Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Transl Vis Sci Technol ; 13(1): 20, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38252520

ABSTRACT

Purpose: To evaluate the feasibility of using DARC (detection of apoptosing retinal cells) technology as a biomarker for preclinical assessment of glaucomatous damage in a non-human primate (NHP) model of ocular hypertension (OHT). Methods: Elevated intraocular pressure (IOP) was induced by applying a laser to the trabecular meshwork in each eye of NHPs. Changes in DARC counts in the retina, identified as fluorescent-tagged annexin V (ANX776)-positive cells, were evaluated together with optic nerve damage, assessed using spectral domain-optical coherence tomography. The pharmacokinetic properties of ANX776 in both healthy and OHT model monkeys were also examined. Results: Sustained elevation of IOP and subsequent thinning of the retinal nerve fiber layer thickness (RNFLT) around the optic nerve head were confirmed in the OHT model. Increases in DARC counts were also detected after IOP elevation. We identified a statistically significant relationship between cumulative DARC counts and reductions in RNFLT both globally and in each peripapillary sector. Intravenous administration of ANX776 increased blood annexin V in a dose-dependent manner, which was subsequently eliminated. Conclusions: This study revealed that DARC technology can effectively assess glaucomatous damage in an NHP OHT model. We obtained the fundamental data that could serve as a reference for developing preclinical models to evaluate the pharmacodynamics of neuroprotective agents using DARC technology in NHP OHT models. Translational Relevance: Our basic data in a monkey OHT model could be useful for future preclinical studies using DARC technology to estimate the pharmacodynamic response of neuroprotective agents.


Subject(s)
Glaucoma , Neuroprotective Agents , Ocular Hypertension , Animals , Annexin A5 , Primates , Apoptosis
2.
Int J Mol Sci ; 24(5)2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36902377

ABSTRACT

PURPOSE: Bladder cancer (BCa) is one of the most common cancer types worldwide and is characterized by a high rate of recurrence. In previous studies, we and others have described the functional influence of plasminogen activator inhibitor-1 (PAI1) in bladder cancer development. While polymorphisms in PAI1 have been associated with increased risk and worsened prognosis in some cancers, the mutational status of PAI1 in human bladder tumors has not been well defined. METHODS: In this study, we evaluated the mutational status of PAI1 in a series of independent cohorts, comprised of a total of 660 subjects. RESULTS: Sequencing analyses identified two clinically relevant 3' untranslated region (UTR) single nucleotide polymorphisms (SNPs) in PAI1 (rs7242; rs1050813). Somatic SNP rs7242 was present in human BCa cohorts (overall incidence of 72%; 62% in Caucasians and 72% in Asians). In contrast, the overall incidence of germline SNP rs1050813 was 18% (39% in Caucasians and 6% in Asians). Furthermore, Caucasian patients with at least one of the described SNPs had worse recurrence-free survival and overall survival (p = 0.03 and p = 0.03, respectively). In vitro functional studies demonstrated that SNP rs7242 increased the anti-apoptotic effect of PAI1, and SNP rs1050813 was related to a loss of contact inhibition associated with cellular proliferation when compared to wild type. CONCLUSION: Further investigation of the prevalence and potential downstream influence of these SNPs in bladder cancer is warranted.


Subject(s)
Plasminogen Activator Inhibitor 1 , Polymorphism, Single Nucleotide , Urinary Bladder Neoplasms , Humans , Neoplasm Recurrence, Local , Plasminogen Activator Inhibitor 1/genetics , Urinary Bladder Neoplasms/genetics
3.
Gan To Kagaku Ryoho ; 47(5): 815-818, 2020 May.
Article in Japanese | MEDLINE | ID: mdl-32408326

ABSTRACT

A 70-year-old woman was diagnosed with cT2N2M0 left triple negative breast cancer. She received neoadjuvant chemotherapy. Around 30 months after the surgery, she developed carcinomatous pericardial tamponade, carcinomatous pleurisy, and metastatic thyroid cancer due to the breast cancer. Around the same time, a BRCA2 mutation was confirmed. Thus, olaparib therapy was started. Around 45 days after the start of olaparib administration, the therapeutic effect was cPR. The treatment remains effective even after approximately 1 year.


