Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 302
Filter
1.
Virchows Arch ; 484(5): 827-836, 2024 May.
Article in English | MEDLINE | ID: mdl-38561462

ABSTRACT

Urothelial carcinoma (UC) of the bladder is a common cause of cancer-related death worldwide. Vasculogenic mimicry (VM) is a process by which the malignant cells can generate vascular-like structures formed of periodic acid-Schiff (PAS) positive/CD31 negative extracellular matrix independent of angiogenesis and thus promotes tumor progression. N-myc downstream-regulated gene 1 (NDRG1) is a protein that can modulate tumor angiogenesis; however, its role in regulating tumor angiogenesis and VM formation has not been previously investigated in UC. This study aims to evaluate the role of intra-tumor microvessel density (MVD) (as a surrogate measure of angiogenesis), VM, and NDRG1 in UC and their correlation with different clinicopathologic features, then assess the correlation between them in UC. Sixty specimens of UC of the bladder were included. PAS-CD31 immunohistochemical double staining method was used to evaluate the intra-tumor MVD and VM. Immunohistochemical expression of NDRG1 was also examined. VM and NDRG1 expression were detected in 41.7% and 83.3% of UC specimens respectively. The mean of intra-tumor MVD, VM area, and NDRG1 was significantly higher in tumors with higher grade, lymphovascular invasion, and higher T stage. NDRG1 expression was positively correlated with MVD and VM. We can suggest that MVD, VM, and NDRG1 may serve as poor prognostic markers for UC. The positive correlation between NDRG1 and both MVD and VM may provide the first evidence that NDRG1 can induce tumor angiogenesis and VM in UC which may offer a novel pathway for further therapeutic strategies.


Subject(s)
Cell Cycle Proteins , Intracellular Signaling Peptides and Proteins , Microvascular Density , Neovascularization, Pathologic , Urinary Bladder Neoplasms , Humans , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism , Male , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/metabolism , Middle Aged , Female , Aged , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Adult , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/blood supply , Aged, 80 and over , Immunohistochemistry , Urothelium/pathology , Angiogenesis
2.
Biosensors (Basel) ; 12(6)2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35735546

ABSTRACT

Laser speckle contrast imaging (LSCI) is a powerful visualization tool for quantifying blood flow in tissues, providing simplicity of configuration, ease of use, and intuitive results. With recent advancements, smartphone and camera technologies are suitable for the development of smartphone-based LSCI applications for point-of-care (POC) diagnosis. A smartphone-based portable LSCI endoscope system was validated for POC diagnosis of vascular disorders. The endoscope consisted of compact LED and laser illumination, imaging optics, and a flexible fiberscope assembled in a 3D-printed hand-held cartridge for access to body cavities and organs. A smartphone's rear camera was mounted thereto, enabling endoscopy, LSCI image acquisition, and processing. Blood flow imaging was calibrated in a perfused tissue phantom consisting of a microparticle solution pumped at known rates through tissue-mimicking gel and validated in a live rat model of BBN-induced bladder cancer. Raw LSCI images successfully visualized phantom flow: speckle flow index showed linearity with the pump flow rate. In the rat model, healthy and cancerous bladders were distinguishable in structure and vasculature. The smartphone-based low-cost portable mobile endoscope for monitoring blood flow and perfusion shows promise for preclinical applications and may be suitable for primary diagnosis at home or as a cost-effective POC testing assay.


Subject(s)
Laser Speckle Contrast Imaging , Point-of-Care Systems , Regional Blood Flow/physiology , Smartphone , Vascular Diseases/diagnosis , Animals , Endoscopes , Phantoms, Imaging , Rats , Urinary Bladder Neoplasms/blood supply , Vascular Diseases/physiopathology
3.
Comput Math Methods Med ; 2021: 1047948, 2021.
Article in English | MEDLINE | ID: mdl-34675991

