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










Database
Language
Publication year range
1.
Front Oncol ; 11: 665420, 2021.
Article in English | MEDLINE | ID: mdl-33959512

ABSTRACT

Although many cancer patients are administered radiotherapy for their treatment, the interaction between tumor cells and macrophages in the tumor microenvironment attenuates the curative effects of radiotherapy. The enhanced activation of mTOR signaling in the tumors promotes tumor radioresistance. In this study, the effects of rapamycin on the interaction between tumor cells and macrophages were investigated. Rapamycin and 3BDO were used to regulate the mTOR pathway. In vitro, tumor cells cocultured with macrophages in the presence of each drug under normoxic or hypoxic conditions were irradiated with γ-rays. In vivo, mice were irradiated with γ-radiation after injection with DMSO, rapamycin and 3BDO into tumoral regions. Rapamycin reduced the secretion of IL-4 in tumor cells as well as YM1 in macrophages. Mouse recombinant YM1 decreased the enhanced level of ROS and the colocalized proportion of both xCT and EEA1 in irradiated tumor cells. Human recombinant YKL39 also induced results similar to those of YM1. Moreover, the colocalized proportion of both xCT and LC3 in tumor tissues was elevated by the injection of rapamycin into tumoral regions. Overall, the suppression of mTOR signaling in the tumor microenvironment might be useful for the improvement of tumor radioresistance.

2.
Cells ; 9(12)2020 12 17.
Article in English | MEDLINE | ID: mdl-33348858

ABSTRACT

The treatment of tumors requires the induction of cell death. Radiotherapy, chemotherapy, and immunotherapy are administered to kill cancer cells; however, some cancer cells are resistant to these therapies. Therefore, effective treatments require various strategies for the induction of cell death. Regulated cell death (RCD) is systematically controlled by intracellular signaling proteins. Apoptosis and autophagy are types of RCD that are morphologically different from necrosis, while necroptosis, pyroptosis, and ferroptosis are morphologically similar to necrosis. Unlike necrosis, regulated necrotic cell death (RNCD) is caused by disruption of the plasma membrane under the control of specific proteins and induces tissue inflammation. Various types of RNCD, such as necroptosis, pyroptosis, and ferroptosis, have been used as therapeutic strategies against various tumor types. In this review, the mechanisms of necroptosis, pyroptosis, and ferroptosis are described in detail, and a potential effective treatment strategy to increase the anticancer effects on apoptosis- or autophagy-resistant tumor types through the induction of RNCD is suggested.


Subject(s)
Neoplasms/pathology , Regulated Cell Death , Antineoplastic Agents/therapeutic use , Autophagy , Ferroptosis , Humans , Inflammation/metabolism , Inflammation/pathology , Necroptosis , Neoplasms/metabolism , Neoplasms/therapy , Pyroptosis , Regulated Cell Death/drug effects
3.
Mol Cells ; 43(12): 989-1001, 2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33250450

ABSTRACT

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a facultative intracellular pathogen that causes salmonellosis and mortality worldwide. S. Typhimurium infects macrophages and survives within phagosomes by avoiding the phagosome-lysosome fusion system. Phagosomes sequentially acquire different Rab GTPases during maturation and eventually fuse with acidic lysosomes. Lysophosphatidylcholine (LPC) is a bioactive lipid that is associated with the generation of chemoattractants and reactive oxygen species (ROS). In our previous study, LPC controlled the intracellular growth of Mycobacterium tuberculosis by promoting phagosome maturation. In this study, to verify whether LPC enhances phagosome maturation and regulates the intracellular growth of S. Typhimurium, macrophages were infected with S. Typhimurium. LPC decreased the intracellular bacterial burden, but it did not induce cytotoxicity in S. Typhimuriuminfected cells. In addition, combined administration of LPC and antibiotic significantly reduced the bacterial burden in the spleen and the liver. The ratios of the colocalization of intracellular S. Typhimurium with phagosome maturation markers, such as early endosome antigen 1 (EEA1) and lysosome-associated membrane protein 1 (LAMP-1), were significantly increased in LPC-treated cells. The expression level of cleaved cathepsin D was rapidly increased in LPCtreated cells during S. Typhimurium infection. Treatment with LPC enhanced ROS production, but it did not affect nitric oxide production in S. Typhimurium-infected cells. LPC also rapidly triggered the phosphorylation of IκBα during S. Typhimurium infection. These results suggest that LPC can improve phagosome maturation via ROS-induced activation of NF-κB pathway and thus may be developed as a therapeutic agent to control S. Typhimurium growth.


