Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
BMC Cancer ; 22(1): 530, 2022 May 11.
Article in English | MEDLINE | ID: mdl-35545767

ABSTRACT

BACKGROUNDS: Salivary biomarkers hold huge potential for the non-invasive diagnosis of oral squamous cell carcinoma. Angiogenic factors and matrix-metalloproteinases (MMPs) are highly expressed in OSCC tissue, but their expression patterns in the saliva are unknown. This study aimed to analyze the levels of angiogenic factors and MMPs in tumor tissue and saliva of OSCC patients. METHODS: OSCC-tissue, adjacent normal tissue (ANT), saliva from OSCC patients, and healthy controls were obtained. The expression patterns of angiogenic factors and MMPs were analyzed by immunohistochemistry, protein chip array, and RT-qPCR. RESULTS: Results showed higher expression of ANG, ANG-2, HGF, PIGF, VEGF, MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-13, TIMP-1, and TIMP-2 in OSCC-tissues compared to the ANT. Among the overexpressed markers in OSCC-tissues, HGF, VEGF, PIGF, PDGF-BB, MMP-1, MMP-3, MMP-8, MMP-9, MMP-10, MMP-13, and TIMP-2 were significantly upregulated in the saliva of OSCC patients compared to healthy controls. CONCLUSIONS: The levels of HGF, VEGF, PIGF, MMP-1, MMP-3, MMP-8, MMP-9, MMP-10, MMP-13, and TIMP-2 were upregulated both in OSCC tissue and saliva of OSCC patients. Bioinformatic analysis revealed the correlation of these factors with patient survival and cancer functional states in head and neck cancer, indicating these factors as possible saliva-based non-invasive diagnostic/prognostic markers and therapeutic targets of OSCC.


Subject(s)
Biomarkers, Tumor , Mouth Neoplasms , Squamous Cell Carcinoma of Head and Neck , Biomarkers, Tumor/metabolism , Humans , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Mouth Neoplasms/diagnosis , Mouth Neoplasms/pathology , Placenta Growth Factor/metabolism , Saliva/metabolism , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/pathology , Tissue Inhibitor of Metalloproteinase-2/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
J Transl Med ; 18(1): 109, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32122379

ABSTRACT

BACKGROUND: Myelin sheaths surrounding axons are critical for electrical signal transmission in the central nervous system (CNS). Diseases with myelin defects such as multiple sclerosis (MS) are devastating neurological conditions for which few effective treatments are available. Dysfunction of the dopaminergic system has been observed in multiple neurological disorders. Its role in myelin pathogenesis, however, is unclear. METHODS: This work used a combination of literature curation, bioinformatics, pharmacological and genetic manipulation, as well as confocal imaging techniques. Literature search was used to establish a complete set of genes which is associated with MS in humans. Bioinformatics analyses include pathway enrichment and crosstalk analyses with human genetic association studies as well as gene set enrichment and causal relationship analyses with transcriptome data. Pharmacological and CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genetic manipulation were applied to inhibit the dopaminergic signaling in zebrafish. Imaging techniques were used to visualize myelin formation in vivo. RESULTS: Systematic analysis of human genetic association studies revealed that the dopaminergic synapse signaling pathway is enriched in candidate gene sets. Transcriptome analysis confirmed that expression of multiple dopaminergic gene sets was significantly altered in patients with MS. Pathway crosstalk analysis and gene set causal relationship analysis reveal that the dopaminergic synapse signaling pathway interacts with or is associated with other critical pathways involved in MS. We also found that disruption of the dopaminergic system leads to myelin deficiency in zebrafish. CONCLUSIONS: Dopaminergic signaling may be involved in myelin pathogenesis. This study may offer a novel molecular mechanism of demyelination in the nervous system.


Subject(s)
Myelin Sheath , Zebrafish , Animals , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Signal Transduction , Zebrafish/genetics
3.
Development ; 143(6): 1005-15, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26893349

