98%
921
2 minutes
20
Skin allografts are prone to rejection because of their high immunogenicity. By achieving immune tolerance, the long-term survival of skin allografts can be extended without the need for immunosuppressants or with only short-term low-dose dependency. Tolerogenic dendritic cells (tolDCs) show a strong potential for graft tolerance. We explored the mechanism whereby rapamycin-modified CD169low/-tolDCs regulate interleukin-10 (IL-10) B regulatory (IL-10+ Breg) cell production and mediate the long-term survival of skin allografts in mice. CD169low/-tolDCs were obtained through flow cytometry sorting after treating the mesenchymal stem cell (MSC)-derived dendritic cells with a low dose of GM-CSF. A treatment regimen combining preoperative stimulation and postoperative adoptive infusion of CD169low/-tolDCs was used to treat an acute rejection (AR) mouse skin transplantation model-the adoptive infusion group. An equivalent dose of saline was administered to the control group. Survival and graft rejection rates were assessed. Mixed lymphocyte culture, flow cytometry, immunohistochemistry (IHC), and western blotting (WB) were used to elucidate the expression of different IL-10+ Breg subsets in mice treated with adoptive infusion therapy and the molecular mechanisms whereby CD169low/-tolDCs induce IL-10+ Breg production to mediate tolerance. Adoptive infusion of CD169low/-tol DCs markedly prolonged the rejection time after skin transplantation in mice and promoted graft survival. A significant increase was observed in local blood flow signals in the transplanted skin, along with mild local inflammation. Flow cytometric analysis revealed a positive correlation between high expression of IL-10+ Breg and the changes in Foxp3+ Tregs in vivo, primarily enriched in the CD19 + CD24 + CD27+ mBreg and CD19 + CD23 + CD27-CD24+ Breg subsets, with higher levels of IgM expression. Significant differences were observed compared with control mice. CD79b/NF-κB pathway was found to be involved in Breg production. CD24, CD23, CD79, BTK, NF-κB p50/p65, CD40, and IKKα levels in the adoptive infusion group were significantly increased compared with those in the control group. Adoptive infusion of CD169low/-MSCs/tolDCs may activate the NF-κB pathway through CD79/BTK-dependent and CD40/IKKα-independent pathways, inducing high expression of IL-10 + Breg and promoting graft survival of mouse skin transplantation.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1016/j.trim.2025.102244 | DOI Listing |
Rinsho Ketsueki
September 2025
Department of Hematology, Japanese Red Cross Medical Center.
A 58-year-old woman received chimeric antigen receptor T-cell (CAR-T) therapy for triple-class refractory multiple myeloma. Following CAR-T infusion, she developed severe cytokine release syndrome (CRS) and was promptly admitted to the intensive care unit (ICU). Subsequently, she developed immune effector cell-associated neurotoxicity syndrome (ICANS), and then progressed to cardiac arrest.
View Article and Find Full Text PDFHematology
December 2025
Bone Marrow Transplantation Center, The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
Chimeric antigen receptor (CAR)-T therapy has shown significant success in the treatment of relapsed or refractory acute lymphoblastic leukemia (r/r ALL). However, its role in patients with the TCF3-PBX1 fusion gene - which generally exhibit poor prognostic indicators - remains uncertain. From September 2016 to March 2023, 7 patients with r/r ALL positive for the TCF3-PBX1 fusion gene underwent CD19 CAR-T-cell therapy at the First Affiliated Hospital of Zhejiang University School of Medicine.
View Article and Find Full Text PDFAbnormal blood vessels limit the delivery and function of endogenous T cells as well as adoptively transferred Chimeric Antigen Receptor (CAR)-T cells in the tumor microenvironment (TME). We recently showed that vascular normalization using anti-VEGF therapy can overcome these challenges and improve the outcome of CAR-T therapy in glioblastoma models in mice. Here, we developed a physiologically based pharmacokinetic model to simulate the dynamics of both adoptively transferred CAR-T cells and endogenous immune cells in solid tumors following vascular normalization.
View Article and Find Full Text PDFAngew Chem Int Ed Engl
September 2025
Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P.R. China.
The therapeutic efficacy of adoptive cell therapy is highly dependent on the status and function of the infused cells. However, insufficient nutrient availability within the immunosuppressive tumor microenvironment (TME) often impedes these cells from fully exerting their cytotoxic potential against solid tumors. Here, we present a strategy of integrating adoptively transferred macrophages with intracellular nutrient depots composed of L-arginine-based nanomicelles to provide a sustainable supply of essential metabolite and optimize the cellular activity in the nutrient-deprived TME.
View Article and Find Full Text PDFCytotherapy
August 2025
Division of Hematology and Cellular Therapy, Allegheny Health Network Cancer Institute, Pittsburgh, Pennsylvania, USA; Allegheny Health Network Research Institute, Pittsburgh, Pennsylvania, USA; Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA. Electroni
Background Aims: Cancers of many different tissue origins can metastasize to the pleura, a space with a unique immune environment that predisposes to aggressive tumor behavior and the development of effusions, an exudative leakage of serous fluid accompanied by an immune infiltrate. Effusions are drained therapeutically to relieve dyspnea, often several times per week. Characteristically, they contain 50-1000 × 10 viable pleural T cells (PITs), which can be reliably activated and expanded in culture, making them an ideal source for generation of a cellular therapeutic.
View Article and Find Full Text PDF