Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Carcinog ; 63(8): 1486-1499, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780182

ABSTRACT

Lactate dehydrogenase A (LDHA) is known to promote the growth and invasion of various types of tumors, affects tumor resistance, and is associated with tumor immune escape. But how LDHA reshapes the tumor microenvironment and promotes the progression of renal cell carcinoma (RCC) remains unclear. In this study, we found that LDHA was highly expressed in clear cell RCC (ccRCC), and this high expression was associated with macrophage infiltration, while macrophages were highly infiltrated in ccRCC, affecting patient prognosis via M2-type polarization. Our in vivo and in vitro experiments demonstrated that LDHA and M2-type macrophages could enhance the proliferation, invasion, and migration abilities of ccRCC cells. Mechanistically, high expression of LDHA in ccRCC cells upregulated the expression of EPHA2 in exosomes derived from renal cancer. Exosomal EPHA2 promoted M2-type polarization of macrophages by promoting activation of the PI3K/AKT/mTOR pathway in macrophages, thereby promoting the progression of ccRCC. All these findings suggest that EPHA2 may prove to be a potential therapeutic target for advanced RCC.


Subject(s)
Carcinoma, Renal Cell , Disease Progression , Exosomes , Kidney Neoplasms , Macrophages , Receptor, EphA2 , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/genetics , Receptor, EphA2/metabolism , Receptor, EphA2/genetics , Humans , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/genetics , Exosomes/metabolism , Animals , Macrophages/metabolism , Macrophages/pathology , Mice , Cell Line, Tumor , Cell Proliferation , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics , Cell Movement , Gene Expression Regulation, Neoplastic , Male , Tumor Microenvironment , Prognosis , TOR Serine-Threonine Kinases/metabolism , Female , Signal Transduction , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism
2.
Oncologist ; 28(6): 553-e472, 2023 06 02.
Article in English | MEDLINE | ID: mdl-36940261

ABSTRACT

BACKGROUND: The MORPHEUS platform comprises multiple open-label, randomized, phase Ib/II trials designed to identify early efficacy and safety signals of treatment combinations across cancers. Atezolizumab (anti-programmed cell death 1 ligand 1 [PD-L1]) was evaluated in combination with PEGylated recombinant human hyaluronidase (PEGPH20). METHODS: In 2 randomized MORPHEUS trials, eligible patients with advanced, previously treated pancreatic ductal adenocarcinoma (PDAC) or gastric cancer (GC) received atezolizumab plus PEGPH20, or control treatment (mFOLFOX6 or gemcitabine plus nab-paclitaxel [MORPHEUS-PDAC]; ramucirumab plus paclitaxel [MORPHEUS-GC]). Primary endpoints were objective response rates (ORR) per RECIST 1.1 and safety. RESULTS: In MORPHEUS-PDAC, ORRs with atezolizumab plus PEGPH20 (n = 66) were 6.1% (95% CI, 1.68%-14.80%) vs. 2.4% (95% CI, 0.06%-12.57%) with chemotherapy (n = 42). In the respective arms, 65.2% and 61.9% had grade 3/4 adverse events (AEs); 4.5% and 2.4% had grade 5 AEs. In MORPHEUS-GC, confirmed ORRs with atezolizumab plus PEGPH20 (n = 13) were 0% (95% CI, 0%-24.7%) vs. 16.7% (95% CI, 2.1%-48.4%) with control (n = 12). Grade 3/4 AEs occurred in 30.8% and 75.0% of patients, respectively; no grade 5 AEs occurred. CONCLUSION: Atezolizumab plus PEGPH20 showed limited clinical activity in patients with PDAC and none in patients with GC. The safety of atezolizumab plus PEGPH20 was consistent with each agent's known safety profile. (ClinicalTrials.gov Identifier: NCT03193190 and NCT03281369).


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Stomach Neoplasms , Humans , Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Pancreatic Ductal/drug therapy , Hyaluronoglucosaminidase/adverse effects , Paclitaxel/adverse effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Stomach Neoplasms/drug therapy
3.
Cancer Sci ; 113(2): 446-458, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34813676

ABSTRACT

Growing evidence supports that N6-methyladenosine (m6A) modification acts as a critical regulator involved in tumorigenesis at the mRNA level. However, the role of m6A modification at the noncoding RNA level remains largely unknown. We found that methyltransferase-like 14 (METTL14) was significantly downregulated in renal cell carcinoma (RCC) tissues (n = 580). Gain-of-function and loss-of-function experiments revealed that METTL14 attenuated the proliferation and migration ability of RCC cells in vivo and in vitro. The methylated RNA immunoprecipitation experiments identified that METTL14 decreased the expression of long noncoding RNA nuclear enriched abundant transcript 1_1 (NEAT1_1) in an m6A-dependent manner. Mechanistically, RNA pull-down assay and RNA immunoprecipitation identified NEAT1_1 directly bound to m6A reader YTH N6-methyladenosine RNA binding protein 2 (YTHDF2). Notably, YTHDF2 accelerated the degradation of NEAT1_1 by selectively recognizing METTL14-mediated m6A marks on NEAT1_1. Multivariate analysis suggested that METTL14 downregulation was associated with malignant characteristics and predicted poor prognosis in RCC patients. In conclusion, our results uncover a newly identified METTL14-YTHDF2-NEAT1_1 signaling axis, which facilitates RCC growth and metastasis and provides fresh insight into RCC therapy.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Methyltransferases/metabolism , RNA, Long Noncoding/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Methyltransferases/genetics , Mice , Prognosis , RNA-Binding Proteins/metabolism
4.
Biochem Biophys Res Commun ; 516(3): 880-887, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31270029

