Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(10): 2649-2664.e18, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33848463

ABSTRACT

Receptor tyrosine kinase (RTK)-mediated activation of downstream effector pathways such as the RAS GTPase/MAP kinase (MAPK) signaling cascade is thought to occur exclusively from lipid membrane compartments in mammalian cells. Here, we uncover a membraneless, protein granule-based subcellular structure that can organize RTK/RAS/MAPK signaling in cancer. Chimeric (fusion) oncoproteins involving certain RTKs including ALK and RET undergo de novo higher-order assembly into membraneless cytoplasmic protein granules that actively signal. These pathogenic biomolecular condensates locally concentrate the RAS activating complex GRB2/SOS1 and activate RAS in a lipid membrane-independent manner. RTK protein granule formation is critical for oncogenic RAS/MAPK signaling output in these cells. We identify a set of protein granule components and establish structural rules that define the formation of membraneless protein granules by RTK oncoproteins. Our findings reveal membraneless, higher-order cytoplasmic protein assembly as a distinct subcellular platform for organizing oncogenic RTK and RAS signaling.


Subject(s)
Biomolecular Condensates/metabolism , Cytoplasmic Granules/metabolism , Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Enzyme Activation , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HEK293 Cells , Humans , SOS1 Protein/metabolism , Signal Transduction
2.
J Org Chem ; 88(14): 9946-9958, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37410072

ABSTRACT

3-O-ß-Glucuronide triterpenes are plant-derived compounds. Some of them have been used as herbal medicine and in pharmaceuticals, such as chikusetsu saponins and Quillaja saponins. However, the demand for these materials has remained largely a challenge owing to their natural scarcity and low-yielding purification process. Therefore, a chemical triterpene 3-O-glucuronidation was conducted in this study to alleviate the surging demand on natural source. Various glucuronyl imidate donors and oleanane-type triterpene acceptors were synthesized, and the relative reactivity values (RRV) and acceptor nucleophilic constants (Aka) were systematically measured to study their influence on glucuronidation yield. As a result, applying donors in higher RRV value generally improved the production of 3-O-glucuronide triterpenes. Meanwhile, a bulky pivaloyl group was an ideal 2-O-protection to provide ß-selectivity and prevented side reactions, including orthoester formation and acyl-transfer reaction. Collectively, a positive correlation was observed between reactive donors/acceptors and improved glucuronidation yields. These findings offered insights on the influence of donors' and acceptors' reactivities on 3-O-ß-glucuronide triterpenes synthesis, and this knowledge would help to access saponins of interest to address future needs.


Subject(s)
Plants, Medicinal , Saponins , Triterpenes , Triterpenes/chemistry , Glucuronides , Plants, Medicinal/chemistry , Saponins/chemistry , Plant Extracts/chemistry
3.
Int J Cancer ; 151(9): 1611-1625, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35762443

ABSTRACT

High-grade neuroendocrine tumors (NETs) of the lung consist of small-cell lung cancer (SCLC) and large-cell neuroendocrine carcinoma (LCNEC). Both exhibit aggressive malignancy with poor prognosis. The transformation of lung adenocarcinoma (ADC) to SCLC or LCNEC also contributes to acquired resistance to epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs). Despite initially being responsive to chemotherapy, high-grade NET patients inevitably develop drug resistance; thus, novel therapeutic targets are urgently needed for these patients. Our study reported that VGF (nerve growth factor inducible), a factor mainly expressed in neurons during neural development, is highly expressed in SCLC and LCNEC as well as in a subset of ADCs, whereas targeting VGF attenuates cancer cell growth and tumor formation. High VGF expression was associated with advanced stage SCLC and predicted poor prognosis in lung ADC. In addition, EGFR-TKI selection enriched VGF expression in TKI-resistant ADC under epigenetic control. The VGF locus possessed the HDAC1 binding site, and treatment of ADC cells with the HDAC1 inhibitor induced VGF expression. High VGF expression was associated with chemoresistance, and silencing VGF induced BMF and BCL2L11 expression and rendered lung cancer cells sensitive to chemotherapy drugs. These findings suggested the potential of VGF as a prognostic factor and therapeutic target in lung cancers with neuroendocrine feature.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Large Cell , Carcinoma, Neuroendocrine , Lung Neoplasms , Small Cell Lung Carcinoma , Adenocarcinoma of Lung/drug therapy , Carcinoma, Large Cell/pathology , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Nerve Growth Factors , Prognosis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology
4.
J Cell Sci ; 133(8)2020 04 28.
Article in English | MEDLINE | ID: mdl-32220979

