Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Sci Rep ; 13(1): 18327, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37884590

ABSTRACT

Scribble is a master scaffold protein in apical-basal polarity. Current knowledge about the biological function of Scribble in colonic epithelial plasticity/regeneration during intestinal inflammation is limited. Here, we showed that the level of Scribble is decreased in inflammatory bowel disease (IBD) patients and mice with DSS-induced colitis. ScribΔIEC mice develops severe acute colitis with disrupted epithelial barrier integrity and impaired crypt stem cell's function. Mechanistically, Scribble suppressed the process of autophagy by modulating the stability of caspase-dependent degradation of Atg16L1 by directly interacting with Atg16L1 in a LRR domain-dependent manner in IECs and led to an accumulation of ROS both in intestinal stem cells and epithelial cells. In addition, further study indicates that dietary sphingomyelin alleviates DSS-induced colitis by increase the expression of Scribble, which suggests that Scribble may be the critical marker of IBD. Our study shows that Scribble deficiency is associated with the dysregulated autophagy and impaired maintenance of colonic stemness, and it may be a target for diagnosis and treatment of IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Mice , Animals , Colitis/chemically induced , Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Colon/metabolism , Autophagy , Oxidative Stress , Inflammation/metabolism , Dextran Sulfate/toxicity , Intestinal Mucosa/metabolism , Disease Models, Animal
2.
Biochim Biophys Acta Mol Basis Dis ; 1869(8): 166823, 2023 12.
Article in English | MEDLINE | ID: mdl-37632981

ABSTRACT

BACKGROUNDS AND AIMS: Liver cancer is the sixth most common type of cancer and the fifth leading cause of cancer mortality worldwide. Scribble has been shown to function as a neoplastic tumor suppressor gene in most tumors. Our previous studies reported that down-regulation or mislocalization of Scribble was sufficient to initiate mammary tumorigenesis and NSCLC. Recently, it was reported that Scribble was highly expressed in hepatocellular carcinoma (HCC). We aim to study how it was up-regulated and the contradictory role of Scribble in HCC. METHODS AND RESULTS: Using a mouse model of carbon tetrachloride (CCl4)-induced liver fibrosis system, we showed that Scribble was over-expressed and which may protect the mice against hepatic fibrosis. Unexpectedly, we found out the potential for Scribble to act as a tumor driver at the advanced stage of N-nitrosodiethylamine (DEN) plus CCl4 induced HCC mice model in vivo. In addition, we observed even higher expression of Scribble in HCC tumors harboring elevated levels of wild-type p53. Most importantly, nuclear translocated Scribble could interact with p53, which lead to enhanced stability and transcriptional activity of p53. Mechanistically, our data suggested that Scribble might drive HCC progression by promoting metabolic regulation of p53 through p53-upregulated modulator of apoptosis (PUMA)-mediated Warburg effect. CONCLUSIONS: Our data identified the molecular basis of hepatic fibrosis-specific gene expression of polarity gene, such as Scribble. Interestingly, with the progression from fibrosis to cirrhosis to HCC, its nuclear translocation promoted a wild-type p53-mediated cancer metabolic switch and tumor progression in HCC. Taken together, we demonstrated that Scribble was up-regulated and served a protective role in liver fibrosis, while also apparently acting as a tumor driver in fibrosis-dependent hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Tumor Suppressor Protein p53/genetics , Liver Neoplasms/genetics , Apoptosis Regulatory Proteins , Carcinogenesis/genetics , Liver Cirrhosis/genetics , Apoptosis , Disease Models, Animal , Glycolysis
3.
Nat Commun ; 13(1): 897, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35173168