ABSTRACT

Angiopoietin-like proteins (ANGPTLs) comprise a group of proteins that are structurally similar to angiopoietins. In our previous studies, we found that ANGPTL3 can inhibit sorafenib resistance in renal cell carcinoma (RCC). According to bioinformatics analysis based on data in the Cancer Genome Atlas (TCGA), we found that expression of ANGPTL3 was significantly lower in RCC tissues than in adjacent tissues and that disease-free survival and overall survival were significantly shorter in patients with lower ANGPTL3 levels than in those with higher ANGPTL3 levels. Consistent with these results, we demonstrated that RCC tissues exhibited lower ANGPTL3 mRNA and protein expression levels than paired adjacent tissues. Moreover, we found that ANGPTL3 upregulation was associated with better clinical outcomes in RCC patients. ANGPTL3 overexpression inhibited the metastatic ability in RCC cells. Mechanistically, ANGPTL3 binds to vasodilator-stimulated phosphoprotein (VASP) and inhibits its phosphorylation at amino acid 157 in RCC cells. Finally, ANGPTL3 expression and VASP-157 phosphorylation may be combined to predict the prognosis of RCC patients. Overall, our findings describe the role of ANGPTL3 in inhibiting RCC metastasis and thus provide new molecular markers for RCC treatment and prognosis.


Subject(s)
Angiopoietin-like Proteins/genetics , Carcinoma, Renal Cell/genetics , Cell Adhesion Molecules/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Lung Neoplasms/genetics , Microfilament Proteins/genetics , Phosphoproteins/genetics , Aged , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins/metabolism , Animals , Atlases as Topic , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Profiling , Heterografts , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Microfilament Proteins/metabolism , Middle Aged , Phosphoproteins/metabolism , Phosphorylation , Protein Array Analysis , Signal Transduction , Survival Analysis , Tumor Burden
5.
Am J Physiol Renal Physiol ; 314(2): F269-F279, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28446458

ABSTRACT

Renal tubular injury is the hallmark of cisplatin-induced nephrotoxicity. Caspase-11, a member of the caspase family, plays an important role in inflammation and cell death. However, its role in cisplatin-induced renal tubular injury remains unclear. In cisplatin-treated mice, caspase-11 expression was significantly elevated and the expression of caspase-11 was mainly located in renal tubule. Inhibition of caspase-11 by small-interference RNA or its inhibitor wedelolactone attenuated cisplatin-induced renal dysfunction and tubular injury. In cultured primary renal tubular epithelial cells, cisplatin significantly promoted the expression and activation of caspase-11. Inhibition of caspase-11 by small-interference RNA reduced cisplatin-induced cell apoptosis. Overexpression of caspase-11 promoted cell apoptosis by activating the caspase-3-related cell apoptosis. Furthermore, coimmunoprecipitation results showed there was a direct interaction between caspase-11 and caspase-3, and the interaction was enhanced by cisplatin. The fluorescence confocal microscopy results showed that caspase-11 and caspase-3 were colocalized in the cytoplasm of renal tubular epithelial cells. These results demonstrate that caspase-11 plays an important role in cisplatin-induced renal tubular injury. Caspase-11 promotes renal epithelial cell apoptosis by activating the caspase-3-dependent apoptotic pathway. Caspase-11 might be a potential target for therapeutic treatment against cisplatin-induced nephrotoxicity.


Subject(s)
Acute Kidney Injury/enzymology , Apoptosis , Caspase 3/metabolism , Caspases/metabolism , Cisplatin , Epithelial Cells/enzymology , Kidney Tubules/enzymology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Caspase 3/genetics , Caspases/genetics , Caspases, Initiator , Cells, Cultured , Disease Models, Animal , Epithelial Cells/pathology , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Protein Binding , Signal Transduction
6.
Acta Pharmacol Sin ; 39(9): 1513-1521, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30150789