Subject(s)
Antineoplastic Agents/therapeutic use , BRCA2 Protein/genetics , Breast Neoplasms , Phthalazines/therapeutic use , Piperazines/therapeutic use , Aged , Breast Neoplasms/drug therapy , Female , Humans , Mutation , Neoplasm Recurrence, Local
4.
J Transl Med ; 18(1): 57, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024545

ABSTRACT

BACKGROUND: Accumulating evidence suggests that plasminogen activator inhibitor-1 (PAI-1) plays an important role in bladder tumorigenesis by regulating cell cycle. However, it remains unclear whether and how inhibition of PAI-1 suppresses bladder tumorigenesis. METHODS: To elucidate the therapeutic effect of PAI-1 inhibition, we tested its tumorigenicity in PAI-1 knockout (KO) mice exposed to a known bladder carcinogen. RESULTS: PAI-1 deficiency did not inhibit carcinogen-induced bladder cancer in mice although carcinogen-exposed wild type mice significantly increased PAI-1 levels in bladder tissue, plasma and urine. We found that PAI-1 KO mice exposed to carcinogen tended to upregulate protein C inhibitor (PAI-3), urokinase-type plasminogen activator (uPA) and tissue-type PA (tPA), and significantly increased PAI-2, suggesting a potential compensatory function of these molecules when PAI-1 is abrogated. Subsequent studies employing gene expression microarray using mouse bladder tissues followed by post hoc bioinformatics analysis and validation experiments by qPCR and IHC demonstrated that SERPING1 is further downregulated in PAI-1 KO mice exposed to BBN, suggesting that SERPING1 as a potential missing factor that regulate PAI-2 overexpression (compensation pathway). CONCLUSIONS: These results indicate that serpin compensation pathway, specifically PAI-2 overexpression in this model, supports bladder cancer development when oncoprotein PAI-1 is deleted. Further investigations into PAI-1 are necessary in order to identify true potential targets for bladder cancer therapy.


Subject(s)
Plasminogen Activator Inhibitor 1 , Plasminogen Activator Inhibitor 2 , Urinary Bladder Neoplasms , Animals , Mice , Mice, Knockout , Nitrosamines , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 2/genetics , Serpin E2 , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/genetics
5.
PLoS One ; 13(10): e0205888, 2018.
Article in English | MEDLINE | ID: mdl-30335858

ABSTRACT

Adipose tissue, which is conserved in higher eukaryotes, plays central roles in controlling the body's energy balance, including excess energy storage and energy expenditure during starvation. In adipogenesis, intranuclear receptor, peroxisome proliferator-activated receptor gamma (PPARγ) is a key molecule, and PPARγ agonists can promote adipogenesis. Many studies on the in vitro screening of PPARγ agonists with compounds derived from various materials have been reported; however, in vivo assays for quick examination of these feeding effects have not been established. In this study, we developed a technique using a lipophilic fluorescent reagent, Nile red to quantitatively estimate the adipose tissue volumes by using Japanese rice fish, medaka (Oryzias latipes) and studied effects of dietary soy sauce oil (SSO), which is a discarded by-product from Japanese traditional food and is known to have PPARγ-agonistic activity, on adipogenesis. We found that SSO feeding increased the adipose tissue volumes, and the expression levels of adipogenesis-related genes increased in these medaka larvae. These results suggest that SSO feeding increases the adipose tissue volumes through adipogenesis promotion by PPARγ-agonistic activity in medaka, and medaka is a powerful model for studying adipogenesis. Furthermore, our study also demonstrates the availability of SSO as a dietary additive for farmed fish.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/drug effects , Animal Feed/analysis , Dietary Fats, Unsaturated/administration & dosage , Larva/drug effects , Oryzias/genetics , PPAR gamma/genetics , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Adipogenesis/genetics , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue/metabolism , Animals , Animals, Genetically Modified , Aquaculture , Diet/methods , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Oryzias/growth & development , Oryzias/metabolism , Oxazines/administration & dosage , Oxazines/chemistry , PPAR gamma/agonists , PPAR gamma/metabolism , Soy Foods/analysis
6.
Carcinogenesis ; 39(1): 47-55, 2018 01 12.
Article in English | MEDLINE | ID: mdl-28968647

