Current Stem Cell Research & Therapy - Volume 20, Issue 5, 2025
Volume 20, Issue 5, 2025
-
-
Molecular Mechanisms and Pathways of Mesenchymal Stem Cell-mediated Therapy in Brain Cancer
Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach in the treatment of brain cancer due to their unique biological properties, including their ability to home tumor sites, modulate the tumor microenvironment, and exert anti-tumor effects. This review delves into the molecular mechanisms and pathways underlying MSC-mediated therapy in brain cancer. We explore the various signalling pathways activated by MSCs that contribute to their therapeutic efficacy, such as the PI3K/Akt, Wnt/β-catenin, and Notch pathways. Additionally, we discuss the role of exosomes and microRNAs secreted by MSCs in mediating anti-tumor effects. The review also addresses the challenges and future directions in optimizing MSC-based therapies for brain cancer, including issues related to MSC sourcing, delivery methods, and potential side effects. Through a comprehensive understanding of these mechanisms and pathways, we aim to highlight the potential of MSCs as a viable therapeutic option for brain cancer and to guide future research in this field.
-
-
-
Exploring the Dual Roles of Neural Stem Cells in Glioblastoma: Therapeutic Implications and Opportunities
Glioblastoma (GBM) is recognized as the most aggressive and lethal form of primary brain tumor, characterized by rapid proliferation and significant resistance to conventional therapies. Recent studies have illuminated the complex role of Neural Stem Cells (NSCs) in both the progression and treatment of GBM. This review examines the specific molecular pathways influenced by NSCs, focusing on critical signaling cascades such as Notch, P13K, and SHH, which are implicated in tumor development and maintenance. Furthermore, we explore the dual role of NSCs in glioblastoma, where they can act as both facilitators of tumorigenesis and potential agents of tumor suppression, depending on the microenvironmental context. Understanding these intricate interactions is essential for developing innovative therapeutic strategies that target NSCs in GBM. This review aims to provide a comprehensive overview of current knowledge and to identify future research directions in this promising field, ultimately contributing to the advancement of personalized treatment approaches for patients with glioblastoma.
-
-
-
Adipose-derived Stem Cells for Treatment of Diabetic Foot Ulcers: A Review
This study investigates the therapeutic potential of adipose-derived stem cells (ASCs) in diabetic foot ulcers (DFUs). The goal is to further research regenerative medicine by improving knowledge of ASC-based therapies in diabetic wound management. A comprehensive literature review included studies from reputable databases, including PubMed and the Cochrane Library. We paid particular attention to the clinical, in vivo, and in vitro investigations of the utility and effectiveness of ASCs in treating DFU. We also highlighted novel isolation techniques and application methods for ASCs in chronic wound management. ASCs have shown great potential in regenerative interventions for diabetes, especially in DFU management. These cells facilitate wound repair by differentiating into different cell types, promoting angiogenesis, secreting growth factors, reducing inflammation, and increasing wound perfusion. However, the current body of research on ASC applications for DFU still requires further investigation. This shows the importance of thoroughly studying their biological mechanisms and therapeutic uses. This review establishes that ASC-based treatments effectively enhance outcomes for patients suffering from DFU. We recommend further investigation of the functionality of ASCs and therapeutic approaches to maximize their therapeutic potential in managing diabetic wounds, thereby advancing the development of regenerative medicine.
-
-
-
The Mechanisms of Mesenchymal Stem Cells in the Treatment of Experimental Autoimmune Encephalomyelitis
Authors: Chunran Xue, Haojun Yu, Ye Sun, Xiying Wang, Xuzhong Pei, Yi Chen and Yangtai GuanMultiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system and is a leading cause of disability in young adults. Most therapeutic strategies are based on immunosuppressant effects. However, none of the drugs showed complete remission and may result in serious adverse events such as infection. Mesenchymal stem cells (MSCs) have gained much attention and are considered a potential therapeutic strategy owing to their immunomodulatory effects and neuroprotective functions. Experimental autoimmune encephalomyelitis (EAE), a classical animal model for MS, is widely used to explore the efficacy and mechanism of MSC transplantation. This review summarises the therapeutic mechanism of MSCs in the treatment of EAE, including the effects on immune cells (T cells, B cells, dendritic cells, natural killer cells) and central nervous system-resident cells (astroglia, microglia, oligodendrocytes, neurons) as well as various strategies to improve the efficacy of MSCs in the treatment of EAE. Additionally, we discuss the clinical application of MSCs for MS patients as well as the challenges and prospects of MSC transplantation.
