Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
STAR Protoc ; 4(4): 102679, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37910511

ABSTRACT

Here, we present a protocol for collecting, dissociating, isolating, staining, and analyzing immune cells from pancreatic cancer tissues for flow cytometry. The isolated cells can also be used for single-cell RNA sequencing and other related procedures. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2023).1.


Subject(s)
Pancreatic Neoplasms , Humans , Flow Cytometry , Pancreatic Neoplasms/genetics , Staining and Labeling
2.
Mol Ther ; 31(10): 2929-2947, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37515321

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is not sensitive to immune checkpoint blockade therapy, and negative feedback of tumor immune evasion might be partly responsible. We isolated CD8+ T cells and cultured them in vitro. Proteomics analysis was performed to compare changes in Panc02 cell lines cultured with conditioned medium, and leucine-rich repeat kinase 2 (LRRK2) was identified as a differential gene. LRRK2 expression was related to CD8+ T cell spatial distribution in PDAC clinical samples and upregulated by CD8+ T cells via interferon gamma (IFN-γ) simulation in vitro. Knockdown or pharmacological inhibition of LRRK2 activated an anti-pancreatic cancer immune response in mice, which meant that LRRK2 acted as an immunosuppressive gene. Mechanistically, LRRK2 phosphorylated PD-L1 at T210 to inhibit its ubiquitination-mediated proteasomal degradation. LRRK2 inhibition attenuated PD-1/PD-L1 blockade-mediated, T cell-induced upregulation of LRRK2/PD-L1, thus sensitizing the mice to anti-PD-L1 therapy. In addition, adenosylcobalamin, the activated form of vitamin B12, which was found to be a broad-spectrum inhibitor of LRRK2, could inhibit LRRK2 in vivo and sensitize PDAC to immunotherapy as well, which potentially endows LRRK2 inhibition with clinical translational value. Therefore, PD-L1 blockade combined with LRRK2 inhibition could be a novel therapy strategy for PDAC.

3.
Cell Rep ; 42(6): 112620, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37285267

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer that typically demonstrates resistance to chemotherapy. Tumor-associated macrophages (TAMs) are essential in tumor microenvironment (TME) regulation, including promoting chemoresistance. However, the specific TAM subset and mechanisms behind this promotion remain unclear. We employ multi-omics strategies, including single-cell RNA sequencing (scRNA-seq), transcriptomics, multicolor immunohistochemistry (mIHC), flow cytometry, and metabolomics, to analyze chemotherapy-treated samples from both humans and mice. We identify four major TAM subsets within PDAC, among which proliferating resident macrophages (proliferating rMφs) are strongly associated with poor clinical outcomes. These macrophages are able to survive chemotherapy by producing more deoxycytidine (dC) and fewer dC kinases (dCKs) to decrease the absorption of gemcitabine. Moreover, proliferating rMφs promote fibrosis and immunosuppression in PDAC. Eliminating them in the transgenic mouse model alleviates fibrosis and immunosuppression, thereby re-sensitizing PDAC to chemotherapy. Consequently, targeting proliferating rMφs may become a potential treatment strategy for PDAC to enhance chemotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , Drug Resistance, Neoplasm , Multiomics , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Macrophages/metabolism , Fibrosis , Tumor Microenvironment , Pancreatic Neoplasms
4.
J Immunother Cancer ; 11(2)2023 02.
Article in English | MEDLINE | ID: mdl-36849200