ABSTRACT

The formation of pre-metastatic niche is a key step in the metastatic burden. The pluripotent factor Lin28B is frequently expressed in breast tumors and is particularly upregulated in the triple negative breast cancer subtype. Here, we demonstrate that Lin28B promotes lung metastasis of breast cancer by building an immune-suppressive pre-metastatic niche. Lin28B enables neutrophil recruitment and N2 conversion. The N2 neutrophils are then essential for immune suppression in pre-metastatic lung by PD-L2 up-regulation and a dysregulated cytokine milieu. We also identify that breast cancer-released exosomes with low let-7s are a prerequisite for Lin28B-induced immune suppression. Moreover, Lin28B-induced breast cancer stem cells are the main sources of low-let-7s exosomes. Clinical data further verify that high Lin28B and low let-7s in tumors are both indicators for poor prognosis and lung metastasis in breast cancer patients. Together, these data reveal a mechanism by which Lin28B directs the formation of an immune-suppressive pre-metastatic niche.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Exosomes/metabolism , Lung Neoplasms/secondary , RNA-Binding Proteins/metabolism , Animals , Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Immune Tolerance/immunology , Lung/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Prognosis , RNA-Binding Proteins/genetics
4.
Cell Death Dis ; 13(1): 25, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934045

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) remains the most common gastrointestinal cancer and a leading cause of cancer deaths worldwide, with most showing pathologies indicating the malignant transformation of early stage intestinal stem cells. The long non-coding RNA Meg3, which functions as a tumor suppressor, has been reported to be abnormal in multiple tumorigenesis events; however, the underlying mechanism by which Meg3 contributes to the malignant proliferation of colonic stem cells remains unclear. METHODS: We analyzed the expression levels of Meg3, miR-708, and SOCS3 in samples from Apc loss-of-function (Apcmin) mice and patients with CRC, particularly in colonic crypt cells. Apcmin mice and AMO/DSS-induced mice model (in vivo) and organoid culture system (in vitro) were used to explore the effect of the Meg3/miR-708/SOCS3 axis on tumorigenesis in the colon. In vitro, we performed RNApull-down, RNA immunoprecipitation, and luciferase reporter assays using DLD1 and RKO cell lines. FINDINGS: The Meg3/miR-708/SOCS3 signaling axis plays a critical role in the early stage of CRC development. Our data showed Meg3 levels negatively correlate with miR-708 levels both in clinical samples and in the Apcmin mouse model, which indicated that Meg3 acts as a competitive endogenous RNA (ceRNA) of miR-708. Then, miR-708 served as an oncogene, inducing neoplasia in both Apcmin mice and cultured colonic organoids. Put together, miR-708 appears to promote malignant proliferation of colonic stem cells by targeting SOCS3/STAT3 signaling. INTERPRETATION: These data revealed that Meg3 sponges miR-708 to inhibit CRC development via SOCS3-mediated repression of the malignant proliferation of colonic stem cells. The Meg3/miR-708/SOCS3 signaling axis provides potential targets for the diagnosis and treatment of CRC, particularly early stage CRC.


Subject(s)
MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Humans , Male , Mice
5.
iScience ; 24(8): 102819, 2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34381963

ABSTRACT

The endoplasmic reticulum (ER)-resident transmembrane protein kinase/RNase Ire1 is a conserved sensor of the cellular unfolded protein response and has been implicated in lipid homeostasis, including lipid synthesis and transport, across species. Here we report a novel catabolic role of Ire1 in regulating lipid mobilization in Drosophila. We found that Ire1 is activated by nutrient deprivation, and, importantly, fat body-specific Ire1 deficiency leads to increased lipid mobilization and sensitizes flies to starvation, whereas fat body Ire1 overexpression results in the opposite phenotypes. Genetic interaction and biochemical analyses revealed that Ire1 regulates lipid mobilization by promoting Xbp1s-associated FoxO degradation and suppressing FoxO-dependent lipolytic programs. Our results demonstrate that Ire1 is a catabolic sensor and acts through the Xbp1s-FoxO axis to hamper the lipolytic response during chronic food deprivation. These findings offer new insights into the conserved Ire1 regulation of lipid homeostasis.