ABSTRACT

OBJECTIVE: To study the clinical diagnostic value of contrast-enhanced ultrasound (CEUS) in bladder occupied lesions. METHODS: 38 cases of conventional-ultrasound-found bladder occupied lesions did color Doppler flow imaging (CDFI) and CEUS checks. By comparing the difference between two types of blood flow imaging technologies in displaying the flow of bladder occupied lesions and observing the perfusion modes of contrast agents to enter lesions, the perfusion characteristics of CEUS were analyzed. Finally, they were contrasted with the surgical pathology results. RESULTS: Of all the 38 cases, there were 51 bladder occupied lesions, including 43 bladder malignant tumors, 2 bladder inverted papillomas, and 6 glandular cystitis lesions. The blood flow display rate of bladder occupied lesions was 100% using CEUS. Apparently, it was higher than that of CDFI (62.7%), and the result of these showed a statistically significant difference (P < 0.05). Using CEUS, 46 malignant lesions and 5 glandular cystitis lesions were indicated, and the diagnostic accuracy rate was 86.3%. CONCLUSION: CEUS can improve the blood flow display rate of bladder occupied lesions, and it can also observe the real-time blood flow of these lesions. It can help judge their nature and has a higher clinical value in differentiating the benign from the malignant.


Subject(s)
Ultrasonography, Doppler, Color/methods , Ultrasonography/methods , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/diagnostic imaging , Adult , Aged , Aged, 80 and over , Computational Biology , Contrast Media , Cystitis/diagnostic imaging , Diagnosis, Computer-Assisted , Diagnosis, Differential , Diagnostic Errors , Female , Humans , Male , Middle Aged , Retrospective Studies
4.
Cancer Lett ; 523: 10-28, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34597712

ABSTRACT

Tumour angiogenesis is an independent risk factor for bladder cancer (BCa) progression, but viable and promising antiangiogenic targets are understudied. Secretory autophagy has received increasing interest recently, while the roles and executing mechanisms in the tumour microenvironment (TME) remain unclear. Herein, we found that active cathepsin B (CTSB) was upregulated in tumour tissues and serum EVs of 241 BCa patients from four cohorts and was significantly associated with poor prognosis. Starving TME (STME)-induced conventional autophagy in BCa cells elevated active CTSB levels by facilitating the expression and nuclear translocation of NFATC2. In addition, STME-induced secretory autophagy simultaneously led to markedly increased secretion of LC3-conjugated EVs loaded with active CTSB (EV-CTSB) into the TME. The increased exogenous active CTSB in endothelial cells by directly ingesting EV-CTSB prominently activated the TPX2-mediated phosphorylation of the AURKA-PI3K-AKT axis, increased VEGFA expression, and promoted angiogenesis. Our findings not only verify that EV-CTSB can be a promising target for antiangiogenic strategies in bladder cancer, but also reveal a novel action pattern based on secretory autophagy-induced EV secretion which is enlightening to explore crosstalk in the TME from various perspectives.


Subject(s)
Autophagy/physiology , Cell Cycle Proteins/physiology , Extracellular Vesicles/physiology , Microtubule-Associated Proteins/physiology , Neovascularization, Pathologic/etiology , Tumor Microenvironment/physiology , Urinary Bladder Neoplasms/blood supply , Adult , Aged , Animals , Aurora Kinase A/metabolism , Cathepsin B/physiology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Urinary Bladder Neoplasms/pathology
5.
Exp Mol Pathol ; 121: 104665, 2021 08.
Article in English | MEDLINE | ID: mdl-34216584

