Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 780
Filtrer
1.
Vet Immunol Immunopathol ; 271: 110741, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38520894

RÉSUMÉ

Tumor-infiltrating lymphocyte (TIL) density plays an important role in anti-tumor immunity and is associated with patient outcome in various human and canine malignancies. As a first assessment of the immune landscape of the tumor microenvironment in canine renal cell carcinoma (RCC), we retrospectively analyzed clinical data and quantified CD3, FoxP3, and granzyme B immunostaining in formalin-fixed paraffin-embedded tumor samples from 16 dogs diagnosed with renal cell carcinoma treated with ureteronephrectomy. Cell density was low for all markers evaluated. Increased numbers of intratumoral FoxP3 labelled (+) cells, as well as decreased granzyme B+: FoxP3+ TIL ratio, were associated with poor patient outcomes. Our initial study of canine RCC reveals that these tumors are immunologically cold and Tregs may play an important role in immune evasion.


Sujet(s)
Antigènes CD3 , Néphrocarcinome , Maladies des chiens , Facteurs de transcription Forkhead , Granzymes , Tumeurs du rein , Lymphocytes TIL , Animaux , Chiens , Néphrocarcinome/médecine vétérinaire , Néphrocarcinome/immunologie , Néphrocarcinome/enzymologie , Antigènes CD3/analyse , Antigènes CD3/métabolisme , Maladies des chiens/immunologie , Maladies des chiens/enzymologie , Facteurs de transcription Forkhead/analyse , Facteurs de transcription Forkhead/métabolisme , Granzymes/métabolisme , Granzymes/analyse , Immunohistochimie/médecine vétérinaire , Tumeurs du rein/médecine vétérinaire , Tumeurs du rein/immunologie , Tumeurs du rein/enzymologie , Lymphocytes TIL/immunologie , Études rétrospectives
2.
J Biomed Nanotechnol ; 18(4): 1001-1008, 2022 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-35854457

RÉSUMÉ

The aim of this study was to examine the impact of Resveratrol nanoparticles on migration/invasion capacity of renal cell carcinoma (RCC) cells and its mechanism. Human RCC cells were exposed to dimethyl sulfoxide or gradient concentrations of Resveratrol nanoparticles respectively, and U0126 were also added in some experiments. We examined renal cell viability by MTT assay, and wound healing test and Transwell assays were used detect invasion and migration capability of RCC cells. We used Western blotting assay to analyze the protein levels in extracellular signal-regulated kinase (ERK) signaling. We also detected the enzymatic capacity of matrix metalloproteinase 2 (MMP-2) in cells by gelatin enzymatic profiling. Resveratrol nanoparticles treatment significantly suppressed cell viability to migrate and invade RCC cells in a dose-dependent manner. Also, notably were reduced MMP-2 activity and expression, and elevated TIMP-2 level were observed in RCC cells exposed with Resveratrol nanoparticles. Further, Resveratrol nanoparticles treatment significantly decreased only the expression of p-ERK1/2, but not p-p38 and p-JNK. Moreover, U0126, which is the ERK inhibitor, exerted similar role as Resveratrol nanoparticles did. Of note was that, combined use of U0126 and Resveratrol nanoparticles displayed a more intense suppression of MMP-2 activity and expression, and also the viability to migrate and invade the RCC cells, compared with Resveratrol nanoparticles treatment alone. The Resveratrol nanoparticles inhibited RCC cells migration and invasion by regulating MMP2 expression and ERK pathways.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Système de signalisation des MAP kinases , Matrix metalloproteinase 2 , Nanoparticules , Resvératrol , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/enzymologie , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Humains , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/enzymologie , Tumeurs du rein/anatomopathologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Matrix metalloproteinase 2/biosynthèse , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/métabolisme , Inhibiteurs de métalloprotéinases matricielles/administration et posologie , Inhibiteurs de métalloprotéinases matricielles/pharmacologie , Nanoparticules/administration et posologie , Invasion tumorale , Resvératrol/administration et posologie , Resvératrol/pharmacologie
4.
Mol Cell ; 82(7): 1249-1260.e7, 2022 04 07.
Article de Anglais | MEDLINE | ID: mdl-35216667

RÉSUMÉ

Fumarate is an oncometabolite. However, the mechanism underlying fumarate-exerted tumorigenesis remains unclear. Here, utilizing human type2 papillary renal cell carcinoma (PRCC2) as a model, we show that fumarate accumulates in cells deficient in fumarate hydratase (FH) and inhibits PTEN to activate PI3K/AKT signaling. Mechanistically, fumarate directly reacts with PTEN at cysteine 211 (C211) to form S-(2-succino)-cysteine. Succinated C211 occludes tethering of PTEN with the cellular membrane, thereby diminishing its inhibitory effect on the PI3K/AKT pathway. Functionally, re-expressing wild-type FH or PTEN C211S phenocopies an AKT inhibitor in suppressing tumor growth and sensitizing PRCC2 to sunitinib. Analysis of clinical specimens indicates that PTEN C211 succination levels are positively correlated with AKT activation in PRCC2. Collectively, these findings elucidate a non-metabolic, oncogenic role of fumarate in PRCC2 via direct post-translational modification of PTEN and further reveal potential stratification strategies for patients with FH loss by combinatorial AKTi and sunitinib therapy.


