Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Clin Cancer Res ; 28(1): 175-186, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34667024

ABSTRACT

PURPOSE: Dedifferentiated liposarcoma (DDLS), one of the most common and aggressive sarcomas, infrequently responds to chemotherapy. DDLS survival and growth depend on underexpression of C/EBPα, a tumor suppressor and transcriptional regulator controlling adipogenesis. We sought to screen and prioritize candidate drugs that increase C/EBPα expression and may therefore serve as differentiation-based therapies for DDLS. EXPERIMENTAL DESIGN: We screened known bioactive compounds for the ability to restore C/EBPα expression and inhibit proliferation selectively in two DDLS cell lines but not in normal adipose-derived stem cells (ASC). Selected hits' activity was validated, and the mechanism of the most potent, SN-38, was investigated. The in vivo efficacy of irinotecan, the prodrug of SN-38, was evaluated in DDLS xenograft models. RESULTS: Of 3,119 compounds, screen criteria were met by 19. Validation experiments confirmed the DDLS selectivity of deguelin, emetine, and SN-38 and showed that they induce apoptosis in DDLS cells. SN-38 had the lowest IC50 (approximately 10 nmol/L), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53. Irinotecan significantly inhibited tumor growth at well-tolerated doses, induced nuclear expression of C/EBPα, and inhibited HIF1α expression in DDLS patient-derived and cancer cell line xenograft models. In contrast, doxorubicin, the most common treatment for nonresectable DDLS, reduced tumor growth by 30% to 50% at a dose that caused weight loss. CONCLUSIONS: This high-content screen revealed potential treatments for DDLS. These include irinotecan, which induces apoptosis of DDLS cells in a C/EBPα-dependent, p53-independent manner, and should be clinically evaluated in patients with advanced DDLS.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha , CCAAT-Enhancer-Binding Proteins , Liposarcoma , Adipocytes/metabolism , CCAAT-Enhancer-Binding Protein-alpha/analysis , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Proteins/analysis , Genes, Tumor Suppressor , Humans , Liposarcoma/drug therapy , Liposarcoma/genetics , Liposarcoma/pathology , Stem Cells/metabolism
2.
Cancer Invest ; 38(4): 240-249, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32212938

ABSTRACT

We evaluated the value of UHRF1, a regulator of methylation, as a biomarker for lung cancer. UHRF1 is expressed at higher levels in both lung adenocarcinoma (AD) and squamous cell carcinoma (SQ); however, a meta-analysis showed that UHRF1 expression is correlated with worse survival in patients with AD but not in those with SQ. UHRF1 knockdown suppressed the growth of lung cancer cell lines through G1 cell cycle arrest in some cell lines. These results suggest that UHRF1 may server as a diagnostic marker for AD and SQ and as a prognostic marker for AD in lung cancer.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Biomarkers, Tumor/analysis , CCAAT-Enhancer-Binding Proteins/analysis , Carcinoma, Squamous Cell/diagnosis , Lung Neoplasms/diagnosis , Ubiquitin-Protein Ligases/analysis , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Computational Biology , DNA Methylation , Datasets as Topic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Prognosis , RNA Interference , Survival Analysis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Ann Hematol ; 97(10): 1757-1765, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29785446

ABSTRACT

Allogeneic hematopoietic stem cell transplantation is an established consolidation therapy for patients with acute myeloid leukemia. However, relapse after transplantation remains a major clinical problem resulting in poor prognosis. Thus, detection of measurable ("minimal") residual disease to identify patients at high risk of relapse is essential. A feasible method to determine measurable residual disease may be digital droplet PCR (ddPCR) that allows absolute quantification with high sensitivity and specificity without the necessity of standard curves. Using ddPCR, we analyzed pre-transplant peripheral blood and bone marrow of 51 NPM1-mutated acute myeloid leukemia patients transplanted in complete remission or complete remission with incomplete recovery. Mutated NPM1 measurable residual disease-positive patients had higher cumulative incidence of relapse (P < 0.001) and shorter overall survival (P = 0.014). Restricting the analyses to patients receiving non-myeloablative conditioning, mutated NPM1 measurable residual disease positivity is associated with higher cumulative incidence of relapse (P < 0.001) and shorter overall survival (P = 0.006). Positive mutated NPM1 measurable residual disease status determined by ddPCR before allogeneic stem cell transplantation is associated with worse prognosis independent of other known prognostic markers-also for those receiving non-myeloablative conditioning. In the future, mutated NPM1 measurable residual disease status determined by ddPCR might guide treatment and improve patients' outcomes.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Polymerase Chain Reaction/methods , Preoperative Care , Adult , Aged , Allografts , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Bone Marrow/chemistry , Bone Marrow Transplantation , CCAAT-Enhancer-Binding Proteins/analysis , Combined Modality Therapy , DNA, Neoplasm/genetics , Female , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/blood , Neoplasm, Residual , Nuclear Proteins/analysis , Nuclear Proteins/blood , Nucleophosmin , Peripheral Blood Stem Cell Transplantation , Prognosis , Recurrence , Remission Induction , Retrospective Studies , Sensitivity and Specificity , Transplantation Conditioning/methods , Treatment Outcome , fms-Like Tyrosine Kinase 3/analysis
4.
Theranostics ; 8(4): 1084-1105, 2018.
Article in English | MEDLINE | ID: mdl-29464001