ABSTRACT

Insulin inhibits transcription factor Forkhead box O (FoxO) activity, and the steroid hormone 20-hydroxyecdysone (20E) activates FoxO; however, the mechanism is unclear. We hypothesized that 20E upregulates phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase (PTEN) expression to activate FoxO, thereby promoting proteolysis during molting in the lepidopteran insect Helicoverpa armigera. FoxO expression is increased during molting and metamorphosis. The knockdown of FoxO in fifth instar larvae results in larval molting failure. 20E inhibits FoxO phosphorylation, resulting in FoxO nuclear translocation. Insulin, via Akt, induces FoxO phosphorylation and cytoplasmic localization. 20E represses insulin-induced Akt phosphorylation and FoxO phosphorylation. 20E, via ecdysone receptor B1 (EcRB1) and the ultraspiracle protein (USP1), upregulates PTEN expression, which represses Akt phosphorylation, thereby repressing FoxO phosphorylation. The non-phosphorylated FoxO enters the nucleus and attaches to a FoxO-binding element in the upstream region of the Broad isoform 7 (BrZ7) gene to regulate BrZ7 transcription under 20E induction. 20E upregulates FoxO expression via EcRB1 and USP1. FoxO regulation of BrZ7 expression regulates Carboxypeptidase A expression for final proteolysis during insect molting. Hence, 20E activates FoxO via upregulating PTEN expression to counteract insulin activity and promote proteolysis.


Subject(s)
Ecdysterone/pharmacology , Forkhead Transcription Factors/metabolism , Molting/drug effects , Moths/growth & development , Proteolysis/drug effects , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Conserved Sequence , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Insect Proteins/metabolism , Insulin/pharmacology , Larva/physiology , Models, Biological , Moths/drug effects , Moths/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects
4.
Lipids Health Dis ; 18(1): 138, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31186008

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) infection remains a global health issue associated with substantial morbidity and mortality. Serum apolipoprotein C3 (ApoC3) and apolipoprotein A5 (ApoA5) levels were decreased in chronic hepatitis B (CHB) patients, however the relationship between ApoC3 or ApoA5 and HBV DNA load remains elusive. METHODS: A total of 384 CHB patients including 194 HBsAg(+) HBeAg(-) and 190 HBsAg(+) HBeAg(+) and 154 healthy individuals were recruited in our study. Serum levels of alanine aminotransferase (ALT), aspartate transaminase (AST), total cholesterol (Chol), triglycerides (TG), apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), high-density lipoproteins cholesterol (HDL-C), low-density lipoproteins cholesterol (LDL-C) and lipoprotein a (Lpa) were examined in an automatic biochemical analyzer. Apolipoprotein A5 (ApoA5) and apolipoprotein C3 (ApoC3) were detected via ELISA. RESULTS: Serum ApoA1, ApoB, ApoC3 and ApoA5 levels were reduced in CHB patients. In HBeAg(-) CHB patients, plasma ApoC3 levels were negatively associated with HBV DNA load (r = 0.219, P < 0.001). But no correlation between ApoA5 and HBV DNA load was observed in CHB patients. CONCLUSIONS: These data showed that HBV infection inhibits lipid metabolism and ApoC3 is negatively associated with HBV DNA load in HBeAg (-) CHB patients. These findings provided new evidence about the link between ApoC3-related lipid metabolism and immune response.


Subject(s)
Cholesterol, HDL/blood , Cholesterol, LDL/blood , DNA, Viral/genetics , Hepatitis B e Antigens/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/blood , Hepatitis B, Chronic/virology , Triglycerides/blood , Adult , Apolipoprotein A-V/blood , Apolipoprotein C-III , Apolipoproteins B/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
5.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 39(4): 499-505, 2017 Aug 20.
Article in English | MEDLINE | ID: mdl-28877827

ABSTRACT

Objective To investigate the effects of gender and age on the prevalence and complications of nonalcoholic fatty liver disease(NAFLD). Methods A total of 8429 NAFLD patients were selected from the Health Check-up Center and Outpatient Departments of Qilu Hospital of Shandong University(Qingdao).The questionnaire-based survey,physical examinations,biochemical tests,and liver ultrasonography were performed for all cases.Patients were divided into young group(<45 years),middle aged group(45 years≤age<60 years),and old group(≥60 years)according to age,and the clinical features and laboratory findings were analyzed. Results The proportion of male patients gradually decreased with age,while the proportion of female patients increased(P<0.01);The incidences of metabolic diseases showed significant difference among young group,middle aged group,and old group(P<0.01).Except for hyperlipidemia,the proportion of male patients with NAFLD-accompanied metabolic symdrome was significantly higher than that of female patients in all three age groups(all P<0.01). Conclusions The prevalence of NAFLD-accompanied metabolic syndrome disease is associated with age and gender.This finding is useful for the prevention and treatment of NAFLD.