ABSTRACT

Myoblast fusion is required for myotube formation during myogenesis, and defects in myoblast differentiation and fusion have been implicated in a number of diseases, including human rhabdomyosarcoma. Although transcriptional regulation of the myogenic program has been studied extensively, the mechanisms controlling myoblast fusion remain largely unknown. This study identified and characterized the dynamics of a distinct class of blebs, termed bubbling blebs, which are smaller than those that participate in migration. The formation of these bubbling blebs occurred during differentiation and decreased alongside a decline in phosphatidylinositol-(3,4,5)-trisphosphate (PIP3) at the plasma membrane before myoblast fusion. In a human rhabdomyosarcoma-derived (RD) cell line that exhibits strong blebbing dynamics and myoblast fusion defects, PIP3 was constitutively abundant on the membrane during myogenesis. Targeting phosphatase and tensin homolog (PTEN) to the plasma membrane reduced PIP3 levels, inhibited bubbling blebs and rescued myoblast fusion defects in RD cells. These findings highlight the differential distribution and crucial role of PIP3 during myoblast fusion and reveal a novel mechanism underlying myogenesis defects in human rhabdomyosarcoma.


Subject(s)
Muscle Development , Rhabdomyosarcoma , Cell Differentiation , Cell Fusion , Humans , Muscle Development/genetics , Muscle Fibers, Skeletal , Myoblasts , Rhabdomyosarcoma/genetics
5.
Stem Cells ; 39(10): 1298-1309, 2021 10.
Article in English | MEDLINE | ID: mdl-34182610

ABSTRACT

Programmed death-ligand 1 (PD-L1), an immune checkpoint ligand, is recognized as a potential target for cancer immunotherapy as well as for the induction of transplantation tolerance. However, how the crosstalk between stem cell programming and cytokine signaling regulates PD-L1 expression during stem cell differentiation and cancer cell plasticity remains unclear. Herein, we reported that PD-L1 expression was regulated by SOX2 during embryonic stem cell (ESC) differentiation and lung cancer cell plasticity. PD-L1 was induced during ESC differentiation to fibroblasts and was downregulated during SOX2-mediated reprogramming of fibroblasts to induced pluripotent stem cells (iPSCs). Furthermore, SOX2 activation affected cancer cell plasticity and inhibited PD-L1 expression in lung cancer cells. We discovered that the H3K27ac signal at the PD-L1 locus was enhanced during ESC differentiation to fibroblasts as well as during cancer plasticity of SOX2-positive lung cancer cells to SOX2-negative counterparts. Romidepsin, an epigenetic modifier, induced PD-L1 expression in lung cancer cells, whereas TGF-ß stimulation downregulated SOX2 but upregulated PD-L1 expression in lung cancer cells. Furthermore, in addition to PD-L1, the expressions of EGFR and its ligand HBEGF were downregulated by activation of endogenous SOX2 expression during lung cancer cell plasticity and iPSC reprogramming, and the activation of EGFR signaling by HBEGF upregulated PD-L1 expression in lung cancer cells. Together, our results reveal the crosstalk between SOX2 programming and cytokine stimulation influences PD-L1 expression, and these findings may provide insights into PD-L1-mediated therapeutics.


Subject(s)
B7-H1 Antigen , Epigenesis, Genetic , Lung Neoplasms , B7-H1 Antigen/metabolism , Cell Differentiation/genetics , Cell Plasticity/genetics , Cytokines/metabolism , ErbB Receptors/metabolism , Humans , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Stem Cells/cytology
6.
Int J Mol Sci ; 23(16)2022 Aug 15.
Article in English | MEDLINE | ID: mdl-36012420

