Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
BMC Anesthesiol ; 24(1): 172, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720250

ABSTRACT

BACKGROUND: Low immune function after laparoscopic total gastrectomy puts patients at risk of infection-related complications. Low-dose naloxone (LDN) can improve the prognosis of patients suffering from chronic inflammatory diseases or autoimmune diseases. The use of LDN during perioperative procedures may reduce perioperative complications. The purpose of this study was to examine the effects of LDN on endogenous immune function in gastric cancer patients and its specific mechanisms through a randomized controlled trial. METHODS: Fifty-five patients who underwent laparoscopic-assisted total gastrectomy were randomly assigned to either a naloxone group (n = 23) or a nonnaloxone group (n = 22). Patients in the naloxone group received 0.05 µg/kg-1.h- 1naloxone from 3 days before surgery to 5 days after surgery via a patient-controlled intravenous injection (PCIA) pump, and patients in the nonnaloxone group did not receive special treatment. The primary outcomes were the rates of postoperative complications and immune function assessed by NK cell, CD3+ T cell, CD4+ T cell, CD8+ T cell, WBC count, neutrophil percentage, and IL-6 and calcitonin levels. The secondary outcomes were the expression levels of TLR4 (Toll-like receptor), IL-6 and TNF-α in gastric cancer tissue. RESULTS: Compared with the nonnaloxone group, the naloxone group exhibited a lower incidence of infection (in the incision, abdomen, and lungs) (P < 0.05). The numbers of NK cells and CD8+ T cells in the naloxone group were significantly greater than those in the nonnaloxone group at 24 h after surgery (P < 0.05) and at 96 h after surgery (P < 0.05). Compared with those in the nonnaloxone group, the CD3 + T-cell (P < 0.05) and CD4 + T-cell (P < 0.01) counts were significantly lower in the naloxone group 24 h after surgery. At 24 h and 96 h after surgery, the WBC count (P < 0.05) and neutrophil percentage (P < 0.05) were significantly greater in the nonnaloxone group. The levels of IL-6 (P < 0.05) and calcitonin in the nonnaloxone group were significantly greater at 24 h after surgery. At 24 h following surgery, the nonnaloxone group had significantly greater levels of IL-6 (P < 0.05) and calcitonin than did the naloxone group. Compared with those in the naloxone group, the expression levels of TLR4 (P < 0.05) in gastric cancer tissue in the naloxone group were greater; however, the expression levels of IL-6 (P < 0.01) and TNF-α (P < 0.01) in the naloxone group were greater than those in the nonnaloxone group. CONCLUSION: Laparoscopic total gastrectomy patients can benefit from 0.05 ug/kg- 1. h- 1 naloxone by reducing their risk of infection. It is possible that LDN alters the number of cells in lymphocyte subpopulations, such as NK cells, CD3 + T cells, and CD4 + T cells, and the CD4+/CD8 + T-cell ratio or alters TLR4 receptor expression in immune cells, thereby altering immune cell activity. TRIAL REGISTRATION: The trial was registered at the Chinese Clinical Trial Registry on 24/11/2023 (ChiCTR2300077948).


Subject(s)
Gastrectomy , Laparoscopy , Naloxone , Postoperative Complications , Stomach Neoplasms , Humans , Naloxone/administration & dosage , Gastrectomy/methods , Male , Female , Laparoscopy/methods , Middle Aged , Stomach Neoplasms/surgery , Postoperative Complications/prevention & control , Aged , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Perioperative Care/methods , Interleukin-6 , Toll-Like Receptor 4
2.
Regen Ther ; 24: 351-360, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37674692