Subject(s)
Age Factors , Metabolic Syndrome/epidemiology , Non-alcoholic Fatty Liver Disease/epidemiology , Sex Factors , Female , Humans , Male , Middle Aged , Prevalence , Risk Factors , Ultrasonography
6.
J Biol Chem ; 289(19): 13026-41, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24692553

ABSTRACT

In addition to the classical nuclear receptor pathway, there is a nongenomic pathway in the cell membrane that regulates gene expression in animal steroid hormone signaling; however, this mechanism is unclear. Here, we report that the insect steroid hormone 20-hydroxyecdysone (20E) regulates calcium influx via phospholipase Cγ1 (PLCG1) to modulate the protein kinase C phosphorylation of the transcription factor ultraspiracle (USP1) in the lepidopteran insect Helicoverpa armigera. The PLCG1 mRNA levels are increased during the molting and metamorphic stages. The depletion of PLCG1 by RNA interference can block 20E-enhanced pupation, cause larvae death and pupation defects, and repress 20E-induced gene expression. 20E may induce the tyrosine phosphorylation of PLCG1 at the cytosolic tyrosine kinase (Src) homology 2 domains and then determine the migration of PLCG1 toward the plasma membrane. The G-protein-coupled receptor (GPCR) inhibitor suramin, Src family kinase inhibitor PP2, and the depletions of ecdysone-responsible GPCR (ErGPCR) and Gαq restrain the 20E-induced tyrosine phosphorylation of PLCG1. PLCG1 participates in the 20E-induced Ca(2+) influx. The inhibition of GPCR, PLC, inositol 1,4,5-trisphosphate receptor, and calcium channels represses the 20E-induced Ca(2+) influx. Through calcium signaling, PLCG1 mediates the transcriptional activation driven by the ecdysone-response element. Through PLCG1 and calcium signaling, 20E regulates PKC phosphorylation of USP1 at Ser-21 to determine its ecdysone-response element binding activity. These results suggest that 20E activates PLCG1 via the ErGPCR and Src family kinases to regulate Ca(2+) influx and PKC phosphorylation of USP1 to subsequently modulate gene transcription for metamorphosis.


Subject(s)
Calcium Signaling/physiology , Ecdysterone/metabolism , Insect Proteins/metabolism , Moths/metabolism , Phospholipase C gamma/metabolism , Receptors, Steroid/metabolism , Animals , Antinematodal Agents/pharmacology , Base Sequence , Calcium Signaling/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Ecdysterone/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Insect Proteins/genetics , Molecular Sequence Data , Moths/genetics , Phospholipase C gamma/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Steroid/genetics , Suramin/pharmacology
7.
J Biol Chem ; 289(38): 26630-26641, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25096576

ABSTRACT

The steroid hormone 20-hydroxyecdysone (20E) initiates insect molting and metamorphosis. By contrast, juvenile hormone (JH) prevents metamorphosis. However, the mechanism by which JH inhibits metamorphosis remains unclear. In this study, we propose that JH induces the phosphorylation of Broad isoform Z7 (BrZ7), a newly identified protein, to inhibit 20E-mediated metamorphosis in the lepidopteran insect Helicoverpa armigera. The knockdown of BrZ7 in larvae inhibited metamorphosis by repressing the expression of the 20E response gene. BrZ7 was weakly expressed and phosphorylated during larval growth but highly expressed and non-phosphorylated during metamorphosis. JH regulated the rapid phosphorylation of BrZ7 via a G-protein-coupled receptor-, phospholipase C-, and protein kinase C-triggered pathway. The phosphorylated BrZ7 bound to the 5'-regulatory region of calponin to regulate its expression in the JH pathway. Exogenous JH induced BrZ7 phosphorylation to prevent metamorphosis by suppressing 20E-related gene transcription. JH promoted non-phosphorylated calponin interacting with ultraspiracle protein to activate the JH pathway and antagonize the 20E pathway. This study reveals one of the possible mechanisms by which JH counteracts 20E-regulated metamorphosis by inducing the phosphorylation of BrZ7.


Subject(s)
Ecdysterone/pharmacology , Insect Proteins/metabolism , Juvenile Hormones/physiology , Moths/growth & development , Protein Processing, Post-Translational , Transcription Factors/metabolism , 5' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cells, Cultured , Ecdysterone/physiology , Gene Expression Regulation , Insect Proteins/genetics , Juvenile Hormones/pharmacology , Larva/growth & development , Metamorphosis, Biological/drug effects , Molecular Sequence Data , Pest Control , Phosphorylation , Phylogeny , Protein Transport , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic
8.
Biochim Biophys Acta ; 1830(11): 5184-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23850472