ABSTRACT

Although sepsis and acute kidney injury (AKI) have a bidirectional interplay, the pathophysiological mechanisms between AKI and sepsis are not clarified and worthy of a comprehensive and updated review. The primary pathophysiology of sepsis-associated AKI (SA-AKI) includes inflammatory cascade, macrovascular and microvascular dysfunction, cell cycle arrest, and apoptosis. The pathophysiology of sepsis following AKI contains fluid overload, hyperinflammatory state, immunosuppression, and infection associated with kidney replacement therapy and catheter cannulation. The preventive strategies for SA-AKI are non-specific, mainly focusing on infection control and preventing further kidney insults. On the other hand, the preventive strategies for sepsis following AKI might focus on decreasing some metabolites, cytokines, or molecules harmful to our immunity, supplementing vitamin D3 for its immunomodulation effect, and avoiding fluid overload and unnecessary catheter cannulation. To date, several limitations persistently prohibit the understanding of the bidirectional pathophysiologies. Conducting studies, such as the Kidney Precision Medicine Project, to investigate human kidney tissue and establishing parameters or scores better to determine the occurrence timing of sepsis and AKI and the definition of SA-AKI might be the prospects to unveil the mystery and improve the prognoses of AKI patients.


Subject(s)
Acute Kidney Injury , Sepsis , Apoptosis , Humans , Kidney , Renal Replacement Therapy , Sepsis/complications
7.
Int J Mol Sci ; 23(7)2022 Mar 26.
Article in English | MEDLINE | ID: mdl-35409017

ABSTRACT

Acute kidney injury (AKI) and gut dysbiosis affect each other bidirectionally. AKI induces microbiota alteration in the gastrointestinal (GI) system, while gut dysbiosis also aggravates AKI. The interplay between AKI and gut dysbiosis is not yet well clarified but worthy of further investigation. The current review focuses on the pathophysiology of this bidirectional interplay and AKI treatment in this base. Both macrophages and neutrophils of the innate immunity and the T helper type 17 cell from the adaptive immunity are the critical players of AKI-induced gut dysbiosis. Conversely, dysbiosis-induced overproduction of gut-derived uremic toxins and insufficient generation of short-chain fatty acids are the main factors deteriorating AKI. Many novel treatments are proposed to deter AKI progression by reforming the GI microbiome and breaking this vicious cycle. Data support the benefits of probiotic treatment in AKI patients, while the results of postbiotics are mainly limited to animals. Prebiotics and synbiotics are primarily discussed in chronic kidney disease patients rather than AKI patients. The effect of adsorbent treatment seems promising, but more studies are required before the treatment can be applied to patients. Immune therapy and some repurposed drugs such as allopurinol are prospects of future treatments and are worth more discussion and survey.


Subject(s)
Acute Kidney Injury , Gastrointestinal Microbiome , Probiotics , Synbiotics , Acute Kidney Injury/etiology , Acute Kidney Injury/therapy , Animals , Dysbiosis/metabolism , Dysbiosis/therapy , Humans , Prebiotics , Probiotics/therapeutic use
8.
Int J Mol Sci ; 23(14)2022 Jul 17.
Article in English | MEDLINE | ID: mdl-35887232

ABSTRACT

Ribose-5-phosphate isomerase A (RPIA) regulates tumorigenesis in liver and colorectal cancer. However, the role of RPIA in lung cancer remains obscure. Here we report that the suppression of RPIA diminishes cellular proliferation and activates autophagy, apoptosis, and cellular senescence in lung cancer cells. First, we detected that RPIA protein was increased in the human lung cancer versus adjust normal tissue via tissue array. Next, the knockdown of RPIA in lung cancer cells displayed autophagic vacuoles, enhanced acridine orange staining, GFP-LC3 punctae, accumulated autophagosomes, and showed elevated levels of LC3-II and reduced levels of p62, together suggesting that the suppression of RPIA stimulates autophagy in lung cancer cells. In addition, decreased RPIA expression induced apoptosis by increasing levels of Bax, cleaved PARP and caspase-3 and apoptotic cells. Moreover, RPIA knockdown triggered cellular senescence and increased p53 and p21 levels in lung cancer cells. Importantly, RPIA knockdown elevated reactive oxygen species (ROS) levels. Treatment of ROS scavenger N-acetyl-L-cysteine (NAC) reverts the activation of autophagy, apoptosis and cellular senescence by RPIA knockdown in lung cancer cells. In conclusion, RPIA knockdown induces ROS levels to activate autophagy, apoptosis, and cellular senescence in lung cancer cells. Our study sheds new light on RPIA suppression in lung cancer therapy.


