Sort by
Matrine Enhances the Antitumor Efficacy of Chidamide in CTCL by Promoting Apoptosis.

Cutaneous T-cell Lymphoma (CTCL) is a rare group of non-Hodgkin lymphoma originating from the skin, which is characterized by T-cell lymphoproliferative disorders. Chidamide, a Chinese original antineoplastic agent with independent intellectual property rights, and matrine, an extract of Chinese herbal medicine, both have been reported to exert effects on the treatment of tumors individually. However, chidamide combined with matrine has not been tested for the treatment of CTCL. Both HH and Hut78 CTCL cell lines were treated with chidamide (0.4 μmol/L), matrine (0.6 g/L), or chidamide combined with matrine for 24, 48, and 72 h. Cell viability was estimated by MTS assay at each time point. Flow cytometry was then conducted to detect cell apoptosis. The exact mechanism of chidamide combined with matrine on CTCL cells was detected by Western blotting and further validated in xenograft models of NOD/SCID mice. Compared to the single drug, chidamide combined with matrine showed a more significant effect on proliferation inhibition and apoptosis induction on CTCL cells both in vitro and in vivo. The results from the in vitro and in vivo studies suggested that matrine could enhance the anti-tumor effect of chidamide by increasing the protein expression of cleaved caspase- 3 and decreasing the expression of E-cadherin, NF-κB, p-Bad, and Bcl-2 to activate apoptosis. Our data have demonstrated chidamide combined with matrine to exhibit elevated antitumor activity in both CTCL cells and xenograft models of NOD/SCID mice, which may be a potential treatment option for CTCL.

Just Published
Relevant
Bioinformatic Analysis and Experimental Validation of HMGA2-AS1 as a Prognostic Biomarker Associated with Immune Infiltration in Gastric Cancer.

Natural antisense long noncoding RNAs (lncRNAs) have the ability to modulate the expression of their corresponding sense genes. Consequently, any dysregulation of these lncRNAs can contribute to the development of pathological processes. The ambiguity surrounding the role of HMGA2-AS1 in gastric cancer (GC) requires further investigation. The aim of this study was to examine the involvement of HMGA2-AS1 in GC. The Kaplan-Meier method, Cox regression analysis, gene set enrichment analysis (GSEA), and immune infiltration analysis were used in this study. These methods were used to evaluate the relationship between clinical characteristics and HMGA2-AS1 expression, prognostic factors, and the significant functional impact of HMGA2-AS1. HMGA2-AS1 levels in GC cell lines were validated using quantitative real-time polymerase chain reaction (qRT-PCR). In patients diagnosed with GC, a significant correlation was observed between high expression of HMGA2-AS1 and the T stage (p = 0.01). Furthermore, the high expression of HMGA2- AS1 was identified as a prognostic indicator for poorer OS (p = 0.004), PFS (p = 0.006), and DSS (p = 0.011). Furthermore, the expression of HMGA2-AS1 (p < 0.001) demonstrated an independent association with OS in patients with GC. The presence of a low expression phenotype of HMGA2-AS1 was associated with differential enrichment of various pathways, including the focal adhesion-PI3K-Akt-mTOR signaling pathway, focal adhesion, ECM glycoproteins, MET promoting cell motility, among others. Furthermore, the expression of HMGA2-AS1 exhibited correlations with B cells, CD56 bright cells, and TFH and Th17 cells. Furthermore, GC cell lines demonstrated significantly higher expression of HMGA2-AS1. Elevated expression of HMGA2-AS1 in GC patients exhibited a significant correlation with unfavorable survival outcomes and increased immune infiltration. This suggests that HMGA2- AS1 holds promise as a potential prognostic biomarker and target for immunotherapy in GC.

Just Published
Relevant
UBE2L3 Promotes Oxidative Stress-regulated Necroptosis to Accelerate Osteosarcoma Progression.