ABSTRACT

SND p102 was first described as a transcriptional co-activator, and subsequently determined to be a co-regulator of Pim-1, STAT6 and STAT5. We previously reported that SND p102 expression was increased in high glucose-treated mesangial cells (MCs) and plays a role in the extracellular matrix (ECM) accumulation of MCs by regulating the activation of RAS. In this study, we further examined the roles of SND p102 in diabetic nephropathy (DN)-induced glomerulosclerosis. Rats were injected with STZ (50 mg/kg, ip) to induce diabetes. MCs or isolated glomeruli were cultured in normal glucose (NG, 5.5 mmol/L)- or high glucose (HG, 25 mmol/L)-containing DMEM. We found that SND p102 expression was significantly increased in the diabetic kidneys, as well as in HG-treated isolated glomeruli and MCs. In addition, HG treatment induced significant fibrotic changes in MCs evidenced by enhanced protein expression of TGF-ß, fbronectin and collagen IV, and significantly increased the proliferation of MCs. We further revealed that overexpression of SND p102 significantly increased the protein expression of angiotensin II (Ang II) type 1 receptor (AT1R) in MCs by increasing its mRNA levels via directly targeting the AT1R 3'-UTR, which resulted in activation of the ERK/Smad3 signaling and subsequently promoted the up-regulation of fbronectin, collagen IV, and TGF-ß in MCs, as well as the cell proliferation. These results demonstrate that SND p102 is a key regulator of AT1R-mediating ECM synthesis and cell proliferation in MCs. Thus, small molecule inhibitors of SND p102 may be a novel therapeutic strategy for DN.


Subject(s)
Cell Proliferation/physiology , Diabetic Nephropathies/physiopathology , Extracellular Matrix/metabolism , Kidney/physiopathology , Mesangial Cells/physiology , Nuclear Proteins/metabolism , Animals , Collagen Type IV/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Down-Regulation , Endonucleases , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/metabolism , Fibrosis/physiopathology , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Signaling System/physiology , Male , Nuclear Proteins/genetics , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
7.
Kidney Int ; 92(4): 888-899, 2017 10.
Article in English | MEDLINE | ID: mdl-28483378

ABSTRACT

Fibrogenesis involves the activation of renal fibroblasts upon kidney injury. However, the mechanisms underlying renal fibroblast activation are poorly characterized. c-Myc is a predominant oncogene encoding a pleiotropic transcription factor that participates in the regulation of various genes, including genes vital for regulating the cell cycle, cell proliferation, and apoptosis. Here we tested whether renal fibrosis in unilateral ureteral obstruction and folic acid-induced renal fibrosis mouse models are associated with the overexpression of c-Myc. Transforming growth factor-ß (TGF-ß) has been identified as a key mediator of renal fibrosis, and it is secreted in an inactive form as a complex with latency-associated peptide and latent TGF-ß-binding proteins. Five αv-containing integrins with different ß -subunits can activate TGF-ß, and consistent with this we found that c-Myc bound directly to the promoter of integrin αv in renal fibroblasts activating its transcription. This, in turn, induced activation of TGF-ß signaling. Pharmacological blockade of c-Myc attenuated renal fibrosis in vivo in the ureteral obstruction and folic acid-treated mouse models and inhibited the proliferation and activation of renal fibroblasts in vitro. Thus, c-Myc overexpression stimulated proliferation and activation of renal fibroblasts by inducing integrin αv -mediated TGF-ß signaling. Hence, targeting c-Myc may have clinical utility in the treatment of renal fibrosis.


Subject(s)
Fibroblasts/pathology , Integrin alphaV/metabolism , Kidney/pathology , Proto-Oncogene Proteins c-myc/metabolism , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta/metabolism , Angiotensin II/metabolism , Animals , Extracellular Matrix/metabolism , Fibrosis , Folic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Renal Insufficiency, Chronic/etiology , Signal Transduction , Thiazoles/pharmacology , Up-Regulation , Ureteral Obstruction/complications
8.
Acta Pharmacol Sin ; 37(5): 637-44, 2016 May.
Article in English | MEDLINE | ID: mdl-27041464

ABSTRACT

AIM: To investigate the effects of ROS scavenger N-acetylcysteine (NAC) on angiotensin II (Ang II)-mediated renal fibrosis in vivo and in vitro. METHODS: Mice were subjected to unilateral ureteral obstruction (UUO), and then treated with vehicle or NAC (250 mg/kg, ip) for 7 days. Histological changes of the obstructed kidneys were observed with Masson's trichrome staining. ROS levels were detected with DHE staining. The expression of relevant proteins in the obstructed kidneys was assessed using Western blotting assays. Cultured rat renal fibroblast NRK-49F cells were used for in vitro experiments. RESULTS: In the obstructed kidneys, Ang II levels were significantly elevated, and collagen I was accumulated in the interstitial spaces. Furthermore, ROS production and the expression of p47 (a key subunit of NADPH oxidase complexes) were increased in a time-dependent manner; the expression of fibronectin, α-SMA and TGF-ß were upregulated. Administration of NAC significantly alleviated the fibrotic responses in the obstructed kidneys. In cultured NRK-49F cells, treatment with Ang II (0.001-10 µmol/L) increased the expression of fibronectin, collagen I, α-SMA and TGF-ß in dose-dependent and time-dependent manners. Ang II also increased ROS production and the phosphorylation of Smad3. Pretreatment with NAC (5 µmol/L) blocked Ang II-induced oxidative stress and ECM production in the cells. CONCLUSION: In mouse obstructed kidneys, the fibrotic responses result from Ang II upregulation can be alleviated by the ROS scavenger N-acetylcysteine.