-
-
-
Breast Milk Stem Cells in the Treatment of Neonatal Diseases
Authors: Lu He, Zhihong Sun, Congcong Zhao and Huiqing SunBreast milk plays an important role in preventing and remission various acute and chronic neonatal diseases. However, the mechanisms through which breast milk performs these effects are still unclear. Recently, research has shown that breast milk contains previously undiscovered cells, called breast milk stem cells (BMSCs), which have the potential for multiple differentiation. These cells have been shown to be expressed in multiple organs and systems of infants after ingestion. This study found that newborns who received BMSCs required a shorter treatment duration and had a better prognosis than those who did not receive BMSCs. This suggests that BMSCs may be able to treat a variety of neonatal diseases, and breast milk may play a role in preventing and mitigating neonatal diseases through BMSCs. In this paper, we summarized the research progress on BMSCs and their therapeutic effects on organs, the possible ways in which BMSCs enter different organs and tissues of the human body, and detailed the types of diseases, modes of action, and mechanisms that BMSCs may play a role in. We aim to pave the way for further clinical and preclinical research on BMSCs, as well as the treatment of neonatal diseases with BMSCs, to provide possible new therapeutic means for treating neonatal diseases and a variety of other diseases.
-
-
-
Modulating Proliferation, Migration and Differentiation of Mesenchymal Stem Cells Using Interleukins
Authors: Sankaranarayanan Srinivasan and Nalinkanth V. GhoneMesenchymal Stem Cells (MSCs) are multipotent stem cells that are obtained from various tissue sources such as bone marrow, adipose tissues, umbilical cords, dental pulps, and peripheral blood has high regenerative potential, migratory abilities, and immunosuppressive properties. These properties make them attractive candidates for tissue engineering, immunosuppressive therapies, and in vivo drug deliveries. MSCs, because of their high propensity to home in an injured tissue microenvironment, are exposed to various cytokines. These cytokines modulate the activity of MSCs to help in the regeneration of injured tissue. Interleukins are one such cytokine that is present in injured tissue microenvironment and plays significant roles in the activation, differentiation, proliferation, maturation, migration, and adhesion of not only immune cells but also MSCs. Interleukins, through both autocrine and paracrine signaling mechanisms, modulate the functioning of MSCs. This article reviews how interleukins influence MSCs by discussing their signaling pathways, their effect on differentiation and other biological effects. A comprehensive understanding of the influence of interleukins on MSCs may provide insights to manipulate improving the therapeutic potential of MSCs or reducing potential risks such as undesirable immune response and tumor formation.
-
-
-
Long Non-coding RNA DANCR Regulates the Proliferation and Migration of Human Adipose-derived Mesenchymal Stromal Cells in Inflammation Conditions
More LessBackgroundMesenchymal stromal cells (MSCs) and Dexamethasone (Dex) are both effective methods to treat inflammatory diseases. However, the interaction between inflammatory factors, Dex, and MSCs in repair is not fully understood. The purpose of this study is to clarify the effects and mechanisms of glucocorticoids on the tissue repair characteristics of MSCs in an inflammatory environment.
MethodsThis is an experimental study. Human adipose-derived mesenchymal stromal cells (hASCs) were cultured, and Long non-coding RNA (lncRNA) differentiation antagonizing non-protein coding RNA (DANCR) expression was detected after treatment with Dex and inflammation factors. Additionally, DANCR was knockdown or overexpressed before Dex or tumor necrosis factor-alpha (TNF-α) treatments, respectively. hASC proliferation, cell cycle, and migration ability were analyzed to evaluate the effects of DANCR in hASCs treated with Dex or TNF-α. Nuclear factor-kB (NF-κB) pathway inhibitors were used to clarify the signal pathway that DANCR involved. All data are presented as the mean ± standard deviation. The two-tailed Student's t-test or one-way analysis of variance (ANOVA) was used to determine the statistical differences between groups.
