Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Thorac Cancer ; 2024 May 08.
Article En | MEDLINE | ID: mdl-38720472

BACKGROUND: Solute carrier family 34 member 2 (SLC34A2) has been implicated in the development of various malignancies. However, the clinical significance and underlying molecular mechanisms of SLC34A2 in esophageal squamous cell carcinoma (ESCC) remain elusive. METHODS: Western blotting, quantitative real-time PCR and immunohistochemistry were utilized to evaluate the expression levels of SLC34A2 mRNA/protein in ESCC cell lines or tissues. Kaplan-Meier curves were employed for survival analysis. CCK-8, colony formation, EdU and xenograft tumor model assays were conducted to determine the impact of SLC34A2 on ESCC cell proliferation. Cell cycle was examined using flow cytometry. RNA-sequencing and enrichment analysis were carried out to explore the potential signaling pathways. The autophagic flux was evaluated by western blotting, mRFP-GFP-LC3 reporter system and transmission electron microscopy. Immunoprecipitation and mass spectrometry were utilized for identification of potential SLC34A2-interacting proteins. Cycloheximide (CHX) chase and ubiquitination assays were conducted to test the protein stability. RESULTS: The expression of SLC34A2 was significantly upregulated in ESCC and correlated with unfavorable clinicopathologic characteristics particularly the Ki-67 labeling index and poor prognosis of ESCC patients. Overexpression of SLC34A2 promoted ESCC cell proliferation, while silencing SLC34A2 had the opposite effect. Moreover, SLC34A2 induced autophagy to promote ESCC cell proliferation, whereas inhibition of autophagy suppressed the proliferation of ESCC cells. Further studies showed that SLC34A2 interacted with an autophagy-related protein STX17 to promote autophagy and proliferation of ESCC cells by inhibiting the ubiquitination and degradation of STX17. CONCLUSIONS: These findings indicate that SLC34A2 may serve as a prognostic biomarker for ESCC.

2.
ChemSusChem ; : e202301722, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38487956

Nickel oxide (NiOX) is an ideal inorganic hole transport material for the fabrication of inverted perovskite solar cells owing to its excellent optical and semiconductor properties. Currently, the main research on developing the performance of NiOX-based perovskite solar cells focuses on improving the conductivity of NiOX thin films and preventing the redox reactions between metal cations (Ni3+ on the surface of NiOX) and organic cations (FA+ or MA+ in the perovskite precursors) at the NiOX/perovskite interface. In this study, a new type of interface defects in NiOX-based CsPbI2Br solar cells is reported. That is the Pb2+ from CsPbI2Br perovskites can diffuse into the lattice of NiOX surface as the annealing temperature of perovskites changes. The diffusion of Pb2+ increases the ratio of Ni3+/Ni2+ on the surface of NiOX, leading to an increase in the density of trap state at the interface between NiOX and perovskites, which eventually results in a serious decline in the photovoltaic performance of solar cells.

3.
PLoS Pathog ; 19(11): e1011764, 2023 Nov.
Article En | MEDLINE | ID: mdl-37948460

Subacute ruminal acidosis (SARA) has been demonstrated to promote the development of mastitis, one of the most serious diseases in dairy farming worldwide, but the underlying mechanism is unclear. Using untargeted metabolomics, we found hexadecanamide (HEX) was significantly reduced in rumen fluid and milk from cows with SARA-associated mastitis. Herein, we aimed to assess the protective role of HEX in Staphylococcus aureus (S. aureus)- and SARA-induced mastitis and the underlying mechanism. We showed that HEX ameliorated S. aureus-induced mastitis in mice, which was related to the suppression of mammary inflammatory responses and repair of the blood-milk barrier. In vitro, HEX depressed S. aureus-induced activation of the NF-κB pathway and improved barrier integrity in mouse mammary epithelial cells (MMECs). In detail, HEX activated PPARα, which upregulated SIRT1 and subsequently inhibited NF-κB activation and inflammatory responses. In addition, ruminal microbiota transplantation from SARA cows (S-RMT) caused mastitis and aggravated S. aureus-induced mastitis, while these changes were reversed by HEX. Our findings indicate that HEX effectively attenuates S. aureus- and SARA-induced mastitis by limiting inflammation and repairing barrier integrity, ultimately highlighting the important role of host or microbiota metabolism in the pathogenesis of mastitis and providing a potential strategy for mastitis prevention.