ABSTRACT

BACKGROUND: Heat shock protein 90 (Hsp90) interacts with steroid hormone receptors, signaling kinases, and various transcription factors. However, the mechanism by which Hsp90 interacts with different proteins in various pathways remains unclear. METHODS: Western blot was used to study Hsp90 expression profile in Helicoverpa armigera (Lepidoptera). RNA interference was performed to investigate the function of Hsp90 in 20-hydroxyecdysone (20E) and juvenile hormone (JH) signal pathways. The binding of Hsp90 to the transcription factor ultraspiracle protein (USP1) and JH candidate receptor methoprene-tolerant (Met1) was analyzed by co-immunoprecipitation. Phospho-(Ser) PKC substrate antibody was used to detect Hsp90 phosphorylation. RESULTS: Hsp90 participated in 20E- or JH-induced gene expression. 20E induced the interaction between Hsp90 and USP1, whereas JH III and methoprene induced the interaction between Hsp90 and Met1, respectively. 20E and JH counteracted each other for these protein interactions. Both JH III and methoprene induced protein kinase C (PKC) phosphorylation of Hsp90. This process could be inhibited by phospholipase C (PLC) and PKC inhibitors. 20E suppressed JH III- or methoprene-induced PKC phosphorylation of Hsp90. CONCLUSION: 20E maintained the non-PKC-phosphorylation status of Hsp90. Hsp90 interacted with USP1 to induce gene expression in the 20E pathway. JH regulated the PKC-phosphorylation status of Hsp90. Hsp90 also interacted with Met1 to induce gene expression in the JH pathway. GENERAL SIGNIFICANCE: Our study describes a novel mechanism of Hsp90 action by altering phosphorylation and protein interaction in various hormonal signaling pathways.


Subject(s)
Ecdysterone/metabolism , HSP90 Heat-Shock Proteins/metabolism , Insecta/metabolism , Juvenile Hormones/metabolism , Protein Interaction Domains and Motifs/genetics , Animals , Ecdysterone/genetics , Gene Expression , HSP90 Heat-Shock Proteins/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Insecta/genetics , Juvenile Hormones/genetics , Lepidoptera/genetics , Lepidoptera/metabolism , Methoprene/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation
9.
Cell Commun Signal ; 12: 9, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24507557

ABSTRACT

BACKGROUND: Animal steroid hormones are conventionally known to initiate signaling via a genomic pathway by binding to the nuclear receptors. The mechanism by which 20E initiates signaling via a nongenomic pathway is unclear. RESULTS: We illustrate that 20E triggered the nongenomic pathway through a plasma membrane G-protein-coupled receptor (named ErGPCR) in the lepidopteran insect Helicoverpa armigera. The transcript of ErGPCR was increased at the larval molting stage and metamorphic molting stage by 20E regulation. Knockdown of ErGPCR via RNA interference in vivo blocked larval-pupal transition and suppressed 20E-induced gene expression. ErGPCR overexpression in the H. armigera epidermal cell line increased the 20E-induced gene expression. Through ErGPCR, 20E modulated Calponin nuclear translocation and phosphorylation, and induced a rapid increase in cytosolic Ca2+ levels. The inhibitors of T-type voltage-gated calcium channels and canonical transient receptor potential calcium channels repressed the 20E-induced Ca2+ increase. Truncation of the N-terminal extracellular region of ErGPCR inhibited its localization on the plasma membrane and 20E-induced gene expression. ErGPCR was not detected to bind with the steroid hormone analog [3H]Pon A. CONCLUSION: These results suggest that ErGPCR participates in 20E signaling on the plasma membrane.


Subject(s)
Calcium Signaling , Cell Membrane/metabolism , Ecdysterone/metabolism , Insect Proteins/metabolism , Lepidoptera/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Insect Proteins/genetics , Microfilament Proteins/metabolism , Molecular Sequence Data , Receptors, G-Protein-Coupled/genetics , Calponins
10.
Oncol Rep ; 52(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-39027990

ABSTRACT

Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that certain of the immunohistochemical data shown in Fig. 5C were strikingly similar to data appearing in different form in another article written by different authors at different research institutes that had already been submitted for publication elsewhere prior to the submission of this paper to Oncology Reports [Wu X, Cai D, Zhang F, Li M and Wan Q: Long noncoding RNA TUSC7 inhibits cell proliferation, migration and invasion by regulating SOCS4 (SOCS5) expression through targeting miR­616 in endometrial carcinoma. Life Sci 231: 116549, 2019]. In addition, the CACNA203 western blot data shown in Fig. 2A­c and B­C respectively looked strikingly similar, even though different experiments were intended to have been shown in these figure parts. In view of the fact that the contentious data had already apparently been submitted for publication prior to the receipt of this paper at Oncology Reports, and owing to a overall lack of confidence in the presentation of the data, the Editor of has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Oncology Reports 43: 121­132, 2020; DOI: 10.3892/or.2019.7396].

