Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 705361, 2021.
Article in English | MEDLINE | ID: mdl-34489957

ABSTRACT

Immunogenic cell death (ICD) is a form of regulated cell death (RCD) induced by various stresses and produces antitumor immunity via damage-associated molecular patterns (DAMPs) release or exposure, mainly including high mobility group box 1 (HMGB1), calreticulin (CRT), adenosine triphosphate (ATP), and heat shock proteins (HSPs). Emerging evidence has suggested that ionizing radiation (IR) can induce ICD, and the dose, type, and fractionation of irradiation influence the induction of ICD. At present, IR-induced ICD is mainly verified in vitro in mice and there is few clinical evidence about it. To boost the induction of ICD by IR, some strategies have shown synergy with IR to enhance antitumor immune response, such as hyperthermia, nanoparticles, and chemotherapy. In this review, we focus on the molecular mechanisms of ICD, ICD-promoting factors associated with irradiation, the clinical evidence of ICD, and immunogenic forms of cell death. Finally, we summarize various methods of improving ICD induced by IR.


Subject(s)
Immunogenic Cell Death/radiation effects , Alarmins/physiology , Animals , Antigens, Neoplasm/immunology , Biomarkers , Combined Modality Therapy , Cytokines/physiology , Dose-Response Relationship, Radiation , Ferroptosis/radiation effects , HMGB1 Protein/physiology , Humans , Hyperthermia, Induced , Mice , Morpholines/therapeutic use , Necroptosis/radiation effects , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/radiotherapy , Piperazines/therapeutic use , Pyrroles/therapeutic use , Radiation Tolerance , Radiation, Ionizing
2.
Biomed Res Int ; 2021: 8880179, 2021.
Article in English | MEDLINE | ID: mdl-33532500

ABSTRACT

Regulated necrosis (necroptosis) is crucially involved in cardiac ischaemia-reperfusion injury (MIRI). The aim of our study is to investigate whether shock wave therapy (SWT) is capable of exerting protective effects by inhibiting necroptosis during myocardial ischaemia-reperfusion (I/R) injury and the possible role of autophagy in this process. We established a hypoxia/reoxygenation (H/R) model in vitro using HL-1 cells to simulate MIRI. MTS assays and LDH cytotoxicity assay were performed to measure cell viability and cell damage. Annexin V/PI staining was used to determine apoptosis and necrosis. Western blotting was performed to assess the changes in cell signaling pathways associated with autophagy, necroptosis, and apoptosis. Reactive oxygen species (ROS) production was detected using DHE staining. Autophagosome generation and degradation (autophagic flux) were analysed using GFP and RFP tandemly tagged LC3 (tfLC3). HL-1 cells were then transfected with p62/SQSTM1 siRNA in order to analyse its role in cardioprotection. Our results revealed that SWT increased cell viability in the H/R model and decreased receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 expression. ROS production was also inhibited by SWT. Moreover, SWT decreased Beclin1 expression and the ratio of LC3-II/LC3-I following H/R. Simultaneously, in the tfLC3 assay, the SWT provoked a decrease in the cumulative autophagosome abundance. siRNA-mediated knockdown of p62 attenuated H/R-induced necroptosis, and SWT did not exert additive effects. Taken together, SWT ameliorated H/R injury by inhibiting necroptosis. SWT also relieved the blockade of autophagic flux in response to H/R injury. The restoration of autophagic flux by SWT might contribute to its cardioprotective effect on necroptosis following H/R injury.