6.
Mol Med Rep ; 23(5)2021 05.
Article in English | MEDLINE | ID: mdl-33760113

ABSTRACT

Pancreatic mucinous cystadenocarcinoma (MCC) is a rare malignant tumor, with a limited number of studies. The present study aimed to investigate the function and mechanism of microRNA (miR)­224­5p on proliferation, migration and invasion of MCC of the pancreas. Reverse transcription­quantitative PCR was used to explorethe expression of miR­224­5p and the PTEN gene. MTT, wound healing, Transwell and tumorigenesis assays were conducted to investigate the proliferation, migration and invasion of MCC1 cells in vitro and in vivo. Western blot analysis was employed to test the protein expression of PTEN. The target gene of miR­224­5p was assessed and verified by luciferase assay. miR­224­5p expression was notably higher, while PTEN expression was lower, in MCC1 cells compared with normal tissues and cells. Overexpression of miR­224­5p promoted the proliferation, migration and invasion of MCC and knockdown of miR­224­5p inhibited these functions. Bioinformatics analysis and luciferase assay indicated that PTEN was the direct target gene of miR­224­5p. The negative correlation between miR­224­5p and PTEN was confirmed both in vitro and in vivo. PTEN reversed the effects of miR­224­5p on proliferation, migration and invasion of MCC1 cells. The present study revealed for the first time, to the best of the authors' knowledge, that miR­224­5p was highly expressed and served an oncogenic role in MCC. miR­224­5p not only regulated the proliferation, migration and invasion of pancreatic MCC but may also be a potential therapeutic target for MCC.


Subject(s)
Cystadenocarcinoma, Mucinous/genetics , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/genetics , Aged , Animals , Apoptosis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cystadenocarcinoma, Mucinous/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/pathology
7.
Commun Biol ; 4(1): 213, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594251

ABSTRACT

The mucosa microenvironment is critical for intestinal stem cell self-renewal and reconstruction of the epithelial barrier in inflammatory bowel disease (IBD), where the mechanisms underlying cross-talk between intestinal crypts and the microenvironment remain unclear. Here, we firstly identified miR-494-3p as an important protector in colitis. miR-494-3p levels were decreased and negatively correlated with the severity in human IBD samples, as well as in colitis mice. In colitis crypts, a notable cytokine-cytokine receptor, miR-494-3p-targeted EDA2R and the ligand EDA-A2, suppressed colonic stemness and epithelial repair by inhibiting ß-catenin/c-Myc. In differentiated IECs, miR-494-3p inhibits macrophage recruitment, M1 activation and EDA-A2 secretion by targeting IKKß/NF-κB in colitis. A miR-494-3p agomir system notably ameliorated the severity of colonic colitis in vivo. Collectively, our findings uncover a miR-494-3p-mediated cross-talk mechanism by which macrophage-induced intestinal stem cell impairment aggravates intestinal inflammation.


Subject(s)
Colitis/metabolism , Colon/metabolism , Ectodysplasins/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Paracrine Communication , Stem Cells/metabolism , Xedar Receptor/metabolism , Animals , Antagomirs/administration & dosage , Cells, Cultured , Chemotaxis , Colitis/genetics , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Disease Models, Animal , Ectodysplasins/genetics , Humans , I-kappa B Kinase/metabolism , Intestinal Mucosa/pathology , Macrophage Activation , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Organoids , Stem Cell Niche , Stem Cells/pathology , Wnt Signaling Pathway , Xedar Receptor/genetics
8.
Journal of Preventive Medicine ; (12): 1117-1121, 2021.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-905053

ABSTRACT

@#Contamination of foodborne pathogens is the main cause of related diseases. Escherichia coli O157:H7 (E.coli O157:H7), as a representative of pathogenic E.coli, is one of the most severe and commonly reported E.coli in the world, but there is still no effective clinical treatment against the infection. Antibiotics show effective in the early infection of E.coli O157:H7. However, their extensive use has led to drug-resistant bacteria and genes in recent years, which becomes a great threat to public health. This article reviews the research progress of E.coli O157:H7 from the prevalence of antibiotic-resistant bacteria, the resistance mechanism, and the prevention and control methods, in order to provide a reference for its prevention, early clinical treatment and related research.