ABSTRACT

Objective: Senile osteoporosis (SOP) is an aging-related disease. The depalmitoylating enzyme Acyl-protein thiesterase1 (APT1) is involved in disease regulation. This study explored the mechanism of APT1 in SOP. Methods: Eight-week-old SAMP6 mice were selected as SOP models and SAMR1 mice were controls, while osteoblasts were isolated from the femoral surface-soft tissues of SOP and control mice as in vitro models. Mouse femur morphological, bone mineral density (BMD), femur maximum elastic stress and maximum load, and APT1 expression were detected by HE staining, X-ray bone densitometer, material testing machine, and RT-qPCR and Western blot (WB). Osteoprotegrin (OPG)-labeled osteoblasts and APT1 localization in bone tissues were detected by immunohistochemical staining. APT1 expression was promoted in SOP mice by tail vein injection of APT1 lentivirus or promoted/silenced in osteoblasts by transfection of pcDNA3.1-APT1 overexpression or si-APT1 plasmids. SOP mouse osteoblast differentiation (OD), OD-related protein levels, osteoblast proliferation, BMPR1a palmitoylation level, and BMP/Smad pathway were detected by alizarin red staining, ALP activity detection, WB, CCK-8, and IP-ABE method. The effects of the pathway inhibitor LDN-193189 on OD were detected. Results: APT1 was under-expressed in osteoblasts of bone tissue in SOP mice and mainly localized in osteoblasts. SOP mice manifested increased bone marrow cavity and bone trabecular space, thinned trabecular bone, decreased BMD, maximum elastic stress, maximum load, and reduced OPG-positive osteoblasts in bone tissues, which were averted by APT1 overexpression, thus alleviating SOP. APT1 overexpression increased osteoblast calcium nodules, ALP activity, OD-related protein levels, and cell proliferation. In mechanism, APT1 overexpression inhibited BMPR1a palmitoylation in SOP mouse osteoblasts and activated the BMP/Smad pathway, thus promoting OD. Conclusion: APT1 activated the BMP/Smad pathway and promoted OD by regulating BMPR1a depalmitoylation, thus alleviating mouse SOP.

3.
Biochem Pharmacol ; 197: 114907, 2022 03.
Article in English | MEDLINE | ID: mdl-35007523

ABSTRACT

Phosphorylation of proteins is one of the most extensively investigated post-translational protein modifications. Threonine, serine and tyrosine in proteins are the most commonly phosphorylated amino acids. Dysregulated cancer-related signaling pathways due to aberrant phosphorylation status of the key protein(s) in these pathways exist in most malignancies. Intensive studies in the recent decade have implicated long non-coding RNAs (lncRNAs) in the precise regulation of protein phosphorylation in cancers. In this review, we systematically delve into recent advance that underlines the multidimensional role of lncRNAs in modulating protein phosphorylation, regulating cancerous signaling and impacting prognosis of gastrointestinal (GI) cancers including hepatocellular carcinoma, colorectal cancer, gastric cancer, esophageal cancer, and pancreatic cancer. LncRNAs regulate protein phosphorylation via directly binding to the target protein(s), interacting with the partner protein(s) of the target protein(s) or lncRNAs-encoded small peptides. Although there are still extensive studies on disclosing the intricate interactions between lncRNAs and proteins and their impacts on protein phosphorylation, we believe that targeting lncRNAs controlling phosphorylation of key protein(s) in cancerous signaling pathways might provide novel paths for precision therapeutics of GI cancers in the future.


Subject(s)
Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Protein Processing, Post-Translational/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/drug therapy , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Processing, Post-Translational/drug effects
4.
Arch Toxicol ; 95(9): 3063-3070, 2021 09.
Article in English | MEDLINE | ID: mdl-34251499