ABSTRACT

Rationale: PIWI-interacting RNAs (piRNAs), a class of newly discovered small RNA molecules that function by binding to the Argonaute protein family (i.e., the PIWIL protein subfamily), and long noncoding RNAs (lncRNA) are implicated in several cancers. However, the detailed roles of ncRNAs in glioma remain unclear. Methods: The expression of PIWIL3, piR-30188, OIP5-AS1, miR-367, CEBPA and TRAF4 were measured in glioma tissues and cells. The role of PIWIL3/OIP5-AS1/miR-367-3p/CEBPA feedback loop was evaluated in cell and animal models. The association of the above molecules was analyzed. Results: Over-expression of PIWIL3, piR-30188 and miR-367-3p or knockdown of OIP5-AS1 resulted in inhibition of glioma cells progression. Binding sites between piR-30188 and OIP5-AS1 as well as between OIP5-AS1 and miR-367-3p were confirmed by RNA immunoprecipitation and luciferase assays. OIP5-AS1 knockdown or miR-367-3p over-expression contributed to a decrease in CEBPA (CCAAT/enhancer binding protein alpha) protein. Furthermore, CEBPA was detected as a target of miR-367-3p and played an oncogenic role in glioma. Treatment with CEBPA and miR-367-3p resulted in the modulation of downstream TRAF4 (TNF receptor-associated factor 4). PIWIL3 was also a target of CEBPA, forming a positive feedback loop in the growth regulation of glioma cells. Significantly, knockdown of OIP5-AS1 combined with over-expression of PIWIL3 and miR-367-3p resulted in tumor regression and extended survival in vivo. Conclusion: These results identified a novel molecular pathway in glioma cells that may provide a potential innovative approach for tumor therapy.


Subject(s)
Argonaute Proteins/analysis , CCAAT-Enhancer-Binding Proteins/analysis , Glioma/pathology , MicroRNAs/analysis , Neuroglia/physiology , RNA, Long Noncoding/analysis , Animals , Cell Line, Tumor , Disease Models, Animal , Feedback, Physiological , Gene Expression Profiling , Heterografts , Humans , Mice, Nude , Neoplasm Transplantation
5.
Acta Cir Bras ; 33(1): 1-13, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29412228

ABSTRACT

PURPOSE: To investigate the effect of dexmedetomidine (Dex) in a rat ex vivo lung model of ischemia-reperfusion injury. METHODS: An IL-2 ex vivo lung perfusion system was used to establish a rat ex vivo lung model of ischemia-reperfusion injury. Drugs were added to the perfusion solution for reperfusion. Lung injury was assessed by histopathological changes, airway pressure (Res), lung compliance (Compl), perfusion flow (Flow), pulmonary venous oxygen partial pressure (PaO2), and lung wet/dry (W/D) weight ratio. The levels of superoxide dismutase (SOD), malondialdehyde (MDA), 78 kDa glucose-regulated protein (GRP78) and CCAAT/enhancer-binding protein homologous protein (CHOP) were measured, respectively. RESULTS: The introduction of Dex attenuated the post-ischemia-reperfusion lung damage and MDA level, improved lung histology, W/D ratio, lung injury scores and SOD activity. Decreased mRNA and protein levels of GRP78 and CHOP compared with the IR group were observed after Dex treatment. The effect of Dex was dosage-dependence and a high dose of Dex (10 nM) was shown to confer the strongest protective effect against lung damage (P<0.05). Yohimbine, an α2 receptor antagonist, significantly reversed the protective effect of Dex in lung tissues (P<0.05). CONCLUSION: Dex reduced ischemia-reperfusion injury in rat ex vivo lungs.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Ischemia/prevention & control , Lung/blood supply , Reperfusion Injury/prevention & control , Animals , Blotting, Western , CCAAT-Enhancer-Binding Proteins/analysis , Disease Models, Animal , Heat-Shock Proteins/analysis , Lung/pathology , Male , Malondialdehyde/analysis , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reference Values , Reperfusion Injury/pathology , Reproducibility of Results , Superoxide Dismutase/analysis , Time Factors , Treatment Outcome
6.
Acta cir. bras ; 33(1): 1-13, Jan. 2018. tab, graf
Article in English | LILACS | ID: biblio-886254