ABSTRACT

Abnormal metabolism and uncontrolled angiogenesis are two important characteristics of malignant tumors. Although HBXIP is known to be associated with a poor prognosis for bladder cancer (BC), its effects on glycolysis and angiogenesis in BC have not been investigated. BC prognosis and relative gene expression of HBXIP were analyzed using the GEPIA, UALCAN, and STRING databases. BC cell angiogenesis and glycolysis were assessed by vasculogenic mimicry and glycolysis assay. Human umbilical vein endothelial cell (HUVEC) viability, migration, and angiogenesis were assessed by CCK8, transwell, wound healing, and tube formation assays. The results showed that HBXIP was highly expressed in BC tissues and cells. Knockdown of HBXIP expression decreased the levels of glucose uptake, lactate production, and glycolytic enzyme expression in BC cells, and decreased cell viability and migration of HUVECs. Additionally, silencing HBXIP reduced the total length of tubes and number of intersections, and EPO and VEGF protein expression in BC cells and HUVECs. Furthermore, knockdown of HBXIP expression reversed cell viability, migration, tube formation, and vasculogenic mimicry under high glucose and lactate conditions. Mechanistically, silencing of HBXIP reduced the protein expression levels of pAKT-ser473 and pmTOR, and inhibition of HBXIP, AKT, and mTOR expression decreased glycolytic enzyme protein expression. Our findings suggest that HBXIP reduces glycolysis in BC cells via regulation of AKT/mTOR signaling, thereby blocking BC angiogenesis. Collectively, this study provides a potential strategy to target HBXIP and AKT/mTOR for regulating glycolysis progression concurrently with anti-angiogenesis effects, and thereby develop novel therapeutics for the treatment of BC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glycolysis , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/blood supply , Adaptor Proteins, Signal Transducing/genetics , Cell Movement , Cell Proliferation , Cell Survival , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
6.
Acta Biochim Pol ; 68(4): 547-555, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34314132

ABSTRACT

Matrix metalloproteinases (MMPs) play an important role in many physiological and pathological processes, including neoplastic processes. They belong to a group of enzymes called endopeptidases and have the ability to hydrolyze all proteins in the extracellular matrix (ECM). They are produced in most connective tissue cells, macrophages, leukocytes, endothelial cells, microglial cells and in cancer cells. Neoplastic diseases are one of the main causes of death in Poland and in the world, therefore learning about the process of carcinogenesis seems to be particularly important. The process of carcinogenesis is currently widely studied and MMPs play one of the key roles in the development of cancer. They do this by regulating local tumor growth, stromal invasion, stimulating angiogenesis and metastasis formation. Bladder cancer is the 7th most common cancer in the male population and the 11th most common cancer in the world. In bladder cancer, most studies have been devoted to MMP-2 and MMP-9, that are enzymes responsible for the degradation of type IV collagen in the first place, which through the destruction of basement membranes and ECM, play an essential role in the tumor invasion process. Since bladder cancer is characterized by the ability to relapse, from the point of view of clinical practice it seems particularly important to develop a marker of early bladder tumor recurrence. MMPs detected in the urine and serum of patients with bladder cancer are potential factors that could play such a role.


Subject(s)
Matrix Metalloproteinases/metabolism , Urinary Bladder Neoplasms/enzymology , Biomarkers, Tumor/metabolism , Humans , Neovascularization, Pathologic , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/pathology
7.
J Clin Oncol ; 39(22): 2486-2496, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33989025

ABSTRACT

PURPOSE: The combination of gemcitabine and cisplatin (GC) is a standard therapy for metastatic urothelial carcinoma. Based on data that angiogenesis plays a role in urothelial carcinoma growth and progression, a randomized placebo-controlled trial was performed with the primary objective of testing whether patients treated with GC and bevacizumab (GCB) have superior overall survival (OS) than patients treated with GC and placebo (GCP). PATIENTS AND METHODS: Between July 2009 and December 2014, 506 patients with metastatic urothelial carcinoma without prior chemotherapy for metastatic disease and no neoadjuvant or adjuvant chemotherapy within 12 months were randomly assigned to receive either GCB or GCP. The primary end point was OS, with secondary end points of progression-free survival, objective response, and toxicity. RESULTS: With a median follow-up of 76.3 months among alive patients, the median OS was 14.5 months for patients treated with GCB and 14.3 months for patients treated with GCP (hazard ratio for death = 0.87; 95% CI, 0.72 to 1.05; two-sided stratified log-rank P = .14). The median progression-free survival was 8.0 months for GCB and 6.7 months for GCP (hazard ratio = 0.77; 95% CI, 0.63 to 0.95; P = .016). The proportion of patients with grade 3 or greater adverse events did not differ significantly between both arms, although increased bevacizumab-related toxicities such as hypertension and proteinuria occurred in the bevacizumab-treated arm. CONCLUSION: The addition of bevacizumab to GC did not result in improved OS. The observed median OS of about 14 months is consistent with prior phase III trials of cisplatin-based chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Urologic Neoplasms/drug therapy , Aged , Angiogenesis Inhibitors/administration & dosage , Bevacizumab/administration & dosage , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Double-Blind Method , Female , Humans , Male , Neovascularization, Pathologic/drug therapy , Survival Analysis , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/drug therapy , Urologic Neoplasms/blood supply , Gemcitabine
8.
Int J Mol Sci ; 22(9)2021 Apr 25.
Article in English | MEDLINE | ID: mdl-33923108

