Numerous studies have shown that carbon ion radiotherapy (CIRT) induces anti-cancer immune responses in melanoma patients, yet the mechanism remains elusive. The abundance of myeloid-derived suppressor cells (MDSC) in the tumour microenvironment is associated with therapeutic efficacy and disease outcome. This study analysed the changes in the immune contexture in response to the carbon ion treatment. The murine melanoma B16, MelanA, and S91 tumour models were established in syngeneic immunocompetent mice. Then, the tumours were irradiated with carbon ion beams, and flow cytometry was utilised to observe the immune contexture changes in the bone marrow, peripheral blood, spleen, and tumours. The immune infiltrates in the tumour tissues were further assessed using haematoxylin/eosin staining and immunohistochemistry. The immunoblot detected the expression of proteins associated with the JAK/STAT signalling pathway. The secretion of immune-related cytokines was examined using ELISA. Compared to conventional radiotherapy, particle beams have distinct advantages in cancer therapy. Here, the use of carbon ion beams (5 GyE) for melanoma-bearing mice was found to reduce the population of MDSC in the bone marrow, peripheral blood, and spleen of the animals via a JAK2/STAT3-dependent mechanism. The percentage of CD3+, CD4+, CD8+ T cells, macrophages, and natural killer cells increased after radiation, resulting in reduced tumour growth and prolonged overall survival in the three different mouse models of melanoma. This study, therefore, substantiated that CIRT boosts anti-tumour immune responses via the inhibition of MDSC.
Aim: To identify the methylated-differentially expressed genes (MDEGs) that may serve as diagnostic markers and therapeutic targets in Epstein-Barr virus-associated gastric cancer (EBVaGC) and to explore the methylation-based pathways for elucidating biological mechanisms of EBVaGC. Materials & methods: Gene expression and methylation profiles were downloaded from GEO database. MDEGs were identified by GEO2R. Pathway enrichment analyses were conducted based on DAVID database. Hub genes were identified by Cytoscape, which were further verified by The Cancer Genome Atlas database. Results: A total of 367 hypermethylated, lowly expressed genes were enriched in specific patterns of cell differentiation. 31 hypomethylated, highly expressed genes demonstrated enrichment in regulation of immune system process. After validation using The Cancer Genome Atlas database, seven genes were confirmed to be significantly different hub genes in EBVaGC. Conclusion: EBVaGC-specific MDEGs and pathways can be served as potential biomarkers for precise diagnosis and treatment of EBVaGC and provide novel insights into the mechanisms involved.
Ubiquitin-specific peptidase 10 (USP10), a typical de-ubiquitinase, has been found to play a double-edged role in human cancers. Previously, we reported that the expression of USP10 was negatively correlated with the depth of gastric wall invasion, lymph node metastasis, and prognosis in gastric cancer (GC) patients. However, it remains unclear whether USP10 can regulate the metastasis of GC cells through its de-ubiquitination function.
Abstract Adipose-derived stromal cells (ADSCs) have excellent capacities for regeneration and tissue protection, while sevoflurane, as a requisite component of surgical procedures, has shown therapeutic benefit in animal models of sepsis. This study therefore determined if the combination of sevoflurane and ADSCs exerted additional protective effects against acute lung injury (ALI) induced by cecal ligation and puncture (CLP) in rats. The animals were randomized into five groups: (sham operation (group I), CLP followed by mechanical ventilation (group II), CLP plus sevoflurane at 0.5 minimum alveolar concentration (group III), CLP plus intravenous autologous 5 × 10 6 ADSCs (group IV), and CLP plus sevoflurane and ADSCs (group V). Levels of the pro-inflammatory cytokines tumor necrosis factor-α, transforming growth factor-β1, interleukin-1β and interleukin-6 were significantly increased in CLP rats. Moreover, epithelial sodium channel expression levels and activities of Na/K-ATPase and alveolar fluid clearance were significantly reduced in CLP-induced ALI rats. ADSCs improved all these parameters, and these effects were further enhanced by the addition of sevoflurane. In conclusion, combined treatment with ADSCs and sevoflurane is superior to either ADSCs or sevoflurane therapy alone for preventing ALI. This beneficial effect may be partly due to improved alveolar fluid clearance by the paracrine or systemic production of keratinocyte growth factor and via anti-inflammatory properties.