Subject(s)
Autophagy/radiation effects , Cell Hypoxia/radiation effects , Extracorporeal Shockwave Therapy , Myocytes, Cardiac , Necroptosis/radiation effects , Animals , Cell Line , Cell Survival/radiation effects , Heart/radiation effects , Mice , Models, Biological , Myocardial Reperfusion Injury , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/radiation effects
3.
Cell Death Dis ; 11(12): 1070, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318476

ABSTRACT

Lack of effective treatments for aggressive breast cancer is still a major global health problem. We have previously reported that photodynamic therapy using methylene blue as photosensitizer (MB-PDT) massively kills metastatic human breast cancer, marginally affecting healthy cells. In this study, we aimed to unveil the molecular mechanisms behind MB-PDT effectiveness and specificity towards tumor cells. Through lipidomics and biochemical approaches, we demonstrated that MB-PDT efficiency and specificity rely on polyunsaturated fatty acid-enriched membranes and on the better capacity to deal with photo-oxidative damage displayed by non-tumorigenic cells. We found out that, in tumorigenic cells, lysosome membrane permeabilization is accompanied by ferroptosis and/or necroptosis. Our results also pointed at a cross-talk between lysosome-dependent cell death (LDCD) and necroptosis induction after photo-oxidation, and contributed to broaden the understanding of MB-PDT-induced mechanisms and specificity in breast cancer cells. Therefore, we demonstrated that efficient approaches could be designed on the basis of lipid composition and metabolic features for hard-to-treat cancers. The results further reinforce MB-PDT as a therapeutic strategy for highly aggressive human breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Light , Antioxidants/pharmacology , Breast Neoplasms/drug therapy , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Female , Ferroptosis/drug effects , Ferroptosis/radiation effects , Humans , Lipids/chemistry , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/radiation effects , Methylene Blue/pharmacology , Methylene Blue/therapeutic use , Models, Biological , Necroptosis/drug effects , Necroptosis/radiation effects , Oxidation-Reduction , Photochemotherapy , Triple Negative Breast Neoplasms/pathology
4.
Sci Rep ; 10(1): 18946, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33144600

ABSTRACT

Ultraviolet radiation (UVR) is a major environmental genotoxic agent. In skin, it can lead to the formation of mutagenic DNA damage. Several mechanisms are in place to prevent the conversion of these DNA damage into skin cancer-driver mutations. An important mutation prevention mechanism is the programmed cell death, which can safely dispose of the damaged cells. Apoptosis is the most studied and best characterised programmed cell death, but an increasing amount of new cell death pathways are emerging. Using different pharmacological cell death inhibitors and antioxidants, we have evaluated the implication of apoptosis, necroptosis, ferroptosis and parthanatos in UVB-induced cell death in human diploid dermal fibroblasts. Our results show that apoptosis is the only known cell death mechanism induced by UVB irradiation in fibroblasts. We also showed that lethal UVB irradiation induces a PARP-dependent drastic loss of cellular metabolic activity caused by an overused of NAD+.


Subject(s)
Apoptosis/radiation effects , Cell Death/radiation effects , Fibroblasts/cytology , Fibroblasts/drug effects , Ultraviolet Rays , DNA Damage/radiation effects , Ferroptosis/radiation effects , Humans , Necroptosis/radiation effects , Parthanatos/radiation effects , Reactive Oxygen Species/metabolism , Signal Transduction/radiation effects
5.
Radiat Res ; 193(5): 435-450, 2020 05.
Article in English | MEDLINE | ID: mdl-32134361