ABSTRACT

In 2018, 550,000 people were diagnosed with bladder cancer (BC), of which nearly 200,000 people died. Moreover, men are 4 times more likely than women to be diagnosed with BC. The risk factors include exposure to environmental and occupational chemicals, especially tobacco smoke, benzidine and genetic factors. Despite numerous studies, the molecular basis of BC development remains unclear. A growing body of evidence suggests that inflammation, oxidant-antioxidant imbalance and angiogenesis disorders may play a significant role in the development and progression of bladder cancer. The patients with bladder cancer were characterised by an increased level of reactive oxygen species (ROS), the products of lipid peroxidation, proinflammatory cytokines and proangiogenic factors as compared to controls. Furthermore, it was shown that polymorphisms localised in genes associated with these pathways may modulate the risk of BC. Interestingly, ROS overproduction may induce the production of proinflammatory cytokines, which finally activated angiogenesis. Moreover, the available literature shows that both inflammation and oxidative stress may lead to activation of angiogenesis and tumour progression in BC patients.


Subject(s)
Inflammation/complications , Neovascularization, Pathologic/etiology , Oxidative Stress/physiology , Urinary Bladder Neoplasms/etiology , Cytokines/metabolism , Humans , Inflammation/pathology , Mycobacterium bovis , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/metabolism , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/therapy
9.
Cancer Res ; 80(14): 2996-3008, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32536602

ABSTRACT

Tumor endothelial cells (TEC) lining tumor blood vessels actively contribute to tumor progression and metastasis. In addition to tumor cells, TEC may develop drug resistance during cancer treatment, allowing the tumor cells to survive chemotherapy and metastasize. We previously reported that TECs resist paclitaxel treatment via upregulation of ABCB1. However, whether TEC phenotypes are altered by anticancer drugs remains to be clarified. Here, we show that ABCB1 expression increases after chemotherapy in urothelial carcinoma cases. The ratio of ABCB1-positive TEC before and after first-line chemotherapy in urothelial carcinoma tissues (n = 66) was analyzed by ABCB1 and CD31 immunostaining. In 42 cases (64%), this ratio increased after first-line chemotherapy. Chemotherapy elevated ABCB1 expression in endothelial cells by increasing tumor IL8 secretion. In clinical cases, ABCB1 expression in TEC correlated with IL8 expression in tumor cells after first-line chemotherapy, leading to poor prognosis. In vivo, the ABCB1 inhibitor combined with paclitaxel reduced tumor growth and metastasis compared with paclitaxel alone. Chemotherapy is suggested to cause inflammatory changes in tumors, inducing ABCB1 expression in TEC and conferring drug resistance. Overall, these findings indicate that TEC can survive during chemotherapy and provide a gateway for cancer metastasis. Targeting ABCB1 in TEC represents a novel strategy to overcome cancer drug resistance. SIGNIFICANCE: These findings show that inhibition of ABCB1 in tumor endothelial cells may improve clinical outcome, where ABCB1 expression contributes to drug resistance and metastasis following first-line chemotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Interleukin-8/metabolism , Neovascularization, Pathologic/pathology , Paclitaxel/pharmacology , Urinary Bladder Neoplasms/mortality , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Drug Resistance, Multiple , Female , Gene Expression Regulation, Neoplastic , Humans , Induction Chemotherapy , Interleukin-8/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neovascularization, Pathologic/chemically induced , Prognosis , Survival Rate , Tumor Cells, Cultured , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
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
11.
Cancer Lett ; 473: 118-129, 2020 03 31.
Article in English | MEDLINE | ID: mdl-31843555