ABSTRACT

Abstract Purpose: To investigate the effect of dexmedetomidine (Dex) in a rat ex vivo lung model of ischemia-reperfusion injury. Methods: An IL-2 ex vivo lung perfusion system was used to establish a rat ex vivo lung model of ischemia-reperfusion injury. Drugs were added to the perfusion solution for reperfusion. Lung injury was assessed by histopathological changes, airway pressure (Res), lung compliance (Compl), perfusion flow (Flow), pulmonary venous oxygen partial pressure (PaO2), and lung wet/dry (W/D) weight ratio. The levels of superoxide dismutase (SOD), malondialdehyde (MDA), 78 kDa glucose-regulated protein (GRP78) and CCAAT/enhancer-binding protein homologous protein (CHOP) were measured, respectively. Results: The introduction of Dex attenuated the post-ischemia-reperfusion lung damage and MDA level, improved lung histology, W/D ratio, lung injury scores and SOD activity. Decreased mRNA and protein levels of GRP78 and CHOP compared with the IR group were observed after Dex treatment. The effect of Dex was dosage-dependence and a high dose of Dex (10 nM) was shown to confer the strongest protective effect against lung damage (P<0.05). Yohimbine, an α2 receptor antagonist, significantly reversed the protective effect of Dex in lung tissues (P<0.05). Conclusion: Dex reduced ischemia-reperfusion injury in rat ex vivo lungs.


Subject(s)
Animals , Male , Reperfusion Injury/prevention & control , Dexmedetomidine/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Ischemia/prevention & control , Lung/blood supply , Reference Values , Superoxide Dismutase/analysis , Time Factors , Reperfusion Injury/pathology , Blotting, Western , Reproducibility of Results , Treatment Outcome , Rats, Sprague-Dawley , CCAAT-Enhancer-Binding Proteins/analysis , Disease Models, Animal , Real-Time Polymerase Chain Reaction , Heat-Shock Proteins/analysis , Lung/pathology , Malondialdehyde/analysis
7.
Int. braz. j. urol ; 43(2): 224-229, Mar.-Apr. 2017. tab, graf
Article in English | LILACS | ID: biblio-840826

ABSTRACT

ABSTRACT Introduction Recently, expression of the UHRF1 gene was found to be up-regulated in numerous neoplasms, including the urinary bladder transitional cell carcinoma (TCC). Objective The aim of our study was to determine if the expression levels of UHRF1 gene correlates with the major pathological characteristics of the tumor and patients’ clinical outcome. Materials and Methods In our study, we have analyzed the tissue samples derived from group of 70 patients with histologically confirmed TCC of the urinary bladder, while normal urinary bladder mucosa obtained from 40 patients with nonmalignant diseases was used as a negative control group. Expression of UHRF1 gene in each patient sample was determined using reverse transcriptase-polymerase chain reaction. Results UHRF1 gene expression was found to be app. 2.5 times higher in samples from patients with TCC in comparison with normal epithelium derived from control group patients. Analysis show that gene expression correlates with the malignancy of the tumor. A highly significant differences were found between the expression values of samples from low and high grade TCC, as well as between the high grade and control group. UHRF1 expression was higher in patients with non-muscle invasive disease than in those with muscle invasive disease. Conclusions The result of this study indicates that UHRF1 gene expression levels correlates with the major pathological characteristics of TCC samples and with the clinical outcome of those patients. Determination of UHRF1 gene expression could have a potential to be used as a sensitive molecular marker in patients with urinary bladder cancer.


Subject(s)
Humans , Male , Female , Adult , Aged , Aged, 80 and over , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Gene Expression Regulation, Neoplastic , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/genetics , Reference Values , Urinary Bladder/pathology , Genetic Markers , Statistics, Nonparametric , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases , Tumor Burden , Neoplasm Grading , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging
8.
Int Braz J Urol ; 43(2): 224-229, 2017.
Article in English | MEDLINE | ID: mdl-28128913

ABSTRACT

INTRODUCTION: Recently, expression of the UHRF1 gene was found to be up-regulated in numerous neoplasms, including the urinary bladder transitional cell carcinoma (TCC). OBJECTIVE: The aim of our study was to determine if the expression levels of UHRF1 gene correlates with the major pathological characteristics of the tumor and patients' clinical outcome. MATERIALS AND METHODS: In our study, we have analyzed the tissue samples derived from group of 70 patients with histologically confirmed TCC of the urinary bladder, while normal urinary bladder mucosa obtained from 40 patients with nonmalignant diseases was used as a negative control group. Expression of UHRF1 gene in each patient sample was determined using reverse transcriptase-polymerase chain reaction. RESULTS: UHRF1 gene expression was found to be app. 2.5 times higher in samples from patients with TCC in comparison with normal epithelium derived from control group patients. Analysis show that gene expression correlates with the malignancy of the tumor. A highly significant differences were found between the expression values of samples from low and high grade TCC, as well as between the high grade and control group. UHRF1 expression was higher in patients with non-muscle invasive disease than in those with muscle invasive disease. CONCLUSIONS: The result of this study indicates that UHRF1 gene expression levels correlates with the major pathological characteristics of TCC samples and with the clinical outcome of those patients. Determination of UHRF1 gene expression could have a potential to be used as a sensitive molecular marker in patients with urinary bladder cancer.