Abstract MicroRNAs (miRNAs) are non-coding RNAs of 19-22 nucleotides which regulate a diverse set of physiological processes including immune cell development by post-transcriptional gene regulation. Circulating miRNAs in blood have been assessed as biomarkers of disease diagnosis and are capable of modulating a wide-range of biological functions through targeting the molecules of recipient cells. In our previous work, some radiosensitive circulating miRNAs have been identified, and most of them were closely associated with immune and hematopoietic system. Here, we further investigated the effects of radioprotector on the changes of miRNA expression levels after exposure of mice to X-rays or carbon ion beam, aiming to explore novel potential therapeutic targets. Kunming mice were whole-body exposed to 2 and 4 Gy of X-rays generated by Faxitron RX650 or 0.1 and 0.5 Gy of carbon ion beam generated by the HIRFL (Institute of Modern Physics, Lanzhou, China). Angelica sinensis polysaccharide (ASP) which is able to promote the hematopoietic function in vivo and GANRA agent which can scavenge the ROS were administrated to mice by gavage for 7 days before exposure. The expression of circulating miRNAs were detected by qRT-PCR analysis. Our results confirmed that ASP significantly protected the immune and hematopoietic systems of irradiated mice by recovering the numbers of bone marrow hematopoietic cells, peripheric leucocytes, lymphocytes and atrophy of the spleen. GANRA increased the number of lymphocytes, leucocytes and the ratio of CD4+/CD8+ cells in the spleen of irradiated mice. Moreover, GANRA could eliminate ROS which was produced by X-rays and carbon ion beam, and increased the cell viability. Meanwhile, the expression levels of circulating miRNAs such as let-7a, miR-34a, miR-223, miR-150 and miR-574 were distinctively different between ASP or GANRA treated and the control group. ASP or GANRA treatment could rescue the up-regulation of let-7a, miR-34a and miR-223, and the down-regulation of miR-150 and miR-574 that were induced by radiation, indicating that circulating miRNAs could be possible targets of radioprotector responding to radiation exposure. In conclusion, ASP and GANRA treatments have significant protective effects on the immune and hematopoietic system. The differential expressions of circulating miRNAs in radioprotector treated and non-treated groups indicate that miRNAs are potential targets for the protection of radiation-induced immunologic injury. By further exploring the function of these miRNA and their influence on the immune and hematopoietic systems, it will accelerate the development of new biomarkers and drug targets. Citation Format: Heng Zhou, Wenjun Wei, Jufang Wang. Circulating miRNAs as potential targets for the protection of radiation-induced immunologic injury [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5389.
Cavity ring-down is an essential test to measure ultra-high quality factor (UHQ) optical cavities, which is, however, frequently misinterpreted due to lacking of a specified analysis guideline. Here we clarify the basic property of cavity ring down and present a step-by-step method that enables extraction of the overall quality factor, as well as the intrinsic loss and coupling state of UHQ cavities with better fidelity and simplicity than prior schemes. Our work can facilitate acurrate design and characterization of UHQ cavities for ultra-low noise lasers, high finesse reference cavities, and ultra-narrow optical filters.
Our aim was to study the clinical features of postoperative meningitis after neurosurgery and identify the factors affecting the duration of treatment.This retrospective study assessed 283 patients with postoperative bacterial meningitis in the Neurosurgery Department of Beijing Tiantan Hospital, Beijing, People's Republic of China, between January and December 2012. Patients' clinical data were reviewed, and multivariate logistic regression analysis was used to identify the factors associated with a prolonged treatment course.The mortality rate was 0.4% in these patients, of whom 12.4% were found with pathogens in the cerebrospinal fluid. Among the three most common pathogens detected were methicillin-resistant Staphylococcus aureus, Acinetobacter baumannii, and Pseudomonas aeruginosa. The mean treatment course was 13.5±2.1 days. Interestingly, the treatment duration for postoperative meningitis was significantly longer in patients with intracranial malignant tumors than in those with benign lesions. Single-factor analysis showed that male sex (P=0.042) and malignant (rather than benign) lesions (P<0.001) were significantly associated with prolonged treatment duration. Multivariate analysis further confirmed that malignant intracranial lesions represented an independent risk factor for prolonged treatment duration (odds ratio: 2.5962; 95% confidence interval: 1.1092-6.6134).The nature of the intracranial lesion is an independent risk factor for the duration of treatment in postoperative meningitis after neurosurgery.