9.
Oncol Lett ; 20(6): 276, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33029204

ABSTRACT

The prognosis of invasive pancreatic mucinous cystadenocarcinoma (MCC) is poor, and the molecular mechanism underlying its development remains unclear. The present study aimed to explore the potential role of autophagy in pancreatic MCC. The results demonstrated an increase in autophagy signaling in pancreatic MCC tissues and the MCC1 cell line compared with adjacent tissues and normal human pancreatic ductal epithelium (HPDE) cells. In addition, abnormal autophagy activation facilitated the migration and invasion of MCC1 cells. MicroRNA (miR)-224-5p expression levels were significantly higher in MCC1 cells compared with those in HPDE cells. Treatment with rapamycin further demonstrated that high levels of autophagy elevated miR-224-5p expression in MCC1 cells in a time-dependent manner. BCL2 was identified as a downstream target gene of miR-224-5p, which binds to the 3'-untranslated region of BCL2. In addition, the results of the present study demonstrated that BCL2 knockdown reversed the inhibition of autophagy mediated by the miR-224-5p inhibitor. To the best of our knowledge, this is the first study to evaluate the role of autophagy in pancreatic MCC. Thus, these results suggested that autophagy may be hyperactivated in pancreatic MCC. In addition, the present study identified a positive feedback loop between autophagy signaling and miR-224-5p, which may promote the aggressive migration and invasion of MCC1. These results may provide a new insight into the relationship between autophagy and tumor metastasis in pancreatic MCC.

10.
Oncol Lett ; 20(3): 2757-2762, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32782592

ABSTRACT

Gastric cancer (GC) is the third most common cause of cancer-associated mortality in China. Aberrant microRNA (miR) expression can occur through multiple biological processes and has been implicated in cancer development. However, to the best of our knowledge, the function of miR-502-5p in GC is currently unclear. In the present study, the expression and function of miR-502-5p in GC was evaluated. Reverse transcription-quantitative (RT-q) PCR was used to measure the expression levels of miR-502-5p in GC tissues, normal adjacent tissues, a normal human gastric epithelial cell line (GES-1) and two GC cell lines. miR-502-5p expression levels were significantly lower in GC tissues and GC cell lines compared with those in adjacent normal tissues and GES-1 cells, respectively. Subsequently, the target genes of miR-502-5p were predicted, and it was demonstrated that the transcription factor SP1 was a direct target. SP1 expression, cell viability, migration and invasion, and SP1 protein levels were examined using RT-qPCR, an MTT assay, Transwell assay and western blotting, respectively. Human GC cells were then transfected with an miR-502-5p mimic to emulate miR-502-5p overexpression, resulting in inhibition of the proliferation, migration and invasion capacities of human GC cells. Compared with the negative control, cells overexpressing miR-502-5p had decreased levels of SP1 mRNA and protein. These data suggest that miR-502-5p serves as a tumor suppressor gene by targeting SP1 to regulate the proliferation, migration and invasion of GC cells.

11.
Cell Death Dis ; 11(2): 150, 2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32094323

ABSTRACT

Since online publication of this article, the authors noticed that some of the fly stocks were mislabelled in the methods and in Supplementary Figure 5. The corrected methods text is provided below.

12.
Cell Death Dis ; 10(11): 800, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641108

ABSTRACT

Abnormal aggregation of misfolded pathological proteins in neurons is a prominent feature of neurodegenerative disorders including Parkinson's disease (PD). Perturbations of proteostasis at the endoplasmic reticulum (ER) triggers ER stress, activating the unfolded protein response (UPR). Chronic ER stress is thought to underlie the death of neurons during the neurodegenerative progression, but the precise mechanism by which the UPR pathways regulate neuronal cell fate remains incompletely understood. Here we report a critical neurodegenerative role for inositol-requiring enzyme 1 (IRE1), the evolutionarily conserved ER stress sensor, in a Drosophila model of PD. We found that IRE1 was hyperactivated upon accumulation of α-synuclein in the fly photoreceptor neurons. Ectopic overexpression of IRE1 was sufficient to trigger autophagy-dependent neuron death in an XBP1-independent, JNK-dependent manner. Furthermore, IRE1 was able to promote dopaminergic neuron loss, progressive locomotor impairment, and shorter lifespan, whereas blocking IRE1 or ATG7 expression remarkably ameliorated the progression of α-synuclein-caused Parkinson's disease. These results provide in vivo evidence demonstrating that the IRE1 pathway drives PD progression through coupling ER stress to autophagy-dependent neuron death.