Subject(s)
CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/genetics , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Female , Genetic Markers , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Tumor Burden , Ubiquitin-Protein Ligases , Urinary Bladder/pathology
9.
Thorax ; 71(1): 52-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26543090

ABSTRACT

BACKGROUND: Sensitisation with Aspergillus fumigatus (Af) is known to be associated with severe allergic lung inflammation, but the mechanism remains to be clarified. Phosphoinositide 3-kinase (PI3K)-δ and endoplasmic reticulum (ER) stress are suggested to be involved in steroid-resistant lung inflammation. We aimed to elucidate the role of PI3K-δ and its relationship with ER stress in fungus-induced allergic lung inflammation. METHODS: Using Af-exposed in vivo and in vitro experimental systems, we examined whether PI3K-δ regulates ER stress, thereby contributing to steroid resistance in fungus-induced allergic lung inflammation. Moreover, we checked expression of an ER stress marker in lung tissues isolated from patients with allergic bronchopulmonary aspergillosis. RESULTS: Af-exposed mice showed that ER stress markers, unfolded protein response (UPR)-related proteins, phosphorylated Akt, generation of mitochondrial reactive oxygen species (mtROS), eosinophilic allergic inflammation, and airway hyperresponsiveness (AHR) were increased in the lung. Similarly, glucose-regulated protein 78 was increased in lung tissues of patients with ABPA. A PI3K-δ inhibitor reduced Af-induced increases in ER stress markers, UPR-related proteins, allergic inflammation and AHR in mice. However, dexamethasone failed to reduce Af-induced allergic inflammation, AHR and elevation of ER stress. Administration of an ER stress inhibitor or a mtROS scavenger improved Af-induced allergic inflammation. The PI3K-δ inhibitor reduced Af-induced mtROS generation and the mtROS scavenger ameliorated ER stress. In primary cultured tracheal epithelial cells, Af-induced ER stress was inhibited by blockade of PI3K-δ. CONCLUSIONS: These findings suggest that PI3K-δ regulates Af-induced steroid-resistant eosinophilic allergic lung inflammation through ER stress.


Subject(s)
Aspergillosis, Allergic Bronchopulmonary/enzymology , Aspergillosis, Allergic Bronchopulmonary/etiology , Endoplasmic Reticulum Stress/physiology , Phosphatidylinositol 3-Kinases/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Biomarkers/analysis , Blotting, Western , Bronchoalveolar Lavage , CCAAT-Enhancer-Binding Proteins/analysis , Female , Glutathione/analysis , Glutathione Disulfide/analysis , Immunoglobulin E/blood , Inflammation/enzymology , Inflammation/etiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Quinazolines/pharmacology , RNA, Small Interfering/analysis
10.
Mol Cell Biol ; 36(5): 693-713, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26667036

ABSTRACT

The integrated stress response (ISR) controls cellular adaptations to nutrient deprivation, redox imbalances, and endoplasmic reticulum (ER) stress. ISR genes are upregulated in stressed cells, primarily by the bZIP transcription factor ATF4 through its recruitment to cis-regulatory C/EBP:ATF response elements (CAREs) together with a dimeric partner of uncertain identity. Here, we show that C/EBPγ:ATF4 heterodimers, but not C/EBPß:ATF4 dimers, are the predominant CARE-binding species in stressed cells. C/EBPγ and ATF4 associate with genomic CAREs in a mutually dependent manner and coregulate many ISR genes. In contrast, the C/EBP family members C/EBPß and C/EBP homologous protein (CHOP) were largely dispensable for induction of stress genes. Cebpg(-/-) mouse embryonic fibroblasts (MEFs) proliferate poorly and exhibit oxidative stress due to reduced glutathione levels and impaired expression of several glutathione biosynthesis pathway genes. Cebpg(-/-) mice (C57BL/6 background) display reduced body size and microphthalmia, similar to ATF4-null animals. In addition, C/EBPγ-deficient newborns die from atelectasis and respiratory failure, which can be mitigated by in utero exposure to the antioxidant, N-acetyl-cysteine. Cebpg(-/-) mice on a mixed strain background showed improved viability but, upon aging, developed significantly fewer malignant solid tumors than WT animals. Our findings identify C/EBPγ as a novel antioxidant regulator and an obligatory ATF4 partner that controls redox homeostasis in normal and cancerous cells.