Subject(s)
Lysophosphatidylcholines/metabolism , Macrophages/metabolism , Macrophages/microbiology , NF-kappa B/metabolism , Phagosomes/metabolism , Salmonella Infections/metabolism , Signal Transduction , Animals , Male , Mice , Mice, Inbred BALB C , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/physiology
4.
Front Microbiol ; 11: 1684, 2020.
Article in English | MEDLINE | ID: mdl-32765474

ABSTRACT

Autophagy is a lysosomal self-digestion pathway that maintains internal homeostasis inside cells and critical process by which the innate immune system eliminates intracellular bacteria. In this study, we showed that stimulation of toll-like receptor 7 (TLR7) with imiquimod (IMQ) triggered autophagic cell death in macrophages by enhancing the generation of reactive oxygen species (ROS) via the p38- or MEK/ERK1/2-mediated signaling pathway in the early phase. IMQ significantly increased mitochondrial ROS and targeted autophagosomes to the mitochondria. Stimulation of TLR7 with IMQ enhanced the expression of BNIP3, which was localized to mitochondria and interacted with beclin-1, leading to mitophagy. In addition, IMQ substantially induced NO production through the GSK-3ß-mediated signaling pathway, which led to autophagy in the late stage. We further examined whether the induction of autophagy by IMQ effectively eliminated intracellular microbes. Macrophages were infected with a virulent Mycobacterium tuberculosis (Mtb) strain, H37Rv, and then treated with IMQ. IMQ suppressed intracellular Mtb growth by inducing autophagy in a dose-dependent manner and increased NO production. Inhibition of autophagy using 3-methyladenine (3-MA) prevented autophagosome formation and control of intracellular Mtb growth in macrophages. These findings revealed a novel mechanism by which IMQ induces selective autophagy to promote intracellular killing machinery against Mtb infection in macrophages.

5.
Microorganisms ; 8(9)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854338

ABSTRACT

Phagocytosis is an essential mechanism in innate immune defense, and in maintaining homeostasis to eliminate apoptotic cells or microbes, such as Mycobacterium tuberculosis, Salmonella enterica, Streptococcus pyogenes and Legionella pneumophila. After internalizing microbial pathogens via phagocytosis, phagosomes undergo a series of 'maturation' steps, to form an increasingly acidified compartment and subsequently fuse with the lysosome to develop into phagolysosomes and effectively eliminate the invading pathogens. Through this mechanism, phagocytes, including macrophages, neutrophils and dendritic cells, are involved in the processing of microbial pathogens and antigen presentation to T cells to initiate adaptive immune responses. Therefore, phagocytosis plays a role in the bridge between innate and adaptive immunity. However, intracellular bacteria have evolved diverse strategies to survive and replicate within hosts. In this review, we describe the sequential stages in the phagocytosis process. We also discuss the immune evasion strategies used by pathogens to regulate phagosome maturation during intracellular bacterial infection, and indicate that these might be used for the development of potential therapeutic strategies for infectious diseases.

6.
J Oncol ; 2019: 5956867, 2019.
Article in English | MEDLINE | ID: mdl-31929797

ABSTRACT

Radiotherapy is widely used for the treatment of cancer patients, but tumor radioresistance presents serious therapy challenges. Tumor radioresistance is closely related to high levels of mTOR signaling in tumor tissues. Therefore, targeting the mTOR pathway might be a strategy to promote solid tumor sensitivity to ionizing radiation. Radioresistance is associated with enhanced antioxidant mechanisms in cancer cells. Therefore, examination of the relationship between mTOR signaling and antioxidant mechanism-linked radioresistance is required for effective radiotherapy. In particular, the effect of mTOR signaling on antioxidant glutathione induction by the Keap1-NRF2-xCT pathway is described in this review. This review is expected to assist in the identification of therapeutic adjuvants to increase the efficacy of radiotherapy.