ABSTRACT

BACKGROUND: Solid tumors pose unique roadblocks to treatment with chimeric antigen receptor (CAR) T cells, including limited T-cell persistence, inefficient tumor infiltration, and an immunosuppressive tumor microenvironment. To date, attempts to overcome these roadblocks have been unsatisfactory. Herein, we reported a strategy of combining Runx3 (encoding RUNX family transcription factor 3)-overexpression with ex vivo protein kinase B (AKT) inhibition to generate CAR-T cells with both central memory and tissue-resident memory characteristics to overcome these roadblocks. METHODS: We generated second-generation murine CAR-T cells expressing a CAR against human carbonic anhydrase 9 together with Runx3-overexpression and expanded them in the presence of AKTi-1/2, a selective and reversible inhibitor of AKT1/AKT2. We explored the influence of AKT inhibition (AKTi), Runx3-overexpression, and their combination on CAR-T cell phenotypes using flow cytometry, transcriptome profiling, and mass cytometry. The persistence, tumor-infiltration, and antitumor efficacy of CAR-T cells were evaluated in subcutaneous pancreatic ductal adenocarcinoma (PDAC) tumor models. RESULTS: AKTi generated a CD62L+central memory-like CAR-T cell population with enhanced persistence, but promotable cytotoxic potential. Runx3-overexpression cooperated with AKTi to generate CAR-T cells with both central memory and tissue-resident memory characteristics. Runx3-overexpression enhanced the potential of CD4+CAR T cells and cooperated with AKTi to inhibit the terminal differentiation of CD8+CAR T cells induced by tonic signaling. While AKTi promoted CAR-T cell central memory phenotype with prominently enhanced expansion ability, Runx3-overexpression promoted the CAR-T cell tissue-resident memory phenotype and further enhanced persistence, effector function, and tumor-residency. These novel AKTi-generated Runx3-overexpressing CAR-T cells exhibited robust antitumor activity and responded well to programmed cell death 1 blockade in subcutaneous PDAC tumor models. CONCLUSIONS: Runx3-overexpression cooperated with ex vivo AKTi to generate CAR-T cells with both tissue-resident and central memory characteristics, which equipped CAR-T cells with better persistence, cytotoxic potential, and tumor-residency ability to overcome roadblocks in the treatment of solid tumors.


Subject(s)
Carcinoma, Pancreatic Ductal , Internship and Residency , Pancreatic Neoplasms , Humans , Animals , Mice , Proto-Oncogene Proteins c-akt , Signal Transduction , Tumor Microenvironment , Core Binding Factor Alpha 3 Subunit , Pancreatic Neoplasms
5.
Bioact Mater ; 20: 259-270, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35702611

ABSTRACT

Rationale: Hypoxia in tumor microenvironment (TME) represents an obstacle to the efficacy of immunotherapy for pancreatic ductal adenocarcinoma (PDAC) through several aspects such as increasing the expression of immune checkpoints or promoting fibrosis. Reversing hypoxic TME is a potential strategy to improve the validity of immune checkpoint blockade (ICB). Methods: Here, we synthesized polydopamine-nanoparticle-stabilized oxygen microcapsules with excellent stabilization, bioavailability, and biocompatibility for direct oxygen delivery into tumor sites by interfacial polymerization. Results: We observed oxygen microcapsules enhanced the oxygen concentration in the hypoxia environment and maintained the oxygen concentration for a long period both in vitro and in vivo. We found that oxygen microcapsules could significantly improve the efficiency of ICB against PDAC in vivo. Mechanismly, combined treatments using oxygen microcapsules and ICB could reduce the infiltration of tumor-associated macrophages (TAMs) and polarized pro-tumor M2 macrophages into anti-tumor M1 macrophages. In addition, combined treatments could elevate the proportion of T helper subtype 1 cells (Th1 cells) and cytotoxic T lymphocytes cells (CTLs) to mediate anti-tumor immune response in TME. Conclusion: In summary, this pre-clinical study indicated that reversing hypoxia in TME by using oxygen microcapsules was an effective strategy to improve the performances of ICB on PDAC, which holds great potential for treating PDAC in the future.