11.
Cardiooncology ; 10(1): 56, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232830

ABSTRACT

BACKGROUND: Cardiovascular toxicity represents a significant adverse consequence of cancer therapies, yet there remains a paucity of effective biomarkers for its timely monitoring and diagnosis. To give a first evidence able to elucidate the role of Growth Differentiation Factor 15 (GDF15) in the context of cancer diagnosis and its specific association with cardiac indicators in cancer patients, thereby testing its potential in predicting the risk of CTRCD (cancer therapy related cardiac dysfunction). METHODS: Analysis of differentially expressed genes (DEGs), including GDF15, was performed by utilizing data from the public repositories of the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). Cardiomyopathy is the most common heart disease and its main clinical manifestations, such as heart failure and arrhythmia, are similar to those of CTRCD. Examination of GDF15 expression was conducted in various normal and cancerous tissues or sera, using available database and serum samples. The study further explored the correlation between GDF15 expression and the combined detection of cardiac troponin-T (c-TnT) and N-terminal prohormone of brain natriuretic peptide (NT-proBNP), assessing the combined diagnostic utility of these markers in predicting risk of CTRCD through longitudinal electrocardiograms (ECG). RESULTS: GDF15 emerged as a significant DEG in both cancer and cardiomyopathy disease models, demonstrating good diagnostic efficacy across multiple cancer types compared to healthy controls. GDF15 levels in cancer patients correlated with the established cardiac biomarkers c-TnT and NT-proBNP. Moreover, higher GDF15 levels correlated with an increased risk of ECG changes in the cancer cohort. CONCLUSION: GDF15 demonstrated promising diagnostic potential in cancer identification; higher GDF15, combined with elevated cardiac markers, may play a role in the monitoring and prediction of CTRCD risk.

12.
Dev Biol ; 371(1): 13-22, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22824427

ABSTRACT

The insulin and 20-hydroxyecdysone (20E) pathways coordinately regulate insect growth and metamorphosis. However, the molecular mechanism of the interaction of these two pathways in regulating insect development is not well understood. In the present study, we found that a small GTPase Rab4b from a lepidopteran insect Helicoverpa armigera participates in gene transcription in the two pathways. The results show that RNA interference of Rab4b in larvae results in a decrease in glycogen levels, small pupae, abnormal metamorphic transition, or larval death. The molecular mechanisms are demonstrated that knockdown of Rab4b in the larvae suppresses the transcription of glycogen synthase (GS), as well as the metamorphic-initiating factor (Br) and hormone receptor 3 (HR3), but increases the transcription of Forkhead box class O (FOXO). Further studies in the cell line confirm that Rab4b is necessary for gene transcription in the insulin and 20E pathways. Rab4b locates in the cytoplasm and takes part in regulation on FOXO cytoplasmic location by insulin induction, but travels toward the cell membrane upon 20E induction without affecting the FOXO location. The transcription of Rab4b could be upregulated by insulin injection or glucose feeding to the larvae, but not by 20E or juvenile hormone analogy methoprene. Our data suggest that Rab4b takes part in metamorphosis by regulating gene transcription and glycogen level in the insulin and 20E pathways.


Subject(s)
Ecdysterone/metabolism , Glycogen/metabolism , Insulin/metabolism , Metamorphosis, Biological/physiology , Moths/growth & development , Signal Transduction/physiology , Transcription, Genetic/physiology , rab4 GTP-Binding Proteins/metabolism , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers/genetics , Forkhead Transcription Factors/metabolism , Glycogen Synthase/metabolism , Immunohistochemistry , Larva/growth & development , Larva/metabolism , Molecular Sequence Data , Moths/metabolism , Phylogeny , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , rab4 GTP-Binding Proteins/genetics
13.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 41(4): 471-477, 2023 Aug 01.
Article in English, Zh | MEDLINE | ID: mdl-37474480

ABSTRACT

When the use of root canal retreatment and apical surgery experiences difficulty in treating endodontic diseases, intentional replantation is an optional clinical technique used to retain the tooth. A 28-year-old female complained of chewing discomfort at the mandibular second molar after undergoing root canal treatment 3 month ago. History record and radiographic examination revealed that a C-shaped root canal system was filled with gutta-percha in the mandibular second molar. A radiolucency area existed at the root furcal area with a thin canal wall in the distal and mesial roots. Intentional replantation was used to treat this tooth. The clinical and radiographic results showed that intentional replantation and nano-biomaterial application facilitated infection control, tooth retention, and periodontal tissue regeneration.


