figures

Figure 1

15 pages, 602 KB  
Review
Exploring the Interaction of Tumor-Derived Exosomes and Mesenchymal Stem Cells in Tumor Biology
by Konstantinos S. Papadopoulos, Penelope Korkolopoulou and Christina Piperi
Int. J. Mol. Sci. 2025, 26(7), 3095; https://doi.org/10.3390/ijms26073095 - 27 Mar 2025
Cited by 3 | Viewed by 1645
Abstract
Exosomes are actively produced extracellular vesicles, released from different cell types, that exert important regulatory roles in vital cellular functions. Tumor-derived exosomes (TDEs) have received increasing attention because they enable intercellular communication between the neoplastic and non-neoplastic cells present in the microenvironment of [...] Read more.
Exosomes are actively produced extracellular vesicles, released from different cell types, that exert important regulatory roles in vital cellular functions. Tumor-derived exosomes (TDEs) have received increasing attention because they enable intercellular communication between the neoplastic and non-neoplastic cells present in the microenvironment of tumors, affecting important functions of different types of mesenchymal stem cells (MSCs) with the ability to self-renew and differentiate. MSC-derived exosomes (MSC-exos) carry a variety of bioactive molecules that can interact with specific cellular targets and signaling pathways, influencing critical processes in tumor biology, and exhibiting properties that either promote or inhibit tumor progression. They can regulate the tumor microenvironment by modulating immune responses, enhancing or suppressing angiogenesis, and facilitating tumor cells’ communication with distant sites, thus altering the behavior of non-cancerous cells present in the microenvironment. Herein, we explore the main functions of TDEs and their intricate interactions with MSC-exos, in terms of enhancing cancer progression, as well as their promising clinical applications as tumor microenvironment modulators. Full article
(This article belongs to the Special Issue Mesenchymal Stem Cells and Cancer)
Show Figures

Figure 1

25 pages, 2456 KB  
Review
The Molecular and Cellular Mechanisms of Endometriosis: From Basic Pathophysiology to Clinical Implications
by Heidi Mariadas, Jie-Hong Chen and Kuo-Hu Chen
Int. J. Mol. Sci. 2025, 26(6), 2458; https://doi.org/10.3390/ijms26062458 - 10 Mar 2025
Cited by 13 | Viewed by 7415
Abstract
Endometriosis is a complex gynecological disorder characterized by endometrial-like tissue growing outside the uterus, leading to chronic pain, infertility, and reduced quality of life. Its pathophysiology involves genetic, epigenetic, immune, and molecular factors. Theories such as retrograde menstruation, coelomic metaplasia, and stem cell [...] Read more.
Endometriosis is a complex gynecological disorder characterized by endometrial-like tissue growing outside the uterus, leading to chronic pain, infertility, and reduced quality of life. Its pathophysiology involves genetic, epigenetic, immune, and molecular factors. Theories such as retrograde menstruation, coelomic metaplasia, and stem cell involvement explain lesion formation. Endometrial mesenchymal stem cells (eMSCs) and epithelial progenitors (eEPs) contribute to lesion establishment by adhering to peritoneal surfaces, proliferating, and differentiating into ectopic tissue. Aberrant adhesion molecules, inflammatory cytokines, and molecular pathways like PI3K/Akt and Wnt/β-catenin drive proliferation, angiogenesis, and resistance to apoptosis. Elevated estrogen levels and progesterone resistance further promote lesion growth and immune evasion. Immune dysfunction, including altered macrophage activity and reduced natural killer (NK) cell function, contributes to inflammation and lesion persistence. Pain is linked to prostaglandin E2 (PGE2) and nerve infiltration, emphasizing the need for targeted pain management. Current therapies, such as GnRH agonists, suppress ovarian hormone production but face limitations in long-term efficacy and side effects. Integrating molecular insights into clinical practice may advance diagnostics and treatment, with emerging approaches focusing on molecular pathways, immune modulation, and hormonal regulation for more effective, personalized therapies. Future research should unravel the complex mechanisms driving endometriosis to improve patient outcomes. Full article
(This article belongs to the Special Issue Female Infertility and Fertility)
Show Figures

