Through the mechanism of action, we observed that DSF activated the STING signaling pathway by inhibiting the Poly(ADP-ribose) polymerases (PARP1). Our findings, when considered collectively, underscore the potential for this novel combination strategy, incorporating DSF and chemoimmunotherapy, to be clinically applied in the treatment of patients with pancreatic ductal adenocarcinoma (PDAC).
The capacity of chemotherapy to eradicate laryngeal squamous cell carcinoma (LSCC) is frequently curtailed by the emergence of resistance in affected patients. The superfamily member, Lymphocyte antigen 6, subfamily D (Ly6D), shows a high presence in numerous cancers; however, its contribution to the chemoresistance of LSCC cells, along with the intricate molecular mechanisms involved, is not yet fully understood. We report in this study that elevated levels of Ly6D contribute to chemoresistance in LSCC cells, a resistance that is reversed by silencing Ly6D. Subsequent bioinformatics analysis, PCR array screening, and functional characterization revealed that Wnt/-catenin pathway activation contributes to Ly6D-induced chemoresistance. Ly6D overexpression-mediated chemoresistance is hampered by the genetic and pharmacological inhibition of β-catenin. The overexpression of Ly6D has a mechanistic effect on reducing miR-509-5p expression, thereby liberating CTNNB1, its target gene, to activate the Wnt/-catenin pathway and ultimately bolster chemoresistance. By introducing miR-509-5p, the chemoresistance in LSCC cells, augmented by Ly6D and -catenin, was reversed. Ultimately, the ectopic expression of miR-509-5p exhibited a substantial decrease in the expression of the two further targets, MDM2 and FOXM1. The integrated analysis of these data underscores the key function of Ly6D/miR-509-5p/-catenin in chemotherapy resistance and unveils a prospective strategy for the clinical management of refractory LSCC.
Renal cancer therapy often incorporates vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) as essential anti-angiogenic drugs. While Von Hippel-Lindau dysfunction is fundamental to the effectiveness of VEGFR-TKIs, the influence of singular and joint mutations in the genes encoding the chromatin remodeling proteins Polybromo-1 (PBRM1) and Lysine Demethylase 5C (KDM5C) is not well characterized. A study investigated the tumor mutation and expression profiles of 155 unselected clear cell renal cell carcinoma (ccRCC) patients undergoing first-line VEGFR-TKI treatment. The IMmotion151 trial's clear cell renal cell carcinoma cases provided further support for the observations. In a proportion of cases (4-9%), a co-occurrence of PBRM1 and KDM5C (PBRM1&KDM5C) mutations was observed, particularly enriched within the favorable-risk group at Memorial Sloan Kettering Cancer Center. structured medication review Our cohort study found that tumors which mutated only in PBRM1, or in both PBRM1 and KDM5C, showed an increase in angiogenesis (P=0.00068 and 0.0039, respectively), and a similar trend appeared in tumors with only KDM5C mutations. Significant VEGFR-TKIs responses were observed in patients with PBRM1 and KDM5C mutations, followed by patients with isolated PBRM1 or KDM5C mutations. A statistically significant correlation between the presence of these mutations and progression-free survival (PFS) was found (P=0.0050, 0.0040, and 0.0027, respectively). Specifically, a trend of extended PFS was observed in the PBRM1-only mutated group (HR=0.64; P=0.0059). IMmotion151 trial validation showcased a similar relationship between angiogenesis increase and patient progression-free survival (PFS). The VEGFR-TKI arm saw the longest PFS in patients with both PBRM1 and KDM5C mutations, an intermediate PFS in those with only one of these mutations, and the shortest PFS in patients without these mutations (P=0.0009 and 0.0025, for PBRM1/KDM5C and PBRM1 versus non-mutated, respectively). In conclusion, somatic mutations in PBRM1 and KDM5C genes are commonly found in patients with metastatic clear cell renal cell carcinoma (ccRCC), and these mutations may contribute to increased tumor angiogenesis and potentially improve the efficacy of anti-angiogenic treatment strategies based on VEGFR-TKIs.