Osteosarcoma is a highly invasive bone marrow stromal tumor with limited treatment options. Oxidative stress plays a crucial role in the development and progression of tumors, but the underlying regulatory mechanisms are not fully understood. Recent studies have revealed the significant involvement of UBE2L3 in oxidative stress, but its specific role in osteosarcoma remains poorly investigated. This study aimed to explore the molecular mechanisms by which UBE2L3 promotes oxidative stress-regulated necroptosis to accelerate the progression of osteosarcoma using in vitro cell experiments. Human osteoblast hFOB1.19 cells and various human osteosarcoma cell lines (MG-63, U2OS, SJSA-1, HOS, and 143B) were cultured in vitro. Plasmids silencing UBE2L3 and negative control plasmids were transfected into U2OS and HOS cells. The cells were divided into the following groups: U2OS cell group, HOS cell group, si-NC-U2OS cell group, si-UBE2L3-U2OS cell group, si-NC-HOS cell group, and si-UBE2L3-HOS cell group. Cell viability and proliferation capacity were measured using the Tunnel method and clonogenic assay. Cell migration and invasion abilities were assessed by Transwell and scratch assays. Cell apoptosis was analyzed by flow cytometry, and ROS levels were detected using immunofluorescence. The oxidative stress levels in various cell groups and the expression changes of necroptosis-related proteins were assessed by PCR and WB. Through these experiments, we aim to evaluate the impact of oxidative stress on necroptosis and uncover the specific mechanisms by which targeted regulation of oxidative stress promotes tumor cell necroptosis as a potential therapeutic strategy for osteosarcoma. The mRNA expression levels of UBE2L3 in human osteosarcoma cell lines were significantly higher than those in human osteoblast hFOB1.19 cells (p <0.01). UBE2L3 expression was significantly decreased in U2OS and HOS cells transfected with si-UBE2L3, indicating the successful construction of stable cell lines with depleted UBE2L3. Tunnel assay results showed a significant increase in the number of red fluorescent-labeled cells in si-UBE2L3 groups compared to si-NC groups in both cell lines, suggesting a pronounced inhibition of cell viability. Transwell assay demonstrated a significant reduction in invasion and migration capabilities of si-UBE2L3 groups in osteosarcoma cells. The clonogenic assay revealed significant suppression of proliferation and clonogenic ability in both U2OS and HOS cells upon UBE2L3 knockdown. Flow cytometry confirmed that UBE2L3 knockdown significantly enhanced apoptosis in U2OS and HOS cells. Immunofluorescence results showed that UBE2L3 silencing promoted oxidative stress levels in osteosarcoma cells and facilitated tumor cell death. WB analysis indicated a significant increase in phosphorylation levels of necroptosis-related proteins, RIP1, RIP3, and MLKL, in both osteosarcoma cell lines after UBE2L3 knockdown. In addition, the expression of necrosis-associated proteins was inhibited by the addition of the antioxidant N-acetylcysteine (NAC). UBE2L3 is upregulated in osteosarcoma cells, and silencing of UBE2L3 promotes oxidative stress in these cells, leading to enhanced necroptosis and delayed progression of osteosarcoma.

Relevant
Survival Benefits of Ganoderma Lucidum in Early-stage Triple-negative Breast Cancer: A Real World Study.

Ganoderma lucidum extracts are widely used as adjuvants in the treatment of triple-negative breast cancers (TNBC) in China. However, its clinical value in TNBC remains unclear. Therefore, we investigated the clinical effect of Ganoderma lucidum spore powder (GLSP) on prognosis in patients with early-stage TNBC in this study. A total of 388 patients who were diagnosed with TNBC at the Sun Yat-sen University Cancer Center from February 2012 to December 2017 were retrospectively reviewed. The propensity score matching (PSM) method was applied to balance baseline data. Kaplan-Meier method and Cox proportional hazards model were used to evaluate the relationship between GLSP and prognosis. Of the 388 patients, 72 (18.6%) patients took GLSP. After PSM, 208 patients were selected for analysis, including 71 (34.1%) patients who took the powder. The median followup period was 51 months. The patients who took GLSP (the treatment group) and those who did not take GLSP (the control group) were similar in most clinico-pathological features before being matched. However, the proportion of patients who received breast-conserving surgery in the treatment group was higher (27.8% vs. 16.1%; p =0.021) than in the control group. No significant difference was found in the baseline data between the two groups for the matched cohort (all p >0.05). Univariate analysis and multivariate analysis showed that patients taking GLSP benefited from improved overall survival (OS) (HR=0.159, p = 0.002) and disease-free survival (DFS) (HR=0.232, p = 0.005) before being matched. The main result of the survival analysis after matching was similar to that described above. Patients in the treatment group achieved both greater OS and DFS benefits than patients in the control group (all p < 0.05). In stratified analysis according to TNM stages, after adjusting for the significant prognostic factors, multivariate analysis revealed that the treatment group had better OS than the control group for patients in stages II and III (HR=0.172, p =0.004). The results of this real-world propensity-score-matched study suggest that GLSP can improve OS and DFS in early-stage TNBC patients. A higher OS was observed for patients taking GLSP, particularly in stage II and stage III.