Figure 1

14 pages, 2027 KB  
Article
Mesenchymal Stem/Stromal Cells (MSCs) from Mouse Pelvic vs. Long Bones Exhibit Disparate Critical Quality Attributes: Implications for Translational Studies
by Siddaraju V. Boregowda, Cori N. Booker, Jacqueline Strivelli and Donald G. Phinney
Cells 2025, 14(4), 274; https://doi.org/10.3390/cells14040274 - 13 Feb 2025
Cited by 2 | Viewed by 975
Abstract
Mesenchymal stem/stromal cells (MSCs) have been exploited as an experimental cell therapy in a broad array of clinical applications but have underperformed based on results from pre-clinical studies due to gaps in translating pre-clinical findings to human patients. Herein, we isolated mouse MSCs [...] Read more.
Mesenchymal stem/stromal cells (MSCs) have been exploited as an experimental cell therapy in a broad array of clinical applications but have underperformed based on results from pre-clinical studies due to gaps in translating pre-clinical findings to human patients. Herein, we isolated mouse MSCs from pelvic bone marrow (BMP), a preferred source for human MSCs, and compared their growth, differentiation, and immuno-modulatory activity to those derived from long bone marrow (BML), the traditional source of mouse MSCs. We report that BMP-MSCs exhibit significantly enhanced growth kinetics in 5% and 21% oxygen saturation and superior bi-lineage differentiation and hematopoiesis-supporting activity as compared to BML-MSCs. Additionally, we show that TNF upregulates inducible nitric oxide synthase (NOS2) in BML- and BMP- MSCs and augments their immune suppressive activity in cell-based assays, while interferon-gamma (INFG) upregulates indoleamine, 2-3, dioxygenase (IDO1) and enhances the immune suppressive activity of only BMP-MSCs. These results indicate that mouse MSCs sourced from different bone compartments exhibit measurable differences in critical quality attributes, and these differences are comparable to those observed across species. Based on these differences, BMP- MSCs represent a useful resource to model the behavior of human BM-derived MSCs. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Figure 1

17 pages, 2557 KB  
Article
Establishment of iPSC-Derived MSCs Expressing hsa-miR-4662a-5p for Enhanced Immune Modulation in Graft-Versus-Host Disease (GVHD)
by Susie Lee, Eung-Won Kim, Hae-Ri Lee, Sun-Ung Lim, Chan Kwon Jung, Young-Ju Kang, Gyung-Ah Jung and Il-Hoan Oh
Int. J. Mol. Sci. 2025, 26(2), 847; https://doi.org/10.3390/ijms26020847 - 20 Jan 2025
Viewed by 1461
Abstract
The immune-modulatory effects of mesenchymal stromal cells (MSCs) are widely used to treat inflammatory disorders, with indoleamine 2,4-dioxygenase-1 (IDO-1) playing a pivotal role in suppressing stimulated T-cell proliferation. Taking that three-dimensional (3D) cultures enhance MSCs’ anti-inflammatory properties compared with two-dimensional (2D) cultures, the [...] Read more.
The immune-modulatory effects of mesenchymal stromal cells (MSCs) are widely used to treat inflammatory disorders, with indoleamine 2,4-dioxygenase-1 (IDO-1) playing a pivotal role in suppressing stimulated T-cell proliferation. Taking that three-dimensional (3D) cultures enhance MSCs’ anti-inflammatory properties compared with two-dimensional (2D) cultures, the differentially expressed miRNAs were examined. Thus, we identified hsa-miR-4662a-5p (miR-4662a) as a key inducer of IDO-1 via its suppression of bridging integrator-1 (BIN-1), a negative regulator of the IDO-1 gene. The IDO-1-inducing potential of miR-4662a was conserved across primary MSCs from various donors and sources but exhibited variability. Notably, iPSC-derived MSCs (iMSCs) demonstrated superior IDO-1 induction and immune-modulatory efficacy compared with their donor-matched primary MSCs. Accordingly, iMSCs expressing miR-4662a (4662a/iMSC) exhibited stronger suppressive effects on T-cell proliferation and more potent suppressive effects on graft-versus-host disease (GVHD), improving survival rates and reducing tissue damage in the liver and gut. Our results point to the therapeutic potential of standardized, off-the-shelf 4662a/iMSC as a robust immune-modulating cell therapy for GVHD. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