Subject(s)
Autophagy , Lung Neoplasms , Aldose-Ketose Isomerases , Apoptosis , Cell Line, Tumor , Cellular Senescence , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Reactive Oxygen Species/metabolism
9.
Carcinogenesis ; 42(7): 951-960, 2021 07 16.
Article in English | MEDLINE | ID: mdl-33993270

ABSTRACT

Inhibitors of DNA binding and cell differentiation (ID) proteins regulate cellular differentiation and tumor progression. Whether ID family proteins serve as a linkage between pathological differentiation and cancer stemness in colorectal cancer is largely unknown. Here, the expression of ID4, but not other ID family proteins, was enriched in LGR5-high colon cancer stem cells. Its high expression was associated with poor pathological differentiation of colorectal tumors and shorter survival in patients. Knockdown of ID4 inhibited the growth and dissemination of colon cancer cells, while enhancing chemosensitivity. Through gene expression profiling analysis, brain-derived neurotrophic factor (BDNF) was identified as a downstream target of ID4 expression in colorectal cancer. BDNF knockdown decreased the growth and migration of colon cancer cells, and its expression enhanced dissemination, anoikis resistance and chemoresistance. ID4 silencing attenuated the epithelial-to-mesenchymal transition pattern in colon cancer cells. Gene cluster analysis revealed that ID4 and BDNF expression was clustered with mesenchymal markers and distant from epithelial genes. BDNF silencing decreased the expression of mesenchymal markers Vimentin, CDH2 and SNAI1. These findings demonstrated that ID4-BDNF signaling regulates colorectal cancer survival, with the potential to serve as a prognostic marker in colorectal cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inhibitor of Differentiation Proteins/metabolism , Neoplastic Stem Cells/pathology , Apoptosis , Biomarkers, Tumor/genetics , Brain-Derived Neurotrophic Factor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Humans , Inhibitor of Differentiation Proteins/genetics , Neoplastic Stem Cells/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured
10.
PLoS Biol ; 16(1): e2003714, 2018 01.
Article in English | MEDLINE | ID: mdl-29337987

ABSTRACT

Altered metabolism is one of the hallmarks of cancers. Deregulation of ribose-5-phosphate isomerase A (RPIA) in the pentose phosphate pathway (PPP) is known to promote tumorigenesis in liver, lung, and breast tissues. Yet, the molecular mechanism of RPIA-mediated colorectal cancer (CRC) is unknown. Our study demonstrates a noncanonical function of RPIA in CRC. Data from the mRNAs of 80 patients' CRC tissues and paired nontumor tissues and protein levels, as well as a CRC tissue array, indicate RPIA is significantly elevated in CRC. RPIA modulates cell proliferation and oncogenicity via activation of ß-catenin in colon cancer cell lines. Unlike its role in PPP in which RPIA functions within the cytosol, RPIA enters the nucleus to form a complex with the adenomatous polyposis coli (APC) and ß-catenin. This association protects ß-catenin by preventing its phosphorylation, ubiquitination, and subsequent degradation. The C-terminus of RPIA (amino acids 290 to 311), a region distinct from its enzymatic domain, is necessary for RPIA-mediated tumorigenesis. Consistent with results in vitro, RPIA increases the expression of ß-catenin and its target genes, and induces tumorigenesis in gut-specific promotor-carrying RPIA transgenic zebrafish. Together, we demonstrate a novel function of RPIA in CRC formation in which RPIA enters the nucleus and stabilizes ß-catenin activity and suggests that RPIA might be a biomarker for targeted therapy and prognosis.


Subject(s)
Aldose-Ketose Isomerases/metabolism , Aldose-Ketose Isomerases/physiology , beta Catenin/physiology , Adenomatous Polyposis Coli/metabolism , Adult , Aged , Animals , Animals, Genetically Modified , Carcinogenesis , Cell Line, Tumor , Cell Nucleus , Cell Proliferation/physiology , Cell Transformation, Neoplastic , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Male , Middle Aged , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Domains , RNA, Messenger/genetics , Ubiquitination , Zebrafish , beta Catenin/genetics
11.
Int J Mol Sci ; 22(16)2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34445544