Subject(s)
Activating Transcription Factor 4/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Oxidative Stress , Activating Transcription Factor 4/analysis , Activating Transcription Factor 4/genetics , Animals , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Female , Fetus/abnormalities , Fetus/metabolism , Gene Deletion , Gene Expression Regulation , Glutathione/metabolism , Humans , Male , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/metabolism , Protein Multimerization , Response Elements , Transcription Factor CHOP/metabolism
11.
Am J Clin Pathol ; 139(6): 818-25, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23690127

ABSTRACT

The classification of acute myeloid leukemia (AML) has evolved to the most recent World Health Organization (WHO) schema, which integrates genetic, morphologic, and prognostic data into a single system. However, this system was devised using adult data and how this system applies to a pediatric cohort is unknown. Performing a retrospective chart review, we examined our single-center experience with AML in 115 children and classified their leukemia using the WHO 2008 schema. We examined patient samples for mutations of FLT3, NPM1, and CEBPA. Overall survival was calculated within categories. In our pediatric population, most cases of AML had recurrent genetic abnormalities of favorable prognosis. More than 10% of patients in our series were categorized as AML, with myelodysplasia-related changes, an entity not well-described in pediatric patients. In addition, a large proportion of patients were categorized with secondary, therapy-related AML. To our knowledge, this is the first application of the WHO 2008 classification to a pediatric cohort. In comparison to adult studies, AML in the pediatric population shows a distinct distribution within the WHO 2008 classification.


Subject(s)
Leukemia, Myeloid, Acute/classification , Adolescent , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/genetics , Child , Child, Preschool , Down Syndrome/complications , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/genetics , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Nucleophosmin , Retrospective Studies , Risk , World Health Organization , Young Adult , fms-Like Tyrosine Kinase 3/analysis , fms-Like Tyrosine Kinase 3/genetics
12.
J Agric Food Chem ; 60(4): 1094-101, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22224971

ABSTRACT

Xanthigen is a source of punicic acid and fucoxanthin derived from pomegranate seed and brown seaweed, respectively with recognized triacylglycerol-lowering effects in humans, yet the mechanism remains to be fully elucidated. The present study investigated the inhibitory effects of Xanthigen, fucoxanthin, and punicic acid (70% in pomegranate seed oil) on the differentiation of 3T3-L1 preadipocytes. Xanthigen potently and dose-dependently suppressed accumulation of lipid droplets in adipocytes compared to its individual components, fucoxanthin and pomegranate seed oil. Western blot analysis revealed that Xanthigen markedly down-regulated the protein levels of key adipogenesis transcription factors peroxisome proliferator-activated receptor (PPAR)γ, CCAAT/enhancer binding protein (C/EBP) ß, and C/EBPδ as well as a key enzyme involved in adipogenesis, fatty acid synthase (FAS). Xanthigen up-regulated the NAD(+)-dependent histone deacetylases (SIRT1) and activated AMP-activated protein kinase (AMPK) signaling in differentiated 3T3-L1 adipocytes. In addition, Xanthigen may also stimulate insulin trigger signaling and result in Akt-dependent phosphorylation of forkhead/winged helix O (FoxO)1 and FoxO3a. These results indicate that Xanthigen suppresses adipocyte differentiation and lipid accumulation through multiple mechanisms and may have applications for the treatment of obesity.


Subject(s)
Adipocytes/cytology , Adipogenesis/drug effects , CCAAT-Enhancer-Binding Proteins/analysis , Cell Differentiation/drug effects , PPAR gamma/analysis , Plant Extracts/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Animals , Down-Regulation , Forkhead Transcription Factors/metabolism , Humans , Infant , Linolenic Acids/pharmacology , Mice , Signal Transduction , Sirtuin 1/metabolism , Xanthophylls/pharmacology
13.
Br J Cancer ; 103(2): 217-22, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20517312

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer deaths worldwide. As the sensitivity and specificity of current diagnostic markers are not perfect, we examined whether ubiquitin-like with PHD and ring finger domains 1 (UHRF1), which is overexpressed in various cancers but not yet examined in lung cancer in large scale, can be a novel diagnostic marker of lung cancer. METHODS: Immunohistochemical analysis using surgical specimens obtained from 56 US and 322 Japanese patients with lung cancer was performed. RESULTS: The UHRF1 was stained specifically in the nuclei of cancer cells, but not in the other cells. The UHRF1 expression was observed in all histological types of lung cancer, especially in non-adenocarcinomas (non-ADCs), both in the US and Japanese cases. In 322 Japanese non-small cell lung cancer (NSCLC) cases, UHRF1 expression was associated with the histological type (higher in non-ADCs; P<0.00001), gender (higher in male; P=0.00082), smoking (higher in smokers; P=0.00004), pT factor (higher in advanced stage; P=0.00010), and pN factor (higher in cancers with metastasis in regional lymph nodes; P=0.00018). The UHRF1 expression was also associated with poor prognosis for NSCLC patients (P=0.0364). Although UHRF1 overexpression was associated with these malignant indicators, UHRF1 was detectable in half of lung cancer patients in an early pathological stage. CONCLUSION: The UHRF1 is overexpressed in various types of lung cancer from early pathological stage. Therefore, detection of UHRF1 expression in tissue specimens by immunohistochemistry can be useful for diagnosis of lung cancer in all pathological stages.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/analysis , CCAAT-Enhancer-Binding Proteins/analysis , Lung Neoplasms/diagnosis , Neoplasms, Squamous Cell/diagnosis , Adenocarcinoma/metabolism , Aged , Female , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Male , Neoplasms, Squamous Cell/metabolism , Smoking , Ubiquitin-Protein Ligases
14.
Biomed Pharmacother ; 64(7): 451-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20359855