Mastitis , Staphylococcus aureus , Humans , Female , Animals , Mice , Cattle , Staphylococcus aureus/metabolism , NF-kappa B/metabolism , Milk , Mastitis/metabolism
4.
Microbiome ; 11(1): 78, 2023 04 17.
Article En | MEDLINE | ID: mdl-37069691

BACKGROUND: Mastitis is one of the most severe diseases in humans and animals, especially on dairy farms. Mounting evidence indicates that gastrointestinal dysbiosis caused by induction of subacute ruminal acidosis (SARA) by high-grain diet consumption and low in dietary fiber is associated with mastitis initiation and development, however, the underlying mechanism remains unknown. RESULTS: In the present study, we found that cows with SARA-associated mastitis have altered metabolic profiles in the rumen, with increased sialic acids level in particular. Consumption of sialic acid (SA) in antibiotic-treated mice, but not healthy mice, induced marked mastitis. SA treatment of antibiotic-treated mice also induced mucosal and systemic inflammatory responses, as evidenced by increased colon and liver injuries and several inflammatory markers. In addition, gut dysbiosis caused by antibiotic impaired gut barrier integrity, which was aggravated by SA treatment. SA potentiated serum LPS level caused by antibiotic treatment, leading to increased activation of the TLR4-NF-κB/NLRP3 pathways in the mammary gland and colon. Moreover, SA facilitated gut dysbiosis caused by antibiotic, and especially enhanced Enterobacteriaceae and Akkermansiaceae, which correlated with mastitis parameters. Fecal microbiota transplantation from SA-antibiotic-treated mice mimicked mastitis in recipient mice. In vitro experiments showed that SA prompted Escherichia coli growth and virulence gene expression, leading to higher proinflammatory cytokine production in macrophages. Targeting the inhibition of Enterobacteriaceae by sodium tungstate or treating with the commensal Lactobacillus reuteri alleviated SA-facilitated mastitis. In addition, SARA cows had distinct ruminal microbial structure by the enrichment of SA-utilizing opportunistic pathogenic Moraxellaceae and the depletion of SA-utilizing commensal Prevotellaceae. Treating mice with the specific sialidase inhibitor zanamivir reduced SA production and Moraxellaceae abundance, and improved mastitis in mice caused by ruminal microbiota transplantation from cows with SARA-associated mastitis. CONCLUSIONS: This study, for the first time, indicates that SA aggravates gut dysbiosis-induced mastitis by promoting gut microbiota disturbance and is regulated by commensal bacteria, indicating the important role of the microbiota-gut-mammary axis in mastitis pathogenesis and suggesting a potential strategy for mastitis intervention based on gut metabolism regulation. Video Abstract.


Gastrointestinal Microbiome , Mastitis , Microbiota , Humans , Female , Animals , Cattle , Mice , Gastrointestinal Microbiome/physiology , N-Acetylneuraminic Acid , Dysbiosis/chemically induced , Enterobacteriaceae , Escherichia coli
5.
Exp Cell Res ; 421(1): 113375, 2022 12 01.
Article En | MEDLINE | ID: mdl-36208716

MRG domain binding protein (MRGBP) has been proposed to participate in the development of multiple tumors. However, the role of MRGBP in colorectal cancer (CRC) still remains largely unknown. Here, we found that MRGBP expression is significantly elevated in CRC, and that higher MRGBP expression correlates with poorer survival in CRC patients. Experiments in vivo and in vitro indicated that MRGBP promotes CRC cells proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) and xenograft tumor growth. Mechanically, for one thing, we discovered that MRGBP suppresses DKK1 expression, thus further activating the Wnt/ß-catenin pathway in CRC cells. For another, MRGBP also enhances acetylation of NF-kB/p65 pathway. Treatment with Wnt/ß-catenin and NF-kB pathways inhibitors further confirmed the mediation of these two pathways in MRGBP-promoted CRC cell processes. In conclusion, these findings together suggest that MRGBP promotes CRC progression via DKK1/Wnt/ß-catenin and NF-kB/p65 pathways mediated EMT, identifying MRGBP as a promising prognostic and therapeutic target for CRC.


Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Histone Acetyltransferases , Humans , beta Catenin/genetics , beta Catenin/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Metastasis , NF-kappa B/genetics , NF-kappa B/metabolism , Wnt Signaling Pathway/genetics , Nuclear Proteins/metabolism , Histone Acetyltransferases/metabolism
6.
IEEE Trans Image Process ; 31: 4405-4416, 2022.
Article En | MEDLINE | ID: mdl-35759599

While humans can effortlessly transform complex visual scenes into simple words and the other way around by leveraging their high-level understanding of the content, conventional or the more recent learned image compression codecs do not seem to utilize the semantic meanings of visual content to their full potential. Moreover, they focus mostly on rate-distortion and tend to underperform in perception quality especially in low bitrate regime, and often disregard the performance of downstream computer vision algorithms, which is a fast-growing consumer group of compressed images in addition to human viewers. In this paper, we (1) present a generic framework that can enable any image codec to leverage high-level semantics and (2) study the joint optimization of perception quality and distortion. Our idea is that given any codec, we utilize high-level semantics to augment the low-level visual features extracted by it and produce essentially a new, semantic-aware codec. We propose a three-phase training scheme that teaches semantic-aware codecs to leverage the power of semantic to jointly optimize rate-perception-distortion (R-PD) performance. As an additional benefit, semantic-aware codecs also boost the performance of downstream computer vision algorithms. To validate our claim, we perform extensive empirical evaluations and provide both quantitative and qualitative results.


Data Compression , Algorithms , Humans , Image Enhancement/methods , Perception , Semantics
7.
Microbiol Spectr ; 10(4): e0081122, 2022 08 31.
Article En | MEDLINE | ID: mdl-35727038

Intestinal microbiota-mediated aryl hydrocarbon receptor (AhR) activation plays an important role in host-microbiota interactions and disease development. However, whether AhR activation mediates infection-induced inflammation in remote organs is not clear. The purpose of this study is to assess the effects and underlying mechanism of AhR activation and gut microbiota-mediated dietary tryptophan (Trp) metabolism on infection-induced inflammation using an Escherichia coli (E. coli)-induced endometritis model in mice. We found that AhR activation by 6-formylindolo (3,2-b) carbazole (Ficz), which is an AhR agonist derived from the photooxidation of Trp, alleviated E. coli-induced endometritis by repairing barrier function and inhibiting inflammatory responses, while inhibition of AhR by CH223191, which is a synthetic AhR antagonist, aggravated E. coli-induced endometritis. Gut dysbiosis damaged AhR activation and exacerbated E. coli-induced endometritis in mice, which responded to the reduced abundance of AhR ligand producers, such as Lactobacillus spp. Supplementation with dietary Trp ameliorated E. coli-induced endometritis in a microbiota-dependent manner, which was associated with the production of AhR ligands. Administration of AhR ligands, including indole and indole aldehyde, but not indole-3-propionic acid, rescued the protective effect of Trp on E. coli-induced endometritis in dysbiotic mice. Moreover, consumption of Lactobacillus reuteri (L. reuteri) containing AhR ligand-producing capability also alleviated E. coli-induced endometritis in mice in an AhR-dependent manner. Our results demonstrate that microbiota-mediated AhR activation is a key factor in fighting pathogen-caused inflammation, which leads to a potential strategy to regulate the gut microbiota and metabolism by dietary Trp or probiotics for the intervention of infectious diseases and reproductive health. IMPORTANCE Infection-induced endometritis is a common and frequently occurring disease in humans and animals. Accumulating evidence suggests an important role of the gut microbiota in the development of infection-induced inflammation. Whether and how gut microbiota-mediated AhR activation regulates the pathogenesis of pathogen-induced endometritis remains unknown. The current study found that AhR activation ameliorated E. coli-induced endometritis, and inhibition of AhR produced negative results. Gut dysbiosis reduced the abundance of AhR ligand producers including Lactobacillus spp., damaged AhR activation, and exacerbated E. coli-induced endometritis. Supplementation with dietary Trp, AhR ligands, and L. reuteri containing AhR ligand-producing capability alleviated E. coli-induced endometritis in mice. Our results suggest an important role of microbiota-mediated AhR activation in the pathogenesis of endometritis and provide potential strategies for the intervention of infectious diseases and reproductive health by regulating the gut microbiota and metabolism.