ABSTRACT

Patients with advanced-stage non-small-cell lung cancer (NSCLC) are susceptible to malnutrition and develop folate deficiency (FD). We previously found that folate deprivation induces drug resistance in hepatocellular carcinoma; here, we assessed whether disrupted cytoplasmic folate metabolism could mimic FD-induced metastasis and affect the sensitivity of NSCLC cells to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). We examined whether cytosolic folate metabolism in NSCLC cells was disrupted by FD or the folate metabolism blocker pemetrexed for 1-4 weeks. Our results revealed an increase in NF-κB overexpression-mediated epithelial-mesenchymal transition biomarkers: N-cadherin, vimentin, matrix metalloproteinases (MMPs), SOX9, and SLUG. This finding suggests that the disruption of folate metabolism can drastically enhance the metastatic properties of NSCLC cells. Cytosolic FD also affected EGFR-TKI cytotoxicity toward NSCLC cells. Because SLUG and N-cadherin are resistance effectors against gefitinib, the effects of SLUG knockdown in folate antagonist-treated CL1-0 cells were evaluated. SLUG knockdown prevented SLUG/NF-κB/SOX9-mediated invasiveness and erlotinib resistance acquisition and significantly reduced pemetrexed-induced gelatinase activity and MMP gene expression. To summarize, our data reveal two unprecedented adverse effects of folate metabolism disruption in NSCLC cells. Thus, the folic acid status of patients with NSCLC under treatment can considerably influence their prognosis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/secondary , Cytoplasm/metabolism , Drug Resistance, Neoplasm , Folic Acid/metabolism , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Tumor Cells, Cultured
12.
Molecules ; 26(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34443472

ABSTRACT

Feruloylacetone (FER) is a natural degradant of curcumin after heating, which structurally reserves some functional groups of curcumin. It is not as widely discussed as its original counterpart has been previously; and in this study, its anticancer efficacy is investigated. This study focuses on the suppressive effect of FER on colon cancer, as the efficacious effect of curcumin on this typical cancer type has been well evidenced. In addition, demethoxy-feruloylacetone (DFER) was applied to compare the effect that might be brought on by the structural differences of the methoxy group. It was revealed that both FER and DFER inhibited the proliferation of HCT116 cells, possibly via suppression of the phosphorylated mTOR/STAT3 pathway. Notably, FER could significantly repress both the STAT3 phosphorylation and protein levels. Furthermore, both samples showed capability of arresting HCT116 cells at the G2/M phase via the activation of p53/p21 and the upregulation of cyclin-B. In addition, ROS elevation and changes in mitochondrial membrane potential were revealed, as indicated by p-atm elevation. The apoptotic rate rose to 36.9 and 32.2% after being treated by FER and DFER, respectively. In summary, both compounds exhibited an anticancer effect, and FER showed a greater proapoptotic effect, possibly due to the presence of the methoxy group on the aromatic ring.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Curcumin/pharmacology , Mitochondria/drug effects , Styrenes/pharmacology , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Apoptosis/drug effects , Cell Division/drug effects , Colonic Neoplasms/drug therapy , Curcumin/chemistry , Curcumin/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/agonists , G2 Phase/drug effects , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Phenol/chemistry , Phenol/pharmacology , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Styrenes/chemistry , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/agonists
13.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32948086

ABSTRACT

A strategy was described to design antimicrobial peptides (AMPs) with enhanced salt resistance and antiendotoxin activities by linking two helical AMPs with the Ala-Gly-Pro (AGP) hinge. Among the designed peptides, KR12AGPWR6 demonstrated the best antimicrobial activities even in high salt conditions (NaCl ~300 mM) and possessed the strongest antiendotoxin activities. These activities may be related to hydrophobicity, membrane-permeability, and α-helical content of the peptide. Amino acids of the C-terminal helices were found to affect the peptide-induced permeabilization of LUVs, the α-helicity of the designed peptides under various LUVs, and the LPS aggregation and size alternation. A possible model was proposed to explain the mechanism of LPS neutralization by the designed peptides. These findings could provide a new approach for designing AMPs with enhanced salt resistance and antiendotoxin activities for potential therapeutic applications.


Subject(s)
Endotoxemia/drug therapy , Lipopolysaccharides/antagonists & inhibitors , Pore Forming Cytotoxic Proteins/pharmacology , Salt Tolerance/drug effects , Sodium Chloride/pharmacology , Amino Acid Sequence , Animals , Colony Count, Microbial , Drug Evaluation, Preclinical , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Limulus Test , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Pore Forming Cytotoxic Proteins/chemical synthesis , Pore Forming Cytotoxic Proteins/therapeutic use , Protein Conformation, alpha-Helical , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/blood , Unilamellar Liposomes
14.
Carcinogenesis ; 40(3): 461-473, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30418535