Subject(s)
Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Animals , Animals, Genetically Modified , Autophagy/physiology , Disease Models, Animal , Drosophila Proteins , Drosophila melanogaster , Endoplasmic Reticulum Stress , Endoribonucleases , Humans , Neurons/pathology , Parkinson Disease/genetics , Signal Transduction , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics
13.
J Cancer Res Clin Oncol ; 145(11): 2637-2647, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31598791

ABSTRACT

PURPOSE: Malignant ascites (MA) is a common manifestation in advanced gastric cancer with peritoneal carcinomatosis and usually indicates a poor prognosis. However, lack of in vitro models that can faithfully recapitulate the characteristics of tumour cells in ascites hinders related researches. Tumour organoids have emerged as a robust in vitro model for tumour research and drug screening. Hence, we aimed to generate a 3-D in vitro organoid cultures from malignant ascites of gastric cancer for disease modelling and drug screening. METHODS: Eleven MADOs were generated from the MA tumour cells of gastric cancer patients. We made comparisons between MADOs and original MA tumour cells in histopathology by immunohistochemistry and genomics by whole-exome sequencing. In order to evaluate MADOs as functional in vitro disease models, we tested whether MADOs could be used for drug sensitivity screens. RESULTS: Eleven MADO cultures from human gastric cancer were established. MADOs demonstrated divergent growth characteristics and morphologies. MADO cultures preserve the histological architecture, genomic landscape of the corresponding MA tumour cells. MADOs exhibited heterogeneous responses to standard-of-care chemotherapeutics. CONCLUSIONS: We generated MADOs modelling characteristics and mutated genes of MA tumour cells. A broad range of intrinsic MADO response to conventional chemotherapeutics suggests MADOs are amenable to drug screening.


Subject(s)
Antineoplastic Agents/pharmacology , Ascites/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/methods , Organ Culture Techniques/methods , Organoids/pathology , Stomach Neoplasms/pathology , Humans , In Vitro Techniques , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Tumor Cells, Cultured , Exome Sequencing
14.
EBioMedicine ; 47: 65-77, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31495720

ABSTRACT

BACKGROUND: Cisplatin resistance remains a major clinical obstacle to the successful treatment of non-small cell lung cancer (NSCLC). Scribble contributes to ROS-induced inflammation and cisplatin-elevated toxic reactive oxygen species (ROS) promotes cell death. However, it is unknown whether and how Scribble is involved in the cisplatin-related cell death and the underlying mechanism of Scribble in response to chemotherapies and in the process of oxidative stress in NSCLC. METHODS: We used two independent cohorts of NSCLC samples derived from patients treated with platinum-containing chemotherapy and xenograft modeling in vivo. We analyzed the correlation between Scribble and Nox2 or Nrf2/PD-L1 both in vivo and in vitro, and explored the role of Scribble in cisplatin-induced ROS and apoptosis. FINDINGS: Clinical analysis revealed that Scribble expression positively correlated with clinical outcomes and chemotherapeutic sensitivity in NSCLC patients. Scribble protected Nox2 protein from proteasomal degradation. Scribble knockdown induced cisplatin resistance by blocking Nox2/ROS and apoptosis in LRR domain-dependent manner. In addition, low levels of Scribble correlated with high levels of PD-L1 via activation of Nrf2 transcription in vivo and in vitro. INTERPRETATIONS: Our study revealed that polarity protein Scribble increased cisplatin-induced ROS generation and is beneficial to chemotherapeutic outcomes in NSCLC. Although Scribble deficiency tends to lead to cisplatin resistance by Nox2/ROS and Nrf2/PD-L1, it is still possible that Scribble deficiency-induced PD-L1 may yield benefits in immunotherapy. FUND: National Key R&D Program of China, Strategic Priority Research Program of the Chinese Academy of Sciences, National Natural Science Foundation of China, China Postdoctoral Science Foundation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/drug effects , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Disease Models, Animal , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Mice , Mice, Knockout , NADPH Oxidases/metabolism , NF-E2-Related Factor 2/metabolism , Prognosis , Reactive Oxygen Species/metabolism
15.
Diabetes ; 68(8): 1577-1590, 2019 08.
Article in English | MEDLINE | ID: mdl-31127058