ABSTRACT

Early studies suggest that the androgen receptor (AR) may play differential roles in influencing prostate cancer (PCa) and bladder cancer (BCa) metastasis, but the underlying mechanisms remain unclear. Here, we found that the AR might function via differentially altering vasculogenic mimicry (VM) formation to either decrease PCa metastasis or increase BCa metastasis. Mechanism dissection showed that the AR could differentially alter the expression of the VM marker SLPI through miR-525-5p to regulate SLPI; moreover, it could either increase miR-525-5p transcription in PCa or decrease it in BCa via binding to different androgen-response-elements (AREs) located at different positions in the miR-525 precursor promoter. Further, results from liquid chromatography-mass spectrometry (LC-MS) showed that the co-factors of AR in PCa and BCa are NFIX and HDAC2, respectively. Together, these results provide the first detailed mechanism of how the AR can differentially alter PCa and BCa metastasis; thus, targeting the newly identified AR-miR-525-5p-SLPI axis may help suppress metastasis.


Subject(s)
MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Secretory Leukocyte Peptidase Inhibitor/metabolism , Urinary Bladder Neoplasms/genetics , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Histone Deacetylase 2/metabolism , Humans , Male , NFI Transcription Factors/metabolism , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , RNA, Small Interfering/metabolism , Receptors, Androgen/genetics , Secretory Leukocyte Peptidase Inhibitor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/pathology
12.
J Nutr Biochem ; 74: 108230, 2019 12.
Article in English | MEDLINE | ID: mdl-31683101

ABSTRACT

Carnosine, a naturally occurring dipeptide, was recently reported to exhibit anticancer activity; however, the molecular mechanisms and regulators underlying its activity against tumor-associated angiogenesis remain unidentified. In this study, we evaluated the in vitro and in vivo antitumor effects of carnosine in EJ bladder cancer cells and EJ-xenografted BALB/c nude mice, respectively. In addition, in vitro capillary tube formation of HUVECs, ex vivo aortic ring and in vivo Matrigel plug assays were employed to examine the antiangiogenic potential of carnosine. Carnosine significantly inhibited EJ cell proliferation. Flow cytometric and immunoblot analyses indicated that carnosine modulated regulators of the G1 cell cycle phase, including cyclin D1, CDK4 and p21WAF1. The mitogen-activated protein kinases, ERK and p38, but not JNK or AKT, responded to carnosine. Carnosine inhibited the migratory and invasive potential of EJ cells by inhibiting MMP-9 activity, which was associated with suppression of binding activity of NF-κB, SP-1 and AP-1. In xenograft tumors, carnosine exhibited antitumor activity equivalent to cisplatin, but no weight loss occurred in carnosine-treated mice. In HUVECs, carnosine inhibited VEGF-mediated proliferation, colony tube formation, migration and invasion. The antiangiogenic activity of carnosine was partially due to the suppression of VEGFR-2-mediated ERK/AKT/eNOS signaling and MMP-2. Furthermore, using aortic ring and Matrigel plug assays, we confirmed the antiangiogenic activity of carnosine. Given that targeting tumor-associated angiogenesis is a proven effective therapeutic strategy, our results may provide valuable information for the development of preventive or therapeutic agents for bladder cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carnosine/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Microvessels/drug effects , Microvessels/pathology , Neovascularization, Pathologic/drug therapy , Nitric Oxide Synthase Type III/metabolism , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/pathology , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
13.
J Mater Chem B ; 7(42): 6623-6629, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31591622

ABSTRACT

Benefiting from high spatial resolution and large penetration depth, NIR-II (second near-infrared spectral region, 900-1700 nm) fluorescence imaging based on US Food and Drug Administration (FDA)-approved indocyanine green (ICG) is expected to be a good approach for clinical applications. As of now, nearly all reported works on ICG-assisted NIR-II fluorescence imaging are macro-imaging while micro-angiography is also a significant imaging modality, especially during the diagnosis and treatment of cerebrovascular diseases. Herein, based on NIR-II fluorescence wide-field microscopy, the high-resolution observation of cerebral vasculature was performed at deep brain tissues in mice via intramuscular (IM) injection of ICG. Altered cerebral vessels in mice after brain embolism were further noticed by means of noninvasive through-skull NIR-II fluorescence microscopy. Moreover, ICG-assisted NIR-II fluorescence confocal microscopy was executed to observe cerebral vasculature, presenting optical sectioning capability and higher spatial resolution.