7.
PLoS One ; 13(12): e0209653, 2018.
Article in English | MEDLINE | ID: mdl-30586442

ABSTRACT

Macrophages are one of the major cell types that produce IL-1ß. IL-1ß maturation occurs via inflammasome activation, and mature IL-1ß is then released from the cell. Secreted IL-1ß mediates inflammatory reactions in various pathological environments, such as those in infectious, autoimmune, and cancerous diseases. Although the mechanism of IL-1ß production has been discovered in infectious and autoimmune diseases, its production mechanism in the tumor microenvironment is unclear. Therefore, the mechanism of IL-1ß production in macrophages in the tumor microenvironment was investigated in this study. First, bone marrow-derived macrophages obtained from C57BL/6 mice were treated with B16F10 tumor-conditioned media (TCM) in vitro. TCM increased the levels of IL-1ß via glucose-mediated activation of the inflammasome. Moreover, TCM enhanced the activation of both NF-κB and mTOR pathways in a glucose-dependent manner. In particular, the expression levels of mTORC1 component proteins were dependent on the TCM-induced activation of NF-κB signaling. In addition, TCM affected ASC-ASC interactions through increasing intracellular reactive oxygen species levels. Finally, glucose inhibition by inoculation with 2-deoxy-D-glucose in vivo decreased the IL-1ß levels in both the blood and tumor region of B16F10-bearing C57BL/6 mice relative to those in PBS-injected tumor-bearing mice. These results suggest that glucose supplied from blood vessels might be important for IL-1ß production in tumor-associated macrophages via the integrated signals of the NF-κB and mTOR pathways in the tumor microenvironment.


Subject(s)
Inflammation/genetics , Interleukin-1beta/genetics , Melanoma, Experimental/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Blood Vessels/metabolism , Bone Marrow Cells/drug effects , Culture Media, Conditioned/pharmacology , Glucose/antagonists & inhibitors , Glucose/metabolism , Humans , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammation/pathology , Macrophages/drug effects , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , NF-kappa B/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
8.
Oncol Lett ; 13(5): 3579-3585, 2017 May.
Article in English | MEDLINE | ID: mdl-28529582

ABSTRACT

Angiotensin II receptor blockers (ARBs) are anti-hypertensive drugs that competitively inhibit the binding of angiotensin II to its receptor, resulting in blood vessel dilation and the reduction of blood pressure. These antagonists are also known as sartans, and are a group of pharmaceuticals that possess tetrazole or imidazole groups. In the present study, the anticancer and antimetastatic effects of the ARBs fimasartan, losartan, eprosartan and valsartan on the human prostate cancer PC-3, DU-145 and LNCap-LN3 cell lines were investigated in vitro. The proliferation of the prostate cancer cells was inhibited following treatment with 100 µM ARB. In particular, treatment with fimasartan resulted in marked anti-proliferative activity compared with the other ARBs. With respect to the molecular mechanism of the growth inhibition exhibited by the ARBs, 3-methyladenin (3-MA), an autophagy inhibitor, was revealed to increase the survival rate of PC-3 cells when cell death inhibitors were pretreated with fimasartan. In addition, the ARBs induced autophagy with increased expression levels of autophagy protein (Atg) 5-12, Atg 16-like-1, beclin-1 and microtubule-associated protein 1A/1B-light chain 3 (LC3). Notably, the enhanced expression of LC3-II (a 6.7-fold increase at 72 h) was observed in PC3 cells treated with fimasartan. This was supported by the observation of the time-dependent accumulation of LC3-positive foci in PC-3. In addition, a migration assay indicated that the ARBs induced anti-metastatic effects in PC-3 and DU-145 cells. The aforementioned results suggest that ARBs may induce autophagy-associated cell death and anti-metastatic activity in prostate cancer cells. Thus, ARBs may be a potential medication for patients with prostate cancer and hypertension.

SELECTION OF CITATIONS
SEARCH DETAIL
...