Relevant
Metformin Inhibits NLRP3 Inflammasome Expression and Regulates Inflammatory Microenvironment to Delay the Progression of Colorectal Cancer.

Colorectal cancer is a common malignant tumor, with about one million people diagnosed with it worldwide each year. Recent studies have found that metformin can inhibit the production of inflammatory factors and regulate the polarization of immune cells. However, whether metformin can regulate the inflammatory microenvironment and delay the progression of colorectal cancer by inhibiting the inflammatory response has not been deeply studied yet. This study aimed to explore the molecular mechanism by which metformin inhibits the expression of NLRP3 inflammasome, regulates the inflammatory microenvironment, and delays the progression of colorectal cancer through in vitro cell experiments. In this research, NLRP3 was knocked down in human colorectal cancer cells, and metformin was added to them. Cell proliferation ability was detected by CCK8, and cell migration and invasion abilities were assessed by Transwell assay. The apoptosis rate was determined by flow cytometry. In addition, the expression of NLRP3 inflammatory vesicles and inflammatory factors in each group of cells was studied by qRT-PCR and Western blotting. Finally, clinical colorectal cancer samples were analyzed by immunohistochemistry. The results of the study showed that NLRP3 expression was significantly increased in colorectal cancer cell lines and human colorectal cancer tissues. Knockdown of NLRP3 significantly inhibited tumor cell proliferation, migration, and invasion. In addition, the proliferation, migration and invasion of tumor cells were also significantly reduced by the addition of metformin intervention. Furthermore, qRT-PCR and WB results demonstrated that the expression of IL-1β, IL-6, TNF- α, TGF-β, and IL-10 was down-regulated in LS1034 tumor cells after NLRP3 knockdown. In addition, metformin intervention also resulted in different degrees of downregulation of NLRP3 and inflammatory factor expression (p <0.05). Notably, the reduction in inflammatory factors was more pronounced after the combination of NLRP3 knockdown and metformin intervention. Metformin can inhibit the expression of NLRP3 inflammasome, thereby suppressing the expression of inflammation-related factors, reducing the damage of the inflammatory microenvironment to normal cells, and delaying the progression of colorectal cancer.

Relevant
Puerarin: An Anticancer and Anti-Inflammatory Agent

Objective: Puerarin is a well-known anti-inflammatory agent which has demonstrated its potential to cure numerous ailments. Though many plants are reported in the literature, still their mechanisms are unversed. In this review, numerous mechanisms of puerarin to cure cancer and other inflammatory disorders, pharmacokinetics and adverse events, and boulevards of further research are discussed. Methods: Organized research was done using ScienceDirect, PubMed, ResearchGate, Google Scholar, Google Patents and ScienceDirect Medline to quest all the available data to date on puerarin. Different keywords used were “puerarin”, “Pueraria tuberosa”, “cancer”, “antiinflammatory”, “cardiovascular”, “IBD”, “pharmacokinetics” etc. Results: One hundred thirty-six articles and thirteen patents were studied. Puerarin is reported to treat chronic problems like inflammation, sexual dysfunction, cardiovascular diseases and menaces such as colon, stomach, lung and breast cancer by numerous mechanisms, as these ailments do not progress via a single independent pathway. Conclusion: This article will definitely help budding researchers scrutinize the wealth of information on the therapeutic chattels of puerarin and identify the gaps that have forbidden its application as a potential molecule to cure various ailments.