Sujet(s)
Carcinome papillaire , Néphrocarcinome , Fumarates , Tumeurs du rein , Phosphohydrolase PTEN , Carcinogenèse , Carcinome papillaire/traitement médicamenteux , Carcinome papillaire/enzymologie , Carcinome papillaire/génétique , Carcinome papillaire/métabolisme , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/enzymologie , Néphrocarcinome/génétique , Néphrocarcinome/métabolisme , Cystéine/métabolisme , Résistance aux médicaments antinéoplasiques , Fumarate hydratase/génétique , Fumarate hydratase/métabolisme , Fumarates/pharmacologie , Humains , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/enzymologie , Tumeurs du rein/génétique , Tumeurs du rein/métabolisme , Phosphohydrolase PTEN/antagonistes et inhibiteurs , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes c-akt/génétique , Sunitinib/pharmacologie
5.
Front Immunol ; 12: 734646, 2021.
Article de Anglais | MEDLINE | ID: mdl-34795663

RÉSUMÉ

Interferon-gamma (IFN-γ) has a complex role in modulating the tumor microenvironment (TME) during renal cell carcinoma (RCC) development. To define the role of IFN-γ response genes in RCC progression, we characterized the differential gene expression, prognostic implications, and DNA variation profiles of selected IFN-γ response signatures, which exhibited a significant hazard ratio for the overall survival (OS) and progression-free survival (PFS) of papillary, chromophobia, and clear cell RCC (ccRCC) patients (n = 944). Prognostic nomograms were constructed to predict the outcomes for ccRCC patients, highlighting the prognostic implications of RANBP2-type and C3HC4-type zinc finger containing 1 (RBCK1). Interestingly, large-scale pan-cancer samples (n = 12,521) and three single-cell RNA datasets revealed that RBCK1 showed markedly differential expression between cancer and normal tissues and significantly correlated with tumor-infiltrating immune cells, tumor purity, and immune checkpoint molecules, such as PD-L1, CTLA-4, LAG-3, and TIGIT in pan-cancer samples. Notably, the TIDE score was significantly higher in the RBCK1high group compared with the RBCK1low group in both ccRCC and RCC cohorts. Besides, immunohistochemistry staining showed significantly elevated RBCK1 expression in tumors (n = 50) compared with kidney samples (n = 40) from a real-world cohort, Fudan University Shanghai Cancer Center (FUSCC, Shanghai). After RBCK1 expression was confirmed in ccRCC, we found a significantly decreased number of infiltrating CD4+ T cells, CD4+ FOXP3+ Treg cells, M1 macrophages, and CD56bight/dim NK cells in the immune-cold RBCK1high group. In addition to the distinct heterogeneous immune microenvironment, the increased expression of RBCK1 predicted a prominently worse prognosis than the RBCK1low group for 232 ccRCC patients in the FUSCC proteomic cohort. Furthermore, after transfected with siRNA in human ccRCC cells, extraordinarily decreased cell proliferation, migration capacities, and prominently elevated apoptosis tumor cell proportion were found in the siRNA groups compared with the negative control group. In conclusion, this study identified IFN-γ response clusters, which might be used to improve the prognostic accuracy of immune contexture in the ccRCC microenvironment. Immune-cold RBCK1high patients have pro-tumorigenic immune infiltration and significantly worse outcomes than RBCK1low patients based on results from multi-omics to real-world data. Our discovery of novel independent prognostic indicators for RCC highlights the association between tumor alterations and immune phenotype.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Néphrocarcinome/génétique , Génomique , Interféron gamma/génétique , Tumeurs du rein/génétique , Protéomique , Facteurs de transcription/génétique , Microenvironnement tumoral , Ubiquitin-protein ligases/génétique , Marqueurs biologiques tumoraux/métabolisme , Néphrocarcinome/enzymologie , Néphrocarcinome/immunologie , Néphrocarcinome/thérapie , Lignée cellulaire tumorale , Bases de données génétiques , Techniques d'aide à la décision , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Humains , Interféron gamma/métabolisme , Tumeurs du rein/enzymologie , Tumeurs du rein/immunologie , Tumeurs du rein/thérapie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Nomogrammes , Phénotype , Survie sans progression , Cartes d'interactions protéiques , Protéome , RNA-Seq , Transduction du signal , Analyse sur cellule unique , Facteurs temps , Facteurs de transcription/métabolisme , Transcriptome , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Ubiquitin-protein ligases/métabolisme
6.
Pathol Res Pract ; 227: 153642, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34649054