Subject(s)
Acetylcysteine/therapeutic use , Angiotensin II/metabolism , Antioxidants/therapeutic use , Kidney Diseases/drug therapy , Ureteral Obstruction/drug therapy , Acetylcysteine/pharmacology , Angiotensin II/pharmacology , Animals , Antioxidants/pharmacology , Cell Line , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/pathology , Kidney/drug effects , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Male , Mice, Inbred C57BL , Oxidative Stress , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/drug effects , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
9.
Int J Mol Sci ; 17(2)2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26805826

ABSTRACT

Renal fibrosis is the final common pathway of chronic kidney disease (CKD), and no effective medication is available clinically for managing its progression. Metformin was initially developed as an anti-diabetic drug and recently gained attention for its potential in the treatment of other diseases. In this study, we investigated its effects on renal fibrosis in a mouse model of unilateral ureteral obstruction (UUO) in vivo and in angiotensin II (Ang II)-treated renal fibroblast NRK-49F cells in vitro. Our data showed that UUO induced renal fibrosis and combined with the activation of ERK signaling, the upregulation of fibronectin, collagen I, and transforming growth factor-ß (TGF-ß). The administration of metformin inhibited the activation of ERK signaling and attenuated the production of extracellular matrix (ECM) proteins and collagen deposition in the obstructed kidneys. In cultured renal fibroblasts, Ang II increased the expression of fibronectin and collagen I and also activated ERK signaling and TGF-ß in a time-dependent manner. Pretreatment of the cells with metformin blocked Ang II-induced ERK signaling activation and ECM overproduction. Our results show that metformin prevents renal fibrosis, possibly through the inhibition of ERK signaling, and may be a novel strategy for the treatment of renal fibrosis.


Subject(s)
Angiotensin II/pharmacology , Extracellular Matrix Proteins/metabolism , Kidney Diseases/prevention & control , Metformin/administration & dosage , Ureteral Obstruction/drug therapy , Animals , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Kidney Diseases/etiology , Kidney Diseases/metabolism , MAP Kinase Signaling System/drug effects , Metformin/pharmacology , Mice , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
10.
Andrology ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38505906

ABSTRACT

BACKGROUND: Erectile dysfunction has been associated with leisure sedentary behavior in several epidemiological and observational studies. However, the interpretation of these findings is difficult due to residual confounding or reverse causality. OBJECTIVES: To explore the causal association between leisure sedentary behavior and erectile dysfunction, and to explore the underlying mechanism using Mendelian randomization. MATERIALS AND METHODS: In the present study, publicly available large-scale genome-wide association studies of leisure sedentary behaviors (television watching, computer use, and driving), erectile dysfunction, sex hormones (total testosterone, bioactive testosterone, estradiol, follicle-stimulating hormone, luteinizing hormone, prolactin, and sex hormone binding globulin), biomarkers of endothelial function (C reactive protein, E-selectin, and matrix metalloproteinase 7), and psychiatric symptoms (depression and anxiety) were used to perform two-sample Mendelian randomization analyses. The inverse variance weighting method was the main method used to estimate the association, and sensitivity analyses were also performed. RESULTS: A greater risk of erectile dysfunction was significantly associated with a higher genetic susceptibility to leisure computer usage (odds ratio = 3.57; 95% confidence interval = 1.78-7.16; p < 0.001). No evidence was obtained to suggest that watching television or driving for leisure increased the risk of erectile dysfunction. No association was found between computer use and depression, anxiety, C reactive protein, E-selectin, matrix metalloproteinase 7, or other sex hormones, with the exception of follicle-stimulating hormone levels (odds ratio = 0.29; 95% confidence interval = 0.12-0.69; p = 0.01). No indication of heterogeneity or pleiotropy was identified by sensitivity analysis. DISCUSSION: Extended computer usage for leisure raised the likelihood of developing erectile dysfunction, which may be associated to lower follicle-stimulating hormone levels; however, the role of endothelial dysfunction and psychological disorders in the development of erectile dysfunction should not be underestimated. Moderate physical activity may help to correct the dysfunction. CONCLUSION: The present study offered substantial evidence for a positive causal association between computer use and the risk of erectile dysfunction. However, a definitive causal association needs to be established by further research.