Subject(s)
Brain/blood supply , Brain/diagnostic imaging , Cerebral Angiography/methods , Fluorescent Dyes/chemistry , Indocyanine Green/chemistry , Microscopy, Fluorescence/methods , Animals , Cell Line, Tumor , Female , Humans , Intracranial Thrombosis/diagnostic imaging , Male , Mice, Inbred BALB C , Mice, Inbred ICR , Microscopy, Confocal/methods , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/diagnostic imaging
14.
Cancer Sci ; 110(7): 2119-2132, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31141251

ABSTRACT

Locally advanced and metastatic invasive bladder cancer (BC) has a poor prognosis, and no advanced therapies beyond cisplatin-based combination chemotherapy have been developed. Therefore, it is an urgent issue to elucidate the underlying mechanisms of tumor progression and metastasis of invasive BC for the development of new therapeutic strategies. Here, we clarified a novel role of exosomes containing ErbB2 and CRK in a formation of premetastatic niches and subsequent metastases. CRK adaptors were overexpressed in invasive UM-UC-3 BC cells. In an orthotopic xenograft model, metastases to lung, liver, and bone of UM-UC-3 cells were completely abolished by CRK elimination. Mass spectrometry analysis identified that ErbB2 was contained in UM-UC-3-derived exosomes in a CRK-dependent manner; the exosomes significantly increased proliferation and invasion properties of low-grade 5637 BC cells and HUVECs through FAK and PI3K/AKT signaling pathways. In athymic mice educated with UM-UC-3-derived exosomes, i.v. implanted UM-UC-3 cells were trapped with surrounding PKH67-labeled exosomes in lung and led to development of lung metastasis with disordered vascular proliferation. In contrast, exosomes derived from CRK-depleted BC cells failed to induce these malignant features. Taken together, we showed that CRK adaptors elevated the expression of ErbB2/3 in BC cells, and these tyrosine kinase/adaptor units were transferred from host BC cells to metastatic recipient cells by exosomes, leading to vascular leakiness and proliferation and contributing to the formation of distant metastasis. Thus, CRK intervention with ErbB2/3 blockade might be a potent therapeutic strategy for patients with ErbB2 overexpressing advanced and metastatic BC.


Subject(s)
Exosomes/pathology , Proto-Oncogene Proteins c-crk/metabolism , Receptor, ErbB-2/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Exosomes/genetics , Exosomes/metabolism , Humans , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Proto-Oncogene Proteins c-crk/genetics , Receptor, ErbB-2/genetics , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
15.
Nat Commun ; 10(1): 720, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755618

ABSTRACT

Bladder cancer is one of the most common and highly vascularized cancers. To better understand its genomic structure and underlying etiology, we conduct whole-genome and targeted sequencing in urothelial bladder carcinomas (UBCs, the most common type of bladder cancer). Recurrent mutations in noncoding regions affecting gene regulatory elements and structural variations (SVs) leading to gene disruptions are prevalent. Notably, we find recurrent ADGRG6 enhancer mutations and FRS2 duplications which are associated with higher protein expression in the tumor and poor prognosis. Functional assays demonstrate that depletion of ADGRG6 or FRS2 expression in UBC cells compromise their abilities to recruit endothelial cells and induce tube formation. Moreover, pathway assessment reveals recurrent alterations in multiple angiogenesis-related genes. These results illustrate a multidimensional genomic landscape that highlights noncoding mutations and SVs in UBC tumorigenesis, and suggest ADGRG6 and FRS2 as novel pathological angiogenesis regulators that would facilitate vascular-targeted therapies for UBC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Membrane Proteins/genetics , Mutation , Receptors, G-Protein-Coupled/genetics , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/genetics , Cell Line, Tumor , DNA Mutational Analysis , Exome , Gene Dosage , Genetic Predisposition to Disease , Humans , Neovascularization, Pathologic , Regulatory Sequences, Nucleic Acid , Urinary Bladder Neoplasms/pathology , Whole Genome Sequencing
16.
Int J Urol ; 26(3): 414-422, 2019 03.
Article in English | MEDLINE | ID: mdl-30659668