Research on Transmembrane Proteins (TMEMs) has intensified recently, driven by their participation in the development of numerous cancers. Our earlier findings in clear cell renal cell carcinoma (ccRCC) indicated a notable decrease in the mRNA levels of several TMEM proteins, including TMEM213, 207, 116, 72, and 30B. The down-regulation of TMEM genes displayed a greater intensity in advanced ccRCC tumors, possibly linked to clinical parameters like metastasis (TMEM72 and 116), Fuhrman grade (TMEM30B), and overall patient survival (TMEM30B). Investigating these findings further, we initially verified, through experimental means, the membrane association of the selected TMEMs, as predicted computationally. We then validated the presence of signaling peptides on their N-termini, characterized the orientation of the TMEMs within the membrane, and validated their predicted subcellular locations. To evaluate the potential role of selected TMEMs in cellular activities, experiments focusing on overexpression were conducted in HEK293 and HK-2 cell lines. Subsequently, we studied TMEM isoform expression in ccRCC tumors, discovered mutations in TMEM genes, and analyzed chromosomal alterations in their associated locations. Confirmation of membrane-bound status was achieved for all selected TMEMs; TMEM213 and 207 were localized to early endosomes, TMEM72 to both early endosomes and plasma membrane, and TMEM116 and 30B to the endoplasmic reticulum. Cytoplasmic localization was established for the N-terminus of TMEM213; in addition, the C-termini of TMEM207, TMEM116, and TMEM72 were found to face the cytoplasm; finally, both termini of TMEM30B were observed to be directed toward the cytoplasm. While TMEM mutations and chromosomal abnormalities were scarce in ccRCC cases, our analysis revealed potentially harmful mutations in TMEM213 and TMEM30B, and deletions within the TMEM30B gene locus were present in almost 30% of the examined tumors. Investigations of TMEM overexpression hint that specific TMEMs might participate in the processes of carcinogenesis, including cell adhesion, the regulation of epithelial cell proliferation, and the modulation of the adaptive immune response. This could potentially connect these TMEMs to the development and progression of ccRCC.
Within the mammalian brain, the glutamate ionotropic receptor kainate type subunit 3 (GRIK3) is the most prevalent excitatory neurotransmitter receptor. Despite the acknowledged involvement of GRIK3 in normal neurophysiological activities, its precise biological role in tumor progression remains elusive, a consequence of inadequate investigation. Compared to the expression levels found in surrounding paracarcinoma tissue, we first observed a reduction in GRIK3 expression within non-small cell lung cancer (NSCLC) tissues. Our research indicated that GRIK3 expression levels were substantially correlated with the outcome of NSCLC patients. GRIK3 was observed to repress the proliferation and migratory capacity of NSCLC cells, thus hindering xenograft growth and metastasis. Image- guided biopsy A mechanistic link was observed between GRIK3 deficiency and a rise in the expression of ubiquitin-conjugating enzyme E2 C (UBE2C) and cyclin-dependent kinase 1 (CDK1), prompting Wnt signaling pathway activation and augmenting NSCLC progression. Our research suggests a function for GRIK3 in the process of NSCLC advancement, and its expression level might be an independent prognostic factor for NSCLC patients.
Human peroxisome function in fatty acid oxidation is contingent upon the D-bifunctional protein (DBP) enzyme. Despite its potential influence, the contribution of DBP to oncogenesis is currently unclear. Our preceding research has indicated that the elevated expression of DBP drives the proliferation of hepatocellular carcinoma (HCC) cells. The expression of DBP in 75 primary hepatocellular carcinoma (HCC) samples was measured using RT-qPCR, immunohistochemistry, and Western blot, further analyzing its correlation with HCC survival. Besides this, we delved into the processes by which DBP fosters the proliferation of HCC cells. Upregulated DBP expression was detected in HCC tumor tissues, showing a positive correlation with tumor size and TNM stage. Multinomial ordinal logistic regression analysis showed that low DBP mRNA levels were linked to an independent reduced risk of hepatocellular carcinoma (HCC). In particular, the peroxisome, cytosol, and mitochondria of tumor cells exhibited elevated DBP expression. Live xenograft tumor growth was enhanced by the overexpression of DBP, which was positioned outside the peroxisome. DBP upregulation in the cytosol, mechanistically, spurred the activation of the PI3K/AKT signaling axis, consequently driving HCC cell proliferation by curtailing apoptosis through the AKT/FOXO3a/Bim pathway. Tyrphostin B42 cell line DBP overexpression, in addition to its various other effects, facilitated greater glucose uptake and glycogen accumulation through the AKT/GSK3 axis. It simultaneously elevated the activity of mitochondrial respiratory chain complex III, ultimately boosting ATP levels by virtue of AKT-dependent p-GSK3 translocation into the mitochondria. The first report of DBP expression in both peroxisomes and the cytosol, as revealed by this study, establishes the cytosolic DBP as a key element in the metabolic reshaping and adjustment of HCC cells. This discovery provides valuable guidance for the development of HCC treatment.
Tumor cell proliferation and subsequent progression are contingent upon both the cells themselves and the surrounding microenvironment. Identifying therapies that curb cancer cell growth while bolstering the immune system is crucial. Cancer therapy's efficacy is intertwined with arginine's dual modulation. Arginase inhibition, which increased arginine levels in the tumor, thereby activated T-cells, leading to an anti-tumor outcome. Unlike the expected outcome, arginine levels decreased by employing arginine deiminase tagged with polyethylene glycol (20,000 MW) , subsequently leading to an anti-tumor response in ASS1 deficient tumor cells.