Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Sci Immunol ; 8(88): eadh1306, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37862431

ABSTRACT

Overcoming CD8+ T cell exhaustion is critical in cancer immunotherapy. Recently, an intratumor stem/progenitor-like CD8+ T cell (Tprog cell) population that mediates the persistence of antitumor responses has been defined, which can further develop into a terminally differentiated CD8+ T cell (Tterm cell) subpopulation with potent cytotoxic functions. Tprog cells are the main responders to immune checkpoint blockade therapies, yet how extrinsic signals via transcription factors control Tprog cell generation and persistence in tumors is unclear. Here, we found that BCL6 inhibits tumor-specific Tterm cell generation from Tprog cell downstream of TCF1. We show that Bcl6 deficiency reduced the persistence of Tprog cells, without affecting their generation, thus abrogating long-term tumor control. High-level BCL6 expression was observed in tumor-specific T cells in draining lymph nodes (LNs) and was associated with T cell exhaustion. This was observed in TOX+TCF1+ Tprog cells in both LNs and tumors. BCL6 expression in CD8+ T cells was up-regulated by TGF-ß-SMAD2 signaling but down-regulated by the IL-2-STAT5 pathway. Mechanistically, BCL6 transcriptionally repressed the expression of Tterm cell-associated genes and induced those of Tprog cell-related genes, in a manner antagonistic to BLIMP1. Prdm1 deficiency also promoted the Tprog cell program and greatly improved the efficacy of anti-PD-1 therapy. Thus, we identified the TGF-ß-BCL6 and IL-2-BLIMP1 antagonistic pathways in regulation of antitumor CD8+ T cells, which may benefit the development of long-lasting and effective cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Interleukin-2 , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta , Proto-Oncogene Proteins c-bcl-6/genetics
2.
J Exp Med ; 220(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-36688918

ABSTRACT

In cancer, persistent antigens drive CD8+ T cell differentiation into exhausted progenitor (Texprog) and terminally exhausted (Texterm) cells. However, how the extrinsic and intrinsic regulatory mechanisms cooperate during this process still remains not well understood. Here, we found that STAT3 signaling plays essential roles in promoting intratumor Texterm cell development by enhancing their effector functions and survival, which results in better tumor control. In tumor microenvironments, STAT3 is predominantly activated by IL-10 and IL-21, but not IL-6. Besides, STAT3 also plays critical roles in the development and function of terminally differentiated effector CD8+ T cells in acute infection. Mechanistically, STAT3 transcriptionally promotes the expression of effector function-related genes, while it suppresses those expressed by the progenitor Tex subset. Moreover, STAT3 functions in collaboration with BATF and IRF4 to mediate chromatin activation at the effector gene loci. Thus, we have elucidated the roles of STAT3 signaling in terminally differentiated CD8+ T cell development, especially in cancer, which benefits the development of more effective immunotherapies against tumors.


Subject(s)
Neoplasms , Signal Transduction , Humans , Cell Differentiation , Neoplasms/pathology , CD8-Positive T-Lymphocytes , Tumor Microenvironment , STAT3 Transcription Factor/metabolism
3.
Cell Mol Immunol ; 17(3): 227-236, 2020 03.
Article in English | MEDLINE | ID: mdl-31611650

ABSTRACT

Although PD-L1/PD-1 blockade therapy has been approved to treat many types of cancers, the majority of patients with solid tumors do not respond well, but the underlying reason remains unclear. Here, we studied ovarian cancer (OvCa), a tumor type generally resistant to current immunotherapies, to investigate PD-1-independent immunosuppression. We found that PD-L1 was not highly expressed in the tumor microenvironment (TME) of human OvCa. Instead, B7-H3, another checkpoint molecule, was highly expressed by both tumor cells and tumor-infiltrating antigen-presenting cells (APCs), which correlated with T-cell exhaustion in patients. Using ID8 OvCa mouse models, we found that B7-H3 expressed on tumor cells, but not host cells, had a dominant role in suppressing antitumor immunity. Therapeutically, B7-H3 blockade, but not PD-1 blockade, prolonged the survival of ID8 tumor-bearing mice. Collectively, our results demonstrate that tumor-expressed B7-H3 inhibits the function of CD8+ T cells and suggest that B7-H3 may be a target in patients who are not responsive to PD-L1/PD-1 inhibition, particularly OvCa patients.


Subject(s)
B7 Antigens/immunology , Gene Expression Regulation, Neoplastic/immunology , Immune Checkpoint Inhibitors/pharmacology , Neoplasm Proteins/immunology , Neoplasms, Experimental , Ovarian Neoplasms , T-Lymphocytes/immunology , Animals , B7 Antigens/genetics , Female , Humans , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , T-Lymphocytes/pathology
4.
Sheng Wu Gong Cheng Xue Bao ; 35(5): 775-783, 2019 May 25.
Article in Chinese | MEDLINE | ID: mdl-31222996

ABSTRACT

Messenger RNA (mRNA) can be modified by more than 100 chemical modifications. Among these modifications, N6-methyladenosine (m6A) is one of the most prevalent modifications. During the processes of cells differentiation, embryo development or stress, m6A can be modified on key mRNAs and regulate the progress of cells through modulating mRNA metabolism and translation. Other mRNA modifications, including N1-methyladenosine (m¹A), 5-methylcytosine (m5C) and pseudouridine, together with m6A form the epitranscriptome of mRNA that accurately modulate the mRNA translation. Here we review the types and characteristic of mRNA epigenetic modifications, especially the recent progresses of the function of m6A, we also expect the main research direction of m6A epigenetic modification in the future.