RÉSUMÉ

BACKGROUND: This research focuses on exploring RSK4 protein expression within Clear Cell Renal Cell Carcinoma (ccRCC), based on these investigations on level of expressions coupled with the relevance to clinicopathologic features and clinical outcomes. METHODS: The expression of RSK4 in 48 ccRCC and 20 hydronephrosis samples were under the detection of immunohistochemistry; besides, its relevance to the combination of clinicopathologic features with prognosis was committed in virtue of statistical approaches. RESULTS: The 48 ccRCC samples included 36 (75%, 36/48) positive for RSK4, while the positive rate in hydronephrosis samples were 5 (25%, 5/20). Statistical analysis showed that RSK4 in ccRCC samples express higher expression the hydronephrosis samples (P < 0.05). Furthermore, the expression of RSK4 in ccRCC samples weren't correlated with ages and genders (P > 0.05), while WHO/ISUP nucleolar grade harboured relevance to low survival rate (P = 0.018). Molecular researches demonstrated that over-expression of RSK4 was able to upgrade the proliferation capability of ccRCC cell lines. CONCLUSIONS: According to the expression pattern and molecular systems featured RSK4 in ccRCCs, it performed the function of a latent independent prognostic factor performing the function of a newly built latent therapeutic aim oriented with the patients undergoing RCC. Moreover, the specific mechanism of action is expected to be revealed in the future research.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Néphrocarcinome/enzymologie , Tumeurs du rein/enzymologie , Ribosomal Protein S6 Kinases, 90-kDa/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Marqueurs biologiques tumoraux/génétique , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Immunohistochimie , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Mâle , Souris de lignée BALB C , Souris nude , Adulte d'âge moyen , Valeur prédictive des tests , Pronostic , Ribosomal Protein S6 Kinases, 90-kDa/génétique , Charge tumorale , Régulation positive
8.
Cell Death Dis ; 12(7): 621, 2021 06 16.
Article de Anglais | MEDLINE | ID: mdl-34135317

RÉSUMÉ

Clear cell renal cell carcinomas (ccRCC) reprogram carbon metabolism responses to hypoxia, thereby promoting utilization of glutamine. Recently, sirtuin 4 (SIRT4), a novel molecular has turned out to be related to alternating glutamine metabolism and modulating the tumor microenvironment. However, the role of SIRT4 in ccRCC remains poorly understood. Here, we illustrated that the expression of SIRT4 is markedly reduced in cancerous tissues, and closely associated with malignancy stage, grade, and prognosis. In ccRCC cells, SIRT4 exerted its proapoptotic activity through enhancing intracellular reactive oxygen species (ROS). Heme oxygenase-1 (HO-1) is part of an endogenous defense system against oxidative stress. Nevertheless, overexpression of SIRT4 hindered the upregulation of HO-1 in von Hippel-Lindau (VHL)-proficient cells and repressed its expression in VHL-deficient cells. This discrepancy indicated that competent VHL withstands the inhibitory role of SIRT4 on HIF-1α/HO-1. Functionally, overexpression of HO-1 counteracted the promotional effects of SIRT4 on ROS accumulation and apoptosis. Mechanistically, SIRT4 modulates ROS and HO-1 expression via accommodating p38-MAPK phosphorylation. By contrast, downregulation of p38-MAPK by SB203580 decreased intracellular ROS level and enhanced the expression of HO-1. Collectively, this work revealed a potential role for SIRT4 in the stimulation of ROS and the modulation of apoptosis. SIRT4/HO-1 may act as a potential therapeutic target, especially in VHL-deficient ccRCCs.


Sujet(s)
Néphrocarcinome/enzymologie , Heme oxygenase-1/métabolisme , Tumeurs du rein/enzymologie , Protéines mitochondriales/métabolisme , Stress oxydatif , Sirtuines/métabolisme , Protéine Von Hippel-Lindau supresseur de tumeur/métabolisme , Apoptose , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Glutamine/déficit , Heme oxygenase-1/génétique , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Protéines mitochondriales/génétique , Phosphorylation , Espèces réactives de l'oxygène/métabolisme , Transduction du signal , Sirtuines/génétique , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , p38 Mitogen-Activated Protein Kinases/métabolisme
9.
Pathol Res Pract ; 223: 153453, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34022680

RÉSUMÉ

OBJECTIVE: Metastatic renal cell carcinoma (mRCC) is the important factor for patient mortality, meanwhile gene mutation constantly changes cancer prognosis in tumor process. Exploring the driver mutation in mRCC process become more and more important. MATERIALS AND METHODS: We obtained the 15 paired primary and metastatic mRCC samples and analyzed specific mutation genes in the metastatic foci (SMGs) by next generation sequencing. Moreover, we explored the Correlated networks, Pathway and Gene Ontology (GO) enrichment results, prediction analysis of AS sites and prognosis of survival. RESULTS: We identify EPCAM, TMEM127, EZH2, EXT1, CDKN2A, PRF1, AIP, CDK4, PRKARIA as SMGs and find that CDKN2A mutation sites affect the prognosis of mRCC by altering splicing elements. Based on the differential analysis for SMGs in KIRC, we found that EPCAM, PRF1 and EZH2 were differential expression in both primary tumors with metastasis compared to primary tumors without metastasis or metastatic tissues. By the AS prediction analysis, we suggest that CDKN2A mutation sites play an important role for RCC metastasis by affecting the p16/p14 expression. CONCLUSIONS: The SMGs could provide new molecular cues associated with tumor metastasis and have potential clinical implications for cancer prognosis and treatment. Definitive conclusions await further validation and follow up.