11.
Asian Pac J Allergy Immunol ; 31(3): 247-52, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24053708

ABSTRACT

BACKGROUND: IL-13 (interleukin-13) and RANTES (Regulated upon Activation, Normal T cells Expressed and Secreted) are the important asthma inflammatory mediators. OBJECTIVE: The present study aimed to investigate the single and combined associations between the polymorphism (SNP) loci in IL-13 and RANTES genes with the development of asthma in children of Chinese Han nationality. METHODS: The risk associated with genotypes of three IL-13 SNPs and two RANTES SNPs was determined by the Χ2 test in 384 children with asthma and an equal number of healthy controls matched by sex. RESULTS: Between the experimental and control groups, no statistically significant differences (P >0.05) were found in genotype distribution and allele frequency among three loci (IL-13 C-1112T, IL-13 C1923T, and RANTES A-403G). However, significant diversity was observed among IL-13 A2044G (P =0.0001) and RANTES G-28C (P =0.0001). Moreover, the frequency of IL-13 A2044G A/A and RANTES G-28C G/G in the asthma group was significantly higher than in the control group (odds ratio [OR] =2.59, P =0.0001; OR =3.00, P =0.0001, respectively). Carriers of both IL-13 A2044G A/A and RANTES G-28C G/G have a more significant risk for developing asthma than those with only a single polymorphism. CONCLUSIONS: The three loci (IL-13 C-1112T, IL-13 C1923T, and RANTES A-403G) make little contribution to the development of asthma in children of Chinese Han nationality. IL-13 A2044G and RANTES G-28C are significantly associated with childhood asthma. IL-13 A2044G A/A and RANTES G-28C G/G have a significant and combined effect on the development of asthma.


Subject(s)
Alleles , Asthma/genetics , Chemokine CCL5/genetics , Gene Frequency , Interleukin-13/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Asian People , Asthma/epidemiology , Child , Child, Preschool , China/epidemiology , Female , Genetic Loci , Genotype , Humans , Male , Risk Factors
12.
Front Cardiovasc Med ; 10: 1121340, 2023.
Article in English | MEDLINE | ID: mdl-37025676

ABSTRACT

Background: Erection dysfunction has been associated with hypertension in several epidemiological and observational studies. But the causal association between hypertension and erectile dysfunction requires further investigation. Methods: A two-sample Mendelian randomization (MR) was conducted to analyze the causal effect of hypertension on risk of erection dysfunction. Large-scale publicly available genome-wide association study data were used to estimate the putative causality between hypertension and risk of erectile dysfunction. A total of 67 independent single nucleotide polymorphisms were selected as instrumental variables. Inverse-variant weighted, maximum likelihood, weighted median, penalized weighted median, and MR-PRESSO approaches were utilized in MR analyses. Heterogeneity test, horizontal pleiotropy test, and leave-one-out method were used to prove the stability of the results. Results: In total, all P values were less than 0.05, demonstrating a positive causal link between hypertension and risk of erectile dysfunction in multiple MR methods, such as inverse-variant weighted (random and fixed effect) (OR 3.8315, 95% CI 2.3004-6.3817, P = 0.0085), maximum likelihood (OR 3.8877, 95% CI 2.3224-6.5081, P = 0.0085), weighted median (OR 4.9720, 95% CI 2.3645-10.4550, P = 0.0309), penalized weighted median (OR 4.9760, 95% CI 2.3201-10.6721, P = 0.0355), and MR-PRESSO (OR 3.6185, 95% CI 2.2387-5.8488, P = 0.0092). Sensitivity analysis detected no evidence of heterogeneity, pleiotropy, or outlier single nucleotide polymorphisms. Conclusion: The study revealed a positive causal link between the presence of hypertension and the risk of erectile dysfunction. More attention should be paid during the management of hypertension with the purpose of preventing erectile dysfunction or improving erectile function.

13.
Sci Rep ; 13(1): 9778, 2023 06 16.
Article in English | MEDLINE | ID: mdl-37328520

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) with venous tumor thrombus (VTT) is associated with poor prognosis. Our integrative analyses of transcriptome and proteome reveal distinctive molecular features of ccRCC with VTT, and yield the development of a prognostic classifier to facilitate ccRCC molecular subtyping and treatment. The RNA sequencing and mass spectrometry were performed in normal-tumor-thrombus tissue triples of five ccRCC patients. Statistical analysis, GO and KEGG enrichment analysis, and protein-protein interaction network construction were used to interpret the transcriptomic and proteomic data. A six-gene-based classifier was developed to predict patients' survival using Cox regression, which was validated in an independent cohort. Transcriptomic analysis identified 1131 tumorigenesis-associated differentially expressed genes (DEGs) and 856 invasion-associated DEGs. Overexpression of transcription factor EGR2 in VTT indicated its important role in tumor invasion. Furthermore, proteomic analysis showed 597 tumorigenesis-associated differentially expressed proteins (DEPs) and 452 invasion-associated DEPs. The invasion-associated DEPs showed unique enrichment in DNA replication, lysine degradation, and PPAR signaling pathway. Integration of transcriptome and proteome reveals 142 tumorigenesis-associated proteins and 84 invasion-associated proteins displaying changes consistent with corresponding genes in transcriptomic profiling. Based on their different expression patterns among normal-tumor-thrombus triples, RAB25 and GGT5 were supposed to play a consistent role in both tumorigenesis and invasion processes, while SHMT2 and CADM4 might play the opposite roles in tumorigenesis and thrombus invasion. A prognostic classifier consisting of six DEGs (DEPTOR, DPEP1, NAT8, PLOD2, SLC7A5, SUSD2) performed satisfactorily in predicting survival of ccRCC patients (HR = 4.41, P < 0.001), which was further validated in an independent cohort of 40 cases (HR = 5.52, P = 0.026). Our study revealed the transcriptomic and proteomic profiles of ccRCC patients with VTT, and identified the distinctive molecular features associated with VTT. The six-gene-based prognostic classifier developed by integrative analyses may facilitate ccRCC molecular subtyping and treatment.