ABSTRACT

Aberrant sphingolipid metabolism has been reported to promote breast cancer progression. Sphingosine kinase 1 (SphK1) is a key metabolic enzyme for the formation of pro-survival S1P from pro-apoptotic ceramide. The role of SphK1 in breast cancer has been well studied in estrogen receptor (ER)-positive breast cancer; however, its role in human epidermal growth factor 2 (HER2)-positive breast cancer remains unclear. Here, we show that genetic deletion of SphK1 significantly reduced mammary tumor development with reduced tumor incidence and multiplicity in the MMTV-neu transgenic mouse model. Gene expression analysis revealed significant reduction of claudin-2 (CLDN2) expression in tumors from SphK1 deficient mice, suggesting that CLDN2 may mediate SphK1's function. It is remarkable that SphK1 deficiency in HER2-positive breast cancer model inhibited tumor formation by the different mechanism from ER-positive breast cancer. In vitro experiments demonstrated that overexpression of SphK1 in ER-/PR-/HER2+ human breast cancer cells enhanced cell proliferation, colony formation, migration and invasion. Furthermore, immunostaining of SphK1 and CLDN2 in HER2-positive human breast tumors revealed a correlation in high-grade disease. Taken together, these findings suggest that SphK1 may play a pivotal role in HER2-positive breast carcinogenesis. Targeting SphK1 may represent a novel approach for HER2-positive breast cancer chemoprevention and/or treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptor, ErbB-2/genetics , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic
7.
Oncotarget ; 8(59): 99707-99721, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29245935

ABSTRACT

BACKGROUND: We previously reported an accurate urine-based bladder cancer (BCa)-associated diagnostic signature that can be used to non-invasively detect BCa. In this study, we investigated whether a component of this signature could risk stratify patients with BCa. METHODS: Utilizing immunohistochemistry, we investigated angiogenin, MMP-2, p53, RB and PAI-1 expression from 939 patients with BCa. The expression levels were scored by assigning a proportion score and an intensity score to yield a total staining score for each protein. The expressions of each protein individually and as an aggregate were then correlated with progression-free survival (PFS), cancer-specific survival (CSS) and overall survival (OS). RESULTS: Differential expressions of these markers were noted in BCa. With multivariate analysis in non-muscle invasive bladder cancer (NMIBC) age, tumor grade portended a worse PFS, while age, tumor grade, nodal status, MMP2, RB and PAI-1 expression portended a worse OS. As for multivariate analysis in muscle invasive bladder cancer (MIBC), age MMP-2 and RB were associated with a worse PFS, while age, nodal status, MMP-2, RB and PAI-1 were associated with a worse OS. Using Kaplan-Meier survival analysis, we noted a significant reduction in OS as more of the five biomarkers were expressed in a tumor. Thus, overall, high expressions of MMP-2, RB and/or PAI-1 in bladder tumors were markers of poor prognosis. CONCLUSION: Individually, MMP-2, RB and PAI-1, as well as in aggregate correlated with poor survival in patients with BCa. Thus, patients whose bladder tumors express these biomarkers may benefit from early radical treatment and/or neoadjuvant or adjuvant therapies.

8.
Carcinogenesis ; 38(12): 1218-1227, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29028945

ABSTRACT

Accumulating evidence suggests that the sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate (S1P) pathway plays a pivotal role in colon carcinogenesis. Our previous studies indicate that the SphK1/S1P pathway mediates colon carcinogenesis at least by regulating cyclooxygenase 2 (COX-2) expression and prostaglandin E2 (PGE2) production. However, the mechanisms by which this pathway regulates colon carcinogenesis are still unclear. First, we show that SphK1 deficient mice significantly attenuated azoxymethane (AOM)-induced colon carcinogenesis as measured by colon tumor incidence, multiplicity, and volume. We found that AOM activates peritoneal macrophages to induce SphK1, COX-2, and tumor necrosis factor (TNF)-α expression in WT mice. Interestingly, SphK1 knockout (KO) mice revealed significant reduction of COX-2 and TNF-α expression from AOM-activated peritoneal macrophages, suggesting that SphK1 regulates COX-2 and TNF-α expression in peritoneal macrophages. We found that inoculation of WT peritoneal macrophages restored the carcinogenic effect of AOM in Sphk1 KO mice as measured by aberrant crypt foci (ACF) formation, preneoplastic lesions of colon cancer. In addition, downregulation of SphK1 only in peritoneal macrophage by short hairpin RNA (shRNA) reduced the number of ACF per colon induced by AOM. Intraperitoneal injection of sphingolipids demonstrates that S1P enhanced AOM-induced ACF formation, while ceramide inhibited. Finally, we show that SphK inhibitor SKI-II significantly reduced the number of ACF per colon. These results suggest that SphK1 expression plays a pivotal role in the early stages of colon carcinogenesis through regulating COX-2 and TNF-α expression from activated peritoneal macrophages.