Sujet(s)
Épissage alternatif , Néphrocarcinome/génétique , Inhibiteur p16 de kinase cycline-dépendante/génétique , Tumeurs du rein/génétique , Mutation , Protéines oncogènes/génétique , Néphrocarcinome/enzymologie , Néphrocarcinome/secondaire , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Bases de données génétiques , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Gènes suppresseurs de tumeur , Humains , Tumeurs du rein/enzymologie , Tumeurs du rein/anatomopathologie , Protéines oncogènes/métabolisme
10.
Int J Cancer ; 148(7): 1743-1755, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33320958

RÉSUMÉ

Metabolism reprograming is a hallmark of cancer and plays an important role in tumor progression. The aberrant metabolism in renal cell carcinoma (RCC) leads to accumulation of the oncometabolite L-2-hydroxyglurate (L-2HG). L-2HG has been reported to inhibit the activity of some α-ketoglutarate-dependent dioxygenases such as TET enzymes, which mediate epigenetic alteration, including DNA and histone demethylation. However, the detailed functions of L-2HG in renal cell carcinoma have not been investigated thoroughly. In our study, we found that L-2HG was significantly elevated in tumor tissues compared to adjacent tissues. Furthermore, we demonstrated that L-2HG promoted vasculogenic mimicry (VM) in renal cancer cell lines through reducing the expression of PHLDB2. A mechanism study revealed that activation of the ERK1/2 pathway was involved in L-2HG-induced VM formation. In conclusion, these findings highlighted the pathogenic link between L-2HG and VM and suggested a novel therapeutic target for RCC.


Sujet(s)
Néphrocarcinome/métabolisme , Protéines de transport/métabolisme , Tumeurs du rein/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Alcohol oxidoreductases , Néphrocarcinome/enzymologie , Néphrocarcinome/génétique , Néphrocarcinome/mortalité , Protéines de transport/génétique , Lignée cellulaire tumorale , Évolution de la maladie , Régulation négative , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/génétique , Humains , Tumeurs du rein/enzymologie , Tumeurs du rein/génétique , Tumeurs du rein/mortalité , Système de signalisation des MAP kinases/génétique , Mâle , Adulte d'âge moyen , Mixed function oxygenases/métabolisme , Protéines proto-oncogènes/métabolisme , Petit ARN interférent , RNA-Seq , Réaction de polymérisation en chaine en temps réel
11.
Cancer Med ; 10(1): 119-134, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33107222

RÉSUMÉ

Resistance to the mechanistic target of rapamycin (mTOR) inhibitors, which are a standard treatment for advanced clear cell renal cell carcinoma (ccRCC), eventually develops in most cases. In this study, we established a patient-derived xenograft (PDX) model which acquired resistance to the mTOR inhibitor temsirolimus, and explored the underlying mechanisms of resistance acquisition. Temsirolimus was administered to PDX model mice, and one cohort of PDX models acquired resistance after repeated passages. PDX tumors were genetically analyzed by whole-exome sequencing and detected several genetic alterations specific to resistant tumors. Among them, mutations in ANKRD12 and DNMT1 were already identified in the early passage of a resistant PDX model, and we focused on a DNMT1 mutation as a potential candidate for developing the resistant phenotype. While DNMT1 expression in temsirolimus-resistant tumors was comparable with the control tumors, DNMT enzyme activity was decreased in resistant tumors compared with controls. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9-mediated heterozygous knockdown of DNMT1 in the temsirolimus-sensitive ccRCC (786-O) cell line was shown to result in a temsirolimus-resistant phenotype in vitro and in vivo. Integrated gene profiles using methylation and microarray analyses of PDX tumors suggested a global shift for the hypomethylation status including promotor regions, and showed the upregulation of several molecules that regulate the mTOR pathway in temsirolimus-resistant tumors. Present study showed the feasibility of PDX model to explore the mechanisms of mTOR resistance acquisition and suggested that genetic alterations, including that of DNMT1, which alter the methylation status in cancer cells, are one of the potential mechanisms of developing resistance to temsirolimus.


Sujet(s)
Antinéoplasiques/pharmacologie , Néphrocarcinome/traitement médicamenteux , DNA (Cytosine-5-)-methyltransferase 1/génétique , Tumeurs du rein/traitement médicamenteux , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Sirolimus/analogues et dérivés , Animaux , Néphrocarcinome/enzymologie , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , Méthylation de l'ADN , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Humains , Tumeurs du rein/enzymologie , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Souris de lignée BALB C , Souris SCID , Mutation , Transplantation tumorale , Transduction du signal , Sirolimus/pharmacologie , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
12.
J Clin Pathol ; 74(4): 216-222, 2021 Apr.
Article de Anglais | MEDLINE | ID: mdl-32467322