ABSTRACT

BACKGROUND: Gallbladder carcinoma is known to be an aggressive malignancy and nonsensitive to routine chemotherapy. Its prognosis is quite poor. We have illustrated that somatostatin (SST) can enhance chemosensitivity of gallbladder cancer to Doxorubicin (DOX) in our precious studies. Here, we explored the possible mechanisms by which SST used to enhance the cytotoxicity of DOX on gallbladder carcinoma cell line. METHODS: Human gallbladder cancer cells line (GBC-SD cell line) were divided into four groups: control group, SST group, DOX group, SST+DOX co-treated-group. Cell cycle was detected by flow cytometry (FCM). Cell apoptosis index was detected by using Annexin V/Propidium Iodide Binding on FCM. The expressions of certain key cell cycle-related factors, including retinoblastoma protein (Rb) and E2F-1 protein were investigated by western blotting. ICBP90 protein, which could be a new downstream effector of E2F-1, was also detected by western blotting. The expression of Topo IIα protein, target enzyme of DOX, was assessed in synchronized GBC-SD cells by western blotting. RESULTS: After 24h treatment with SST alone, cell cycle was arrested at S phase in GBC-SD cells line, followed by indistinctive increment of apoptosis index. After 24h treatment with SST and DOX, apoptosis index significantly increased than that of DOX alone (P<0.05). Compared with control group, the expressions of Rb and E2F-1 protein were significantly up-regulated at 24h after treatment with SST. Similarly, the expressions of ICBP90 and Topo IIα protein were also enhanced at 24h after treatment with SST. CONCLUSION: These results suggested that SST could induce cell cycle block in S phase in GBC-SD cells line, the most sensitive phase of the cell cycle for DOX, through up-regulating Rb, E2F-1 and ICBP90 protein expression. Furthermore, ICBP90 induced the enhanced expression of Topo IIα protein which is the target enzyme of DOX and enhanced its cytotoxic effect on GBC-SD cells. We concluded that the mechanisms of SST enhanced chemosensitivity of GBC-SD cell line to DOX might be cell cycle arrest plus up-regulated target enzyme.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Cycle/drug effects , Doxorubicin/pharmacology , Gallbladder Neoplasms/drug therapy , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , CCAAT-Binding Factor/metabolism , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Drug Therapy, Combination , E2F1 Transcription Factor/metabolism , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Humans , Retinoblastoma Protein/metabolism , Somatostatin/pharmacology , Ubiquitin-Protein Ligases , Up-Regulation/drug effects
15.
Nucleic Acids Res ; 37(2): 493-505, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056828

ABSTRACT

UHRF1 (ubiquitin-like, containing PHD and RING finger domains 1) is a multi-domain protein associated with cellular proliferation and epigenetic regulation. The UHRF1 binds to methylated CpG dinucleotides and recruits transcriptional repressors DNA methyltransferase 1 (DNMT1) and histone deacetylase 1 (HDAC1) through its distinct domains. However, the molecular basis of UHRF1-mediated transcriptional regulation via chromatin modifications is yet to be fully understood. Here we show that UHRF1 binds histone lysine methyltransferase G9a, and both are co-localized in the nucleus in a cell-cycle-dependent manner. Concurrent with the cell-cycle progression, gradual deposition of UHRF1 and G9a was observed, which mirrored H3K9me2 accumulation on chromatin. Murine Uhrf1-null embryonic stem (ES) cells displayed a reduced amount of G9a and H3K9me2 on chromatin. UHRF1 recruited and cooperated with G9a to inhibit the p21 promoter activity, which correlated with the elevated p21 protein level in both human UHRF1 siRNA-transfected HeLa cells and murine Uhrf1-null ES cells. Furthermore, endogenous p21 promoter remained bound to UHRF1, G9a, DNMT1 and HDAC1, and knockdown of UHRF1 impaired the association of all three chromatin modifiers with the promoter. Thus, our results suggest that UHRF1 may serve as a focal point of transcriptional regulation mediated by G9a and other chromatin modification enzymes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Silencing , Nuclear Proteins/metabolism , Protein Methyltransferases/metabolism , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Proteins/analysis , Cell Line , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , HeLa Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Humans , Mice , Nuclear Proteins/analysis , Promoter Regions, Genetic , Protein Methyltransferases/analysis , Ubiquitin-Protein Ligases
16.
Zebrafish ; 4(3): 187-99, 2007.
Article in English | MEDLINE | ID: mdl-18041923

