Product Citations: 46

Effects of Dexamethasone and Tacrolimus on Mesenchymal Stem Cell Characteristics and Gene Expression.

In International Journal of Stem Cells on 30 May 2025 by Lee, N. K., Na, D. L., et al.

Mesenchymal stem cells (MSCs) are frequently used for therapeutic applications in both pre-clinical and clinical settings owing to their capacity for immune modulation and neuroprotective effects. However, transient fever is commonly observed as an adverse event following MSC injection in patients with Alzheimer's disease (AD). In this study, we investigated the potential impact of immunosuppressants such as dexamethasone and tacrolimus on altering the characteristics of human mesenchymal stem cells (hMSCs). Additionally, we examined whether these immunosuppressants affect the persistence of hMSCs or the immune response upon their administration into the brain parenchyma of AD mice. The exposure of hMSCs to high concentrations of dexamethasone and tacrolimus in vitro did not significantly alter the characteristics of hMSCs. The expression of genes related to innate immune responses, such as Irak1, Irf3, Nod1, and Ifnar1, was significantly downregulated by the additional administration of dexamethasone and tacrolimus to the brain parenchyma of AD mice. However, hMSC persistence in the AD mouse brain was not affected. The results of this study support the use of immunosuppressants to mitigate fever during stem cell therapy in patients with AD.

  • Stem Cells and Developmental Biology

The aim of this study was to investigate the regenerative effect of lyophilized dental follicle mesenchymal stem cells (DF-MSCs) combined with rat platelet-rich fibrin (PRF) on geriatric skin wounds. Human DF-MSCs which were isolated from the wisdom teeth of healthy donors and PRF were mixed and incubated in a 37 °C incubator for 1-2 h containing 1 million cells in 150 mg PRF. The mixture was suspended in a freeze-drying solution and then lyophilized. Wounds were created on the back skin of Wistar albino rats using a 6 mm punch. Lyophilized DF-MSCs, PRF, or PRF + DF-MSCs were applied to the wounds of rats. On the 15th day, the wound area was histopathologically evaluated in rats. Blood samples from rats were analyzed for total antioxidant status (TAOS), and inflammatory cytokine levels using ELISA. In both young and geriatric rats treated with lyophilized PRF + DF-MSCs, wound area began to significantly decrease from the 10th day compared to the untreated group (p < 0.05). Histopathological examination revealed that in the lyophilized PRF + DF-MSCs treated groups, epithelial integrity and scarless healing significantly increased compared to the untreated groups (p < 0.05). There were no significant differences in TAOS, total oxidant status (TOS), tumor necrosis factor (TNF), interleukin-6 (IL6), and hydroxyproline levels in serum samples from young rats on the 15th day. In geriatric rats, hydroxyproline (HYPS) levels were increased in the DF-MSC and PRF + DF-MSC groups (p < 0.01), TNF was significantly elevated in PRF geriatric group and IL6 was increased in the PRF group compared to the control group (p = 0.01). Lyophilized PRF + DF-MSCs, which is a shelf-stable and ready-to-use product, hold promise, especially for traumatic wounds in geriatric individuals with longer healing times.
© 2025. The Author(s).