Subject(s)
Carcinogenesis/metabolism , Colonic Neoplasms/pathology , Macrophages, Peritoneal/enzymology , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Animals , Colonic Neoplasms/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
J Transl Med ; 15(1): 120, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28583134

ABSTRACT

BACKGROUND: Accumulating evidence suggests that sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate pathway plays a pivotal role in colon carcinogenesis. METHODS: To further support the evidence, we investigated the effects of SphK1 using three separate animal models: SphK1 knockout mice, SphK1 overexpressing transgenic mice, and SphK1 overexpression in human colon cancer xenografts. Using azoxymethane (AOM, colon carcinogen), we analyzed colon tumor development in SphK1 KO and SphK1 overexpression in intestinal epithelial cells regulated by a tet-on system. Then, we analyzed subcutaneous tumor growth using xenografts of HT-29 human colon cancer cell. Finally, immunohistochemical analyses for SphK1 and COX-2 were performed on human colon cancer tissue microarray. RESULTS: SphK1 KO mice, compared to wild-type mice, demonstrated a significant inhibition in colon cancer development induced by AOM (58.6% vs. 96.4%, respectively, P < 0.005). Tumor multiplicity (1.00 vs. 1.64 per colon, respectively, P < 0.05) and tumor volume (14.82 mm3 vs. 29.10 mm3, P < 0.05) were both significantly reduced in SphK1 KO mice compared to wild-type mice. Next, SphK1 overexpression in HT-29 enhanced tumor growth as compared to GFP control in nude mice (229.5 mm3 vs. 90.9 mm3, respectively, P < 0.05). Furthermore, overexpression of SphK1 in intestinal epithelial cells significantly enhances AOM-induced colon tumor formation (P < 0.05). Lastly, SphK1 and COX-2 intensity tended to reduce overall survival of late stage colon cancer patients. CONCLUSIONS: SphK1 expression regulates the early stage of colon carcinogenesis and tumor growth, thus inhibition of SphK1 may be an effective strategy for colon cancer chemoprevention.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Aged , Animals , Azoxymethane , Carcinogenesis/pathology , Cell Proliferation , Cyclooxygenase 2/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Female , HT29 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging
10.
J Transl Med ; 14(1): 287, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27717367

ABSTRACT

BACKGROUND: Bladder cancer (BCa) is among the most commonly diagnosed malignancies worldwide, and due the high rate of post-operative disease recurrence, it is one of the most prevalent in many countries. The development of non-invasive molecular assays that can accurately detect and monitor BCa would be a major advance, benefiting both patients and healthcare systems. We have previously identified a urinary protein biomarker panel that is being developed for application in at-risk patient cohorts. Here, we investigated the potential utility of the multiplex assay in a Japanese cohort. METHODS: The Japanese study cohort collected from urology clinics at two institutions was comprised of a total of 288 subjects. The protein biomarker panel (IL8, MMP9, MMP10, ANG, APOE, SDC1, A1AT, PAI1, CA9, VEGFA) was monitored in voided urine samples collected prior to cystoscopy using a custom multiplex ELISA assay. The diagnostic performance of the biomarker panel was assessed using receiver operator curves, predictive modeling and descriptive statistics. RESULTS: Urinary biomarker concentrations were significantly elevated in cases versus controls, and in cases with high-grade and muscle-invasive tumors. The AUC for the 10-biomarker assay was 0.892 (95 % confidence interval 0.850-0.934), with an overall diagnostic sensitivity specificity of 0.85 and 0.81, respectively. A predictive model trained on the larger institutional cohort correctly identified 99 % of the cases from the second institution. CONCLUSIONS: Urinary levels of a 10-biomarker panel enabled discrimination of patients with BCa. The multiplex urinary diagnostic assay has the potential to be developed for the non-invasive detection of BCa in at-risk Japanese patients.


