Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Publication year range
1.
Breast Cancer Res ; 25(1): 115, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37794509

ABSTRACT

BACKGROUND: B7 homology 4 (B7-H4), a potential target for cancer therapy, has been demonstrated to inhibit T cell cytotoxicity in the early stages of breast cancer. However, B7-H4 manipulating breast tumor immune microenvironment (TIME) in the tumor progression remains unknown. METHODS: We engineered T cells with B7-H4-specific chimeric antigen receptors (CARs) and performed a T cell co-culture assay to characterize B7-H4 expression level in breast cancer cells escaping from T cell cytotoxicity. We generated B7-H4 knockout (KO) and overexpression (OE) breast cancer cells to determine the epithelial-to-mesenchymal transition (EMT) and stemness characteristics in vitro and in vivo, including tumor proliferation, migration, metastasis and chemoresistance. The Cancer Genome Atlas breast cancer database was accessed to investigate the correlation between B7-H4 expression levels and EMT characteristics in patients with breast cancer. RESULTS: Our result found that B7-H4 expression level was significantly reduced in a subset of breast cancer cells that escaped from the cytotoxicity of B7-H4 CAR-T cells. Compared with wild type cells, B7-H4 KO cells prompt EMT and stemness characteristics, including migration, invasion and metastasis, and OE cells vice versa. The increase in H3K27me3 in KO cells confirmed the epigenetic reprogramming of cancer stem cells. The IC50 of doxorubicin or oxaliplatin significantly increased in KO cells, which was in agreement with a decrease in OE cells. Moreover, a trend of downregulated B7-H4 from stage I to stage II breast cancer patients indicates that the low-expressing B7-H4 breast cancer cells escaping from TIME have spread to nearby breast lymph nodes in the cancer progression. CONCLUSIONS: Our study illuminates the novel role of renouncing B7-H4 in breast cancer cells through immune escape, which contributes to EMT processes and provides new insights for breast cancer treatments.


Subject(s)
Breast Neoplasms , T-Lymphocytes , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Tumor Microenvironment/genetics
2.
Int Immunopharmacol ; 122: 110616, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37459784

ABSTRACT

Macrophage pyroptosis plays an important role in the development of radiation-induced cell and tissue damage, leading to acute lung injury. However, the underlying mechanisms of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3)-mediated macrophage pyroptosis and the regulatory factors involved in radiation-induced pyroptosis are unclear. In this study, the expression of the NLRP3 inflammasome and pyroptosis-associated factors in murine macrophage cell lines was investigated after ionizing radiation. High-throughput RNA sequencing was performed to identify and characterize miRNAs and mRNA transcripts associated with NLRP3-mediated cell death. Our results demonstrated that cleaved-caspase-1 (p10) and N-terminal domain of gasdermin-D (GSDMD-N) were upregulated, and the number of NLRP3 inflammasomes and pyroptotic cells increased in murine macrophage cell lines after irradiation (8 Gy). Comparativeprofiling of 300miRNAs revealed that 41 miRNAsexhibited significantly different expression after 8 Gy of irradiation. Granulocyte-specific microRNA-223-3p (miR-223-3p) is a negative regulator of NLRP3. In vitro experiments revealed that the expression of miR-223-3p was significantly altered by irradiation. Moreover, miR-223-3p decreased the expression of NLRP3 and proinflammatory factors, resulting in reduced pyroptosis in irradiated murine macrophages. Subsequently, in vivo experiments revealed the efficacy of miR-223-3p supplementation in ameliorating alveolar macrophage (AM) pyroptosis, attenuating the infiltration of inflammatory monocytes, and significantly alleviating the severity of acute radiation-induced lung injury (ARILI). Our findings suggest that the miR-223-3p/NLRP3/caspase-1 axis is involved in radiation-induced AM pyroptosis and ARILI.


Subject(s)
Inflammasomes , MicroRNAs , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Pyroptosis/physiology , Caspase 1/metabolism
3.
Phytomedicine ; 108: 154508, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36332384

ABSTRACT

BACKGROUND: Myeloid cell-mediated immunosuppression is a major obstacle to checkpoint blockade immunotherapy. We previously reported that total biflavonoids extract from Selaginella doederleinii (TBESD) and a flavone monomer isolated from TBESD, named Delicaflavone, have favorable anti-tumor activity. However, whether TBESD and Delicaflavone could affect the tumor microenvironment (TME) remains unclear. PURPOSE: In this study, we focused on the TME to determine whether TBESD and Delicaflavone could restore anti-tumor immune response. METHODS: 4T1 tumor-bearing immunocompetent BALB/c mice and T cell-deficient nude mice were used to examine the effect of TBESD on T cell-mediated immunity in vivo. Multi-parameter flow cytometry was conducted to evaluate the impacts of TBESD on TME. Primary cells, including murine CD8+ T cells, tumor associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) were prepared to investigate the modulatory activities of TBESD on immune cells. It was further determined whether Delicaflavone or Amentoflavone, two typical functional biflavones from TBESD, mediated those effects of TBESD. Finally, the impacts of TBESD and Delicaflavone on Jak1/STAT6 signaling pathway were explored via western blot. RESULTS: We found that TBESD significantly reduced 4T1 tumor growth in immunocompetent BALB/c mice, but not in nude mice. This effect was associated with the regulation of TME, shown as an increase in functional T cells and M1 phenotype TAMs (M1-TAMs), and a decrease in M2 phenotype TAMs (M2-TAMs), monocytic-MDSCs (M-MDSCs) and regulatory T cells (Tregs) in TBESD-treated BALB/c mouse 4T1 tumors. It was found ex vivo that TBESD restrained the viability and immunosuppressive properties of M2-TAMs and M-MDSCs, especially for the loss of arginase-1 expression. Additionally, TBESD re-educated M2-TAMs to an M1 like phenotype. Further investigations determined that Delicaflavone predominantly mediated the immuno-modulatory activities of TBESD both ex vivo and in vivo. Finally, Delicaflavone and TBESD blocked Jak1/STAT6 signaling pathway in M2-TAMs and MDSCs. CONCLUSION: The present study suggests Delicaflavone as a potent natural inhibitor of M2-TAMs and MDSCs, which fills the gap in knowledge on the immuno-modulatory effects of TBESD and Delicaflavone, and could have translational implications to improve the efficacy of cancer immunotherapy.


Subject(s)
Neoplasms , Selaginellaceae , Animals , Mice , Mice, Nude , CD8-Positive T-Lymphocytes , Myeloid Cells , Mice, Inbred BALB C , Immunity , Immunosuppression Therapy , Cell Line, Tumor , Tumor Microenvironment
4.
Int Immunopharmacol ; 118: 110030, 2023 May.
Article in English | MEDLINE | ID: mdl-36989893

ABSTRACT

BACKGROUND: Due to the negative association between inhibitor of nuclear factor-kB kinase-interacting protein (IKBIP) and survival in gliomas, this study aimed to comprehensively analyze the potential function of IKBIP in glioblastoma multiforme (GBM). METHODS: GBM samples were retrieved from The Cancer Genome Atlas and Chinese Glioma Genome Atlas as training and validation cohorts, respectively, and survival and Cox regression analyses were conducted. Based on clinical indicators and IKBIP, three prognostic models were established and then verified using the validation dataset. Infiltrating immune cell analysis and single-sample gene set enrichment analysis were also conducted to explore the underlying mechanisms. Finally, the key findings were validated through molecular biology experiments. RESULTS: Patients in the high IKBIP score group had poorer survival. Based on Cox regression and subgroup analyses, IKBIP was identified as an independent prognostic factor. Among the three models constructed, the model combining the IKBIP signature and clinical features displayed good performance in terms of discrimination, calibration, and model improvement capability in the training cohort. This model was also successfully validated in an external cohort from the CGGA. Further analysis revealed that many immune cells and related pathways were involved in the high-risk group. In vitro experiments revealed that the knockdown of IKBIP inhibited cell invasion and proliferation, and promoted their senescence. CONCLUSIONS: The prognostic value of IKBIP and its positive impact on the invasiveness of GBM were identified, indicating that IKBIP may serve as an underlying target for the treatment of GBM.


Subject(s)
Glioblastoma , Glioma , Humans , Prognosis , Biomarkers
SELECTION OF CITATIONS
SEARCH DETAIL