ABSTRACT

Mitigation of total-body irradiation (TBI) in C57BL/6 mice by two drugs, which target apoptosis and necroptosis respectively, increases survival compared to one drug alone. Here we investigated whether the biomarker (signature)directed addition of a third anti-ferroptosis drug further mitigated TBI effects. C57BL/6NTac female mice (30-33 g) received 9.25 Gy TBI, and 24 h or later received JP4-039 (20 mg/kg), necrostatin-1 (1.65 mg/kg) and/or lipoxygenase-15 inhibitor (baicalein) (50 mg/kg) in single-, dual- or three-drug regimens. Some animals were sacrificed at days 0, 1, 2, 3, 4 or 7 postirradiation, while the majority in each group were maintained beyond 30 days. For those mice sacrificed at the early time points, femur bone marrow, intestine (ileum), lung and blood plasma were collected and analyzed for radiation-induced and mitigator-modified levels of 33 pro-inflammatory and stress response proteins. Each single mitigator administered [JP4-039 (24 h), necrostatin-1 (48 h) or baicalein (24 h)] improved survival at day 30 after TBI to 25% (P = 0.0432, 0.2816 or 0.1120, respectively) compared to 5% survival of 9.25 Gy TBI controls. Mice were administered the drug individually based on weight (mg/kg). Drug vehicles comprised 30% cyclodextrin for JP4-039 and baicalein, and 10% Cremphor-EL/10% ethanol/80% water for necrostatin-1; thus, dual-vehicle controls were also tested. The dual-drug combinations further enhanced survival: necrostatin-1 (delayed to 72 h) with baicalein 40% (P = 0.0359); JP4-039 with necrostatin-1 50% (P = 0.0062); and JP4-039 with baicalein 60% (P = 0.0064). The three-drug regimen, timed to signature directed evidence of onset after TBI of each death pathway in marrow and intestine, further increased the 30-day survival to 75% (P = 0.0002), and there was optimal normalization to preirradiation levels of inflammatory cytokine and stress response protein levels in plasma, intestine and marrow. In contrast, lung protein levels were minimally altered by 9.25 Gy TBI or mitigators over 7 days. Significantly, elevated intestinal proteins at day 7 after TBI were reduced by necrostatin-1-containing regimens; however, normalization of plasma protein levels at day 7 required the addition of JP4-039 and baicalein. These findings indicate that mitigator targeting to three distinct cell death pathways increases survival after TBI.


Subject(s)
Apoptosis/drug effects , Ferroptosis/drug effects , Necroptosis/drug effects , Radiation-Protective Agents/pharmacology , Whole-Body Irradiation/adverse effects , Animals , Apoptosis/radiation effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/radiation effects , Cytokines/metabolism , Drug Interactions , Female , Ferroptosis/radiation effects , Ileum/drug effects , Ileum/metabolism , Ileum/radiation effects , Mice , Mice, Inbred C57BL , Necroptosis/radiation effects , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control , Small Molecule Libraries/pharmacology , Time Factors
6.
J Exp Clin Cancer Res ; 38(1): 461, 2019 Nov 09.
Article in English | MEDLINE | ID: mdl-31706322

ABSTRACT

BACKGROUND: Tumor cell repopulation after radiotherapy is a major cause for the tumor radioresistance and recurrence. This study aims to investigate the underlying mechanism of tumor repopulation after radiotherapy, with focus on whether and how necroptosis takes part in this process. METHODS: Necroptosis after irradiation were examined in vitro and in vivo. And the growth-promoting effect of necroptotic cells was investigated by chemical inhibitors or shRNA against necroptosis associated proteins and genes in in vitro and in vivo tumor repopulation models. Downstream relevance factors of necroptosis were identified by western blot and chemiluminescent immunoassays. Finally, the immunohistochemistry staining of identified necroptosis association growth stimulation factor was conducted in human colorectal tumor specimens to verify the relationship with clinical outcome. RESULTS: Radiation-induced necroptosis depended on activation of RIP1/RIP3/MLKL pathway, and the evidence in vitro and in vivo demonstrated that the inhibition of necroptosis attenuated growth-stimulating effects of irradiated tumor cells on living tumor reporter cells. The JNK/IL-8 were identified as downstream molecules of pMLKL during necroptosis, and inhibition of JNK, IL-8 or IL-8 receptor significantly reduced tumor repopulation after radiotherapy. Moreover, the high expression of IL-8 was associated with poor clinical prognosis in colorectal cancer patients. CONCLUSIONS: Necroptosis associated tumor repopulation after radiotherapy depended on activation of RIP1/RIP3/MLKL/JNK/IL-8 pathway. This novel pathway provided new insight into understanding the mechanism of tumor radioresistance and repopulation, and MLKL/JNK/IL-8 could be developed as promising targets for blocking tumor repopulation to enhance the efficacy of colorectal cancer radiotherapy.


Subject(s)
Interleukin-8/metabolism , Necroptosis , Neoplasms/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Humans , Immunohistochemistry , Luminescent Measurements , Mice , Molecular Imaging , Necroptosis/radiation effects , Neoplasms/genetics , Neoplasms/radiotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...