Subject(s)
Root Canal Therapy , Tooth Replantation , Female , Humans , Adult , Dental Pulp Cavity , Gutta-Percha/therapeutic use , Tooth Root , Molar/surgery , Retreatment
14.
Apoptosis ; 17(12): 1327-39, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23011181

ABSTRACT

The insect midgut undergoes programmed cell death (PCD) during metamorphosis, but the molecular basis for this phenomenon has not been demonstrated. We report a mod(mdg4) protein [designated as mod(mdg4)1A] that is involved in hormonally regulated insect midgut PCD, from the lepidopteran Helicoverpa armigera. Mod(mdg4)1A is localized in the larval midgut and is highly expressed during metamorphosis. Knockdown of mod(mdg4)1a by feeding dsRNA to the larvae suppressed midgut PCD and delayed metamorphosis. The mechanism is that mod(mdg4)1a knockdown decreased the transcript levels of genes involved in PCD and metamorphosis, but increased the transcript level of inhibitor of apoptosis survivin. The transcript level of mod(mdg4)1a is independently upregulated by 20-hydroxyecdysone (20E) or juvenile hormone (JH) analog methoprene. Overlapped 20E and methoprene counteractively regulate the transcript level of mod(mdg4)1a. 20E upregulates the mod(mdg4)1a transcript level not through its nuclear receptor EcRB1. Methoprene upregulates the mod(mdg4)1a transcript level through the juvenile hormone candidate receptor Met. These findings indicate that mod(mdg4)1a participates in midgut PCD and metamorphosis by regulating the transcript levels of a network of genes via different pathways under 20E and JH regulation.


Subject(s)
Apoptosis , Insect Hormones/metabolism , Metamorphosis, Biological , Moths/growth & development , Moths/metabolism , Animals , Digestive System/cytology , Digestive System/growth & development , Digestive System/metabolism , Gene Expression Regulation, Developmental , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Larva/cytology , Larva/genetics , Larva/growth & development , Larva/metabolism , Moths/cytology , Moths/genetics
15.
Immun Inflamm Dis ; 10(2): 235-240, 2022 02.
Article in English | MEDLINE | ID: mdl-34784451

ABSTRACT

INTRODUCTION: Mast cells are regarded as a kind of classical anaphylaxis cells. However autoimmune diseases and allergic reactions have many similarities or overlaps. A large number of papers have proved that mast cells play a significant role in the pathogenesis of systemic lupus erythematosus (SLE). It is speculated that IgE, anti-IgE antibodies, FcεRI, and anti-FcεRI antibodies activate mast cells through autoimmune pathways and participate in the disease process of SLE. Naturally occurring protein molecules not only exist in monomer form, but also in polymer of protein molecules. Therefore, whether IgE, FcεRIα, anti-IgE antibodies, and anti-FcεRI antibodies also exist in polymeric forms in the natural state is worthy of further investigation. METHODS: The serum samples and clinical data of 131 patients with SLE were collected from Qilu Hospital (Qingdao). Sixty healthy individuals were collected as the control group. Serum FcεRIα, anti-IgE, and anti-FcεRI were detected by enzyme-linked immunosorbent assay. Serum IgE was detected by rate scatter nephelometry. A Chinese hamster ovarian cancer cell line CHO3D10 transfected with human FcεRIα was cultured and the cell protein extract was prepared. The existence forms of FcεRIα in the cell protein extract were detected by the native-page method. RESULTS: The serum FcεRIα in SLE patients was significantly higher than that in control group (3.52 [2.18, 4.71] µg/ml and 1.87 [1.52, 2.33] µg/ml, respectively; p < .05). Anti-IgE was significantly lower than that in the control group (0.85 [0.55, 1.21] µg/ml and 1.23 [0.95, 1.58] µg/ml, respectively; p < .05). The CHO3D10 cell line expressed the FcεRIα, which had one kind of monomer (mFcεRIα) and two kinds of polymers (pFcεRIα) in the degeneration conditions. CONCLUSION: In patients with SLE, the expression of FcεRIα was increased and the level of anti-IgE was decreased. FcεRIα had one kind of monomer and two kinds of polymers. Mast cell-associated FcεRIα involved in the inflammatory lesion of SLE.