ABSTRACT

The zebrafish has become a powerful tool for analysis of vertebrate hematopoiesis. Zebrafish, unlike mammals, have a robust primitive myeloid pathway that generates both granulocytes and macrophages. It is not clear how this unique primitive myeloid pathway relates to mammalian definitive hematopoiesis. In this study, we show that the two myeloid subsets can be distinguished using RNA in situ hybridization. Using a morpholino-antisense gene knockdown approach, we have characterized the hematopoietic defects resulting from knockdown of the myeloid transcription factor gene pu.1 and the unique zebrafish gene c/ebp1. Severe reduction of pu.1 resulted in complete loss of primitive macrophage development, with effects on granulocyte development only with maximal knockdown. Reduction of c/ebp1 did not ablate initial macrophage or granulocyte development, but resulted in loss of expression of the secondary granule gene lys C. These data reveal strong functional conservation of pu.1 between zebrafish primitive myelopoiesis and mammalian definitive myelopoiesis. Further, these results are consistent with a conserved role between c/ebp1 and mammalian C/EBPE, whose ortholog in zebrafish has not been identified. These studies validate the examination of zebrafish primitive myeloid development as a model for human myelopoiesis, and form a framework for identification and analysis of myeloid mutants.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Gene Expression Regulation, Developmental/genetics , Myelopoiesis/genetics , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Zebrafish/embryology , Zebrafish/genetics , Animals , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/genetics , Genetic Techniques , Granulocytes/physiology , In Situ Hybridization, Fluorescence , Macrophages/physiology , Metalloendopeptidases/analysis , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Microinjections , Models, Animal , Mutation/genetics , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA/analysis , RNA/metabolism , Trans-Activators/analysis , Trans-Activators/biosynthesis , Trans-Activators/genetics
17.
Proc Natl Acad Sci U S A ; 104(24): 10264-9, 2007 Jun 12.
Article in English | MEDLINE | ID: mdl-17551013

ABSTRACT

FRET is a well established method for cellular and subcellular imaging of protein interactions. However, FRET obligatorily necessitates fluorescence excitation with its concomitant problems of photobleaching, autofluorescence, phototoxicity, and undesirable stimulation of photobiological processes. A sister technique, bioluminescence resonance energy transfer (BRET), avoids these problems because it uses enzyme-catalyzed luminescence; however, BRET signals usually have been too dim to image effectively in the past. Using a new generation electron bombardment-charge-coupled device camera coupled to an image splitter, we demonstrate that BRET can be used to image protein interactions in plant and animal cells and in tissues; even subcellular imaging is possible. We have applied this technology to image two different protein interactions: (i) dimerization of the developmental regulator, COP1, in plant seedlings; and (ii) CCAAT/enhancer binding protein alpha (C/EBPalpha) in the mammalian nucleus. This advance heralds a host of applications for imaging without fluorescent excitation and its consequent limitations.


Subject(s)
Arabidopsis/cytology , Fluorescence Resonance Energy Transfer/methods , Luminescent Proteins/analysis , Nicotiana/cytology , Plant Proteins/analysis , Proteins/analysis , Animals , Arabidopsis/chemistry , Arabidopsis Proteins/analysis , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Culture Techniques , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Dimerization , Humans , Luminescent Measurements , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Pituitary Gland/cytology , Pituitary Gland/metabolism , Protein Binding , Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Seedlings/chemistry , Seedlings/cytology , Spectrometry, Fluorescence , Nicotiana/chemistry
18.
J Nutr Biochem ; 16(10): 617-24, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16081269