Subject(s)
Asian People , Immunoassay/methods , Urinary Bladder Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/urine , Cohort Studies , Demography , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , ROC Curve , Sensitivity and Specificity , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/urine , Young Adult
11.
Oncotarget ; 7(28): 43109-43123, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27317771

ABSTRACT

Epigenetic-mediated gene activation/silencing plays a crucial role in human tumorigenesis. Eliciting the underlying mechanism behind certain epigenetic changes is essential for understanding tumor biology. Previous studies in human cancers revealed an unrecognized interplay between Angiogenin (ANG) and matrix metalloproteinase-2 (MMP2) leading to pronounced tumorigenesis. Here we provide multiple lines of evidence further indicating ANG oncogenic potential. ANG expression resulted in the hypomethylated state of the MMP2 gene, which led to increased gene expression of MMP2. More than that, our global DNA methylation microarray analysis showed that gene manipulation of ANG affected a variety of pathways, such as cell migration, angiogenesis and specifically, tumor suppressor genes. Mechanistically, ANG negatively regulated DNA methyltransferase 3b (DNMT3b) enzymatic activity by down-regulating its expression and inhibiting its recruitment to the MMP2 promoter. Consistent with this, ANG-MMP2 overexpression and DNMT3b underexpression correlated with reduction in disease free survival of human bladder cancer patients. Together, the results continue to establish ANG as an oncoprotein and further reveal that ANG contributes to oncogenesis by the activation of MMP2 through modulation of DNMT3b functions.


Subject(s)
Carcinogenesis/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 2/metabolism , Ribonuclease, Pancreatic/metabolism , Urinary Bladder Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , DNA Methylation , Disease-Free Survival , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Epigenesis, Genetic , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Neovascularization, Pathologic/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Ribonuclease, Pancreatic/genetics , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , DNA Methyltransferase 3B
12.
Oncotarget ; 7(15): 20869-89, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26989079

ABSTRACT

Resident mesenchymal stem cells (MSCs) promote cancer progression. However, pathways and mechanisms involved in recruiting MSCs into breast tumors remain largely undefined. Here we show that geminin-dependent acetylation releases HMGB1 from the chromatin to the cytoplasm and extracellular space. Extracellular acetylated HMGB1 (Ac-HMGB1) promotes geminin overexpressing (GemOE) cells survival by binding to RAGE and activating NF-κB signaling. Extracellular Ac-HMGB1 also triggers expression and activation of RAGE in the non-expressing MSCs. RAGE activation induces expression of CXCR4 in MSCs and directional migration towards SDF1 (aka CXCL12)-expressing GemOE cells in vitro and in vivo. These effects augmented by the necrotic and hypoxic environment in GemOE tumors, especially within their cores. Reciprocal interactions between newly recruited MSCs and GemOE tumor cells elevate tumor-initiating (TIC), basal and epithelial-to-mesenchymal transition (EMT) traits and enhance aggressiveness in vitro and in vivo in GemOE tumor cells. Indeed, faster, larger and more aggressive tumors develop when GemOE cells are co-injected with MSCs in orthotopic breast tumor model. Concurrently, inhibiting c-Abl (and thus geminin function), RAGE or CXCR4 prevented MSCs recruitment to GemOE cells in vitro and in vivo, and decreased the TIC, basal and EMT phenotypes in these tumor cells. Accordingly, we propose that GemOE tumor cells present within tumor cores represent metastatic precursors, and suppressing the GemOE→HMGB1/RAGE→SDF1/CXCR4 signaling circuit could be a valid target for therapies to inhibit GemOE tumors and their metastases.


Subject(s)
Breast/pathology , Carcinoma, Ductal, Breast/secondary , Cell Transformation, Neoplastic/pathology , Geminin/metabolism , Mesenchymal Stem Cells/pathology , Triple Negative Breast Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Breast/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Chemokine CXCL12/metabolism , Epithelial-Mesenchymal Transition , Female , HMGB1 Protein/metabolism , Humans , Lymphatic Metastasis , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Invasiveness , Prognosis , Receptors, CXCR4/metabolism , Signal Transduction , Survival Rate , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
J Transl Med ; 14: 31, 2016 Jan 30.
Article in English | MEDLINE | ID: mdl-26830497