Endometritis , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Animals , Dysbiosis/therapy , Endometritis/therapy , Escherichia coli/metabolism , Female , Humans , Inflammation , Limosilactobacillus reuteri/metabolism , Ligands , Mice , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism
8.
Food Chem Toxicol ; 163: 112909, 2022 May.
Article En | MEDLINE | ID: mdl-35292335

Bisphenol A (BPA) is a common environmental contaminant, whose exposure is associated with the progression of various kidney diseases. BPA exposure has turned out to be associated with cytotoxicity to renal tubular epithelial cells, but its underlying mechanism remains unknown. Herein, we found that BPA induced ferroptosis in kidney and renal tubular epithelial cells, as showed by increased intracellular iron accumulation, lipid peroxidation and cells death upon BPA exposure. Additionally, utilization of ferrostatin-1 and desferrioxamine, typical ferroptosis inhibitors, can fundamentally diminish cells death. Intriguingly, we discovered that autophagy inhibitor chloroquine can shield renal tubular epithelial cells from BPA-caused ferroptosis. Furthermore, we found that ferritinophagy, a phenomenon that degradation of ferritin and inducing subsequent iron overload, occurred after BPA exposure and excessive iron promoted ferroptosis through Fenton reaction. We next demonstrated that BPA activated the AMPK-mTOR-ULK1 signaling pathway. In turn, AMPK, mTOR, and ULK1 knockdown dramatically mitigated BPA-induced TCMK-1 cells death, and decreased MDA and LC3 levels, but increased FTH protein content. These results indicate that activation of the AMPK-mTOR-ULK1 signaling is involved in BPA-induced ferritinophagy. In conclusion, renal dysfunction and renal tubular epithelial damage induced by BPA are linked to ferroptosis, which depends on the activation of ferritinophagy through AMPK-mTOR-ULK1 axis.


Ferroptosis , AMP-Activated Protein Kinases , Autophagy , Benzhydryl Compounds , Epithelial Cells/metabolism , Iron/metabolism , Phenols , TOR Serine-Threonine Kinases/genetics
9.
IEEE Trans Neural Netw Learn Syst ; 33(4): 1441-1451, 2022 04.
Article En | MEDLINE | ID: mdl-33400656

By redefining the conventional notions of layers, we present an alternative view on finitely wide, fully trainable deep neural networks as stacked linear models in feature spaces, leading to a kernel machine interpretation. Based on this construction, we then propose a provably optimal modular learning framework for classification that does not require between-module backpropagation. This modular approach brings new insights into the label requirement of deep learning (DL). It leverages only implicit pairwise labels (weak supervision) when learning the hidden modules. When training the output module, on the other hand, it requires full supervision but achieves high label efficiency, needing as few as ten randomly selected labeled examples (one from each class) to achieve 94.88% accuracy on CIFAR-10 using a ResNet-18 backbone. Moreover, modular training enables fully modularized DL workflows, which then simplify the design and implementation of pipelines and improve the maintainability and reusability of models. To showcase the advantages of such a modularized workflow, we describe a simple yet reliable method for estimating reusability of pretrained modules as well as task transferability in a transfer learning setting. At practically no computation overhead, it precisely described the task space structure of 15 binary classification tasks from CIFAR-10.


Deep Learning , Neural Networks, Computer
11.
Exp Cell Res ; 406(1): 112722, 2021 09 01.
Article En | MEDLINE | ID: mdl-34242623

Breast cancer is one of the most frequently diagnosed cancers and the leading cause of cancer death in women. MIER3 (Mesoderm induction early response 1, family member3) is considered as a potential oncogene for breast cancer. However, the role of MIER3 in breast cancer remain largely unknown. The expression of MIER3 was detected and the relationship between its expression and clinicopathological characteristics was also analyzed. The effect of MIER3 on proliferation and migration of breast cancer cells was detected in vitro and in vivo. Western blot, IF, and Co-IP were employed to detect the relationship between MIER3, HDAC1, HDAC2, and Snail. ChIP assay was performed to determine the binding of MIER3/HDAC1/HDAC2/Snail complex to the promoter of E-cadherin. In this study, we found that MIER3 was upregulated in breast cancer tissue and closely associated with poor prognosis of patients. MIER3 could promote the proliferation, migration, and epithelial-mesenchymal transition (EMT) of breast cancer cells. Further studies showed that MIER3 interacted with HDAC1/HDAC2 and Snail to form a repressive complex which could bind to E-cadherin promoter and was related to its deacetylation. Our study concluded that MIER3 was involved in forming a co-repressor complex with HDAC1/HDAC2/Snail to promote EMT by silencing E-cadherin.