ABSTRACT

Insulin resistance is a major contributing factor in the development of metabolic disease. Although numerous functions of the polarity protein AF6 (afadin and MLLT4) have been identified, a direct effect on insulin sensitivity has not been previously described. We show that AF6 is elevated in the liver tissues of dietary and genetic mouse models of diabetes. We generated liver-specific AF6 knockout mice and show that these animals exhibit enhanced insulin sensitivity and liver glycogen storage, whereas overexpression of AF6 in wild-type mice by adenovirus-expressing AF6 led to the opposite phenotype. Similar observations were obtained from in vitro studies. In addition, we discovered that AF6 directly regulates IRS1/AKT kinase-mediated insulin signaling through its interaction with Src homology 2 domain-containing phosphatase 2 (SHP2) and its regulation of SHP2's tyrosine phosphatase activity. Finally, we show that knockdown of hepatic AF6 ameliorates hyperglycemia and insulin resistance in high-fat diet-fed or db/db diabetic mice. These results demonstrate a novel function for hepatic AF6 in the regulation of insulin sensitivity, providing important insights about the metabolic role of AF6.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Homeostasis/physiology , Insulin Resistance/physiology , Insulin/metabolism , Kinesins/metabolism , Liver/metabolism , Myosins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Cell Line , Diet, High-Fat , Glucose Tolerance Test , Hepatocytes/metabolism , Insulin Receptor Substrate Proteins/metabolism , Kinesins/genetics , Male , Mice , Mice, Knockout , Myosins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
16.
Mol Carcinog ; 58(2): 206-218, 2019 02.
Article in English | MEDLINE | ID: mdl-30294829

ABSTRACT

Pancreatic cancer is regarded as the most fatal and aggressive malignancy cancer due to its low 5-year survival rate and poor prognosis. The approaches of early diagnosis and treatment are limited, which makes it urgent to identify the complex mechanism of pancreatic oncogenesis. In this study, we used RNA-seq to investigate the transcriptomic (mRNA and miRNA) profiles of pancreatic cancer in paired tumor and normal pancreatic samples from ten patients. More than 1000 differentially expressed genes were identified, nearly half of which were also found to be differentially expressed in the majority of examined patients. Functional enrichment analysis revealed that these genes were significantly enriched in multicellular organismal and metabolic process, secretion, mineral transport, and intercellular communication. In addition, only 24 differentially expressed miRNAs were found, all of which have been reported to be associated with pancreatic cancer. Furthermore, an integrated miRNA-mRNA interaction network was generated using multiple resources. Based on the calculation of disease correlation scores developed here, several genes present in the largest connected subnetwork, such as albumin, ATPase H+ /K+ exchanging alpha polypeptide and carcinoembryonic antigen-related cell adhesion molecule 1, were considered as novel genes that play important roles in the development of pancreatic cancer. Overall, our data provide new insights into further understanding of key molecular mechanisms underlying pancreatic tumorigenesis.


Subject(s)
Gene Expression Profiling/methods , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , RNA, Messenger/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Sequence Analysis, RNA/methods
17.
Int J Oncol ; 53(6): 2671-2682, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30272271

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, with a marked potential for invasion and metastasis. Emerging evidence has suggested that dysregulation of long non-coding RNAs (lncRNAs) is associated with the development of multiple types of cancer. However, the function of lncRNAs in PDAC is poorly known. In the present study, a microarray assay was used to screen for differently expressed lncRNAs in PDAC and it was identified that cancer upregulated drug resistance (CUDR) was upregulated in PDAC. CUDR increased PDAC cell proliferation, migration and invasion, inhibited apoptosis, and promoted drug resistance; it also regulated the PDAC cell epithelial-mesenchymal transition. The CUDR-induced PDAC malignant phenotypes is via the protein kinase B and extracellular-signal-regulated kinase signaling pathways. Downregulation of CUDR may be a novel therapeutic strategy to prevent PDAC development and drug resistance in the future.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Gene Expression Profiling/methods , MAP Kinase Signaling System , Pancreatic Neoplasms/pathology , RNA, Long Noncoding/genetics , Adult , Aged , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis/methods , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation
18.
Nat Commun ; 9(1): 3486, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154439