ABSTRACT

BACKGROUND: Urine based assays that can non-invasively detect bladder cancer (BCa) have the potential to reduce unnecessary and invasive procedures. The purpose of this study was to develop a multiplex immunoassay that can accurately and simultaneously monitor ten diagnostic urinary protein biomarkers for application as a non-invasive test for BCa detection. METHODS: A custom electrochemiluminescent multiplex assay was constructed (Meso Scale Diagnostics, LLC, Rockville, MD, USA) to detect the following urinary proteins; IL8, MMP9, MMP10, ANG, APOE, SDC1, A1AT, PAI1, CA9 and VEGFA. Voided urine samples from two cohorts were collected prior to cystoscopy and samples were analyzed blinded to the clinical status of the participants. Means (±SD) and receiver operating characteristic (ROC) curve analysis were used to compare assay performance and to assess the diagnostic accuracy of the diagnostic signature. RESULTS: Comparative diagnostic performance analyses revealed an AUROC value of 0.9258 for the multiplex assay and 0.9467 for the combination of the single-target ELISA assays (p = 0.625), so there was no loss of diagnostic utility for the MSD multiplex assay. Analysis of the independent 200-sample cohort using the multiplex assay achieved an overall diagnostic sensitivity of 0.85, specificity of 0.81, positive predictive value 0.82 and negative predictive value 0.84. CONCLUSIONS: It is technically feasible to simultaneously monitor complex urinary diagnostic signatures in a single assay without loss of performance. The described protein-based assay has the potential to be developed for the non-invasive detection of BCa.


Subject(s)
Immunoassay/methods , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor/urine , Cohort Studies , Demography , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Reproducibility of Results , Urinary Bladder Neoplasms/urine
14.
Oncotarget ; 7(13): 15747-56, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26894971

ABSTRACT

Anterior gradient 2 (AGR2) is a cancer-associated secreted protein found predominantly in adenocarcinomas. Given its ubiquity in solid tumors, cancer-secreted AGR2 could be a useful biomarker in urine or blood for early detection. However, normal organs express and might also secrete AGR2, which would impact its utility as a cancer biomarker. Uniform AGR2 expression is found in the normal bladder urothelium. Little AGR2 is secreted by the urothelial cells as no measurable amounts could be detected in urine. The urinary proteomes of healthy people contain no listing for AGR2. Likewise, the blood proteomes of healthy people also contain no significant peptide counts for AGR2 suggesting little urothelial secretion into capillaries of the lamina propria. Expression of AGR2 is lost in urothelial carcinoma, with only 25% of primary tumors observed to retain AGR2 expression in a cohort of lymph node-positive cases. AGR2 is secreted by the urothelial carcinoma cells as urinary AGR2 was measured in the voided urine of 25% of the cases analyzed in a cohort of cancer vs. non-cancer patients. The fraction of AGR2-positive urine samples was consistent with the fraction of urothelial carcinoma that stained positive for AGR2. Since cancer cells secrete AGR2 while normal cells do not, its measurement in body fluids could be used to indicate tumor presence. Furthermore, AGR2 has also been found on the cell surface of cancer cells. Taken together, secretion and cell surface localization of AGR2 are characteristic of cancer, while expression of AGR2 by itself is not.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Transitional Cell/metabolism , Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism , Cell Line, Tumor , Humans , Mucoproteins , Oncogene Proteins
15.
Springerplus ; 4: 610, 2015.
Article in English | MEDLINE | ID: mdl-26543745

ABSTRACT

Chemokines, including chemokine (C-X-C motif) ligand 1 (CXCL1), regulate tumor epithelial-stromal interactions that facilitate tumor growth and invasion. Recently, several studies have linked CXCL1 expression to bladder cancer (BCa). In this study, we aimed to determine if increased levels of urinary CXCL1 were found in BCa patients. Voided urines from 86 subjects, cancer subjects (n = 43), non-cancer subjects (n = 43) were analyzed. The protein concentration of CXCL1 was assessed by enzyme-linked immunosorbent assay (ELISA). CXCL1 concentration level was normalized using urinary protein and urinary creatinine concentrations. We used the area under the curve of a receiver operating characteristic (AUROC) to investigate the performance of CXCL1 in detecting BCa. Mean urinary concentrations of CXCL1 were significantly higher in subjects with BCa compared to subjects without BCa (179.8 ± 371.7 pg/mg of creatinine vs. 28.2 ± 71.9 pg/mg, respectively p = 0.0009). Urinary CXCL1 possessed a sensitivity of 55.81 %, specificity of 83.72 %, positive predictive value of 77.42 %, negative predictive value of 65.46 %, and an overall accuracy of 69.77 % (AUROC: 0.7015, 95 % CI 0.5903-0.8126). These results indicate that CXCL1 is elevated in BCa when compared to non-cancer subjects, but lacks robustness as a standalone urinary biomarker. Additional studies into CXCL1 may shed more light on the role of CXCL1 in BCa tumorigenesis as well as ramifications of therapeutically targeting CXCL1.