ABSTRACT

OBJECTIVE: To investigate the effectiveness of combination treatment of vascular targeted photodynamic therapy and anti-cytotoxic T-lymphocyte-associated antigen 4 immunotherapy in a mouse model of urothelial carcinoma. METHODS: We used C57BL/6 mice injected with murine bladder 49 cell line. Mice were randomly allocated into four treatment groups: vascular targeted photodynamic therapy only, anti-cytotoxic T-lymphocyte-associated antigen 4 only, combination therapy and control. We carried out three separate experiments that used distinct cohorts of mice: tumor growth and development of lung metastases monitored with bioluminescent imaging (n = 91); survival evaluated with Kaplan-Meier curves (n = 111); and tumor cell population studied with flow cytometry (n = 20). In a fourth experiment, we re-challenged tumors in previously treated mice and compared tumor growth with that of naïve mice. RESULTS: Combination therapy provided significant benefits over the other three treatment groups: prolonged survival (P < 0.0001), lower tumor signal (P < 0.0001) and decreased lung signal uptake (P ≤ 0.002). We also observed that mice previously treated with vascular targeted photodynamic therapy only or combination therapy did not present tumor growth after re-challenged tumors. CONCLUSIONS: Combination of vascular targeted photodynamic therapy with anti-cytotoxic T-lymphocyte-associated antigen 4 is an effective therapy in a urothelial carcinoma syngeneic mouse model. The present results suggest this therapy as a potential treatment option for both bladder and upper tract tumors in future clinical trials.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Carcinoma, Transitional Cell/therapy , Lung Neoplasms/drug therapy , Photochemotherapy/methods , Urinary Bladder Neoplasms/therapy , Animals , CTLA-4 Antigen/immunology , Carcinoma, Transitional Cell/blood supply , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/secondary , Cell Line, Tumor/transplantation , Disease Models, Animal , Humans , Luminescent Measurements , Lung/blood supply , Lung/diagnostic imaging , Lung/drug effects , Lung/radiation effects , Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Urinary Bladder/blood supply , Urinary Bladder/drug effects , Urinary Bladder/immunology , Urinary Bladder/radiation effects , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
17.
Med Sci Monit ; 24: 5346-5354, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30066698

ABSTRACT

BACKGROUND Collagen type VI alpha 3 chain (COL6A3) has been proven to be a biomarker in the occurrence and development of bladder cancer, which is the most common malignant tumor in the urinary system. This study aimed to explore the effect and molecular mechanism of COL6A3 on EMT in vitro induced by TGF-ß/Smad in bladder carcinoma. MATERIAL AND METHODS There were 42 patients included in the Kaplan-Meier survival analysis. A cell counting kit-8 (CCK-8) assay and an angiogenesis assay were used to measure cell proliferation and tube formation, respectively. Western blot analysis and quantitative reverse transcription-polymerase chain reaction (qPCR) were conducted for the proteins and mRNAs expression. RESULTS COL6A3 was highly expressed in tissues and cells of bladder cancer. COL6A3 silencing could inhibit the cell proliferation and angiopoiesis. In addition, COL6A3 silencing obviously suppressed the levels of matrix metalloproteinase-2 (MMP2), Matrix metalloproteinase-9 (MMP9), and vimentin. On the contrary, the levels of epithelium-specific cell-cell adhesion molecule (E-cadherin) and tumor inhibitor of metalloproteinase-1 (TIMP-1) were significantly increased. Furthermore, we found that COL6A3 silencing reduced the activity of p-Smad2, p-Smad3, and transforming growth factor ß (TGF-ß). CONCLUSIONS COL6A3 could influence the viability and angiogenesis of bladder cancer cells. COL6A3 may have a certain relationship with the TGF-ß/Smad-induced EMT process.