RÉSUMÉ

Renal cell carcinoma (RCC) includes diverse tumour types characterised by various genetic abnormalities. The genetic changes, like mutations, deletions and epigenetic alterations, play a crucial role in the modification of signalling networks, tumour pathogenesis and prognosis. The most prevalent RCC type, clear cell RCC (ccRCC), is asymptomatic in the early stages and has a poorer prognosis compared with the papillary and the chromophobe types RCCs. Generally, ccRCC is refractory to chemotherapy and radiation therapy. Loss of von Hippel-Lindau (VHL) gene and upregulation of hypoxia-inducible factors (HIF), the signature of most sporadic ccRCC, promote multiple growth factors. Hence, VHL/HIF and a variety of pathways, including phosphatase and TEnsin homolog on chromosome 10/phosphatidylinositol-3-kinase (PI3K)/AKT, are closely connected and contribute to the ontogeny of ccRCC. In the recent decade, multiple targeting agents have been developed based on blocking major signalling pathways directly or indirectly involved in ccRCC tumour progression, metastasis, angiogenesis and survival. However, most of these drugs have limitations; either metastatic ccRCC develops resistance to these agents, or despite blocking receptors, tumour cells use alternate signalling pathways. This review compiles the state of knowledge about the PI3K/AKT signalling pathway confined to ccRCC and its cross-talks with VHL/HIF pathway.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Marqueurs biologiques tumoraux , Néphrocarcinome/enzymologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Tumeurs du rein/enzymologie , Phosphatidylinositol 3-kinase/métabolisme , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Animaux , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Humains , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Thérapie moléculaire ciblée , Phosphohydrolase PTEN/métabolisme , Inhibiteurs des phosphoinositide-3 kinases/usage thérapeutique , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Résultat thérapeutique , Hypoxie tumorale , Microenvironnement tumoral
13.
Pathol Res Pract ; 216(11): 153227, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-33027752

RÉSUMÉ

Fatty acid synthase (FASN), a key enzyme essential for fatty acid (FA) synthesis, was reportedly implicated in the initiation and progression of various cancers. However, the clinical significance of FASN in renal cell carcinoma (RCC) has not been fully elucidated yet. Here we compare the expression profile and evaluate the prognostic significance of FASN in clear cell RCC (ccRCC) patients. FASN expression was examined in 3 pairs ccRCC and their adjacent nontumor tissues by western blotting (WB) analysis, and its expression was assessed in 145 ccRCC and 13 nontumor tissues by immunohistochemistry (IHC) analysis with tissue microarrays (TMAs). The prognosis of FASN was further investigated in large-scale database using LinkedOmics (n = 537) and The Cancer Protein Atlas (TCPA, n = 445), respectively. WB detected higher FASN expression in ccRCC than normal tissues, then IHC analysis revealed that FASN expression was positively associated with histological grade, pathological stage, tumor size and metastasis status, and negatively associated with cancer-specific survival (CSS). Univariate survival analysis demonstrated that high grade, advanced stage, large tumor, metastasis, and high FASN expression were significantly associated with a shorter CSS, and multivariate analysis revealed tumor grade, stage, metastasis and FASN were identified as independent predictors for CSS in patients with ccRCC. Further LinkedOmics and TCPA analyses confirmed that high FASN expression was correlated with a poorer overall survival (OS) of ccRCC. Collectively, these findings demonstrated FASN could be a poor prognostic factor in ccRCC patients, which indicated that FA synthesis might be implicated in the tumorigenesis and progression of ccRCC.


Sujet(s)
Marqueurs biologiques tumoraux , Néphrocarcinome/enzymologie , Fatty acid synthases/métabolisme , Tumeurs du rein/enzymologie , Rein/enzymologie , Adulte , Sujet âgé , Néphrocarcinome/mortalité , Néphrocarcinome/anatomopathologie , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Immunohistochimie , Rein/anatomopathologie , Tumeurs du rein/mortalité , Tumeurs du rein/anatomopathologie , Mâle , Adulte d'âge moyen , Stadification tumorale , Pronostic , Taux de survie
14.
Med Sci Monit ; 26: e926755, 2020 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-33024069

RÉSUMÉ

BACKGROUND Protein kinase membrane-associated tyrosine/threonine (PKMYT1) has been found in many tumors, but its association with clear cell renal cell carcinoma (ccRCC) remains unclear. MATERIAL AND METHODS PKMYT1 expression in ccRCC was examined in the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Tumor Immune Estimation Resource databases. The correlation between PKMYT1 expression and clinicopathological parameters was explored via the chi-square test. Receiver operating characteristic curves were used to estimate the diagnostic performance of PKMYT1. Kaplan-Meier curves, a Cox model, nomogram, time-dependent receiver operating characteristic curves, and decision curve analysis (DCA) were used to evaluate the prognostic value and clinical utility of PKMYT1. Genes coexpressed with PKMYT1 in ccRCC were identified based on TCGA, the gene expression profiling interactive, and cBioPortal. Gene Set Enrichment Analysis revealed biological pathways associated with PKMYT1 in ccRCC. RESULTS Weighted gene coexpression network analysis identified PKMYT1 as one of the genes most significantly correlated with progression of histological grade. PKMYT1 was significantly upregulated in ccRCC compared with normal tissue (P<0.001), with a trend toward differentiating between individuals with ccRCC and those who were healthy (area under the curve=0.942). High PKMYT1 expression was correlated with unsatisfactory survival (hazard ratio=1.67, P=0.001), indicating that it is a risk factor for ccRCC. A nomogram incorporating PKMYT1 level was created and showed a clinical net benefit. PKMYT1 was strongly positively correlated with the anti-silencing function of 1B histone chaperone (ASF1B) gene in ccRCC. CONCLUSIONS PKMYT1 is upregulated in ccRCC and its presence indicates poor prognosis, making it a potential therapeutic target for ccRCC.