Breast Neoplasms/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Nuclear Proteins/genetics , Snail Family Transcription Factors/genetics , Aged , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice, Nude , Middle Aged , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Snail Family Transcription Factors/metabolism , Survival Analysis , Tumor Burden , Xenograft Model Antitumor Assays
12.
Biol Trace Elem Res ; 199(2): 594-603, 2021 Feb.
Article En | MEDLINE | ID: mdl-32328968

Selenium (Se) is an essential trace element that maintains normal physiological functions in organisms. Since the discovery of glutathione peroxidase (GSH-PX), public interest in selenoproteins has gradually increased. Based on previous studies, dietary Se maintains erythrocyte homeostasis through selenoprotein-induced mediation of redox reactions. Furthermore, both the surface phosphatidylserine (PS) and intramembrane stomatin contents can be used as indicators of erythrocyte osmotic fragility. This study focused on the mechanism by which dietary Se deficiency increases erythrocyte osmotic fragility. We fed Se-deficient grain to mice for 8 weeks to establish a Se deficiency model in mice. We measured Se levels in the blood as well as the activities of antioxidant enzymes associated with selenoproteins in a Se-deficient environment. We used Western blotting, routine blood analysis, and other methods to detect red blood cell oxidative stress levels, membrane stomatin levels, and PS externalization. Fresh blood was collected to test erythrocyte osmotic fragility. The results showed that antioxidant enzyme activity was affected by dietary Se deficiency. Oxidative stress increased lipid peroxidation and the ROS content in the blood of the mice. Under such conditions, decreased PS exposure and stomatin content in the erythrocyte membrane eventually affected the structure of the erythrocyte membrane and increased erythrocyte osmotic fragility.


Selenium , Animals , Erythrocytes/metabolism , Glutathione Peroxidase/metabolism , Lipid Peroxidation , Mice , Osmotic Fragility , Oxidative Stress , Phosphatidylserines
13.
Theranostics ; 10(24): 11063-11079, 2020.
Article En | MEDLINE | ID: mdl-33042270

Metastasis is a major cause of death in patients with colorectal cancer (CRC). Cysteine-rich protein 2 (CSRP2) has been recently implicated in the progression and metastasis of a variety of cancers. However, the biological functions and underlying mechanisms of CSRP2 in the regulation of CRC progression are largely unknown. Methods: Immunohistochemistry, quantitative real-time polymerase chain reaction (qPCR) and Western blotting (WB) were used to detect the expression of CSRP2 in CRC tissues and paracancerous tissues. CSRP2 function in CRC was determined by a series of functional tests in vivo and in vitro. WB and immunofluorescence were used to determine the relation between CSRP2 and epithelial-mesenchymal transition (EMT). Co-immunoprecipitation and scanning electron microscopy were used to study the molecular mechanism of CSRP2 in CRC. Results: The CSRP2 expression level in CRC tissues was lower than in adjacent normal tissues and indicated poor prognosis in CRC patients. Functionally, CSRP2 could suppress the proliferation, migration, and invasion of CRC cells in vitro and inhibit CRC tumorigenesis and metastasis in vivo. Mechanistic investigations revealed a physical interaction between CSRP2 and p130Cas. CSRP2 could inhibit the activation of Rac1 by preventing the phosphorylation of p130Cas, thus activating the Hippo signaling pathway, and simultaneously inhibiting the ERK and PAK/LIMK/cortactin signaling pathways, thereby inhibiting the EMT and metastasis of CRC. Rescue experiments showed that blocking the p130Cas and Rac1 activation could inhibit EMT induced by CSRP2 silencing. Conclusion: Our results suggest that the CSRP2/p130Cas/Rac1 axis can inhibit CRC aggressiveness and metastasis through the Hippo, ERK, and PAK signaling pathways. Therefore, CSRP2 may be a potential therapeutic target for CRC.


Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Crk-Associated Substrate Protein/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/pharmacology , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colon/pathology , Colon/surgery , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Disease Progression , Down-Regulation , Epithelial-Mesenchymal Transition/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hippo Signaling Pathway , Humans , Kaplan-Meier Estimate , LIM Domain Proteins/genetics , Male , Mice , Middle Aged , Muscle Proteins/genetics , Neoplasm Invasiveness/pathology , Nuclear Proteins/genetics , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Rectum/pathology , Rectum/surgery , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors
14.
Exp Cell Res ; 390(1): 111933, 2020 05 01.
Article En | MEDLINE | ID: mdl-32142855

Current studies have shown that POTE ankyrin domain family members have high expressions as tumor antigens in malignant tumors, such as prostate cancer, ovarian cancer, breast cancer and the like. POTEE is a member of the POTE anchor protein family E. However, its role in colorectal carcinoma (CRC) has not been studied. In this study, the function of POTEE in CRC was examined for the first time and its correlation with CRC cell biological behaviors was analyzed. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry revealed that POTEE was remarkably overexpressed in CRC and associated with an aggressive phenotype. We also found that POTEE was localized in the cytoplasm. In addition, downregulation of POTEE expression can notably inhibit the proliferation, migration, and invasion of CRC cell in vitro, and repressed tumor growth and metastasis in vivo. In contrast, overexpression of POTEE could promote the aggressive behaviors of CRC cells. Mechanistically, POTEE promoted CRC migration, invasion and epithelial-mesenchymal transition (EMT) by increasing the activation of Rac1 and Cdc42. To summarize, these results suggested that POTEE might serve as an oncogene for CRC tumorigenesis and progression, and may become a novel molecular marker for clinical diagnosis and treatment.


Antigens, Neoplasm/genetics , Carcinoma/metabolism , Colorectal Neoplasms/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antigens, Neoplasm/metabolism , Carcinoma/genetics , Carcinoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Female , HCT116 Cells , HT29 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Metastasis , Signal Transduction , Up-Regulation
15.
Biol Trace Elem Res ; 196(1): 145-152, 2020 Jul.
Article En | MEDLINE | ID: mdl-31625053

Zinc (Zn) is an important trace element in the body that has antioxidant effects. It has been proven that Zn deficiency can cause oxidative stress. The purpose of the present study was to clarify the effect and mechanism of Zn deficiency on myocardial fibrosis. Mice were fed with different Zn levels dietary for 9 weeks: Zn-normal group (ZnN, 34 mg Zn/kg), Zn-deficient group (ZnD, 2 mg Zn/kg), and Zn-adequate group (ZnA, 100 mg Zn/kg). We found that the Zn-deficient diet reduced the Zn concentration in myocardial tissue. Moreover, the TUNEL results demonstrated that cardiomyocytes in the ZnD group died in large numbers. Furthermore, ROS levels were significantly increased, and metallothionein (MT) expression levels decreased in the ZnD group. The results of Sirius Red staining indicated an increase in collagen in the ZnD group. Moreover, the ELISA results showed that collagen I, III, and IV and fibronectin (FN) were increased. In addition, the expression of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase (TIMPs) was detected by RT-qPCR. The results showed that the expression of TIMP-1 in the ZnD group was increased, while MMPs were decreased. Immunohistochemical results showed an increase in the content of α-smooth muscle actin (α-SMA), while H&E staining showed an increase in interstitial width and a decrease in the number of cardiac cells. All data suggest that Zn deficiency enhances the oxidative stress response of myocardial tissue and eventually triggers myocardial fibrosis.


Fibrosis/metabolism , Matrix Metalloproteinases/metabolism , Metalloproteases/metabolism , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Zinc/metabolism , Animals , Diet , Male , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/analysis , Zinc/administration & dosage , Zinc/deficiency
16.
Neural Comput ; 32(1): 97-135, 2020 01.
Article En | MEDLINE | ID: mdl-31703172