6.
Front Biosci (Landmark Ed) ; 27(5): 145, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35638412

ABSTRACT

Hypoxia is a typical characteristic of most solid malignancies, which has multiple effects on malignant phenotypes and biological behaviors of tumors including epithelial-mesenchymal-transition (EMT), invasion, migration, metastasis, autophagy, stem cell maintenance, pathological angiogenesis, drug resistance, and immunosuppression. Rcentlyumoand reversing the tumor hypoxic environment via nanotechnology has emerged as a novel therapeutic approach for the treatment of malignancies. The main strategies related to nanotechnology to alleviate or ameliorate hypoxic environment are as follows: (1) Bringing external oxygen to tumor hypoxic microenvironment; (2) Generating oxygen based on nanotechnology in situ; (3) Regulating the structure of the tumor microenvironment; (4) Decreasing oxygen consumption in the tumor microenvironment. In this review, we will discuss these nanotechnologies in detail.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Hypoxia/therapy , Nanotechnology , Neoplasms/genetics , Oxygen
7.
J Exp Clin Cancer Res ; 41(1): 56, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35139879

ABSTRACT

Mortality associated with pancreatic cancer is among the highest of all malignancies, with a 5-year overall survival of 5-10%. Immunotherapy, represented by the blocking antibodies against programmed cell death protein 1 or its ligand 1 (anti-PD-(L)1), has achieved remarkable success in a number of malignancies. However, due to the immune-suppressive tumor microenvironment, the therapeutic efficacy of anti-PD-(L)1 in pancreatic cancer is far from expectation. To address such a fundamental issue, chemotherapy, radiotherapy, targeted therapy and even immunotherapy itself, have individually been attempted to combine with anti-PD-(L)1 in preclinical and clinical investigation. This review, with a particular focus on pancreatic cancer therapy, collects current anti-PD-(L)1-based combination strategy, highlights potential adverse effects of accumulative combination, and further points out future direction in optimization of combination, including targeting post-translational modification of PD-(L)1 and improving precision of treatment.


Subject(s)
B7-H1 Antigen/therapeutic use , Immunotherapy/methods , Pancreatic Neoplasms/drug therapy , B7-H1 Antigen/pharmacology , Humans , Pancreatic Neoplasms/mortality , Survival Analysis , Tumor Microenvironment
8.
Gut ; 71(6): 1176-1191, 2022 06.
Article in English | MEDLINE | ID: mdl-34253573

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) tumour microenvironment (TME) is highly complex with diverse cellular components organising into various functional units, cellular neighbourhoods (CNs). And we wanted to define CN of HCC while preserving the TME architecture, based on which, potential targets for novel immunotherapy could be identified. DESIGN: A highly multiplexed imaging mass cytometry (IMC) panel was designed to simultaneously quantify 36 biomarkers of tissues from 134 patients with HCC and 7 healthy donors to generate 562 highly multiplexed histology images at single-cell resolution. Different function units were defined by topological analysis of TME. CN relevant to the patients' prognosis was identified as specific target for HCC therapy. Transgenic mouse models were used to validate the novel immunotherapy target for HCC. RESULTS: Three major types of intratumour areas with distinct distribution patterns of tumorous, stromal and immune cells were identified. 22 cellular metaclusters and 16 CN were defined. CN composed of various types of cells formed regional function units and the regional immunity was regulated reversely by resident Kupffer cells and infiltrating macrophages with protumour and antitumour function, respectively. Depletion of Kupffer cells in mouse liver largely enhances the T cell response, reduces liver tumour growth and sensitises the tumour response to antiprogrammed cell death protein-1 treatment. CONCLUSION: Our findings reveal for the first time the various topological function units of HCC TME, which also presents the largest depository of pathological landscape for HCC. This work highlights the potential of Kupffer cell-specific targeting rather than overall myeloid cell blocking as a novel immunotherapy for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/pathology , Humans , Image Cytometry , Liver Neoplasms/pathology , Macrophages , Mice , Tumor Microenvironment
10.
Dose Response ; 17(1): 1559325818822938, 2019.
Article in English | MEDLINE | ID: mdl-30828271