ABSTRACT

Dysregulation of the enzymes involved in the pentose phosphate pathway (PPP) is known to promote tumorigenesis. Our recent study demonstrated that ribose-5-phosphate isomerase (RPIA), a key regulator of the PPP, regulates hepatoma cell proliferation and colony formation. Our studies in zebrafish reveal that RPIA-mediated hepatocarcinogenesis requires extracellular signal-regulated kinase (ERK) and ß-catenin signaling. To further investigate RPIA-mediated hepatocarcinogenesis, two independent lines of transgenic zebrafish expressing human RPIA in the liver were generated. These studies reveal that RPIA overexpression triggers lipogenic factor/enzyme expression, steatosis, fibrosis and proliferation of the liver. In addition, the severity of fibrosis and the extent of proliferation are positively correlated with RPIA expression levels. Furthermore, RPIA-mediated induction of hepatocellular carcinoma (HCC) requires the ERK and ß-catenin signaling pathway but is not dependent upon transaldolase levels. Our study presents a mechanism for RPIA-mediated hepatocarcinogenesis and suggests that RPIA represents a valuable therapeutic target for the treatment of HCC.


Subject(s)
Aldose-Ketose Isomerases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms, Experimental/pathology , beta Catenin/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Disease Progression , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/metabolism , Zebrafish/genetics
15.
Gynecol Oncol ; 148(2): 383-392, 2018 02.
Article in English | MEDLINE | ID: mdl-29208367

ABSTRACT

OBJECTIVE: EpCAM is a transmembrane glycoprotein that functions as an epithelial marker in endometrial tissues. However, the correlation between EpCAM and endometrial carcinoma (EC) is not clear. METHODS: This study investigated the association between EpCAM and EC. Immunohistochemistry staining and bioinformatics analysis disclosed the clinical importance of low EpCAM expression. The migratory ability of cells expressing low EpCAM levels was studied in transwell invasion assays in vitro and an orthotopic intra-uterine tumor injection model in vivo. The Connectivity MAP was used to identify drugs that effectively inhibit cells with low EpCAM expression. RESULTS: According to immunohistochemistry analysis results, low EpCAM expression was associated with an advanced stage and lymph node metastasis in patients with endometrioid EC, and high EpCAM expression favored survival. EpCAM silencing promoted cell invasion, and EpCAM re-expression in EpCAM-silenced EC cells attenuated their invasiveness. EpCAM suppression in an orthotopic uterine implantation model promoted the lymph node metastasis of EC cells. According to quantitative PCR and promoter reporter analyses, estrogen receptor alpha signaling regulated EpCAM expression by enhancing its promoter activity. As shown in the Connectivity MAP analysis, transamin inhibited the invasiveness of EpCAM-silenced EC cells. CONCLUSIONS: The loss of EpCAM may increase the malignancy of EC, and these findings provide new insights into the prognostic role of EpCAM in patients with EC.


Subject(s)
Endometrial Neoplasms/etiology , Epithelial Cell Adhesion Molecule/physiology , Animals , Antifibrinolytic Agents/pharmacology , Cell Line, Tumor , Disease Progression , Down-Regulation/physiology , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Epithelial Cell Adhesion Molecule/metabolism , Estrogen Receptor alpha/physiology , Female , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Humans , Kaplan-Meier Estimate , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation/methods , Prognosis , Signal Transduction/physiology , Tranexamic Acid/pharmacology , Transplantation, Heterologous
16.
Respirology ; 23(6): 618-625, 2018 06.
Article in English | MEDLINE | ID: mdl-29462843

ABSTRACT

BACKGROUND AND OBJECTIVE: Craniofacial structure is an important determinant of obstructive sleep apnoea (OSA) syndrome risk. Three-dimensional stereo-photogrammetry (3dMD) is a novel technique which allows quantification of the craniofacial profile. This study compares the facial images of OSA patients captured by 3dMD to three-dimensional computed tomography (3-D CT) and two-dimensional (2-D) digital photogrammetry. Measurements were correlated with indices of OSA severity. METHODS: Thirty-eight patients diagnosed with OSA were included, and digital photogrammetry, 3dMD and 3-D CT were performed. Distances, areas, angles and volumes from the images captured by three methods were analysed. RESULTS: Almost all measurements captured by 3dMD showed strong agreement with 3-D CT measurements. Results from 2-D digital photogrammetry showed poor agreement with 3-D CT. Mandibular width, neck perimeter size and maxillary volume measurements correlated well with the severity of OSA using all three imaging methods. Mandibular length, facial width, binocular width, neck width, cranial base triangle area, cranial base area 1 and middle cranial fossa volume correlated well with OSA severity using 3dMD and 3-D CT, but not with 2-D digital photogrammetry. CONCLUSION: 3dMD provided accurate craniofacial measurements of OSA patients, which were highly concordant with those obtained by CT, while avoiding the radiation associated with CT.