Subject(s)
Anaphylaxis , Lupus Erythematosus, Systemic , Basophils , Humans , Immunoglobulin E , Mast Cells
16.
Oncol Rep ; 43(1): 121-132, 2020 01.
Article in English | MEDLINE | ID: mdl-31746409

ABSTRACT

Endometrial cancer (EC) is one of the most common malignant gynecological tumors in women. The main treatments for EC (surgery, chemotherapy and radiation therapy) produce significant side effects. Thus, it is urgent to identify promising therapeutic targets and prognostic markers. CACNA2D3, as a member of the calcium channel regulatory α2δ subunit family, is reported to exert a tumor suppressive effect in numerous cancers. However, the function of CACNA2D3 in EC is not well known. In the present study, CACNA2D3 was lowly expressed in EC tissues and cells. The overexpression of CACNA2D3 via lentiviral particle injection significantly blocked the tumor growth in an in vivo xenograft model. In vitro, the overexpression of CACNA2D3 markedly inhibited cell proliferation and migration, and promoted cell apoptosis and calcium influx. These data revealed that CACNA2D3 functions as a tumor suppressor in EC. It was also revealed that the addition of progesterone (P4) blocked tumor growth in Ishikawa­injected nude mice. P4 induced the expression of CACNA2D3 in vivo and in vitro, and the silencing of CACNA2D3 affected P4­inhibited cell proliferation and P4­induced cell apoptosis and calcium influx. In Ishikawa cells, P4 enhanced the expression of phosphorylated (p)­p38 MAPK and PTEN, but blocked the levels of p­PI3K and p­AKT. The knockdown of CACNA2D3 blocked the function of P4. These data revealed that P4 promoted cell apoptosis via the activation of the CACNA2D3/Ca2+/p38 MAPK pathway, and blocked cell proliferation via suppression of the PI3K/AKT pathway. Collectively, these findings indicated the antitumor role of CACNA2D3 in EC, and revealed the mechanism of P4 inhibition of EC progression, which provided a new target for EC therapy and new evidence for P4 in EC therapy.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Down-Regulation , Endometrial Neoplasms/drug therapy , Progesterone/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Progesterone/pharmacology , Xenograft Model Antitumor Assays
17.
Exp Anim ; 69(2): 168-177, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-31748426

ABSTRACT

P-element-induced wimpy testis (PIWI)-interacting RNAs (piRNAs), a novel class of noncoding RNAs, are involved in the carcinogenesis. However, the functional significance of piRNAs in oral squamous cell carcinoma (OSCC) remains unknown. In the present study, we used chemical carcinogen 4-nitroquinoline-1-oxide (4NQO) induced OSCC mouse model. piRNAs and mRNAs were profiled using next-generation sequencing in the tongue tumor tissues from 4NQO induction and healthy tongue tissues from control mice. Furthermore, we analyzed the differential gene expression of human OSCC in Gene Expression Omnibus (GEO) database. According to the common differentially expressed genes in the 4NQO model and human OSCC tissues, piRNAs and mRNAs network were established based on informatics method. A total of 14 known piRNAs and 435 novel predicted piRNAs were differently expressed in tumor tissue compared to healthy tissue. Among differently expressed piRNAs 260 were downregulated, and 189 were upregulated. The mRNA targets for the differentially expressed piRNAs were identified using RNAhybrid software. Primary immunodeficiency and herpes simplex infection were the most enriched pathways. A total of 22 mRNAs overlapped in human and mice OSCC. Moreover, we established the regulatory network of 11 mRNAs, including Tmc5, Galnt6, Spedf, Mybl2, Muc5b, Six31, Pigr, Lamc2, Mmp13, Mal, and Mamdc2, and 11 novel piRNAs. Our data showed the interaction between piRNAs and mRNAs in OSCC, which might provide new insights in the development of diagnostic biomarkers and therapeutic targets of OSCC.


Subject(s)
Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/genetics , Mouth Neoplasms/genetics , RNA, Messenger , Testis , Animals , Disease Models, Animal , Male , Mice
18.
Gene ; 754: 144899, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32544494