16 pages, 2538 KB  
Article
Preconditioning with Wound Fluid Enhances Immunosuppressive Properties of Mesenchymal Stromal Cells In Vitro
by Helena Moratin, Isabel Mache, Miguel Goncalves, Totta Ehret Kasemo, Manuel Stöth, Till Jasper Meyer, Stephan Hackenberg, Agmal Scherzad and Marietta Herrmann
Int. J. Mol. Sci. 2025, 26(1), 293; https://doi.org/10.3390/ijms26010293 - 31 Dec 2024
Cited by 1 | Viewed by 906
Abstract
Immunosuppression is one key feature of mesenchymal stromal cells (MSCs) that has high expectations for therapeutic use. The influence of pro-inflammatory stimuli can modify the characteristics of MSCs and enhance immunosuppressive properties. The local postoperative environment contains cytokines, MSCs, and immune cells in [...] Read more.
Immunosuppression is one key feature of mesenchymal stromal cells (MSCs) that has high expectations for therapeutic use. The influence of pro-inflammatory stimuli can modify the characteristics of MSCs and enhance immunosuppressive properties. The local postoperative environment contains cytokines, MSCs, and immune cells in high quantities, and their mutual influence is still unclear. Knowledge of in vivo processes is pivotal for potential therapeutic applications, and therefore, the aim of this study was to investigate the influence of wound fluid (WF) on the immunomodulatory potential of MSCs. CD4+ cells were co-cultured with native or WF-preconditioned MSCs for 5 days. CFSE staining revealed significant suppression of T cell proliferation after co-culture that was even more distinct in co-culture with WF-MSCs. The concentration of IDO-1, TGF-β1 and IFN-γ was higher while TNF-α was reduced in co-culture supernatants, indicating a transition to an anti-inflammatory milieu. In summary, the results provide evidence that the influence of WF alters the immunomodulatory potential of MSCs. These findings should serve as the basis for further investigations with a focus on T cell subpopulations. Full article
(This article belongs to the Special Issue Mesenchymal Stem Cells: Cross-Talk with the Microenvironment)
Show Figures

Figure 1

16 pages, 982 KB  
Review
Targeted Therapy for Severe Sjogren’s Syndrome: A Focus on Mesenchymal Stem Cells
by Carl Randall Harrell, Ana Volarevic, Aleksandar Arsenijevic, Valentin Djonov and Vladislav Volarevic
Int. J. Mol. Sci. 2024, 25(24), 13712; https://doi.org/10.3390/ijms252413712 - 22 Dec 2024
Cited by 3 | Viewed by 3391
Abstract
Primary Sjögren’s syndrome (pSS) is an autoimmune disease characterized by the infiltration of lymphocytes on salivary and lacrimal glands, resulting in their dysfunction. Patients suffering from severe pSS have an increased risk of developing multi-organ dysfunction syndrome due to the development of systemic [...] Read more.
Primary Sjögren’s syndrome (pSS) is an autoimmune disease characterized by the infiltration of lymphocytes on salivary and lacrimal glands, resulting in their dysfunction. Patients suffering from severe pSS have an increased risk of developing multi-organ dysfunction syndrome due to the development of systemic inflammatory response, which results in immune cell-driven injury of the lungs, kidneys, liver, and brain. Therapeutic agents that are used for the treatment of severe pSS encounter various limitations and challenges that can impact their effectiveness. Accordingly, there is a need for targeted, personalized therapy that could address the underlying detrimental immune response while minimizing side effects. Results obtained in a large number of recently published studies have demonstrated the therapeutic efficacy of mesenchymal stem cells (MSCs) in the treatment of severe pSS. MSCs, in a juxtacrine and paracrine manner, suppressed the generation of inflammatory Th1 and Th17 lymphocytes, induced the expansion of immunosuppressive cells, impaired the cross-talk between auto-reactive T and B cells, and prevented the synthesis and secretion of auto-antibodies. Additionally, MSC-derived growth and trophic factors promoted survival and prevented apoptosis of injured cells in inflamed lacrimal and salivary glands, thereby enhancing their repair and regeneration. In this review article, we summarized current knowledge about the molecular mechanisms that are responsible for the beneficial effects of MSCs in the suppression of immune cell-driven injury of exocrine glands and vital organs, paving the way for a better understanding of their therapeutic potential in the targeted therapy of severe pSS. Full article
(This article belongs to the Special Issue Biomedical Applications of Mesenchymal Stem Cells)
Show Figures