Relevant
Advances in the Treatment of Chronic Myeloid Leukemia

Background: The treatment of chronic myeloid leukemia has progressed in recent decades, becoming a model for a disease whose pathogenesis is primarily based on a genetic mutation and has led to survivals comparable to those of the general population. Objective: This review aims to present recent therapeutic advances in this area. Methods: A mini-review was achieved using the articles published in Web of Science and Pub- Med between January 2021 - May 2022, and new patents were made in this field. Results: The three generations of tyrosine kinase inhibitors have transformed chronic myeloid leukemia into a manageable disorder and greatly improved the treatment results of the chronic phase, the prognosis, survival, and quality of life of patients. The therapeutic goals today include achieving a deep and lasting molecular response as soon as possible, successful treatment-free remission, and discovering and applying new therapeutic strategies to act on impaired immune modulation and dormant leukemic stem cells. The allosteric inhibitor asciminib targets the ABL myristoyl pocket, reduces Abl kinase activity, and is effective against most cells that have mutations in the ABL1 kinase domain. Progress and recommendations for achieving long-term treatment- free remission are set out. Nearly 50% of the patients who received first-line tyrosine kinase inhibitors required a change of treatment by 10 years due to intolerance or resistance to treatment. Their main side effects are presented. Conclusion: Obtaining a deep and persistent molecular response contributes to achieving longterm treatment-free remission.

Relevant
The Causal Effects of Cholelithiasis on Acute Pancreatitis and Pancreatic Cancer: A Large Sample Size Mendelian Randomization Analysis.

The aim of two-sample Mendelian randomization (MR) with a large sample size was to explore the causal cholelithiasis impact on acute pancreatitis and pancreatic cancer. We performed the two-sample MR analysis with two models. Publicly available summary- level information for cholelithiasis was acquired from the Genome-Wide Summary Association Studies (GWAS) of FinnGen Biobank. The inverse variance weighted (IVW) method was the main method to obtain the MR estimates. Other methods were also used as supplementary methods, including MR-Egger, maximum likelihood, MR-Robust Adjusted Profile Score (MR-RAPS), weighted median, penalised weighted median method, and Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) method. After the selection of genetic instrumental variables (IVs), 11 single nucleotide polymorphisms (SNPs) (Model 1) and 22 SNPs (Model 2) were used to explore the effect of cholelithiasis on acute pancreatitis, and 10 SNPs (Model 1) and 24 SNPs (Model 2) on pancreatic cancer. The findings obtained by the fixed-effect IVW method with both Model 1 and Model 2 showed that genetically predicted cholelithiasis was significantly related to the elevated acute pancreatitis risk (Model 1: OR: 1.001, 95% CI: 1.000-1.002, p <0.001; Model 2: OR: 1.001, 95% CI: 1.000-1.002, p <0.001). Moreover, cholelithiasis would also raise the pancreatic cancer risk (Model 1: OR: 1.676, 95% CI: 1.228-2.288, p = 0.001; Model 2: OR: 1.432, 95% CI: 1.116-1.839, p = 0.005). Genetically predicted cholelithiasis was significantly related to the elevated risk of acute pancreatitis and pancreatic cancer. More attention should be paid to patients with cholelithiasis for the primary prevention of pancreatic-related diseases.

Relevant
Targeting Ferroptosis as a New Approach for Radiation Protection and Mitigation

Abstract: Radiation-induced normal cell toxicity (RINCT) is a major factor to consider while treating any ailment with radiotherapy. Clinical irradiation of tumors necessitates an understanding of the potential efficacy of radiation protective agents in reducing radiation damage to healthy tissues and their effects on tumor tissue radiosensitivity. Ferroptosis is a relatively new form of iron-dependent cell death that has been linked to a variety of disease pathologies. The key mediators of ferroptosis have been identified as lipid peroxidation and iron metabolism. Lipid peroxidation is the result of a reaction between reactive oxygen (ROS) and reactive nitrogen species (RNS) with phosphatidylethanolamine-containing polyunsaturated fatty acids (PUFAs). Ferroptosis inhibitors have been demonstrated to have anti-inflammatory effects in animal models of disease. It was recently shown that ionizing radiation (IR) generates severe ferroptosis, a critical component of RT-mediated normal cell toxicity. These findings support the use of ferroptosis inhibitor treatments for the treatment of radiation normal cell toxicity. Targeting lipid metabolic substrates and controlling ferroptosis by radiation could reduce toxicity and improve clinical outcomes. In this study, we address the relationships between radiotherapy and various types of radiation-induced cell death, and we discuss the interactions between ferroptosis and other kinds of controlled cell death generated by radiotherapy, and we investigate combination treatment options targeting ferroptosis in radiotherapy. This review will be a foundation for future research on ferroptosis in radiotherapy. Additionally, the relevant patents on ferroptosis inhibitors with various therapeutic potentials have been discussed.

Relevant