ABSTRACT

Flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) are essential coenzymes in redox reactions. For example, FAD is a coenzyme for both glutathione reductase and enzymes that mediate the oxidative folding of secretory proteins. Here we investigated short-term effects of moderately riboflavin-deficient culture medium on flavin-related responses in HepG2 hepatocarcinoma cells. Cells were cultured in riboflavin-deficient (3.1 nmol/l) medium for up to 6 days; controls were cultured in riboflavin-sufficient (532 nmol/l) medium. The activity of glutathione reductase decreased by 98% within 4 days of riboflavin-deficient culture. Transport rates of riboflavin increased in response to riboflavin depletion, whereas expression of enzymes mediating flavocoenzyme synthesis (flavokinase and FAD synthetase) decreased in response to depletion. The oxidative folding and synthesis of plasminogen and apolipoprotein B-100 was impaired within 4 days of culture in riboflavin-deficient medium; this is consistent with impaired processing of secretory proteins in riboflavin-deficient cells. Riboflavin depletion was associated with increased DNA-binding activities of transcription factors with affinity for endoplasmic reticulum stress elements and nuclear factor kappaB (NF-kappaB) consensus elements, suggesting cell stress. Moreover, the abundance of the stress-induced protein GADD153 was greater in riboflavin-deficient cells compared with controls. Riboflavin deficiency was associated with decreased rates of cell proliferation caused by arrest in G1 phase of the cell cycle. These studies are consistent with the hypothesis that HepG2 cells have a great demand for riboflavin and that cell stress develops rapidly if riboflavin supply is marginally low.


Subject(s)
Culture Media/chemistry , Riboflavin Deficiency , Riboflavin/physiology , Biological Transport , CCAAT-Enhancer-Binding Proteins/analysis , Carcinoma, Hepatocellular , Cell Division , Cell Line, Tumor , DNA/metabolism , Endoplasmic Reticulum/metabolism , G1 Phase , Glutathione Reductase/metabolism , Humans , Liver Neoplasms , NF-kappa B/metabolism , Nucleotidyltransferases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Biosynthesis , Protein Folding , Riboflavin/administration & dosage , Time Factors , Transcription Factor CHOP , Transcription Factors/analysis , Transcription Factors/metabolism
19.
Int J Artif Organs ; 28(1): 44-50, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15742309

ABSTRACT

In mammalian cells, cellular differentiation into specific cell types is usually preceded by growth arrest. On the other hand, the induced differentiation may also be preceded by an enhanced G1-S transition of the cell cycle prior to the growth arrest. This suggests that an early increase in proliferation is in some way a prerequisite for subsequent differentiation. We therefore attempted to assess whether we could produce human hepatocytes with further differentiated functions by promoting G1-S transition in a butyrate-treated human hepatocyte cell line. A cyclin E-over-expressing cell line was established by transfecting human cyclin E cDNA. Upon butyrate treatment, the cyclin E-over-expressing cells exhibited a significantly increased albumin-secreting and ammonia-detoxifying capacity when compared to the control cells. In particular, the ornithine transcarbamylase activity was increased in these cells. Collectively, these results implicate that the cyclin E over-expression may augment the hepatocyte-specific functions during the butyrate-induced differentiation process of human hepatocytes by enhancing G1-S cell cycle transition.


Subject(s)
Butyrates/pharmacology , Cyclin E/drug effects , Hepatocytes/drug effects , Albumins/metabolism , Ammonia/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/analysis , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Cyclin E/genetics , DNA-Binding Proteins/analysis , G1 Phase/drug effects , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 4 , Humans , Ornithine Carbamoyltransferase/drug effects , Phosphoproteins/analysis , S Phase/drug effects , Transcription Factor CHOP , Transcription Factors/analysis , Transfection , Urea/analysis
20.
Mol Cell Biol ; 25(8): 2946-56, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15798184

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are a subfamily of basic helix-loop-helix-leucine zipper proteins that regulate lipid metabolism. We show novel evidence of the in vivo occurrence and subnuclear spatial localization of both exogenously expressed SREBP-1a and -2 homodimers and heterodimers obtained by two-photon imaging and spectroscopy fluorescence resonance energy transfer. SREBP-1a homodimers localize diffusely in the nucleus, whereas SREBP-2 homodimers and the SREBP-1a/SREBP-2 heterodimer localize predominantly to nuclear speckles or foci, with some cells showing a diffuse pattern. We also used tethered SREBP dimers to demonstrate that both homo- and heterodimeric SREBPs activate transcription in vivo. Ultrastructural analysis revealed that the punctate foci containing SREBP-2 are electron-dense nuclear bodies, similar or identical to structures containing the promyelocyte (PML) protein. Immunofluorescence studies suggest that a dynamic interplay exists between PML, as well as another component of the PML-containing nuclear body, SUMO-1, and SREBP-2 within these nuclear structures. These findings provide new insight into the overall process of transcriptional activation mediated by the SREBP family.


Subject(s)
CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/physiology , Cell Nucleus/chemistry , DNA-Binding Proteins/analysis , DNA-Binding Proteins/physiology , Transcription Factors/analysis , Transcription Factors/physiology , Transcriptional Activation , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , DNA-Binding Proteins/genetics , Dimerization , Fluorescence Resonance Energy Transfer , Genes, Reporter/genetics , Humans , Lipid Metabolism , Luciferases/analysis , Luciferases/genetics , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Photons , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Protein , Protein Structure, Tertiary , Receptors, LDL/genetics , SUMO-1 Protein/analysis , SUMO-1 Protein/metabolism , Sequence Deletion , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...