Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Publication year range
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(3): 497-504, 2023 May.
Article in Chinese | MEDLINE | ID: mdl-37248574

ABSTRACT

Metabolic reprogramming, an important hallmark of cancer, helps cancer achieve rapid proliferation. Metabolic changes in tumors regulate multiple metabolic pathways of immune cells, thereby suppressing antitumor immunity. Recent studies have been focused on in-depth investigation into the changes in the metabolism of glucose, amino acids, and lipids. Researchers have also conducted in-depth exploration of the interactive metabolic regulation of tumor cells and immune cells. Targeting various metabolic mechanisms while combining available anti-tumor therapies and enhancing the anti-tumor effects of immunotherapy by satisfying the metabolic demands of immune cells has offered new perspectives for therapies targeting the immune metabolism of tumors and enhancing anti-tumor immune responses. Studies on novel immune checkpoint molecules and cellular immunotherapies are also ongoing. Herein, we reviewed the latest findings on the mechanisms of immune metabolism underlying tumor immunosuppression and their application in immunotherapy. We also suggested some ideas for the future development of the regulation of immune metabolism.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Immunotherapy , Tumor Microenvironment
2.
Clin Transl Med ; 11(10): e540, 2021 10.
Article in English | MEDLINE | ID: mdl-34709764

ABSTRACT

Natural killer (NK) cells preferentially accumulate at maternal-foetal interface and are believed to play vital immune-modulatory roles during early pregnancy and related immunological dysfunction may result in pregnant failure such as recurrent miscarriage (RM). However, the mechanisms underlying the establishment of maternal-foetal immunotolerance are complex but clarifying the roles of decidual NK (dNK) cells offers the potential to design immunotherapeutic strategies to assist RM patients. In this report, we analysed RNA sequencing on peripheral NK (pNK) and decidual NK cells during early pregnancy; we identified an immunomodulatory dNK subset CXCR4+ CD56bright dNK and investigated its origin and phenotypic and functional characteristics. CXCR4+ CD56bright dNK displayed a less activated and cytotoxic phenotype but an enhanced immunomodulatory potential relative to the CXCR4 negative subset. CXCR4+ CD56bright dNK promote Th2 shift in an IL-4-dependent manner and can be recruited from peripheral blood and reprogramed by trophoblasts, as an active participant in the establishment of immune-tolerance during early pregnancy. Diminished CXCR4+ dNK cells and their impaired ability to induce Th2 differentiation were found in RM patients and mouse models of spontaneous abortion. Moreover, adoptive transfer of CXCR4+ dNK cells to NK-deficient (Nfil3-/-) mice showed great therapeutic potential of CXCR4+ dNK via recovering the Th2/Th1 bias and reducing embryo resorption rates. The identification of this new dNK cell subset may lay the foundation for understanding NK cell mechanisms in early pregnancy and provide potential prognostic factors for the diagnosis and therapy of RM.


Subject(s)
Abortion, Habitual/prevention & control , Immune Tolerance/immunology , Killer Cells, Natural/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Abortion, Habitual/blood , Abortion, Habitual/immunology , Animals , Decidua/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Neural Cell Adhesion Molecules/blood , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/immunology , Pregnancy , Pregnancy Trimester, First , Receptors, CXCR4/blood
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 52(5): 735-739, 2021 Sep.
Article in Chinese | MEDLINE | ID: mdl-34622585

ABSTRACT

In recent years, immunotherapy, as an emerging anti-tumor therapy, has shown great potential in the treatment of both solid and hematologic tumors. There is increasing preclinical and clinical evidence linking the composition of gut microbiome with the efficacy as well as adverse effects of immune checkpoint inhibitor anti-tumor therapy. We summarized in this review the modulatory role of the gut microbiome in antitumor therapy with different immune checkpoint inhibitors. We also discussed the limitations of existing research and prospective development of the further clinical strategies.


Subject(s)
Gastrointestinal Microbiome , Neoplasms , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms/drug therapy , Prospective Studies
4.
Int J Oncol ; 41(2): 523-32, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22581080

ABSTRACT

Orlistat is an orally administered anti-obesity drug that has shown significant antitumor activity in a variety of tumor cells. To identify the proteins involved in its antitumor activity, we employed a proteomic approach to reveal protein expression changes in the human ovarian cancer cell line SKOV3, following Orlistat treatment. Protein expression profiles were analyzed by 2-dimensional polyacrylamide gel electrophoresis (2-DE) and protein identification was performed on a MALDI-Q-TOF MS/MS instrument. More than 110 differentially expressed proteins were visualized by 2-DE and Coomassie brilliant blue staining. Furthermore, 71 proteins differentially expressed proteins were positively identified via mass spectrometry (MS)/MS analysis. In particular, PKM1/2, a key enzyme involved in tumorigenesis, was found to be significantly downregulated in SKOV3 cells following treatment with Orlistat. Moreover, PKM1/2 was proved to be downregulated in SKOV3 cells by western blot analysis after treatment with Orlistat. Taken together, using proteomic tools, we identified several differentially expressed proteins that underwent Orlistat-induced apoptosis, particularly PKM2. These changes confirmed our hypothesis that Orlistat is a potential inhibitor of ovarian cancer and can be used as a novel adjuvant antitumor agent.


Subject(s)
Antineoplastic Agents/pharmacology , Lactones/pharmacology , Proteome/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cluster Analysis , Electrophoresis, Gel, Two-Dimensional , Fatty Acid Synthase, Type I/metabolism , Female , Gene Expression Profiling , Humans , Membrane Proteins/metabolism , Orlistat , Ovarian Neoplasms , Proteomics , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
5.
J Exp Clin Cancer Res ; 29: 76, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20565783

ABSTRACT

BACKGROUND: Camptothecin (CPT) has recently attracted increasing attention as a promising anticancer agent for a variety of tumors. But the clinical application is largely hampered by its extreme water insolubility and unpredictable side effect. It is essential to establish an efficient and safe protocol for the administration of CPT versus melanoma. METHODS: Camptothecin was encapsulated with N-trimethyl chitosan (CPT-TMC) through microprecipitation and sonication. Its inhibition effect on B16-F10 cell proliferation and induction of apoptosis was evaluated by MTT assay and flow cytometric analysis in vitro. The anti-tumor activity of CPT-TMC was evaluated in C57BL/6 mice bearing B16-F10 melanoma. Tumor volume, tumor weight and survival time were recorded. Assessment of apoptotic cells within tumor tissue was performed by TUNEL assay. Antiangiogenesis and antiproliferation effects of CPT-TMC in vivo were conducted via CD31 and PCNA immunohistochemistry, respectively. RESULTS: CPT-TMC efficiently inhibited B16-F10 cells proliferation and increased apoptosis in vitro. Experiment group showed significant inhibition compared with free CPT-treated group (81.3% vs. 56.9%) in the growth of B16-F10 melanoma xenografts and prolonged the survival time of the treated mice (P < 0.05). Decreased cell proliferation, increased tumor apoptosis as well as a reduction in angiogenesis were observed. CONCLUSIONS: Our data suggest that N-trimethyl chitosan-encapsulated camptothecin is superior to free CPT by overcoming its insolubility and finally raises the potential of its application in melanoma therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Camptothecin/pharmacology , Cell Proliferation/drug effects , Chitosan/pharmacology , Melanoma, Experimental/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/pharmacokinetics , Cell Line, Tumor , Chitosan/pharmacokinetics , Drug Carriers , Female , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Nude , Solubility , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...