Sujet(s)
Néphrocarcinome , Bases de données d'acides nucléiques , Régulation de l'expression des gènes codant pour des enzymes , Régulation de l'expression des gènes tumoraux , Tumeurs du rein , Protéines membranaires/biosynthèse , Protéines tumorales/biosynthèse , Protein-Serine-Threonine Kinases/biosynthèse , Protein-tyrosine kinases/biosynthèse , Néphrocarcinome/diagnostic , Néphrocarcinome/enzymologie , Femelle , Humains , Tumeurs du rein/diagnostic , Tumeurs du rein/enzymologie , Mâle , Adulte d'âge moyen , Pronostic
15.
Ann Diagn Pathol ; 49: 151599, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-32977234

RÉSUMÉ

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) has been incorporated into the recent international histological classification of renal tumors. However, to date, there are limited studies describing the clinicopathological features of fumarate hydratase (FH)-deficient RCC, including the hereditary (HLRCC) and sporadic forms. Herein, we present a clinicopathological study of seven cases with FH-deficient RCC. The age of patients ranged from 26 to 70 years with mean and median age of 51.7 and 57 years, respectively. The follow-up data of all patients were available. One patient was alive without the disease and five patients were alive with active disease. One patient died of the disease. Family history of RCC, or skin or uterine smooth muscle tumor within second degree of kinship was present in four of seven patients. Metastasis was observed in all tumors. Metastatic sites included bone, lungs, liver, peritoneum, ovaries, tonsils, or lymph nodes. Grossly, the cut surface of the tumor usually showed light brown, brown, or whitish color. Microscopically, the cytoplasm of the tumor cells was predominantly eosinophilic and all tumors displayed various architectural patterns such as papillary, tubular, solid, or microcystic patterns. Furthermore, two tumors demonstrated a tubulocystic pattern. Sarcomatoid change and rhabdoid features were seen in five tumors and two tumors, respectively. Large cytomegaloviral (CMV) inclusion-like eosinophilic nucleoli surrounded by a clear halo were identified in all tumors. All tumors showed negative immunohistochemical reaction for FH protein. False positive results of TFE3 protein were observed in three tumors. Furthermore, a germline mutation of FH gene was identified in one patient with family history of the disease. In conclusion, FH-deficient RCC includes hereditary and sporadic forms. Grossly, this tumor is solitary and occurs unilaterally. Histologically, the tumor is characterized by various patterns such as papillary, tubular, solid, tubulocystic, or microcystic, has eosinophilic cytoplasm and CMV-like high-grade nuclei. FH-deficient RCCs frequently metastasize to other anatomic sites. TFE immunoreactivity may occur in some FH-deficient RCCs, and immunohistochemistry can accurately diagnose these tumors and mutational analysis of FH gene.


Sujet(s)
Néphrocarcinome/anatomopathologie , Fumarate hydratase/déficit , Tumeurs du rein/anatomopathologie , Adulte , Sujet âgé , Néphrocarcinome/enzymologie , Femelle , Humains , Tumeurs du rein/enzymologie , Léiomyomatose/anatomopathologie , Mâle , Adulte d'âge moyen , Syndromes néoplasiques héréditaires/anatomopathologie , Tumeurs cutanées/anatomopathologie , Tumeurs de l'utérus/anatomopathologie
16.
Cancer Med ; 9(22): 8624-8638, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32970930

RÉSUMÉ

BACKGROUNDS: Kidney renal clear cell carcinoma (KIRC) is a major pathological type of renal cell carcinoma (RCC), and the prognosis of advanced KIRC patients is often unsatisfactory. Some lysine oxidase (LOX) family genes have been proven to be upregulated in some malignancies and play pivotal roles in the carcinogenesis. However, their roles in KIRC remain unclear. MATERIALS AND METHODS: Here, we used some online databases (eg, ONCOMINE, GEPIA, UALCAN, c-BioPortal, Human Protein Altas) to comprehensively explored the expression levels and the prognostic values of LOX family genes in KIRC using bioinformatic methods. RESULTS: The results revealed that lysyl oxidase (LOX) and lysyl oxidase-like 2 (LOXL2) were significantly overexpressed in KIRC at the level of mRNA expression, protein expression, and RCC cell lines. Further analysis demonstrated that higher mRNA expression of LOX and LOXL2 were significantly correlated with poor survival, tumor grade, individual cancer stages, and nodal metastasis status. DNA copy number amplifications and mRNA upregulation, DNA deep deletion, and mRNA upregulation were the main genetic mutations of LOX and LOXL2, respectively. Prognostic analysis showed that the altered group had significantly poorer overall survival (OS) compared to the unaltered group (p = .0387). Co-expression analysis showed CP, PLOD2, and COL5A1 were significantly correlated with LOX, and COL1A2 was positively correlated with LOXL2. Further analysis confirmed that these co-expressed genes were significantly upregulated and predicted unfavorable prognosis in KIRC. CONCLUSION: Multi-level analysis demonstrated that LOX and LOXL2 were significantly upregulated and predicted poor survival in KIRC, which may apply as promising biomarkers for diagnosis and therapy of KIRC in the future.