ResultsDex decreased the proliferation and migration of hASCs and upregulated DANCR expression in a dosage-dependent relationship. The knockdown of DANCR reversed Dex's repression of hASC proliferation. Moreover, DANCR was decreased by inflammatory cytokines, and overexpressing DANCR alleviated the promotion effects of TNF-α on hASC proliferation and migration. Furthermore, mechanistic investigation validated that DANCR was involved in the NF-κB signaling pathway.
ConclusionWe identified a lncRNA, DANCR, that was involved in Dex and inflammation-affected hASC proliferation and migration. Dex reduced the proliferation and migration of hASCs through DANCR while exerting its anti-inflammatory effects. Thus, it is suggested to avoid the simultaneous application of hASCs and steroids in clinical practice. These results enrich our understanding of the versatile function of lncRNAs in the crosstalk of inflammation conditions and MSCs.
-
-
-
Kartogenin Induces Chondrogenesis in Cartilage Progenitor Cells and Attenuates Cell Hypertrophy in Marrow-Derived Stromal Cells
IntroductionKartogenin (KGN) is a synthetic small molecule that stimulates chondrogenic cellular differentiation by activating smad-4/5 pathways. KGN has been proposed as a feasible alternative to expensive biologic growth factors, such as transforming growth factor β, which remain under strict regulatory scrutiny when it comes to their use in patients. This study reports the previously unexplored effects of KGN stimulation on cartilage-derived mesenchymal progenitor cells (CPCs), which have been shown to be effective in applications of cell-based musculoskeletal tissue regeneration.
MethodsGene expression via RT-qPCR analysis was used to determine the effects of KGN treatment on CPCs and human marrow derived stromal cells (BM-MSCs). The expression of SOX9, COL1, COL2, COL10, RUNX2, and MMP-13 were quantified following 3-10 days of KGN treatment. Additionally, soluble MMP-13 protein was quantified using ELISA. A GAG assay was used to compare proteoglycan production. Cell viability was measured in response to different doses of KGN using an MTT assay.
ResultsOur findings demonstrate that KGN treatment significantly increased markers of chondrogenesis, SOX9 and COL2 following 3-10 days of treatment in human CPCs. KGN treatment also resulted in a significant dose-dependent increase in GAG production in CPCs. The same efficacy was not observed in human BM-MSCs; however, KGN significantly reduced mRNA expression of cell hypertrophy markers, COL10 and MMP-13, in BM-MSCs. Parallel to these mRNA expression results, KGN led to a significant decrease in protein levels of MMP-13 both at 0-5 days and 5-10 days following KGN treatment.
ConclusionIn conclusion, this study demonstrates that KGN can boost the chondrogenicity of CPCs and inhibit hypertrophic terminal differentiation of BM-MSCs.
-
-
-
Muscle-derived Stem Cell Exosomes Enhance Autophagy through the Regulation of the mTOR Signaling Pathway to Attenuate Glucolipotoxicity-induced Pancreatic β-cell Injury
Authors: Xiang-Yu Zeng, Zi-Wen Liu, Hao-Ze Li and Jian-Yu LiuBackgroundDiabetes mellitus (DM) refers to a series of metabolic disorders, including elevated blood glucose level diseases due to insufficient insulin secretion or insulin resistance.
ObjectiveTo investigate the effect and protective mechanism of muscle-derived stem cell exosomes (MDSC-Exo) on glucolipotoxicity-induced pancreatic β cell injury.
MethodsPrimary rat muscle-derived stem cells (MDSCs) were isolated and cultured. After the completion of the third-generation culture for MDSCs, MDSC-Exo was isolated. Then, the morphology and diameter of exosomes were observed by means of electron microscopy and nanoparticle tracking analysis (NTA) instrument. The expression of exosome-related proteins CD63, TSG101 and Calnexin was detected by western blot. After stimulation of rat insulinoma cell line INS-1 with high glucose/palmitic acid (HG/PA) and/or MDSC-Exo, cell viability and apoptosis were measured through MTT and flow cytometry (FCT), respectively. Biochemical reagents were utilized for the examination of the levels of superoxide dismutase (SOD) and malondialdehyde (MDA); enzyme-linked immunosorbent assay (ELISA) for the levels of cellular insulin secretion, and the western blot for the expression level of LC3, p62, AKT, p-AKT, mTOR and p-mTOR.