Subject(s)
Carcinoma, Renal Cell , Carcinoma , Kidney Neoplasms , Thrombosis , Humans , Carcinogenesis , Carcinoma, Renal Cell/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/pathology , Prognosis , Proteome/genetics , Proteomics , Transcriptome
14.
J Immunother Cancer ; 11(12)2023 12 06.
Article in English | MEDLINE | ID: mdl-38056895

ABSTRACT

BACKGROUND: Cancer immunotherapies can induce durable tumor regression, but most patients do not respond. SETD2 mutation has been linked to the efficacy of immune checkpoint inhibitors (ICIs) immunotherapy. The functional importance of the SETD2 inactivation and how to modulate immunotherapy response remains unclear. METHODS: To explore the function of SETD2 in immunotherapy, knockout and subsequent functional experiments were conducted. Bulk RNA-seq, ATAC-seq, Chip-seq and single-cell RNA-seq were performed to dissect the mechanism and explore the immune microenvironment of mouse tumor. Flow cytometry was used to assess cell surface antigen and intratumoral T cell levels. RESULTS: We comprehensively determine the effect of SETD2 inactivation in ICIs therapy and elucidate the mechanistic impact on tumor immunity. Murine syngeneic tumors harboring Setd2 inactivation are sensitive to ICIs. By bulk and single-cell RNA-seq, we further reveal that SETD2 inactivation reprograms intratumoral immune cells and inflames the tumor microenvironment, which is characterized by high infiltration of T cells and enhanced antigen presentation to activate CD8+ T cell-mediated killing. Mechanistically, via an integrated multiomics analysis using ATAC-seq, ChIP-seq and RNA-seq, we demonstrate that SETD2 inactivation reduces NR2F1 transcription by impairing H3K36me3 deposition and chromatin accessibility, which activates the STAT1 signaling pathway to promote chemokines and programmed cell death protein-1 (PD-1) expression and enhance antigen presentation. All these regulatory mechanisms synergistically promote the effects of anti-programmed cell death ligand 1 immunotherapy in Setd2-knockout syngeneic mouse models. The SETD2-NR2F1-STAT1 regulatory axis is conserved in human and murine cancers. Finally, cancer patients harboring SETD2 mutations who received ICIs show increased durable clinical benefits and survival. CONCLUSIONS: These findings provide novel insights into the biology of SETD2 inactivation regulation and reveal a new potential therapeutic biomarker for ICIs immunotherapy in various refractory cancers.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , CD8-Positive T-Lymphocytes , Biomarkers , Immunotherapy , Tumor Microenvironment , COUP Transcription Factor I/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase/metabolism
15.
Cancer Innov ; 1(2): 146-167, 2022 Aug.
Article in English | MEDLINE | ID: mdl-38090653

ABSTRACT

Background: Cancer metastasis and recurrence remain major challenges in renal carcinoma patient management. There are limited biomarkers to predict the metastatic probability of renal cancer, especially in the early-stage subgroup. Here, our study applied robust machine-learning algorithms to identify metastatic and recurrence-related signatures across multiple renal cancer cohorts, which reached high accuracy in both training and testing cohorts. Methods: Clear cell renal cell carcinoma (ccRCC) patients with primary or metastatic site sequencing information from eight cohorts, including one out-house cohort, were enrolled in this study. Three robust machine-learning algorithms were applied to identify metastatic signatures. Then, two distinct metastatic-related subtypes were identified and verified; matrix remodeling associated 5 (MXRA5), as a promising diagnostic and therapeutic target, was investigated in vivo and in vitro. Results: We identified five stable metastasis-related signatures (renin, integrin subunit beta-like 1, MXRA5, mesenchyme homeobox 2, and anoctamin 3) from multicenter cohorts. Additionally, we verified the specificity and sensibility of these signatures in external and out-house cohorts, which displayed a satisfactory consistency. According to these metastatic signatures, patients were grouped into two distinct and heterogeneous ccRCC subtypes named metastatic cancer subtype 1 (MTCS1) and type 2 (MTCS2). MTCS2 exhibited poorer clinical outcomes and metastatic tendencies than MTCS1. In addition, MTCS2 showed higher immune cell infiltration and immune signature expression but a lower response rate to immune blockade therapy than MTCS1. The MTCS2 subgroup was more sensitive to saracatinib, sunitinib, and several molecular targeted drugs. In addition, MTCS2 displayed a higher genome mutation burden and instability. Furthermore, we constructed a prognosis model based on subtype biomarkers, which performed well in training and validation cohorts. Finally, MXRA5, as a promising biomarker, significantly suppressed malignant ability, including the cell migration and proliferation of ccRCC cell lines in vitro and in vivo. Conclusions: This study identified five robust metastatic signatures and proposed two metastatic probability clusters with stratified prognoses, multiomics landscapes, and treatment options. The current work not only provided new insight into the heterogeneity of renal cancer but also shed light on optimizing decision-making in immunotherapy and chemotherapy.