  • Stem Cells and Developmental Biology

Rationale: CD39, a key ectonucleotidase that drives adenosine production, acts as a critical immunosuppressive checkpoint in cancer. Although it has shown promise as a therapeutic target, clinical trials are demonstrating the need for more potent targeting approaches. This need is driving innovation towards the development of novel antibodies and the exploration of strategic combinations with a range of immunotherapies. Methods: An anti-CD39 nanobody was screened and tested for its affinity and binding ability using biolayer interferometry, ELISA and flow cytometry. Blocking ability against soluble and membrane-bound CD39 was measured after CD39 blockade. Internalization was detected using immunofluorescence. The reversal of T-cell function by the anti-CD39 antibody was assessed by CFSE-based T-cell proliferation, CD25 expression and IFN-γ secretion. The in vivo function of tumor growth inhibition was further tested in a mouse model and we also tested the phenotype of immune cells after CD39 antibody administration from tumor tissue, draining lymph nodes and peripheral blood. We inserted the antibody sequence into the chimeric antigen receptor (CAR) construct to induce MSLN CAR-T cells to secret the CD39 antibody, and the efficacy was measured in xenograft models of ovarian cancer. Results: We screened human CD39 antibodies using a VHH library and developed a single-epitope anti-CD39 nanobody, named huCD39 mAb, with high affinity and potent binding and blocking ability. The huCD39 mAb was internalized in a time-dependent manner. The in vitro study revealed that the huCD39 mAb was highly effective in enhancing T-cell proliferation and functionality. In vivo, the huCD39 mAb showed significant anti-tumor efficacy in an immunocompetent mouse model. Flow cytometry analysis demonstrated downregulated CD39 expression in immune cells after antibody administration. We also observed increased CD39 expression in ovarian cancer tissue and in activated CAR T cells. Subsequently, we developed a type of MSLN CAR-T cells secreting huCD39 mAb which showed effective eradication or inhibition in ovarian tumor xenografts. Conclusions: A novel huCD39 mAb with strong blocking ability against human CD39 and potent inhibition of tumor growth has been developed. Furthermore, a modified huCD39 mAb-secreting CAR-T cell has been generated, exhibiting superior efficacy against ovarian cancer. This provides a promising strategy for optimizing immunotherapies in ovarian cancer and potentially other malignancies.
© The author(s).

  • FC/FACS
  • Cancer Research
  • Immunology and Microbiology

Lineage-Specific Mesenchymal Stromal Cells Derived from Human iPSCs Showed Distinct Patterns in Transcriptomic Profile and Extracellular Vesicle Production.

In Advanced Science (Weinheim, Baden-Wurttemberg, Germany) on 1 July 2024 by Winston, T., Song, Y., et al.

Over the past decades, mesenchymal stromal cells (MSCs) have been extensively investigated as a potential therapeutic cell source for the treatment of various disorders. Differentiation of MSCs from human induced pluripotent stem cells (iMSCs) has provided a scalable approach for the biomanufacturing of MSCs and related biological products. Although iMSCs shared typical MSC markers and functions as primary MSCs (pMSCs), there is a lack of lineage specificity in many iMSC differentiation protocols. Here, a stepwise hiPSC-to-iMSC differentiation method is employed via intermediate cell stages of neural crest and cytotrophoblast to generate lineage-specific MSCs with varying differentiation efficiencies and gene expression. Through a comprehensive comparison between early developmental cell types (hiPSCs, neural crest, and cytotrophoblast), two lineage-specific iMSCs, and six source-specific pMSCs, are able to not only distinguish the transcriptomic differences between MSCs and early developmental cells, but also determine the transcriptomic similarities of iMSC subtypes to postnatal or perinatal pMSCs. Additionally, it is demonstrated that different iMSC subtypes and priming conditions affected EV production, exosomal protein expression, and cytokine cargo.
© 2024 The Authors. Advanced Science published by Wiley‐VCH GmbH.

  • FC/FACS

Human umbilical cord mesenchymal stem cells (UC-MSCs)-derived hepatocyte-like cells (HLCs) have shown great promise in the treatment of liver diseases. However, most current induction protocols yield hepatocyte-like cells with limited function as compared with primary hepatocytes. Schisandrin B (Sch B) is one of the main components of Schisandra chinensis, which can prevent fibrosis progression and promote liver cell regeneration. Herein, we investigated the effects of Sch B on hepatic differentiation of UC-MSCs. We found that treatment with 10 μM Sch B from the second stage of the differentiation process increased hepatic marker levels and hepatic function. Additionally, RNA-seq analysis revealed that Sch B promoted hepatic differentiation via activating the JAK2/STAT3 pathway. When transplanted HLCs into mice with CCL4-induced liver fibrosis, Sch B-treated HLCs exhibited significant therapeutic effects. This study provides an optimized hepatic differentiation protocol for UC-MSCs based on Sch B, yielding functioning cells for liver disease treatment.
© 2024 The Author(s).

  • Stem Cells and Developmental Biology
View this product on CiteAb