We propose a novel family of connectionist models based on kernel machines and consider the problem of learning layer by layer a compositional hypothesis class (i.e., a feedforward, multilayer architecture) in a supervised setting. In terms of the models, we present a principled method to "kernelize" (partly or completely) any neural network (NN). With this method, we obtain a counterpart of any given NN that is powered by kernel machines instead of neurons. In terms of learning, when learning a feedforward deep architecture in a supervised setting, one needs to train all the components simultaneously using backpropagation (BP) since there are no explicit targets for the hidden layers (Rumelhart, Hinton, & Williams, 1986). We consider without loss of generality the two-layer case and present a general framework that explicitly characterizes a target for the hidden layer that is optimal for minimizing the objective function of the network. This characterization then makes possible a purely greedy training scheme that learns one layer at a time, starting from the input layer. We provide instantiations of the abstract framework under certain architectures and objective functions. Based on these instantiations, we present a layer-wise training algorithm for an l-layer feedforward network for classification, where l≥2 can be arbitrary. This algorithm can be given an intuitive geometric interpretation that makes the learning dynamics transparent. Empirical results are provided to complement our theory. We show that the kernelized networks, trained layer-wise, compare favorably with classical kernel machines as well as other connectionist models trained by BP. We also visualize the inner workings of the greedy kernelized models to validate our claim on the transparency of the layer-wise algorithm.

19.
Mol Med Rep ; 20(2): 2039-2040, 2019 08.
Article En | MEDLINE | ID: mdl-31257539

Following the publication of the article, the authors noted an error associated with the presentation of Fig. 3B. This Figure part showed the cell cycle analysis as determined by flow cytometry of HT29 cells transfected with either a HNF1A-AS1-specific siRNA or a negative control siRNA at 48 h post-transfection. An error was made in the compilation of this Figure, and the same data were inadvertently selected to represent the cell cycle analysis of both the HT29-SI and the HT29-NC cells (i.e., the data relating to the HT29-NC cells were included in the Figure twice). A corrected version of Fig. 3 is shown on the next page. Note that this correction affects neither the interpretation of the data nor the reported conclusions of this work. The authors all agree to this Corrigendum, and regret that this error went unnoticed during the proofs correction stage. They also wish to thank the Editor for allowing them the opportunity to publish this Corrigendum, and regret any inconvenience this error has caused. [the original article was published in Molecular Medicine Reports 16: 4694-4700, 2017; DOI: 10.3892/mmr.2017.7175].

20.
J Exp Clin Cancer Res ; 37(1): 304, 2018 Dec 05.
Article En | MEDLINE | ID: mdl-30518405

BACKGROUND: Inosine 5'-monophosphate dehydrogenase type II (IMPDH2) was originally identified as an oncogene in several human cancers. However, the clinical significance and biological role of IMPDH2 remain poorly understood in colorectal cancer (CRC). METHODS: Quantitative real-time polymerase chain reaction (qPCR), western blotting analysis, the Cancer Genome Atlas (TCGA) data mining and immunohistochemistry were employed to examine IMPDH2 expression in CRC cell lines and tissues. A series of in-vivo and in-vitro assays were performed to demonstrate the function of IMPDH2 and its possible mechanisms in CRC. RESULTS: IMPDH2 was upregulated in CRC cells and tissues at both mRNA and protein level. High IMPDH2 expression was closely associated with T stage, lymph node state, distant metastasis, lymphovascular invasion and clinical stage, and significantly correlated with poor survival of CRC patients. Further study revealed that overexpression of IMPDH2 significantly promoted the proliferation, invasion, migration and epithelial-mesenchymal transition (EMT) of CRC cells in vitro and accelerated xenograft tumour growth in nude mice. On the contrary, knockdown of IMPDH2 achieved the opposite effect. Gene set enrichment analysis (GSEA) showed that the gene set related to cell cycle was linked to upregulation of IMPDH2 expression. Our study verified that overexpressing IMPDH2 could promote G1/S phase cell cycle transition through activation of PI3K/AKT/mTOR and PI3K/AKT/FOXO1 pathways and facilitate cell invasion, migration and EMT by regulating PI3K/AKT/mTOR pathway. CONCLUSIONS: These results suggest that IMPDH2 plays an important role in the development and progression of human CRC and may serve as a novel prognostic biomarker and therapeutic target for CRC.


Colorectal Neoplasms/metabolism , Forkhead Box Protein O1/metabolism , IMP Dehydrogenase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Female , HCT116 Cells , HEK293 Cells , Heterografts , Humans , IMP Dehydrogenase/genetics , Male , Mice , Mice, Nude , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transfection
...