ResultsMDSC-Exo was successfully isolated and identified, and it was found that MDSC-Exo could reduce HG/PA-induced apoptosis as well as MDA levels in INS-1 cells. Also, MDSC-Exo could significantly increase cell viability, insulin secretion ability within 24 hours and SOD level. Besides, MDSC-Exo was able to significantly increase the LC3-II/I ratio, decrease the expression level of p62, and promote autophagy in the cells. Aside from what has been mentioned, MDSC-Exo showed a significant reduction effect on p-Akt and p-mTOR level as well as p-Akt/Akt and p-mTOR/mTOR ratios.
ConclusionMDSC-Exo can alleviate oxidative stress and enhance autophagy by inhibiting Akt/mTOR signaling pathway activation. Then, the inhibition of apoptosis and the promotion of insulin secretion can be achieved to alleviate glucolipotoxicity-induced pancreatic β cell injury.
-
-
-
Effect of miR-98/IL-6/STAT3 on Autophagy and Apoptosis of Cardiac Stem Cells Under Hypoxic Conditions In vitro
Authors: Xueyuan Li, Yang Zhang and Guangwei ZhangBackgroundThe heavy burden of cardiovascular diseases demands innovative therapeutic strategies dealing with cardiomyocyte loss. Cardiac Stem Cells (CSCs) are renewable cells in the myocardium with differentiation and endocrine functions. However, their functions are significantly inhibited in conditions of severe hypoxia or inflammation. The mechanism of hypoxia affecting CSCs is not clear. Interleukin-6 (IL-6) appears active in both hypoxic and inflammatory microenvironments. The aim of this study was to explore whether IL-6 is related to CSC apoptosis and autophagy under severe hypoxia.
MethodsIn this study, rat CSCs were extracted by alternate digestion. The interaction of miR-98 and IL-6 mRNA was detected by the dual luciferase method, and qPCR was applied to confirm the effect of miR-98 on IL-6 expression. The effect of IL-6 on CSC apoptosis was measured by flow cytometry and the effect of IL-6 on CSC autophagy by transmission electron microscopy. The western blot method was applied to detect the effect of IL-6 on the expressions of proteins related to apoptosis and autophagy. ANOVA and Dunnett T3's test were employed in the statistical analysis. When p < 0.05, the difference was significant.
ResultsUnder severe hypoxia conditions, IL-6 increased CSC apoptosis and decreased p-STAT3 expression significantly. CSC apoptosis increased significantly after inhibition of the STAT3 signaling pathway under severe hypoxia. IL-6 could also significantly inhibit CSCs’ autophagy and block their autophagy flow under severe hypoxic conditions. Meanwhile, it was confirmed that miR-98 had a binding site on IL-6 mRNA and miR-98 significantly inhibited IL-6 mRNA expression in CSCs under severe hypoxic conditions.
ConclusionmiR-98/IL-6/STAT3 has been found to be involved in the regulation of CSCs’ apoptosis and autophagy under severe hypoxic conditions and there might be a mutual linkage between CSCs’ apoptosis and their autophagy.
-
-
-
Corrigendum to: The Renoprotective and Anti-Inflammatory Effects of Human Urine-Derived Stem Cells on Acute Kidney Injury Animals
Authors: Yuanyuan Kuang, Chenyu Fan, Xiaojun Long, Jiajia Zheng, Yunsi Zeng, Yuhui Wei, Jiasheng Zhang, Shuangjin Yu, Tong Chen, Hehuan Ruan, Yi Wang, Ning Na, Yiming Zhou and Jiang Qiu
-
Volumes & issues
-
Volume 20 (2025)
-
Volume 19 (2024)
-
Volume 18 (2023)
-
Volume 17 (2022)
-
Volume 16 (2021)
-
Volume 15 (2020)
-
Volume 14 (2019)
-
Volume 13 (2018)
-
Volume 12 (2017)
-
Volume 11 (2016)
-
Volume 10 (2015)
-
Volume 9 (2014)
-
Volume 8 (2013)
-
Volume 7 (2012)
-
Volume 6 (2011)
-
Volume 5 (2010)
-
Volume 4 (2009)
-
Volume 3 (2008)
-
Volume 2 (2007)
-
Volume 1 (2006)
Most Read This Month