Figure 1

15 pages, 2991 KB  
Article
Elevated IL-6 Expression in Autologous Adipose-Derived Stem Cells Regulates RANKL Mediated Inflammation in Osteoarthritis
by Hyun-Joo Lee, Dae-Yong Kim, Hyeon jeong Noh, Song Yi Lee, Ji Ae Yoo, Samuel Jaeyoon Won, Yoon Sang Jeon, Ji Hoon Baek and Dong Jin Ryu
Cells 2024, 13(24), 2046; https://doi.org/10.3390/cells13242046 - 11 Dec 2024
Cited by 5 | Viewed by 1616
Abstract
Interleukin-6 (IL-6) expression in mesenchymal stem cells (MSCs) has been shown to play a pivotal role in modulating cartilage regeneration and immune responses, particularly in the context of diseases that involve both degenerative processes and inflammation, such as osteoarthritis (OA). However, the precise [...] Read more.
Interleukin-6 (IL-6) expression in mesenchymal stem cells (MSCs) has been shown to play a pivotal role in modulating cartilage regeneration and immune responses, particularly in the context of diseases that involve both degenerative processes and inflammation, such as osteoarthritis (OA). However, the precise mechanism through which IL-6 and other immune-regulatory factors influence the therapeutic efficacy of autologous adipose-derived stem cells (ASCs) transplantation in OA treatment remains to be fully elucidated. This study aims to investigate the relationship between IL-6 expression in autologous ASCs isolated from OA patients and their impact on immune modulation, particularly focusing on the regulation of Receptor Activator of Nuclear factor Kappa-Β Ligand (RANKL), a key mediator of immune-driven cartilage degradation in OA. Autologous ASCs were isolated from the stromal vascular fraction (SVF) of adipose tissue obtained from 22 OA patients. The isolated ASCs were cultured and characterized using reverse transcription polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and flow cytometry to the phenotype and immune regulatory factors of MSCs. Based on IL-6 expression levels, ASCs were divided into high and low IL-6 expression groups. These groups were then co-cultured with activated peripheral blood mononuclear cells (PBMCs) to evaluate their immune-modulatory capacity, including the induction of regulatory T cells, inhibition of immune cell proliferation, and regulation of key cytokines, such as interferon-gamma (IFN-γ). Additionally, RANKL expression, a critical factor in osteoclastogenesis and cartilage degradation, was assessed in both ASC groups. High IL-6-expressing ASCs demonstrated a significantly greater capacity to inhibit immune cell proliferation and IFN-γ production compared to their low IL-6-expressing counterparts under co-culture conditions. Moreover, the group of ASCs with high IL-6 expression showed a marked reduction in RANKL expression, suggesting enhanced potential to control osteoclast activity and subsequent cartilage defect in OA. Conclusion: Autologous ASCs with elevated IL-6 expression exhibit enhanced immunomodulatory properties, particularly in regulating over-activated immune response and reducing osteoclastogenesis through RANKL suppression. These findings indicate that selecting ASCs based on IL-6 expression could enhance the therapeutic efficacy of ASC-based treatments for OA by mitigating immune-driven joint inflammation and cartilage degradation, potentially slowing disease progression. Full article
Show Figures

Figure 1

Back to TopTop