Subject(s)
Adenosine/analogs & derivatives , Epigenesis, Genetic , Gene Expression Regulation , RNA, Messenger , Adenosine/genetics , Adenosine/metabolism , Cell Differentiation/genetics , Embryonic Development/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism
5.
Mol Med Rep ; 18(6): 5409-5416, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30365132

ABSTRACT

Multidrug resistance (MDR) is a major concern when using chemotherapy for the treatment of patients with colorectal cancer. MDR modulators are agents that can reverse MDR and, thus, enhance the chemosensitivity of tumor cells. The development of MDR modulators can improve the therapeutic efficacies of MDR in cancer. However, few effective MDR modulators have been identified so far. Curcumin has been reported to be an effective compound in the reversal of MDR in colorectal cancer cells. However, the mechanisms associated with the reversal effect of curcumin on MDR and its regulation of target factors in MDR cells remain to be fully elucidated. 3­(4,5­dimethyl­2­thiazol)­2,5­diphenyltetrazolium bromide assays, flow cytometer apoptosis assays as well as mRNA and protein expression assays were performed in the present study, and the results confirmed the reversal effect of curcumin on HCT­8/5­Fu cells and provided evidence that activated nuclear factor erythroid 2­related factor (Nrf2) deficiency induced by the curcumin altered the B­cell lymphoma 2 (Bcl­2) associated X protein/Bcl­2 expression ratio, which led to the induction of apoptosis in HCT­8/5­Fu cells. These results indicated that Nrf2 may have a functional in the reversal effect of curcumin and contribute, at least in part, to the outcomes of chemotherapy in patients with MDR.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , NF-E2-Related Factor 2/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Genes, bcl-2 , Humans , Signal Transduction/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
Asian Pac J Trop Med ; 10(12): 1190-1196, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29268977

ABSTRACT

OBJECTIVE: To assess the effect of curcumin on CDDP-induced drug resistance and explore the underlying molecular mechanism through Nrf2 system and autophagy pathway. METHODS: A drug-resistant cell model was established by exposing A549/CDDP cell to 2 µg/mL CDDP. A549/CDDP cell was treated with 20 µg/mL CDDP and 10 µM curcumin. The cell viability and apoptosis level, the signals of Keap1/P62-Nrf2 and autophagy pathway were analyzed. RESULTS: CDDP induction promoted drug-resistant phenotype in A549/CDDP cell and activated autophagy as well as Nrf2 signals in A549/CDDP cell. Meanwhile, curcumin combination attenuated autophagy and Nrf2 activation induced by CDDP, and reversed the drug-resistant phenotype. Notably, curcumin combination augmented Keap1 transcription. Furthermore, Keap1 ablation with short hairpin RNAs hampered the efficacy of curcumin, suggesting Keap1 played a crucial role on reversal effect of curcumin. CONCLUSIONS: The present findings demonstrate that CDDP promotes abnormal activation of Nrf2 pathway and autophagy, leading to drug resistance of A549/CDDP cell. Curcumin attenuates this process and combat drug-resistance through its potent activation on Keap1 transcription, which is essential for interplay between oxidative stress induced Nrf2 activation and autophagy/apoptosis switch.

7.
Article in English | MEDLINE | ID: mdl-28694921

ABSTRACT

Understanding human brain development and disease is largely hampered by the relative inaccessibility of human brain tissues. Recent advances in human induced pluripotent stem cells (hiPSCs) have led to the generation of unlimited human neural cells and thereby facilitate the investigation of human brain development and pathology. Compared with traditional 2-dimensional (2D) culture methods, culturing the hiPSC-derived neural cells in a three-dimensional (3D) free-floating manner generates human central nervous system (CNS) organoids. These 3D CNS organoids possess the unique advantage of recapitulating multi-regional or region-specific cytoarchitecture seen in the early human fetal brain development. The CNS organoids are becoming a strong complement to the animal model in studying brain development and pathology, and developing new therapies to treat neurodevelopmental diseases. Further improvements to the long-term maintenance and neural maturation of the organoids may allow them to model neurodegenerative diseases. In this review, we will summarize the current development of hiPSCs to generate CNS organoids for modeling neurological disorders and future perspective.

8.
Clin Lung Cancer ; 16(2): 86-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25458559

ABSTRACT

The success in identifying the chromosomal rearrangements involving the anaplastic lymphoma kinase (ALK) as an oncogenic driver has thoroughly changed the treatment of non-small-cell lung cancer. In the past decade, targeted drugs have emerged as an efficient personalized strategy for ALK-rearranged non-small-cell lung cancer. The accelerated approval of potent ALK inhibitors, such as crizotinib and more recently ceritinib (LDK378), based on the well designed phase I/II trials has been a landmark success in clinical cancer research and contributes a new era of oncogenic targeted therapy characterized by elegant clinical trial design. In this review, we aim to present the current knowledge on acquired resistance of crizotinib known as a first-in-class ALK inhibitor and potential solutions to improve the cost-effectiveness, and to review the difference between ceritinib and crizotinib; preclinical data and results of the elegant early clinical trial of ceritinib which promoted its accelerated approval, pharmacokinetics, safety profile, and tolerability, the updated results (eg, efficacy on brain metastases), and robust design of ongoing phase II/III trials, and future directions of ceritinib to be a potent alternative to crizotinib for ALK-rearranged non-small-cell lung cancer are also presented.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Pyrimidines/therapeutic use , Sulfones/therapeutic use , Anaplastic Lymphoma Kinase , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Crizotinib , Drug Resistance, Neoplasm , Gene Rearrangement , Humans , Lung Neoplasms/pathology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Sulfones/adverse effects , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...