Subject(s)
Face/diagnostic imaging , Mandible/diagnostic imaging , Maxilla/diagnostic imaging , Neck/diagnostic imaging , Skull Base/diagnostic imaging , Sleep Apnea, Obstructive/diagnostic imaging , Adult , Face/pathology , Humans , Imaging, Three-Dimensional , Male , Mandible/pathology , Maxilla/pathology , Middle Aged , Neck/pathology , Organ Size , Photogrammetry , Photography , Polysomnography , Skull Base/pathology , Sleep Apnea, Obstructive/pathology , Tomography, X-Ray Computed
17.
J Biol Chem ; 291(4): 1877-1889, 2016 01 22.
Article in English | MEDLINE | ID: mdl-26542803

ABSTRACT

We previously demonstrated that the epidermal growth factor receptor (EGFR) up-regulated miR-7 to promote tumor growth during lung cancer oncogenesis. Several lines of evidence have suggested that alterations in chromatin remodeling components contribute to cancer initiation and progression. In this study, we identified SMARCD1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily d, member 1) as a novel target gene of miR-7. miR-7 expression reduced SMARCD1 protein expression in lung cancer cell lines. We used luciferase reporters carrying wild type or mutated 3'UTR of SMARCD1 and found that miR-7 blocked SMARCD1 expression by binding to two seed regions in the 3'UTR of SMARCD1 and down-regulated SMARCD1 mRNA expression. Additionally, upon chemotherapy drug treatment, miR-7 down-regulated p53-dependent apoptosis-related gene BAX (BCL2-associated X protein) and p21 expression by interfering with the interaction between SMARCD1 and p53, thereby reducing caspase3 cleavage and the downstream apoptosis cascades. We found that although SMARCD1 sensitized lung cancer cells to chemotherapy drug-induced apoptosis, miR-7 enhanced the drug resistance potential of lung cancer cells against chemotherapy drugs. SMARCD1 was down-regulated in patients with non-small cell lung cancer and lung adenocarcinoma cell lines, and SMARCD1 and miR-7 expression levels were negatively correlated in clinical samples. Our investigation into the involvement of the EGFR-regulated microRNA pathway in the SWI/SNF chromatin remodeling complex suggests that EGFR-mediated miR-7 suppresses the coupling of the chromatin remodeling factor SMARCD1 with p53, resulting in increased chemo-resistance of lung cancer cells.


Subject(s)
Apoptosis , Chromatin/metabolism , Gene Expression Regulation , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Chromatin/genetics , Chromosomal Proteins, Non-Histone , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , MicroRNAs/genetics , Protein Binding , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics
18.
Sleep Breath ; 21(1): 85-91, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27380033

ABSTRACT

BACKGROUNDS: Obstructive sleep apnea (OSA) is common in patients on hemodialysis, but its correlation with chronic kidney disease (CKD) is not clear. We aimed to clarify the relationship between OSA without hypertension or diabetes and incidence of CKD in Taiwan. METHODS: This population-based cohort study consisted of patients with newly diagnosed OSA between 2000 and 2009. The comparison cohort was matched for age, sex, diabetes mellitus, and hypertension. All subjects previously diagnosed with acute or chronic kidney disease were excluded. The primary end point was newly diagnosed CKD. RESULTS: We identified 6866 subjects with OSA during the 10-year study period. The median duration until development of CKD in the OSA cohort was 3.2 years, 2.5 months earlier than that in the non-OSA cohort. After exclusion of hypertension and diabetes, 4319 OSA patients was identified and the hazard ratio (HR) of CKD with OSA was 1.37 (95 % confidence interval [CI], 1.05-1.77; p = 0.019). In the subgroup analysis, an increased incidence of CKD in OSA was observed in women (HR, 1.41; 95 % CI, 1.12-1.78; p = 0.0036). CONCLUSIONS: This longitudinal population-based cohort study provides evidence that patients with OSA even without diabetes or hypertension are at higher risk of developing CKD over the next 3 years and nearly 2.5 months earlier than the non-OSA cohort, particularly women.