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays a crucial role in colorectal cancer (CRC) metastasis. Soluble E-cadherin (sE-cadherin) is a peptide degradation product of the E-cadherin, a key epithelial molecule of EMT. However, it is not known if elevated levels of sE-cadherin also occur during EMT. And the study of sE-cadherin in colorectal cancer is rare. The purpose of the study was to evaluate the relationship between sE-cadherin and EMT in CRC and to evaluate the diagnostic value of sE-cadherin as a serum marker for CRC. Transforming growth factor-ß1 (TGF-ß1) was used to induce EMT in HT29 and SW480 cells. The cells treated with TGF-ß1 showed morphological and biological behavior changes consistent with EMT. Western blot and ELISA showed the levels of sE-cadherin were increased during EMT in CRC cells. In addition, we intravenously injected luciferase-labeled SW480 cells into nude mice to construct CRC metastasis model. Following the elongation of time, the fluorescence intensity of the experimental group was gradually increased. Correspondingly, the serum concentration of sE-cadherin also increased during CRC metastasis in mice. Furthermore, compared to healthy subjects, significantly higher levels of serum sE-cadherin were also observed in CRC patients and correlated with clinicopathological features. For discriminating CRC from healthy controls, the area under the receiver operating characteristic (ROC) curve (AUC) of sE-cadherin was 0.853, while the optimal cut-off point was set at 5928.16 ng/ml, the diagnostic sensitivity was 73.9% and the specificity was 80%. Compared with current commercial biomarkers (CEA, CA19-9 and CA125), the diagnostic performance of sE-cadherin was highest. Combined sE-cadherin and CEA raised the sensitivity to 82.4%. Serum sE-cadherin level can be used as a potential diagnostic biomarker of CRC.


Subject(s)
Antigens, CD/blood , Biomarkers, Tumor/blood , Cadherins/blood , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Lung Neoplasms/secondary , Animals , Apoptosis , Colorectal Neoplasms/blood , Female , Humans , Lung Neoplasms/blood , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Prognosis , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Int J Clin Exp Pathol ; 12(2): 590-598, 2019.
Article in English | MEDLINE | ID: mdl-31933864

ABSTRACT

BACKGROUND: Hepatocellular carcinoma results in high cancer mortality and is difficult to eradicate because of its late stage at the time of diagnosis, multicentricity, and cirrhotic background. It is therefore urgent to explore effective and economical therapeutic methods to treat this disease. OBJECTIVES: We aimed to investigate the antitumor activity of exogenous opioid growth factor (OGF), as well as the effect of the combination of OGF and cisplatin on hepatocellular carcinoma. The possible underlying mechanisms were also explored. MATERIALS AND METHODS: RT-PCR and immunohistochemistry were employed to determine the expression of OGF receptor (OGFr) in hepatocellular carcinoma. MTT assays were used to explore the effect of OGF on cell migration and proliferation. Animal experiments were performed to explore the effect of OGF and DDP on tumors. RESULTS: OGFr is present in human HCC cells and was differentially expressed between HCC tumor and non-tumor tissues. OGF inhibited HCC cell proliferation and migration. The silencing of OGFr blocked the expression of p21 and p53. Treatment using OGF, DDP and OGF+DDP all suppressed the growth of HCC tumors, with the maximum effect in the OGF+DDP group. CONCLUSION: Our study clarified that OGF inhibits cell migration and proliferation of HCC in animal experiments and that exogenous OGF enhances the anti-tumor activity of cisplatin on HCC by upregulating p21 and p53. These findings may provide a new strategy for future HCC therapeutics.

20.
Int J Clin Exp Pathol ; 12(4): 1194-1203, 2019.
Article in English | MEDLINE | ID: mdl-31933934

ABSTRACT

PURPOSE: Metastasis is the hallmark of gastric cancer (GC) and is the most widely recognized reason for GC-related deaths. However, the underlying mechanism of GC metastasis remains unknown. Herein we sought to investigate the biologic function of Gpx3 in gastric tumor metastasis and the underlying mechanism. METHODS: Cell migration and invasion was determined with Transwell chamber assay. Western blotting was used to determine protein expression levels of Gpx3, EMT markers and Wnt signaling related molecules. In vivo metastasis was determined with experiment lung metastasis model in tumor xenografts. RESULTS: Gpx3 expression was lower in GC patients and GC cell lines when compared with normal tissues and cells. Further studies showed that overexpression of Gpx3 was able to inhibit GC cell migration and invasion whereas Gpx3 knockdown promoted cell migration and invasion. Furthermore, AGS cells overexpressing Gpx3 showed lower metastatic potential when compared with the parental cells. Gpx3 was also found to regulate the expression of EMT markers. Mechanistic study showed that Gpx3 selectively inhibited Wnt/JNK signaling pathway over canonical Wnt/ß-catenin pathway. The data revealed that blockade of NFкB and JNK signaling pathway abolished siGpx3-induced cell migration and invasion. CONCLUSIONS: Taken together, we identify Gpx3 as a suppressor of GC metastasis. Above results provide the rationale that regulation of Gpx3 serves as a potential therapeutic option for GC.

SELECTION OF CITATIONS
SEARCH DETAIL