16.
Chromosome Res ; 23(4): 649-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25947045

ABSTRACT

We have developed a convenient multicolor fluorescent in situ hybridization (FISH) (five-, four-, three-, and two-color FISHs) for detecting specific genes/DNA segments on the human chromosomes. As a foundation of multicolor FISH, we first isolated 80 bacterial artificial chromosome (BAC) probes that specifically detect the peri-centromeres (peri-CEN) and subtelomeres (subTEL) of 24 different human chromosomes (nos. 1~22, X, and Y) by screening our homemade BAC library (Keio BAC library) consisting of 200,000 clones. Five-color FISH was performed using human DNA segments specific for peri-CEN or subTEL, which were labeled with five different fluorescent dyes [7-diethylaminocoumarin (DEAC): blue, fluorescein isothiocyanate (FITC): green, rhodamine 6G (R6G): yellow, TexRed: red, and cyanine5 (Cy5): purple]. To observe FISH signals under a fluorescence microscope, five optic filters were carefully chosen to avoid overlapping fluorescence emission. Five-color FISH and four-color FISH enabled us to accurately examine the numerical anomaly of human chromosomes. Three-color FISH using two specific BAC clones, that distinguish 5' half of oncogene epidermal growth factor receptor (EGFR) from its 3' half, revealed the amplification and truncation of EGFR in EGFR-overproducing cancer cells. Moreover, two-color FISH readily detected a fusion gene in leukemia cells such as breakpoint cluster region (BCR)/Abelson murine leukemia viral oncogene homologue (ABL) on the Philadelphia (Ph') chromosome with interchromosomal translocation. Some other successful cases such as trisomy 21 of Down syndrome are presented. Potential applications of multicolor FISH will be discussed.


Subject(s)
Chromosomes, Human , In Situ Hybridization, Fluorescence/methods , Chromosome Aberrations , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosomes, Artificial, Bacterial , DNA Probes , Genomic Library , Humans , Microscopy, Fluorescence, Multiphoton , Staining and Labeling
17.
Biosci Biotechnol Biochem ; 78(7): 1123-8, 2014.
Article in English | MEDLINE | ID: mdl-25229845

ABSTRACT

Resveratrol was glucosylated to its 3- and 4'-ß-glucosides by cultured cells of Phytolacca americana. On the other hand, cultured P. americana cells glucosylated pterostilbene to its 4'-ß-glucoside. P. americana cells converted piceatannol into its 4'-ß-glucoside. The 3- and 4'-ß-glucosides of resveratrol were further glucosylated to 3- and 4'-ß-maltosides of resveratrol, 4'-ß-maltoside of which is a new compound, by cyclodextrin glucanotransferase. Resveratrol 3-ß-glucoside and 3-ß-maltoside showed low 2,2-diphenyl-1-picrylhydrazyl free-radical-scavenging activity, whereas other glucosides had no radical-scavenging activity. Piceatannol 4'-ß-glucoside showed the strongest inhibitory activity among the stilbene glycosides towards histamine release from rat peritoneal mast cells. Pterostilbene 4'-ß-glucoside showed high phosphodiesterase inhibitory activity.


Subject(s)
Glycosides/chemistry , Stilbenes/chemical synthesis , Stilbenes/pharmacology , Animals , Anti-Allergic Agents/chemical synthesis , Anti-Allergic Agents/chemistry , Anti-Allergic Agents/pharmacology , Biphenyl Compounds , Cell Line , Chemistry Techniques, Synthetic , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Histamine Release/drug effects , Male , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Picrates , Rats , Resveratrol , Stilbenes/chemistry
18.
Cancer Cell Int ; 14(1): 76, 2014.
Article in English | MEDLINE | ID: mdl-25197261