16.
Oxid Med Cell Longev ; 2022: 3617775, 2022.
Article in English | MEDLINE | ID: mdl-35028006

ABSTRACT

METHODS: This study was based on the multiomics data (including mRNA, lncRNA, miRNA, methylation, and WES) of 258 ccRCC patients from TCGA database. Firstly, we screened the feature values that had impact on the prognosis and obtained two subtypes. Then, we used 10 algorithms to achieve multiomics clustering and conducted pseudotiming analysis to further validate the robustness of our clustering method, based on which the two subtypes of ccRCC patients were further subtyped. Meanwhile, the immune infiltration was compared between the two subtypes, and drug sensitivity and potential drugs were analyzed. Furthermore, to analyze the heterogeneity of patients at the multiomics level, biological functions between two subtypes were compared. Finally, Boruta and PCA methods were used for dimensionality reduction and cluster analysis to construct a renal cancer risk model based on mRNA expression. RESULTS: A prognosis predicting model of ccRCC was established by dividing patients into the high- and low-risk groups. It was found that overall survival (OS) and progression-free interval (PFI) were significantly different between the two groups (p < 0.01). The area under the OS time-dependent ROC curve for 1, 3, 5, and 10 years in the training set was 0.75, 0.72, 0.71, and 0.68, respectively. CONCLUSION: The model could precisely predict the prognosis of ccRCC patients and may have implications for drug selection for ccRCC patients.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Data Analysis , Humans , Prognosis
17.
Oxid Med Cell Longev ; 2022: 5910575, 2022.
Article in English | MEDLINE | ID: mdl-35401923

ABSTRACT

Background: Cancer is a major threat to human health worldwide. Although recent innovations and advances in early detection and effective therapies such as targeted drugs and immune checkpoint inhibitors have saved more lives of cancer patients and improved their quality of life, our knowledge about cancer remains largely unknown. CCNA2 belongs to the cell cyclin family and has been demonstrated to be a tumorigenic gene in multiple solid tumor types. The aim of the present study was to make a comprehensive analysis on the role of CCNA2 at a pancancer level. Methods: Multidatabases were collected to evaluate the different expression, prognostic value, DNA methylation, tumor mutation burden, microsatellite instability, mismatch repair, tumor immune microenvironment, and drug sensitivity of CCNA2 across pancancer. IHC was utilized to validate the expression and prognostic value of CCNA2 in ccRCC patients from SMMU cohort. Results: CCNA2 was differentially expressed in most cancer types vs. normal tissues. CCNA2 may significantly influence the prognosis of multiple cancer types, especially clear cell renal cell carcinoma (ccRCC). CCNA2 was also frequently mutated in most cancer types. Notably, CCNA2 was significantly correlated with immune cell infiltration and immune checkpoint inhibitory genes. In addition, CCNA2 was also strongly related to drug resistance. Conclusion: CCNA2 may prove to be a new biomarker for prognostic prediction, tumor immunity assessment, and drug susceptibility evaluation in pancancer level, especially in ccRCC.


Subject(s)
Carcinoma, Renal Cell , Cyclin A2 , Kidney Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Cyclin A2/genetics , Humans , Kidney Neoplasms/genetics , Quality of Life , Tumor Microenvironment/genetics
18.
Front Immunol ; 13: 842069, 2022.
Article in English | MEDLINE | ID: mdl-35281041

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal carcinoma and is associated with poor prognosis and notorious for its immune dysfunction characteristic. SNRPA1 is a spliceosome component responsible for processing pre-mRNA into mRNA, while the biological effect of SNRPA1 in ccRCC remains elusive. The aim of this study was to decipher the effect of SNRPA1 on clinical effect and tumor immunity for ccRCC patients. Multi-databases were collected to evaluate the different expression, prognostic value, DNA methylation, tumor immune microenvironment, and drug sensitivity of SNRPA1 on ccRCC. IHC was utilized to validate the expression and prognostic value of SNRPA1 in ccRCC patients from the SMMU cohort. The knockout expression of SNRPA by sgRNA plasmid inhibited the cell proliferation, migration, and metastasis ability and significantly increased the sensitivity of sunitinib treatment. In addition, we explored the role of SNRPA1 in pan-cancer level. The results indicated that SNRPA1 was differentially expressed in most cancer types. SNRPA1 may significantly influence the prognosis of multiple cancer types, especially in ccRCC patients. Notably, SNRPA1 was significantly correlated with immune cell infiltration and immune checkpoint inhibitory genes. In addition, the aggressive and immune inhibitory effects shown in SNRPA1 overexpression and the effect of SNRPA1 on ccRCC cell line invasion, metastasis, and drug sensitivity in vitro were observed. Moreover, SNRPA1 was related to Myc, MTORC, G2M, E2F, and DNA repair pathways in various cancer types. In all, SNRPA1 may prove to be a new biomarker for prognostic prediction, effect tumor immunity, and drug susceptibility in ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Female , Humans , Kidney Neoplasms/genetics , Male , Prognosis , Sunitinib/pharmacology , Sunitinib/therapeutic use , Tumor Microenvironment
19.
Int J Biol Sci ; 17(12): 3158-3172, 2021.
Article in English | MEDLINE | ID: mdl-34421357