ABSTRACT

Transarterial chemoembolization (TACE) is an effective treatment for unresectable hepatocellular carcinoma (HCC) patients. Although overall survival (OS) of TACE-treated patients has been evidently prolonged, not all unresectable HCC patients can benefit from TACE. Genome-wide association studies identified multiple HCC susceptibility single nucleotide polymorphisms (SNPs). However, it is still unclear how lncRNAs and their functional SNPs impact therapeutic responses of TACE. In the study, we hypothesized that the functional lncRNA H19 SNP(s) might impact H19 expression and, thus, prognosis of TACE-treated HCC patients. We found that the H19 rs3741219 SNP was significantly associated with OS of HCC patients received TACE. Cox proportional hazards model demonstrated that the rs3741219 CC genotype was associated with longer OS and a 37% decreased death risk compared with the TT carriers after TACE therapy (P = 0.001). Interestingly, the rs3741219 T-to-C change led to allelic down-regulation of lncRNA H19 expression via creating the binding sites of miR-146b-3p and miR-1539. Luciferase reporter gene assays indicated that miR-146b-3p and miR-1539 could markedly silence the rs3741219 C-allelic H19 expression but not lncRNA H19 with the T allele. Consistently, there was significantly reduced expression of lncRNA H19 in HCC and normal tissues of the C allele carriers compared with the H19 levels in patients with the T allele. Knock-down of lncRNA H19 significantly promoted the anti-viability efficiency of oxaliplatin (the main chemotherapy drug used in TACE) to HCC cells. In view of these results, we assume that lncRNA H19 might be a potential therapeutic target for unresectable HCC patients.


Subject(s)
Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Liver Neoplasms/therapy , RNA, Long Noncoding/genetics , Adult , Aged , Aged, 80 and over , Alleles , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Genome-Wide Association Study , Genotype , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Middle Aged , Oxaliplatin/pharmacology , Polymorphism, Single Nucleotide , Prognosis
5.
Biochem Pharmacol ; 188: 114558, 2021 06.
Article in English | MEDLINE | ID: mdl-33844983

ABSTRACT

Long non-coding RNAs (lncRNAs) are a class of RNA transcripts longer than 200 nucleotides and mostly cannot be translated into proteins. Next-generation transcriptome sequencing of various cell types has enabled the annotation of tens of thousands of lncRNAs in human genome. Varying levels of evidence supports the implications of lncRNAs in the onset and progression of cancers. Ubiquitin is an evolutionarily conserved protein and could post-translationally mark a number of proteins. The most important proteolytic role of ubiquitination is degradation of substrate proteins by the 26S proteasome. Compiling evidences demonstrated that lncRNAs are involved in the accurate execution of protein stability programs via the ubiquitin-proteasome system. In the current review, we systematically summarize the detailed mechanisms how lncRNAs modulate ubiquitination of target proteins, regulate cancerous signaling pathways and control tumorigenesis of gastrointestinal cancers. Although there are still considerable studies on unraveling the complicated interactions between lncRNAs and proteins, we believe that lncRNAs are promising but challenging molecules which may strongly facilitate precision cancer therapeutics in the future.


Subject(s)
Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Ubiquitinated Proteins/metabolism , Ubiquitination/physiology , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Gastrointestinal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , RNA, Long Noncoding/genetics , Ubiquitinated Proteins/genetics
6.
Drug Deliv ; 28(1): 478-486, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33641554

ABSTRACT

Many strategies have been developed to overcome the stratum corneum (SC) barrier, including functionalized nanostructures. Chemical penetration enhancers (CPEs) and cell-penetrating peptides (CPP) were applied to decorate nanostructured lipid carriers (NLC) for topical anesthetic and pain relief. A novel pyrenebutyrate (PB-PEG-DSPE) compound was synthesized by the amide action of the carboxylic acid group of PB with the amido groups of DSPE-PEG. PB-PEG-DSPE has a hydrophobic group, hydrophilic group, and lipid group. The lipid group can be inserted into NLC to form PB functional NLC. In order to improve the penetrability, TAT and PB multi-decorated NLC were designed for the delivery of lidocaine hydrochloride (LID) (TAT/PB LID NLC). The therapeutic effects of NLC in terms of in vitro skin penetration and in vivo in animal models were further studied. The size of TAT/PB LID NLC tested by DLS was 153.6 ± 4.3 nm. However, the size of undecorated LID NLC was 115.3 ± 3.6 nm. The PDI values of NLC vary from 0.13 ± 0.01 to 0.16 ± 0.03. Zeta potentials of NLC were negative, between -20.7 and -29.3 mV. TAT/PB LID NLC (851.2 ± 25.3 µg/cm2) showed remarkably better percutaneous penetration ability than PB LID NLC (610.7 ± 22.1 µg/cm2), TAT LID NLC (551.9 ± 21.8 µg/cm2) (p < .05) and non-modified LID NLC (428.2 ± 21.4 µg/cm2). TAT/PB LID NLC exhibited the most prominent anesthetic effect than single ligand decorated or undecorated LID NLC in vivo. The resulting TAT/PB LID NLC exhibited good skin penetration and anesthetic efficiency, which could be applied as a promising anesthesia system.