Sujet(s)
Amino-acid oxidoreductases/génétique , Marqueurs biologiques tumoraux/génétique , Néphrocarcinome/génétique , Tumeurs du rein/enzymologie , Mixed function oxygenases/génétique , Amino-acid oxidoreductases/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Néphrocarcinome/enzymologie , Néphrocarcinome/mortalité , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Bases de données d'acides nucléiques , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du rein/génétique , Tumeurs du rein/mortalité , Tumeurs du rein/anatomopathologie , Mixed function oxygenases/métabolisme , Mutation , Pronostic , Cartes d'interactions protéiques , Transduction du signal , Régulation positive
17.
Oncology ; 98(10): 734-742, 2020.
Article de Anglais | MEDLINE | ID: mdl-32726790

RÉSUMÉ

BACKGROUND: There has been no clinically useful diagnostic or prognostic biomarker for renal cell carcinoma (RCC). Serum γ-glutamyltransferase (GGT) activity has been reported to be a prognostic marker for several types of cancer including RCC. Exosomes or small extracellular vesicles present in body fluids have potential as a biomarker. We have recently demonstrated that GGT activity on exosomes isolated from serum is useful for the differential diagnosis of prostate cancer and benign prostate hyperplasia. In this study, we aimed to examine if serum exosomal GGT activity could be a marker for RCC. METHODS: We examined GGT1 expression and GGT activity in cell lysates and exosomes from culture medium of HK-2 proximal tubule epithelial and RCC cell lines. GGT activity was measured using a fluorescent probe for GGT, γ-glutamyl hydroxymethyl rhodamine green. Serum and serum exosomal GGT activities were measured in patients with RCC. GGT1 expression in RCC tissues was evaluated by immunohistochemical staining. RESULTS: GGT1 levels in exosomes from KMRC-1, OS-RC-2 and 786-O cells were elevated compared with those from HK-2 cells. In exosomes, GGT1 expression correlated with GGT activity determined using a fluorescent probe for GGT. In RCC patients, serum exosomal GGT activity was elevated in those with advanced stages (III/IV vs. I/II, p = 0.037) and those with microvascular invasion (with vs. without, p = 0.034). Immunohistochemical analysis showed that membranous GGT1 expression was increased in RCC with microvascular invasion. Notably, preoperative serum exosomal GGT activity could predict the likelihood of having microvascular invasion diagnosed by pathological examination of surgically resected specimens. CONCLUSIONS: Our results suggest that serum exosomal GGT activity could be a clinically useful marker for advanced clinicopathological features of RCC patients, and its combined use with conventional diagnostic modalities may improve the diagnosis and treatment of patients.


Sujet(s)
Néphrocarcinome/enzymologie , Exosomes/enzymologie , Tumeurs du rein/enzymologie , gamma-Glutamyltransferase/sang , Sujet âgé , Néphrocarcinome/sang , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Femelle , Humains , Immunohistochimie , Tumeurs du rein/sang , Tumeurs du rein/anatomopathologie , Mâle , Adulte d'âge moyen , Métastase tumorale , Stadification tumorale , gamma-Glutamyltransferase/biosynthèse
18.
Histopathology ; 77(4): 659-666, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32639054

RÉSUMÉ

AIMS: Renal epithelial neoplasms (RENs) can be difficult to subclassify, owing to overlapping morphological features. Carbonic anhydrase 9 (CA9) is a common biomarker for clear cell renal cell carcinoma (CCRCC); however, the sensitivity and specificity across REN subtypes are less clear. The aim of this study was to investigate CA9 expression in RENs, especially those in the differential diagnosis with CCRCC and less common entities, to determine its reliability as a diagnostic biomarker. METHODS AND RESULTS: CA9 immunostaining was performed on 262 RENs, including 119 CCRCCs and 143 non-CCRCC. Immunostaining was evaluated as negative (0%), rare (1+, 1-10%), focal (2+, 11-50%), or diffuse (3+, >50%). CCRCCs were 3+ CA9-positive in 93% of cases; 4% were CA9-negative. Sixty-seven percent of papillary renal cell carcinomas (RCCs) were 1+/2+ CA9-positive, whereas 33% were CA9-negative. Chromophobe RCCs were nearly always CA9-negative (93%), with 7% showing rare cell reactivity. Clear cell tubulopapillary RCCs (CCTPRCCs) were consistently 3+ CA9-positive, but with a cup-like staining pattern. Fifty-three percent of Xp11.2 RCCs were CA9-negative; however, 6% were 3+ CA9-positive and 12% were 2+ CA9-positive. Two of eight fumarate hydratase-deficient RCCs were 3+ CA9-positive. A small subset of the remaining RCCs showed rare to focal CA9 expression. All oncocytomas and eosinophilic solid and cystic RCCs were CA9-negative. CONCLUSIONS: Overall, diffuse CA9 expression was identified in nearly all CCRCCs and in all CCTPRCCs (high sensitivity); however, CA9 was not entirely specific. At least focal CA9 expression can been seen in a subset of many RCCs, and such findings should be taken into consideration with other morphological, immunophenotypic and clinical findings.