ABSTRACT

DEAD-box protein 39 (DDX39) has been demonstrated to be a tumorigenic gene in multiple tumor types, but its role in the progression and immune microenvironment of clear cell renal cell cancer (ccRCC) remains unclear. The aim of the present study was to investigate the role of DDX39 in the ccRCC tumor progression, immune microenvironment and efficacy of immune checkpoint therapy. The DDX39 expression level was first detected in tumors in the public data and then verified in ccRCC samples from Changzheng Hospital. The prognostic value of DDX39 expression was assessed in the Cancer Genome Atlas (TCGA) and ccRCC patients from Changhai Hospital. The role of DDX39 in promoting ccRCC was analyzed by bioinformatic analysis and in vitro experiments. The association between DDX39 expression and immune cell infiltration and immune inhibitory markers was analyzed, and its value in predicting the immune checkpoint therapy efficacy in ccRCC were evaluated in the public database. DDX39 expression was elevated in Oncomine, GEO and TCGA ccRCC databases, as well as in Changzheng ccRCC samples. In TCGA ccRCC patients, increased DDX39 expression predicted worse overall survival (OS) (p<0.0001) and progression-free interval (PFI) (p<0.0001), and was shown as an independent predictive factor for OS (p=0.002). These findings were consistent with those from Changhai ccRCC patients. In addition, GO and GSEA analysis identified DDX39 as a pro-ccRCC gene. In vitro experiments confirmed the role of DDX39 in promoting ccRCC cell. Finally, DDX39 was found to be positively correlated with a variety of immune inhibitory markers, and could predict the adverse efficacy of immune checkpoint therapy in TIDE analysis. In conclusion, Increased DDX39 in ccRCC patients predicted worse clinical prognosis, promoted ccRCC cell proliferation, migration and invasion, and also predicted adverse efficacy of immune checkpoint therapy.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/drug therapy , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic/physiology , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Blotting, Western , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Cell Proliferation , Computational Biology , Female , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Male , Middle Aged , Prognosis , Real-Time Polymerase Chain Reaction , Tumor Microenvironment
20.
Front Oncol ; 11: 755212, 2021.
Article in English | MEDLINE | ID: mdl-34804944

ABSTRACT

BACKGROUND: Pyroptosis is essential for tumorigenesis and progression of neoplasm. However, the heterogeneity of pyroptosis and its relationship with the tumor microenvironment (TME) in clear cell renal cell carcinoma (ccRCC) remain unclear. The purpose of the present study was to identify pyroptosis-related subtypes and construct a prognosis prediction model based on pyroptosis signatures. METHODS: First, heterogenous pyroptosis subgroups were explored based on 33 pyroptosis-related genes and ccRCC samples from TCGA, and the model established by LASSO regression was verified by the ICGC database. Then, the clinical significance, functional status, immune infiltration, cell-cell communication, genomic alteration, and drug sensitivity of different subgroups were further analyzed. Finally, the LASSO-Cox algorithm was applied to narrow down the candidate genes to develop a robust and concise prognostic model. RESULTS: Two heterogenous pyroptosis subgroups were identified: pyroptosis-low immunity-low C1 subtype and pyroptosis-high immunity-high C2 subtype. Compared with C1, C2 was associated with a higher clinical stage or grade and a worse prognosis. More immune cell infiltration was observed in C2 than that in C1, while the response rate in the C2 subgroup was lower than that in the C1 subgroup. Pyroptosis-related genes were mainly expressed in myeloid cells, and T cells and epithelial cells might influence other cell clusters via the pyroptosis-related pathway. In addition, C1 was characterized by MTOR and ATM mutation, while the characteristics of C2 were alterations in SPEN and ROS1 mutation. Finally, a robust and promising pyroptosis-related prediction model for ccRCC was constructed and validated. CONCLUSION: Two heterogeneous pyroptosis subtypes were identified and compared in multiple omics levels, and five pyroptosis-related signatures were applied to establish a prognosis prediction model. Our findings may help better understand the role of pyroptosis in ccRCC progression and provide a new perspective in the management of ccRCC patients.

SELECTION OF CITATIONS
SEARCH DETAIL