Subject(s)
Anesthetics, Local/administration & dosage , Drug Delivery Systems , Lidocaine/administration & dosage , Pain/drug therapy , Administration, Cutaneous , Anesthetics, Local/pharmacokinetics , Anesthetics, Local/pharmacology , Animals , Cell-Penetrating Peptides/chemistry , Disease Models, Animal , Drug Carriers/chemistry , Excipients/chemistry , Lidocaine/pharmacokinetics , Lidocaine/pharmacology , Lipids/chemistry , Mice , Nanostructures , Rats , Rats, Wistar , Skin Absorption
7.
Cancer Lett ; 499: 255-264, 2021 02 28.
Article in English | MEDLINE | ID: mdl-33242559

ABSTRACT

Despite advancements in therapeutic options, the overall prognosis for non-small-cell lung cancer (NSCLC) remains poor. Further exploration of the etiology and targets for novel treatments is crucial for managing NSCLC. In this study, we revealed the significant potential of EPB41 for inhibiting NSCLC proliferation, invasion and metastasis in vitro and in vivo. Consistent with its tumor suppressor role in NSCLC, the expression of EPB41 in NSCLC specimens evidently decreased compared to that in normal tissues, and low EPB41 expression was associated with poor prognoses for NSCLC patients. We further demonstrated the importance of EPB41 protein as a novel inhibitor of the Wnt signaling, which regulates ß-Catenin stability, and elucidated the crucial role of the EPB41/ALDOC/GSK3ß/ß-Catenin axis in NSCLC. Suppression of EPB41 expression in cancer cells elevated the levels of free ALDOC protein released from the EPB41-ALDOC complex, leading to disassembly of the ß-catenin destruction complex, reduced proteolytic degradation of ß-catenin, elevated cytoplasmic accumulation and nuclear translocation of ß-catenin, thereby activating the expression of multiple oncogenes and, thus, NSCLC pathogenesis. Our study highlights the potential of EPB41 as a future therapeutic target for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cytoskeletal Proteins/metabolism , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/surgery , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytoskeletal Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Kaplan-Meier Estimate , Lung/cytology , Lung/pathology , Lung/surgery , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/surgery , Membrane Proteins/genetics , Mice , Pneumonectomy , Proteolysis , RNA, Small Interfering/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism
8.
Drug Deliv ; 27(1): 1452-1460, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33100057

ABSTRACT

Local anesthetics (LAs) have been widely applied in clinic for regional anesthesia, postoperative analgesia, and management of acute and chronic pain. Nanostructured lipid carriers (NLCs) and lipid-polymer hybrid nanoparticles (LPNs) are reported as good choices for LA therapy. Transactivated transcriptional activator (TAT) was reported as a modifier for the topical delivery of drugs. In the present study, TAT modified, levobupivacaine (LEV) and dexmedetomidine (DEX) co-delivered NLCs (TAT-LEV&DEX-NLCs, T-L&D-N) and LPNs (TAT-LEV&DEX-LPNs, T-L&D-L) were designed and compared for the LA therapy. T-L&D-L exhibited better efficiency in improving the skin permeation, analgesic time, and pain control intensity than T-L&D-N both in vitro and in vivo. On the other side, T-L&D-N also improved the therapeutic effect of drugs to a large extent. These two systems both exhibited superiority in some respects. TAT modified LPNs are more promising platform for the long-term local anesthesia.