Subject(s)
Acute Kidney Injury/physiopathology , Diabetes Mellitus/diagnosis , Diabetes Mellitus/physiopathology , Hypertension/diagnosis , Hypertension/physiopathology , Kidney Failure, Chronic/physiopathology , Kidney Function Tests , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/physiopathology , Acute Kidney Injury/epidemiology , Adult , Aged , Cohort Studies , Diabetes Mellitus/epidemiology , Female , Follow-Up Studies , Humans , Hypertension/epidemiology , Incidence , Kidney Failure, Chronic/epidemiology , Longitudinal Studies , Male , Middle Aged , Retrospective Studies , Risk Factors , Sleep Apnea, Obstructive/epidemiology , Statistics as Topic , Taiwan , Young Adult
19.
J Appl Toxicol ; 37(5): 573-582, 2017 05.
Article in English | MEDLINE | ID: mdl-27813108

ABSTRACT

Long-term exposure to arsenic has been known to induce neoplastic initiation and progression in several organs; however, the role of arsenic (As2 O3 ) in oxidative stress-mediated DNA damage remains elusive. One of the immediate cellular responses to DNA damage is poly(ADP-ribosyl)ation (PARylation), which mediates DNA repair and enhances cell survival. In this study, we found that oxidative stress (H2 O2 )-induced PARylation was suppressed by As2 O3 exposure in different human cancer cells. Moreover, As2 O3 treatment promoted H2 O2 -induced DNA damage and apoptosis, leading to increased cell death. We found that N-ethylmaleimide (NEM), an organic compound derived from maleic acid, could reverse As2 O3 -mediated effects, thus enhancing PARylation with attenuated cell death and increased cell survival. Pharmacologic inhibition of glutathione with l-buthionine-sulfoximine blocked the antagonistic effect of NEM on As2 O3 , thereby continuing As2 O3 -mediated suppression of PARylation and causing DNA damage. Our findings identify NEM as a potential antidote against As2 O3 -mediated DNA damage in a glutathione-dependent manner. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Antidotes/pharmacology , Arsenicals/antagonists & inhibitors , Cell Survival/drug effects , Ethylmaleimide/pharmacology , Oxidative Stress/drug effects , Oxides/antagonists & inhibitors , Poly ADP Ribosylation/drug effects , Apoptosis/drug effects , Arsenic Trioxide , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Colony-Forming Units Assay , Comet Assay , DNA Damage , DNA Repair/drug effects , Ethylmaleimide/antagonists & inhibitors , Humans , Oxides/toxicity
20.
J Formos Med Assoc ; 116(5): 359-365, 2017 May.
Article in English | MEDLINE | ID: mdl-27451274

ABSTRACT

BACKGROUND/PURPOSE: Positional obstructive sleep apnea (OSA) is defined as an apnea hypopnea index at least twice as high in the supine position as in the lateral position. Whether a positional OSA patient persistently has positional OSA in the follow-up period is unknown. This study was conducted to investigate the maintenance of the positional effect on OSA patients and the predictors of changing from positional OSA to nonpositional OSA. METHODS: Patients who were diagnosed to have positional OSA were screened for a follow-up polysomnography (PSG), which evaluated the effect of the same lateral position as baseline PSG on the severity of OSA. Those who met the criteria of positional OSA in both PSGs were classified as the unchanged group, the others were classified as the changed group. RESULTS: Seventy-eight positional OSA patients were enrolled in the final analyses. Twenty-seven of the enrolled patients (35%) were changed to nonpositional OSA patients in the second PSG. A higher apnea index in the lateral position was found in the changed group compared with that in the unchanged group (p = 0.02). Logistic regression also showed that the apnea index in the lateral position was the only independent predictor of changing from positional OSA to nonpositional OSA in the follow-up PSG (odds ratio = 1.13, p = 0.004). CONCLUSION: One-third of positional OSA patients who had a high apnea index in the lateral position tends to become nonpositional OSA patients in the follow-up PSG and must be closely monitored if receiving positional therapy only.


Subject(s)
Health Status Indicators , Posture/physiology , Sleep Apnea, Obstructive/physiopathology , Sleep/physiology , Aged , Female , Follow-Up Studies , Humans , Logistic Models , Male , Middle Aged , Polysomnography , Retrospective Studies , Sleep Apnea, Obstructive/therapy , Supine Position/physiology
SELECTION OF CITATIONS
SEARCH DETAIL