Sujet(s)
Antigènes néoplasiques/analyse , Antigènes néoplasiques/biosynthèse , Marqueurs biologiques tumoraux/métabolisme , Carbonic anhydrase IX/analyse , Carbonic anhydrase IX/biosynthèse , Néphrocarcinome/enzymologie , Tumeurs du rein/enzymologie , Marqueurs biologiques tumoraux/analyse , Néphrocarcinome/classification , Néphrocarcinome/anatomopathologie , Humains , Tumeurs du rein/classification , Tumeurs du rein/anatomopathologie , Sensibilité et spécificité
19.
Transl Res ; 223: 1-14, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32492552

RÉSUMÉ

Nuclear NADPH oxidase-4 (Nox4) is a key component of metabolic reprogramming and is often overexpressed in renal cell carcinoma (RCC). However, its prognostic role in RCC remains unclear. Here we examined the significance of nuclear Nox4 on disease progression and development of drug resistance in advanced RCC. We analyzed human RCC tissue from multiple regions in the primary index tumor, cancer-associated normal adjacent parenchyma, intravascular tumor in locally advanced cancer patients. We found that the higher nuclear Nox4 expression was significantly associated with progression and death. These findings were consistent after controlling for other competing clinical variables. In contrast, patients with lower nuclear Nox4, even in higher stage RCC had better prognosis. We identified a subset of patients with high nuclear Nox4 who had rapid disease progression or died within 6 months of surgery. In addition, higher nuclear Nox4 level correlated with resistance to targeted therapy and immunotherapy. Western blotting performed on fresh human RCC tissue as well as cell-lines revealed increased nuclear Nox4 expression. Our data support an important prognostic role of Nox4 mediated regulation of RCC independent of other competing variables. Nox4 localizes to the nucleus in high-grade, high-stage RCC. Higher nuclear Nox4 has prognostic significance for disease progression, poor survival, and development of drug resistance in RCC.


Sujet(s)
Néphrocarcinome/enzymologie , Néphrocarcinome/anatomopathologie , Noyau de la cellule/enzymologie , Évolution de la maladie , Tumeurs du rein/enzymologie , Tumeurs du rein/anatomopathologie , NADPH Oxidase 4/métabolisme , Lignée cellulaire tumorale , Femelle , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Analyse multifactorielle , Survie sans progression
20.
Biomed Pharmacother ; 128: 110323, 2020 Aug.
Article de Anglais | MEDLINE | ID: mdl-32526457

RÉSUMÉ

Clear cell renal cell carcinoma (ccRCC), the most frequent subtype of renal cell carcinoma (RCC), is characterized by high relapse rate and mortality. Long non-coding RNAs (lncRNAs) are critical participants during cancer development. LncRNA DARS antisense RNA 1 (DARS-AS1), a newly-found lncRNA, is not specifically reported in ccRCC. However, Gene Expression Profiling Interactive Analysis (GEPIA) and starBase databases revealed the up-regulation of DARS-AS1 in ccRCC. Current study investigated the function and mechanism of DARS-AS1 in ccRCC. Functional assays including colony formation assay, EdU assay, caspase-3 activity detection, flow cytometry analysis and JC-1 assay were implemented to identify the role of DARS-AS1 in ccRCC. As a result, silencing of DARS-AS1 retarded proliferation and facilitated apoptosis in ccRCC cells. Moreover, mainly a cytoplasmic localization of lncRNA DARS-AS1 was verified in ccRCC cells. Then, we demonstrated that DARS-AS1 positively regulated its nearby gene, aspartyl-tRNA synthetase (DARS), by sequestering miR-194-5p. Moreover, DARS was testified as the oncogene in ccRCC and DARS-AS1 worked as a tumor-facilitator in ccRCC through miR-194-5p/DARS signaling. In a summary, this study firstly uncovered that DARS-AS1 boosted DARS expression via absorbing miR-194-5p, therefore contributing to malignancy in ccRCC. Our findings may be helpful for opening new strategies for ccRCC treatment.


Sujet(s)
Aspartate-tRNA ligase/métabolisme , Néphrocarcinome/enzymologie , Tumeurs du rein/enzymologie , microARN/métabolisme , ARN long non codant/métabolisme , Apoptose , Aspartate-tRNA ligase/génétique , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Lignée cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , microARN/génétique , ARN long non codant/génétique , Transduction du signal , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE