Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Surg Oncol ; 50(9): 108475, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38870872

ABSTRACT

INTRODUCTION: Epithelial ovarian cancer (EOC) is the most prevalent type of ovarian cancer, yet the impact of ovarian laterality has received limited attention. MATERIALS AND METHODS: We conducted a comprehensive investigation into the impact of laterality (left-right and bilateral-unilateral) on EOC incidence and prognosis, focusing on distinct subtypes. Binomial tests and Pearson's χ2 tests were employed to compare occurrence rates among laterality groups. Cox regression analyses were used to create a proportional hazards model for tumor prognosis. Nomograms were developed and validated, including internal validation via bootstrapping. RESULTS: Our study encompassed 20,790 EOC patients, revealing disparities in incidence and prognosis between unilateral and bilateral cases. Unilateral tumor development was notably predominant in clear cell, endometrioid, brenner, and mucinous subtypes, while bilateral involvement was more frequent in serous ovarian cancer. Laterality differences, reflecting disparities between the left and right sides, were chiefly evident in the incidence rates across various stages and in the prognosis of specific subtypes. Notably, mucinous ovarian cancer exhibited significantly better prognosis on the right side compared to the left (right tumors: HR = 0.745, p = 0.015, CI: 0.587-0.945). CONCLUSION: These findings emphasize the importance of considering ovarian laterality -both left-right and bilateral-unilateral aspects -as a critical factor influencing EOC incidence and prognosis, necessitating attention in clinical practice.

2.
Eur Radiol Exp ; 7(1): 74, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38019353

ABSTRACT

BACKGROUND: We tested the hypothesis that radiofrequency ablation (RFA) for hepatocellular carcinoma (HCC) promotes tumor cell release and explored a method for reducing these effects. METHODS: A green fluorescent protein-transfected orthotopic HCC model was established in 99 nude mice. In vivo flow cytometry was used to monitor circulating tumor cell (CTC) dynamics. Pulmonary fluorescence imaging and pathology were performed to investigate lung metastases. First, the kinetics of CTCs during the periablation period and the survival rate of CTCs released during RFA were investigated. Next, mice were allocated to controls, sham ablation, or RFA with/without hepatic vessel blocking (ligation of the portal triads) for evaluating the postablation CTC level, lung metastases, and survival over time. Moreover, the kinetics of CTCs, lung metastases, and mice survival were evaluated for RFA with/without ethanol injection. Pathological changes in tumors and surrounding parenchyma after ethanol injection were noted. Statistical analysis included t-test, ANOVA, and Kaplan-Meier survival curves. RESULTS: CTC counts were 12.3-fold increased during RFA, and 73.7% of RFA-induced CTCs were viable. Pre-RFA hepatic vessel blocking prevented the increase of peripheral CTCs, reduced the number of lung metastases, and prolonged survival (all p ≤ 0.05). Similarly, pre-RFA ethanol injection remarkably decreased CTC release during RFA and further decreased lung metastases with extended survival (all p ≤ 0.05). Histopathology revealed thrombus formation in blood vessels after ethanol injection, which may clog tumor cell dissemination during RFA. CONCLUSION: RFA induces viable tumor cell dissemination, and pre-RFA ethanol injection may provide a prophylactic strategy to reduce this underestimated effect. RELEVANCE STATEMENT: RFA for HCC promotes viable tumor cell release during ablation, while ethanol injection can prevent RFA induced tumor cell release. KEY POINTS: • RFA induced the release of viable tumor cells during the ablation procedure in an animal model. • Hepatic vessel blocking can suppress tumor cells dissemination during RFA. • Ethanol injection can prevent RFA-induced tumor cell release, presumably because of the formation of thrombosis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/surgery , Mice, Nude , Liver Neoplasms/surgery , Disease Models, Animal , Ethanol
3.
BMC Urol ; 23(1): 32, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36871009

ABSTRACT

BACKGROUND: Primary retroperitoneal mucinous cystic tumours with borderline malignancy (PRMC-BM) are rare and difficult to diagnose preoperatively. We are the first to report two cases of PRMC-BM which mimic a duplex kidney and evaluate the outcomes of different surgical procedures. CASE PRESENTATION: We describe two cases of retroperitoneal cystic tumours. Both were diagnosed with duplex kidney with hydronephrosis on computed tomography scan. The first patient underwent robot-assisted laparoscopic surgery and was found to have a retroperitoneal cystic tumour. The other patient underwent an ultrasound-guided puncture before surgery and was diagnosed with retroperitoneal lymphangioma. Retroperitoneal cystectomy was performed using an open transperitoneal procedure. The final pathologic diagnosis in both cases implies PRMC-BM. The open surgical approach was associated with a shorter operation time, less intraoperative blood loss, and protected cyst wall integrity by comparing the different surgical approaches. During follow-up, the patient in the first case had tumour recurrence six months post-surgery, and the other patient was healthy without recurrence or metastasis 12 months post-surgery. CONCLUSIONS: Primary retroperitoneal mucinous cystic tumours with borderline malignancy can be enclosed within the kidney and misdiagnosed as other cystic diseases of the urinary system. Thus, an open surgical approach may be more suitable for this type of tumour.


Subject(s)
Cysts , Hydronephrosis , Retroperitoneal Neoplasms , Humans , Neoplasm Recurrence, Local , Kidney
4.
BMC Med Imaging ; 20(1): 46, 2020 05 03.
Article in English | MEDLINE | ID: mdl-32362278

ABSTRACT

BACKGROUND: Large cell neuroendocrine carcinoma (LCNEC) of the urinary bladder is an uncommon malignant bladder tumor, and the overall prognosis is poor. Contrast-enhanced ultrasound (CEUS) provides a new effective modality for tumor detection and diagnosis. CASE PRESENTATION: A 30-year-old man complained of repeated painless gross haematuria for half a month. Conventional ultrasound demonstrated a hypoechoic solitary lesion with hyperechoic margins measuring 3.4 × 3.1 cm in the anterior wall of the bladder. Superb microvascular imaging (SMI) showed a strong flow signal in the mass. CEUS revealed that the lesion was characterized by hyper-enhancement in the early phase and hypo-enhancement in the late phase. The entire bladder wall was disrupted by homogeneous hyper-enhanced tumor tissue on CEUS. Time-intensity curves (TICs) showed a rapid wash-in with a high maximum signal intensity (SI) and quick wash-out. Finally, partial cystectomy was performed and the pathological examination confirmed the diagnosis of LCNEC with invasion into the whole layer of the bladder wall. CONCLUSION: This case suggested that CEUS was a valuable imaging method to detect and diagnose LCNEC in the bladder, and that CEUS can provide information related to the depth of wall invasion and the microvasculature.


Subject(s)
Carcinoma, Large Cell/diagnostic imaging , Carcinoma, Neuroendocrine/diagnostic imaging , Urinary Bladder Neoplasms/diagnostic imaging , Adult , Carcinoma, Large Cell/blood supply , Carcinoma, Large Cell/complications , Carcinoma, Large Cell/surgery , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/complications , Carcinoma, Neuroendocrine/surgery , Contrast Media/administration & dosage , Cystectomy , Hematuria/etiology , Humans , Male , Microvessels/diagnostic imaging , Microvessels/pathology , Ultrasonography , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/complications , Urinary Bladder Neoplasms/surgery
5.
Signal Transduct Target Ther ; 5(1): 42, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32327643

ABSTRACT

Tumor necrosis factor alpha-induced protein 1 (TNFAIP1) modulates a plethora of important biological processes, including tumorigenesis and cancer cell migration. However, the regulatory mechanism of TNFAIP1 degradation remains largely elusive. In the present study, with a label-free quantitative proteomic approach, TNFAIP1 was identified as a novel ubiquitin target of the Cullin-RING E3 ubiquitin ligase (CRL) complex. More importantly, Cul3-ROC1 (CRL3), a subfamily of CRLs, was identified to specifically interact with TNFAIP1 and promote its polyubiquitination and degradation. Mechanistically, BTBD9, a specific adaptor component of CRL3 complex, was further defined to bind and promote the ubiquitination and degradation of TNFAIP1 in cells. As such, downregulation of BTBD9 promoted lung cancer cell migration by upregulating the expression of TNFAIP1, whereas TNFAIP1 deletion abrogated this effect. Finally, bioinformatics and clinical sample analyses revealed that BTBD9 was downregulated while TNFAIP1 was overexpressed in human lung cancer, which was associated with poor overall survival of patients. Taken together, these findings reveal a previously unrecognized mechanism by which the CRL3BTBD9 ubiquitin ligase controls TNFAIP1 degradation to regulate cancer cell migration.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , Neoplasms/genetics , Nerve Tissue Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Humans , Multiprotein Complexes , Neoplasms/pathology , Proteolysis , Proteomics , Ubiquitination/genetics
6.
Biomed Res Int ; 2020: 5264265, 2020.
Article in English | MEDLINE | ID: mdl-32280689

ABSTRACT

Osteosarcoma (OS) is the most common primary bone malignancy. Our previous study revealed an association between the level of epidermal growth factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1) and the invasion, metastasis, and poor prognosis of OS. However, the exact correlation between the serum EFEMP1 level and OS diagnosis and progression was unclear. This study is aimed at determining the value of the serum EFEMP1 level in the diagnosis and prognosis of OS. Fifty-one consecutive OS patients were prospectively registered in this study. The serum EFEMP1 levels were measured using ELISA at diagnosis, after neoadjuvant chemotherapy, and before and after surgical treatment. Sixty-nine healthy subjects in the control group, nine patients with chondrosarcoma, and 12 patients with giant cell tumor of the bone were also enrolled in this study. Surgical orthotopic implantation was used to generate a mouse OS model, and the correlation between the circulating EFEMP1 levels and tumor progression was examined. Then, OS patients had significantly higher mean serum EFEMP1 levels (7.61 ng/ml) than the control subjects (1.47 ng/ml). The serum EFEMP1 levels were correlated with the Enneking staging system (r = 0.32, P = 0.021) and lung metastasis (r = 0.50, P < 0.001). There was also a correlation between the serum EFEMP1 level and EFEMP1 expression in the respective OS samples (r = 0.49, P < 0.001). Additionally, patients with either chondrosarcoma or giant cell tumor of the bone had significantly higher serum EFEMP1 levels than OS patients. Surgical and chemotherapeutic treatment led to an increase in the serum EFEMP1 levels. Then, the destruction of bone tissues might be one of the factors about the EFEMP1 levels. In the mouse OS model, the serum EFEMP1 level was correlated with tumor progression. Our results suggested that serum EFEMP1 levels might be used to distinguish OS patients from healthy controls and as an indicator for OS lung metastasis. Serum EFEMP1 levels could serve as a new and assisted biomarker for the auxiliary diagnosis and prognosis of OS.


Subject(s)
Biomarkers, Tumor/blood , Extracellular Matrix Proteins/blood , Extracellular Matrix Proteins/metabolism , Osteosarcoma/diagnosis , Osteosarcoma/metabolism , Adolescent , Adult , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Lung , Lung Neoplasms/diagnosis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Osteosarcoma/pathology , Patients , Prognosis , Young Adult
7.
EBioMedicine ; 45: 81-91, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31208947

ABSTRACT

BACKGROUND: The neddylation pathway is overactivated in human cancers. Inhibition of neddylation pathway has emerged as an attractive anticancer strategy. The mechanisms underlying neddylation overactivation in cancer remain elusive. MLN4924/Pevonedistat, a first-in-class NEDD8-activating enzyme (NAE, E1) inhibitor, exerts significant anti-tumor effects, but its mutagenic resistance remains unresolved. METHODS: The expression of NEDD8-conjugating enzyme UBC12/UBE2M (E2) and NEDD8 were estimated by bioinformatics analysis and western blot in human lung cancer cell lines. The malignant phenotypes of lung cancer cells were evaluated both in vitro and in vivo upon UBC12 knockdown. Cell-cycle arrest was evaluated by quantitative proteomic analysis and propidium iodide stain and fluorescence - activated cell sorting (FACS). The growth of MLN4924 - resistant H1299 cells was also evaluated upon UBC12 knockdown. FINDINGS: The mRNA level of UBC12 in lung cancer tissues was much higher than that in normal lung tissues, increased with disease deterioration, and positively correlated with NEDD8 expression. Moreover, the overexpression of UBC12 significantly enhanced protein neddylation modification whereas the downregulation of UBC12 reduced neddylation modification of target proteins. Functionally, neddylation inactivation by UBC12 knockdown suppressed the malignant phenotypes of lung cancer cells both in vitro and in vivo. The quantitative proteomic analysis and cell cycle profiling showed that UBC12 knockdown disturbed cell cycle progression by triggering G2 phase cell-cycle arrest. Further mechanistical studies revealed that UBC12 knockdown inhibited Cullin neddylation, led to the inactivation of CRL E3 ligases and induced the accumulation of tumor-suppressive CRL substrates (p21, p27 and Wee1) to induce cell cycle arrest and suppress the malignant phenotypes of lung cancer cells. Finally, UBC12 knockdown effectively inhibited the growth of MLN4924-resistant lung cancer cells. INTERPRETATION: These findings highlight a crucial role of UBC12 in fine-tuned regulation of neddylation activation status and validate UBC12 as an attractive alternative anticancer target against neddylation pathway. FUND: Chinese Minister of Science and Technology grant (2016YFA0501800), National Natural Science Foundation of China (Grant Nos. 81401893, 81625018, 81820108022, 81772470, 81572340 and 81602072), Innovation Program of Shanghai Municipal Education Commission (2019-01-07-00-10-E00056), Program of Shanghai Academic/Technology Research Leader (18XD1403800), National Thirteenth Five-Year Science and Technology Major Special Project for New Drug and Development (2017ZX09304001). The funders had no role in study design, data collection, data analysis, interpretation, writing of the report.


Subject(s)
Lung Neoplasms/genetics , NEDD8 Protein/genetics , Proteomics , Ubiquitin-Conjugating Enzymes/genetics , A549 Cells , Animals , Apoptosis/genetics , Cell Proliferation/genetics , China/epidemiology , Cyclopentanes/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Molecular Targeted Therapy , NEDD8 Protein/antagonists & inhibitors , Pyrimidines/pharmacology , Signal Transduction/drug effects , Ubiquitins/genetics
8.
Cell Prolif ; 52(2): e12536, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30341788

ABSTRACT

OBJECTIVES: The present study aimed to reveal expression status of the neddylation enzymes in HNSCC and to elucidate the anticancer efficacy and the underlying mechanisms of inhibiting neddylation pathway. MATERIALS AND METHODS: The expression levels of neddylation enzymes were estimated by Western blotting in human HNSCC specimens and bioinformatics analysis of the cancer genome atlas (TCGA) database. Cell apoptosis was evaluated by Annexin V fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) stain and fluorescence-activated cell sorting (FACS). Small interfering RNA (siRNA) and the CRISPR-Cas9 system were used to elucidate the underlying molecular mechanism of MLN4924-induced HNSCC apoptosis. RESULTS: Expression levels of NAE1 and UBC12 were prominently higher in HNSCC tissues than that in normal tissues. Inactivation of the neddylation pathway significantly inhibited malignant phenotypes of HNSCC cells. Mechanistic studies revealed that MLN4924 induced the accumulation of CRL ligase substrate c-Myc that transcriptionally activated pro-apoptotic protein Noxa, which triggered apoptosis in HNSCC. CONCLUSIONS: These findings determined the over-expression levels of neddylation enzymes in HNSCC and revealed novel mechanisms underlying neddylation inhibition induced growth suppression in HNSCC cells, which provided preclinical evidence for further clinical evaluation of neddylation inhibitors (eg, MLN4924) for the treatment of HNSCC.


Subject(s)
Apoptosis/drug effects , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Head and Neck Neoplasms/drug therapy , NEDD8 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , NEDD8 Protein/agonists , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
9.
Hum Pathol ; 79: 135-143, 2018 09.
Article in English | MEDLINE | ID: mdl-29879402

ABSTRACT

Stress-induced phosphoprotein 1 (STIP1) is an adaptor protein that bridges HSP70 and HSP90 folding and a secretory protein that regulates malignant tumor progression. The aim of the present study was to demonstrate the clinicopathological significance and prognostic role of STIP1 in colorectal cancer (CRC). We used data from The Cancer Genome Atlas (TCGA) to analyze STIP1 expression in CRC and utilized 8 pairs of fresh-frozen tissue samples to investigate STIP1 expression in CRC tissues and adjacent normal tissues using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays. We also used immunohistochemical staining to detect STIP1 expression in 144 formalin-fixed, paraffin-embedded (FFPE) CRC tissue samples and determine the clinical significance of STIP1 expression in CRC. The results of bioinformatics analysis, qRT-PCR, and Western blot showed that STIP1 expression was higher in CRC tissues than in adjacent normal tissues. High STIP1 expression was significantly correlated with advanced T stage (P = .01), N stage (P = .001), M stage (P < .001), and TNM stage (P < .001). Moreover, Kaplan-Meier analyses indicated that higher STIP1 expression predicted a worse prognosis in patients with CRC, and Cox regression analysis revealed that STIP1 was an independent prognostic factor for overall survival and disease-free survival in patients with CRC. In conclusion, our results suggest that STIP1 acts as an oncogene in CRC and can therefore serve as a biomarker for the prognosis of patients with CRC.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Heat-Shock Proteins/analysis , Biomarkers, Tumor/genetics , Blotting, Western , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease-Free Survival , Female , Heat-Shock Proteins/genetics , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Real-Time Polymerase Chain Reaction , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...