ABSTRACT

Quorum sensing inhibitors (QSIs) are being used widely as a promising alternative to antibiotics and drawing attention as potential pollutants. However, the assessment methods of the toxicities of QSIs, including model organism and affecting time, have not been established. To investigate how model organism and acting time impact the toxicities of QSIs, the effect of 4 QSIs to Aliivibrio fischeri and Bacillus subtilis were determined at different exposing time in the present study. The results showed that the toxic effects of QSIs to gram-negative bacteria (A fischeri) and gram-positive bacteria (B subtilis) were different and time dependent. As for A fischeri, QSI (furaneol acetate, FA) merely showed inhibition on the bioluminescence from hours 1 to 2. But from hours 3 to 6, low concentration FA exerted stimulation on the bioluminescence. Then, this stimulation disappeared from hours 7 to 14, and after hour 15 the stimulation appeared again. That is to say, QSIs showed intermittent hormesis effect on the bioluminescence of A fischeri. By contrast, only inhibition was observed in the toxicity test process of QSIs to B subtilis. As exposing time goes, the inhibition weakened gradually when FA was at low concentration regions. What is more, in the present, study toxic mechanisms were also discussed based on model organisms and exposing time. This study demonstrates appreciable impacts of model organism and exposing time on toxicities of QSIs and provides a theoretical basis for risk assessments after QSIs being widely used into the environment.

11.
Sci Total Environ ; 657: 46-55, 2019 Mar 20.
Article in English | MEDLINE | ID: mdl-30530218

ABSTRACT

Hormesis is a dose-response relationship phenomenon characterized by low-dose stimulation and high-dose inhibition. Although hormetic phenomena have been reported in broadly ranging biological areas, there is still no unified mechanism of hormesis. Investigating multiple-species hormesis of one compound and then exploring the possible mechanism may be an effective approach to clarify the reason for the occurrence of hormetic phenomena in a broad range of organisms. In this study, indole was selected as the test chemical due to the broad biological and hormetic effects of indole compounds. The results show that indole induces multiple-species hormetic phenomena in bacteria (Aliivibrio fischeri (A. fischeri), Escherichia coli and Bacillus subtilis), algae (Microcystis aeruginosa and Selenastrum capricornutum), and human cells (human skin fibroblasts and human cervical cancer cells). Through in-depth investigation of the time-dependent hormetic effects of indole, indole derivatives and indole's structural analogs on the bioluminescence of A. fischeri, indole ring has been identified as the potential key structure that causes indole to act on quorum sensing of A. fischeri to induce hormetic effects on the bioluminescence at lag, logarithmic, and stationary phases. Therefore, the occurrence of multiple-species hormetic phenomena is speculated to be derived from the action of indole on the cell-to-cell communication of organism cells. This paper can not only further confirm the generalizability of hormesis but also provide a reasonable explanation for hormesis, which will benefit the development of hormesis and the risk assessment of environmental pollutants.


Subject(s)
Bacteria/drug effects , Hormesis , Indoles/pharmacology , Microalgae/drug effects , Cell Line , HeLa Cells , Humans , Species Specificity
12.
Sci Rep ; 5: 8147, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25634769

ABSTRACT

Chinese white dolphins (Sousa chinensis) inhabiting shallow coastal waters are vulnerable to impacts from human activities in the near shore waters. This study examined the population of Chinese white dolphins occurring off the coast of Zhanjiang in the northern South China Sea. A total of 492 Chinese white dolphins were identified, 176 of which were photographed on more than one occasion. The Zhanjiang Chinese white dolphin population is isolated from populations of conspecifics along the Guangdong coast. It is composed of approximately 1485 individuals (95% CI = 1371-1629; SE = 63.8), with estimates of mean representative range and core area of 168.51 and 44.26 km(2), respectively. The high site fidelity and long-term residence of Chinese white dolphins in the study area are well established. A review of all available data indicates that based on what is currently known, the Zhanjiang Chinese white dolphin population is the second largest of the species and genus in the world. However, the recent industrial boom along the Zhanjiang coast has increased concerns regarding the conservation of the Zhanjiang Chinese white dolphin population. We recommend the designation of a national nature reserve as a most urgent measure for protecting Chinese white dolphins in Zhanjiang waters.


Subject(s)
Conservation of Natural Resources , Dolphins , Animals , China , Demography , Geography , Population Density
SELECTION OF CITATIONS
SEARCH DETAIL
...