ABSTRACT

Loss of WW domain-containing oxidoreductase (Wwox) expression has been observed in breast cancer (BC). However, its regulatory effects are largely unknown, especially in triple-negative breast cancer (TNBC). Herein, gene expression profiling revealed that JAK/STAT3 pathway was one of the most differentially modulated pathways in basal-like BC cells. The lower expression of Wwox was significantly correlated with high activation of STAT3 in basal-like cells and TNBC tissues. Overexpression of Wwox markedly inhibited proliferation and metastasis of BC cells by suppressing STAT3 activation, which is to interact with JAK2 to inhibit JAK2 and STAT3 phosphorylation. Furthermore, Wwox limited STAT3 binding to the interleukin-6 promoter, repressing expression of the IL-6 cytokine. Altogether, our data established that Wwox suppresses BC cell metastasis and proliferation by JAK2/STAT3 pathway. Targeting of Wwox with STAT3 could offer a promising therapeutic strategy for TNBC.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , WW Domain-Containing Oxidoreductase/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunoprecipitation , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinase 2/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , NIH 3T3 Cells , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , STAT3 Transcription Factor/genetics , Signal Transduction , Triple Negative Breast Neoplasms/genetics , WW Domain-Containing Oxidoreductase/genetics
19.
Cancer Sci ; 109(5): 1382-1392, 2018 May.
Article in English | MEDLINE | ID: mdl-29601127

ABSTRACT

Metastasis to distant organs is a particularly ominous feature of malignant cancer. LKB1 (also known as STK11) has been identified as a tumor suppressor in several types of cancers. Here, we show that LKB1 is at low levels and is negatively associated with poor clinical outcomes in pancreatic cancer (PC). LKB1 is inversely correlated with Snail protein in PC, in which the loss of LKB1 facilitates metastasis through elevating Snail protein level. Furthermore, LKB1 boosts Snail's interaction with E3 ligase FBXL14, leading to increasing ubiquitin-mediated Snail degradation. Notably, metformin could increase Snail protein ubiquitination via augmenting the location of LKB1 at cytoplasm as well as increasing LKB1 expression. Altogether, our data established that LKB1 impedes invasion and metastasis by decreasing the Snail protein level in PC. Targeting the LKB1/FBXL14/Snail axis may represent a promising therapeutic strategy and metformin might be beneficial for PC therapy through activating the LKB1-mediated Snail ubiquitination pathway.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Metformin/pharmacology , Pancreatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Snail Family Transcription Factors/chemistry , AMP-Activated Protein Kinase Kinases , Animals , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , F-Box Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/drug therapy , Snail Family Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
20.
ACS Appl Mater Interfaces ; 10(4): 3449-3458, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-29318884

ABSTRACT

Titanium dioxide nanoparticles (TiO2NPs) are among the most widely manufactured nanomaterials with broad applications in food industry, cosmetics, and medicine. Although the toxicity of TiO2NPs at high doses has been extensively explored, the potential health risks of TiO2NPs exposure at nontoxic concentrations remain poorly understood. Epithelial-mesenchymal transition (EMT) plays pivotal roles in a diversity of physiological and pathological processes, including tissue regeneration and cancer metastasis. In this study, we find that the cellular uptake of TiO2NPs inhibits EMT-mediated cell remodeling and cell migration without exhibiting cytotoxicity. Further investigation reveals that TiO2NPs suppress the process of EMT through the blockade of transforming growth factor-ß (TGFß) signaling. Particularly, TiO2NPs interact with the TGFß receptor TßRI/II complex, induce its lysosomal degradation, and thereby downregulate expression of TGFß target genes. Moreover, we show that waterborne TiO2NPs do not elicit toxicity in healthy tissues but hamper EMT-mediated wound healing in two animal models. Long-term exposure of TiO2NPs in environmental water and drinking water impede the regeneration of amputated fin in zebrafish and the recovery of intestinal mucosal damage in colitic mice. Our results reveal the previously unknown effects of TiO2NPs during tissue remodeling and repair, which have significant implications in their risk assessment and management.


Subject(s)
Epithelial-Mesenchymal Transition , Animals , Mice , Nanoparticles , Regeneration , Titanium , Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL
...