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.
Exp Hematol Oncol ; 13(1): 58, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822440

ABSTRACT

BACKGROUND: Gallbladder cancer (GBC) is the most common and lethal malignancy of the biliary tract that lacks effective therapy. In many GBC cases, infiltration into adjacent organs or distant metastasis happened long before the diagnosis, especially the direct liver invasion, which is the most common and unfavorable way of spreading. METHODS: Single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST), proteomics, and multiplexed immunohistochemistry (mIHC) were performed on GBC across multiple tumor stages to characterize the tumor microenvironment (TME), focusing specifically on the preferential enrichment of neutrophils in GBC liver invasion (GBC-LI). RESULTS: Multi-model Analysis reveals the immunosuppressive TME of GBC-LI that was characterized by the enrichment of neutrophils at the invasive front. We identified the context-dependent transcriptional states of neutrophils, with the Tumor-Modifying state being associated with oxidized low-density lipoprotein (oxLDL) metabolism. In vitro assays showed that the direct cell-cell contact between GBC cells and neutrophils led to the drastic increase in oxLDL uptake of neutrophils, which was primarily mediated by the elevated OLR1 on neutrophils. The oxLDL-absorbing neutrophils displayed a higher potential to promote tumor invasion while demonstrating lower cancer cytotoxicity. Finally, we identified a neutrophil-promoting niche at the invasive front of GBC-LI that constituted of KRT17+ GBC cells, neutrophils, and surrounding fibroblasts, which may help cultivate the oxLDL-absorbing neutrophils. CONCLUSIONS: Our study reveals the existence of a subset of pro-tumoral neutrophils with a unique ability to absorb oxLDL via OLR1, a phenomenon induced through cell-cell contact with KRT17+ GBC cells in GBC-LI.

2.
Sci Transl Med ; 15(706): eadg3358, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37494474

ABSTRACT

Organoid models have the potential to recapitulate the biological and pharmacotypic features of parental tumors. Nevertheless, integrative pharmaco-proteogenomics analysis for drug response features and biomarker investigation for precision therapy of patients with liver cancer are still lacking. We established a patient-derived liver cancer organoid biobank (LICOB) that comprehensively represents the histological and molecular characteristics of various liver cancer types as determined by multiomics profiling, including genomic, epigenomic, transcriptomic, and proteomic analysis. Proteogenomic profiling of LICOB identified proliferative and metabolic organoid subtypes linked to patient prognosis. High-throughput drug screening revealed distinct response patterns of each subtype that were associated with specific multiomics signatures. Through integrative analyses of LICOB pharmaco-proteogenomics data, we identified the molecular features associated with drug responses and predicted potential drug combinations for personalized patient treatment. The synergistic inhibition effect of mTOR inhibitor temsirolimus and the multitargeted tyrosine kinase inhibitor lenvatinib was validated in organoids and patient-derived xenografts models. We also provide a user-friendly web portal to help serve the biomedical research community. Our study is a rich resource for investigation of liver cancer biology and pharmacological dependencies and may help enable functional precision medicine.


Subject(s)
Liver Neoplasms , Proteogenomics , Humans , Proteomics , Precision Medicine , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Organoids
3.
Am J Transl Res ; 12(6): 2890-2902, 2020.
Article in English | MEDLINE | ID: mdl-32655817

ABSTRACT

PURPOSE: To detect the expression of AGO1 in cholangiocarcinoma and explore its role and significance in the progression of cholangiocarcinoma. PATIENTS AND METHODS: Using immunohistochemistry, Western blotting and qPCR, we examined the expression of AGO1 in cholangiocarcinoma tissues. Through the analysis of clinical case data, the relationship between AGO1 and clinical prognosis was explored. The effect of AGO1 on cholangiocarcinoma was verified by cell functional experiments. Finally, we examined the effects of AGO1 on EMT-related proteins and signaling pathways. RESULTS: AGO1 is significantly upregulated in cholangiocarcinoma and is associated with the prognosis of cholangiocarcinoma. AGO1 can significantly increase the proliferation, migration and invasion of cholangiocarcinoma cell lines. AGO1 affects the prognosis of cholangiocarcinoma by affecting epithelial-mesenchymal transition (EMT)-related TGF-ß-PI3K-AKT signaling pathways. CONCLUSION: AGO1 is an independent predictor of cholangiocarcinoma prognosis and a potential target for the treatment of cholangiocarcinoma.

4.
Pancreatology ; 19(1): 105-113, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30579733

ABSTRACT

OBJECTIVES: This study aimed to explore double-negative T (DNT) cell cytotoxicity to pancreatic cancer and the effect of the Fas (CD95, APO-1)/FasL (CD178) signaling pathway on this process. METHODS: DNT cells from the peripheral blood of healthy volunteers were expanded in vitro. The inhibitory effect of DNT cells on pancreatic cancer cells was investigated using a CCK-8 assay and nude mouse tumor model. A mechanistic study was performed using pathway blocking assays. RESULTS: DNT cells were amplified in vitro with >90% purity, and the growth of pancreatic cancer in vitro was significantly inhibited by DNT cells. After coculture with DNT cells, Fas, caspase-8 and cleaved caspase-8 showed increased expression in pancreatic cancer cells. When blocking agent decoy receptor 3 (DcR3) was added, the antitumor effect of DNT cells and the expression of Fas, caspase-8 and cleaved caspase-8 were reduced in pancreatic cancer cells. In the nude mouse tumor model, the tumor volume and weight were lower in the DNT cell group and gemcitabine group than in the blank control group. Additionally, the expression of Fas, caspase-8 and cleaved caspase-8 was higher in the DNT cell group than in the blank control group. Moreover, DNT cells promoted apoptosis in cancer cells and animal model tissues. CONCLUSION: DNT cells inhibited the growth of pancreatic cancer, and the Fas/FasL signaling pathway was involved in this process.


Subject(s)
Pancreatic Neoplasms/therapy , T-Lymphocyte Subsets , Animals , Antineoplastic Agents/therapeutic use , Cell Line , Cell Transplantation , Coculture Techniques , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Fas Ligand Protein , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental , fas Receptor , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...