Glutamine metabolism provides energy to tumor cells and also produces reactive oxygen species (ROS). Excessive accumulation of ROS can damage mitochondria and eventually lead to cell death. xCT (SLC7A11) is responsible for the synthesis of glutathione in order to neutralize ROS. In addition, mitophagy can remove damaged mitochondria to keep the cell alive. Ionizing radiation kills tumor cells by causing the accumulation of ROS, which subsequently induces nuclear DNA damage. With this in mind, we explored the mechanism of intracellular ROS accumulation induced by ionizing radiation and hypothesized new methods to enhance the effect of radiotherapy. We used MCF-7 breast cancer cells and HCT116 colorectal cancer cells in our study. The above-mentioned cells were irradiated with different doses of X-rays or carbon ions. Clone formation assays were used to detect cell proliferation, enzyme-linked immunosorbent assay (ELISA) detected ATP, and glutathione (GSH) production, while the expression of proteins was detected by Western blot and quantitative real-time PCR analysis. The production of ROS was detected by flow cytometry, and immunofluorescence was used to track mitophagy-related processes. Finally, BALB/C tumor-bearing nude mice were irradiated with X-rays in order to further explore the protein expression found in tumors with the use of immunohistochemistry. Ionizing radiation increased the protein expressions of ASCT2, GLS, and GLUD in order to upregulate the glutamine metabolic flux in tumor cells. This caused an increase in ATP secretion. Meanwhile, ionizing radiation inhibited the expression of the xCT (SLC7A11) protein and reduced the generation of glutathione, leading to excessive accumulation of intracellular ROS. The mitophagy inhibitor, or knockdown Parkin gene, is able to enhance the ionizing radiation-induced ROS production and increase nucleus DNA damage. This combined treatment can significantly improve the killing effect of radiation on tumor cells. We concluded that ionizing radiation could upregulate the glutamine metabolic flux and enhance ROS accumulation in mitochondria. Ionizing radiation also decreased the SLC7A11 expression, resulting in reduced GSH generation. Therefore, inhibition of mitophagy can increase ionizing radiation-induced cell death.
// Allan Sauvat 1, 2, 3 , Yidan Wang 1, 2, 3, 4 , Florian Segura 1, 2, 3 , Sabrina Spaggiari 1, 2, 3 , Kevin Müller 1, 2, 3 , Heng Zhou 1, 2, 3, 4 , Lorenzo Galluzzi 1, 2, 5, 6 , Oliver Kepp 1, 2, 3 , Guido Kroemer 1, 2, 3, 6, 7 1 Equipe 11 labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France 2 INSERM, U1138, Paris, France 3 Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France 4 Faculté de Medecine, Université Paris-Sud, Le Kremlin-Bicêtre, France 5 Gustave Roussy Cancer Campus, Villejuif, France 6 Faculté de Medecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France 7 Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France Correspondence to: Oliver Kepp, e-mail: oliver.kepp@gustaveroussy.fr Guido Kroemer, e-mail: kroemer@orange.fr Keywords: apoptosis, necrosis, high-throughput screening, drug discovery Received: January 09, 2015 Accepted: January 31, 2015 Published: March 25, 2015 ABSTRACT Cellular viability is usually determined by measuring the capacity of cells to exclude vital dyes such as 4’,6-diamidino-2-phenylindole (DAPI), or by assessing nuclear morphology with chromatinophilic plasma membrane-permeant dyes, such as Hoechst 33342. However, a fraction of cells that exclude DAPI or exhibit normal nuclear morphology have already lost mitochondrial functions and/or manifest massive activation of apoptotic caspases, and hence are irremediably committed to death. Here, we developed a protocol for the simultaneous detection of plasma membrane integrity (based on DAPI) or nuclear morphology (based on Hoechst 33342), mitochondrial functions (based on the mitochondrial transmembrane potential probe DiOC 6 (3)) and caspase activation (based on YO-PRO ® -3, which can enter cells exclusively upon the caspase-mediated activation of pannexin 1 channels). This method, which allows for the precise quantification of dead, dying and healthy cells, can be implemented on epifluorescence microscopy or flow cytometry platforms and is compatible with a robotized, high-throughput workflow.
Current in vitro models for gastric cancer research, such as 2D cell cultures and organoid systems, often fail to replicate the complex extracellular matrix (ECM) found in vivo. For the first time, this study utilizes a gelatin methacryloyl (GelMA) hydrogel, a biomimetic ECM-like material, in 3D bioprinting to construct a physiologically relevant gastric cancer model. GelMA's tunable mechanical properties allow for the precise manipulation of cellular behavior within physiological ranges. Genetic and phenotypic analyses indicate that the 3D bioprinted GelMA (3Db) model accurately mimics the clinical tumor characteristics and reproduces key cancer hallmarks, such as cell proliferation, invasion, migration, angiogenesis, and the Warburg effect. Comparisons of gene expression and drug responses between the 3Db model and patient-derived xenograft models, both constructed from primary gastric cancer cells, validate the model's clinical relevance. The ability of the 3Db model to closely simulate in vivo conditions highlights its crucial role in identifying treatment targets and predicting patient-specific responses, showcasing its potential in high-throughput drug screening and clinical applications. This study is the first to report the pivotal role of GelMA-based 3D bioprinting in advancing gastric cancer research and regenerative medicine.