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is characterized by aggressive loco-regional invasion. Sphingosine kinase1 (SphK1), an enzyme in sphingolipid metabolism, is emerging as a key player in HNSCC pathology. The observation that SphK1 is overexpressed in all HNSCC stages and is associated with depth of tumor invasion, metastasis and clinical failure underscores the importance of SphK1 in HNSCC pathology. Still, the mechanisms underlying SphK1 regulation of invasion have not been delineated. Therefore, we sought to mechanistically describe how SphK1 regulates invasion in HNSCC. METHODS: Invasion assays were used to measure invasive ability of SphK1 overexpressing human tongue squamous cell carcinoma (SCC-25 cells). Western blotting, quantitative qPCR, ELISA and zymography were used to measure the effect of SphK1 and sphingosine 1-phoshate receptor 1 (S1P1) on invasion measures, MMP-2/9, E-cadherin, EGFR, IL-6/STAT3, in SCC-25 cells. RESULTS: SphK1 expression is elevated in cells with an invasive phenotype as compared to non-invasive phenotype. We show SphK1 overexpression increased EGF-induced EGFR/ERK and AKT activity, increased matrix metalloproteinase (MMP)-2/9 mRNA and reduced E-cadherin. SphK1 overexpression also increased IL-6 concentration and EGF-induced STAT3 phosphorylation, exemplifying that SphK1 modulates IL-6/STAT3 signaling. Notably, we show that S1P1 knockdown reduced IL-6/STAT3 signaling, representing another pathway by which SphK1/S1P regulates invasion. CONCLUSIONS: Taken together, our data suggest that SphK1 sits at the hub of multiple key signaling cascades, all which have been implicated in the regulation of invasiveness, making SphK1 an attractive target for the development of HNSCC therapies.

19.
FASEB J ; 27(2): 656-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23109673

ABSTRACT

Accumulating evidence suggests that sphingosine kinase 1 (SphK1) plays a key role in carcinogenesis by regulating cyclooxygenase-2 (COX-2) expression. Recent clinical studies have revealed that COX-2 inhibitors cause adverse cardiovascular side effects, likely due to inhibition of prostacyclin (PGI(2)). In this work, we investigated the roles of SphK1 inhibition on blood pressure (BP). The results show that lack of SphK1 expression did not exacerbate angiotensin II (Ang II)-induced acute hypertension, whereas celecoxib, a COX-2 inhibitor, augmented and sustained higher BP in mice. Interestingly, SphK1-knockout mice inhibited prostaglandin E(2) (PGE(2)) but not PGI(2) production in response to Ang II, whereas celecoxib blocked both PGE(2) and PGI(2) production. Mechanistically, SphK1 down-regulation by siRNA in human umbilical vein endothelial cells decreased cytokine-induced PGE(2) production primarily through inhibition of microsomal PGE synthase-1 (mPGES-1), not COX-2. SphK1 down-regulation also decreased MKK6 expression, which phosphorylates and activates P38 MAPK, which, in turn, regulates early growth response-1 (Egr-1), a transcription factor of mPGES-1. Together, these data indicate that SphK1 regulates PGE(2) production by mPGES-1 expression via the p38 MAPK pathway, independent of COX-2 signaling, in endothelial cells, suggesting that SphK1 inhibition may be a promising strategy for cancer chemoprevention with lack of the adverse cardiovascular side effects associated with coxibs.


Subject(s)
Blood Pressure/physiology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Base Sequence , Celecoxib , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/biosynthesis , Epoprostenol/biosynthesis , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/drug therapy , Hypertension/physiopathology , Intramolecular Oxidoreductases/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mitochondrial Proteins , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Prostaglandin-E Synthases , Pyrazoles/pharmacology , RNA, Small Interfering/genetics , Sulfonamides/pharmacology
20.
Biomolecules ; 3(3): 481-513, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-24970177

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) has a high reoccurrence rate and an extremely low survival rate. There is limited availability of effective therapies to reduce the rate of recurrence, resulting in high morbidity and mortality of advanced cases. Late presentation, delay in detection of lesions, and a high rate of metastasis make HNSCC a devastating disease. This review offers insight into the role of sphingosine kinase-1 (SphK1), a key enzyme in sphingolipid metabolism, in HNSCC. Sphingolipids not only play a structural role in cellular membranes, but also modulate cell signal transduction pathways to influence biological outcomes such as senescence, differentiation, apoptosis, migration, proliferation, and angiogenesis. SphK1 is a critical regulator of the delicate balance between proliferation and apoptosis. The highest expression of SphK1 is found in the advanced stage of disease, and there is a positive correlation between SphK1 expression and recurrent tumors. On the other hand, silencing SphK1 reduces HNSCC tumor growth and sensitizes tumors to radiation-induced death.  Thus, SphK1 plays an important and influential role in determining HNSCC proliferation and metastasis. We discuss roles of SphK1 and other sphingolipids in HNSCC development and therapeutic strategies against HNSCC.

SELECTION OF CITATIONS
SEARCH DETAIL
...