Subject(s)
Collagen Type VI/biosynthesis , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Antigens, CD , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Collagen Type VI/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/metabolism , Urinary Bladder Neoplasms/blood supply , Vimentin/metabolism
18.
Int J Nanomedicine ; 13: 3625-3640, 2018.
Article in English | MEDLINE | ID: mdl-29983558

ABSTRACT

BACKGROUND: Lumbrokinase (LK) is an enzyme complex with antithrombotic, antioxidant, antitumor, and immunomodulatory effects. It has been extensively studied and used in clinical anti-tumor therapy. However, its half-life is short, its bioavailability is low, and its toxicity and side effects are great, which greatly limit its clinical application. Therefore, LK is often combined with other drugs (such as immune agents, hormones, or Chinese herbal medicine) to reduce its dosage and side effects and to improve its anti-tumor effects. METHODS AND RESULTS: Here, we described an LK/paclitaxel (PTX) nanocarrier based on poly(ethylene glycol)-b-(poly(ethylenediamine l-glutamate)-g-poly(ε-benzyoxycarbonyl-l-lysine)-r-poly(l-lysine)) (PEG-b-(PELG-g-(PZLL-r-PLL))). In the present study, LK and PTX were loaded by electrostatic and/or hydrophobic effects under mild conditions, thereby increasing the half-life and bioavailability of the drugs via the sustained release and enhancement of tumor site enrichment by the LK/PTX/PEG-b-(PELG-g-(PZLL-r-PLL)) complex through passive targeting. In this study, using bladder cancer cells (J82 cells) and rat bladder cancer model as the object, the structure of the nanocarrier, the relationship between drugs composition and antitumor properties were systematically studied. CONCLUSION: We propose that the block copolymer PEG-b-(PELG-g-(PZLL-r-PLL)) may function as a potent nanocarrier for augmenting anti-bladder cancer pharmacotherapy, with unprecedented clinical benefits.


Subject(s)
Albumins/therapeutic use , Endopeptidases/therapeutic use , Paclitaxel/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Albumins/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B1/metabolism , Drug Carriers/chemistry , Endopeptidases/blood , Endopeptidases/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Lysine/analogs & derivatives , Lysine/chemical synthesis , Lysine/chemistry , Male , Microvessels/pathology , Molecular Weight , Paclitaxel/blood , Paclitaxel/pharmacology , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polylysine/analogs & derivatives , Polylysine/chemical synthesis , Polylysine/chemistry , Rats, Sprague-Dawley , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism , Urinary Bladder Neoplasms/blood , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/pathology
20.
Cell Signal ; 48: 38-44, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29702203

ABSTRACT

Muscle-invasive or metastatic bladder cancer (BCa) is a life-threatening disease for patients, and tumor angiogenesis is believed to play a critical role in the progression of BCa. However, its underlying mechanism of tumor angiogenesis is still poorly understood. In this study, we discovered that RASAL2, a RAS GTPase activating protein, could inhibit BCa angiogenesis based on our shRNA/siRNA knockdown or ectopic cDNA expression experiments. Mechanistically, RASAL2 downregulation could enhance the phosphorylation of AKT and then subsequently upregulate the expression of ETS1 and VEGFA. Furthermore, there was a negative correlation between RASAL2 and VEGFA or CD31 expression in subcutaneous xenograft and human BCa specimens. Taken together, we provide a new insight into the molecular mechanism of BCa progression, in which RASAL2 can be a new therapeutic target.


Subject(s)
Carrier Proteins/physiology , Neovascularization, Pathologic/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Urinary Bladder Neoplasms/blood supply , Vascular Endothelial Growth Factor A/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , GTPase-Activating Proteins , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred BALB C , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Signal Transduction , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...