Subject(s)
Anesthesia, Local/methods , Anesthetics, Local/administration & dosage , Dexmedetomidine/administration & dosage , Levobupivacaine/administration & dosage , Nanostructures/administration & dosage , Transcriptional Activation/drug effects , Anesthetics, Local/metabolism , Animals , BALB 3T3 Cells , Dexmedetomidine/metabolism , Dose-Response Relationship, Drug , Levobupivacaine/metabolism , Lipids , Mice , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Organ Culture Techniques , Polymers/administration & dosage , Polymers/metabolism , Rats , Rats, Sprague-Dawley , Skin Absorption/drug effects , Skin Absorption/physiology , Transcriptional Activation/physiology
9.
Biomed Pharmacother ; 125: 109861, 2020 May.
Article in English | MEDLINE | ID: mdl-32070872

ABSTRACT

Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality in China. This study aimed to develop a hyaluronic acid (HA) decorated, pH sensitive lipid-polymer hybrid nanoparticles (LPH NPs) to co-deliver erlotinib (ERL) and bevacizumab (BEV) (HA-ERL/BEV-LPH NPs) for targeting and suppressing NSCLC. HA contained pH sensitive nano-materials were synthesized by acylation reaction. HA-ERL/BEV-LPH NPs were prepared using a sonication method. To explore the efficiency of the system, we evaluated the physicochemical parameters and performed a release study, a cellular uptake assay, a cytotoxicity evaluation, and several in vivo anti-tumor studies in comparison with free drugs and single drug systems. All LPH NPs samples have particle sizes of about 100-120 nm, polydispersity index values range from 0.12 to 0.15, and negative zeta potentials. HA-ERL/BEV-LPH NPs contained pH sensitive adipic acid dihydrazide (ADH) showed fast drug release at pH 5.5 than pH 7.4. After 21 days, the tumor volume of the HA-ERL/BEV-LPH NPs group (229.2 ± 13.1 mm3) was significantly smaller than 0.9 % NaCl control group (1126.3 ± 39.4 mm3), with a tumor inhibition rate of 79.7 ± 3.2 %. The maximum plasma ERL concentrations, half life period, and area under the curve of HA-ERL/BEV-LPH NPs were 21.6 µg/mL, 7.57 h, and 290.3 mg/L·h). With the highest tumor tissue accumulation concentration (25.3 µg/mL) and low system toxicity, HA-ERL/BEV-LPH NPs. HA-ERL/BEV-LPH NPs could be used as a promising system for the combination therapy of NSCLC.


Subject(s)
Bevacizumab/pharmacology , Erlotinib Hydrochloride/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/administration & dosage , Bevacizumab/chemistry , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , Disease Models, Animal , Drug Liberation , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/chemistry , Humans , Hydrogen-Ion Concentration , Lipids , Lung Neoplasms/therapy , Mice , Molecular Structure , Molecular Targeted Therapy , Nanoparticles , Polymers , Theranostic Nanomedicine , Xenograft Model Antitumor Assays
10.
Mol Cancer ; 18(1): 147, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31651347

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common malignancy worldwide and the second most lethal human cancer. A portion of patients with advanced HCC can significantly benefit from treatments with sorafenib, adriamycin, 5-fluorouracil and platinum drugs. However, most HCC patients eventually develop drug resistance, resulting in a poor prognosis. The mechanisms involved in HCC drug resistance are complex and inconclusive. Human transcripts without protein-coding potential are known as noncoding RNAs (ncRNAs), including microRNAs (miRNAs), small nucleolar RNAs (snoRNAs), long noncoding RNAs (lncRNAs) and circular RNA (circRNA). Accumulated evidences demonstrate that several deregulated miRNAs and lncRNAs are important regulators in the development of HCC drug resistance which elucidates their potential clinical implications. In this review, we summarized the detailed mechanisms by which miRNAs and lncRNAs affect HCC drug resistance. Multiple tumor-specific miRNAs and lncRNAs may serve as novel therapeutic targets and prognostic biomarkers for HCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Humans , Liver Neoplasms/drug therapy , RNA Interference , Sorafenib/pharmacology , Sorafenib/therapeutic use
11.
J Cell Biochem ; 120(8): 12638-12646, 2019 08.
Article in English | MEDLINE | ID: mdl-30861176

ABSTRACT

OBJECTIVES: Importin-4 (IPO4) is responsible for transporting histones H3 and H4 into the nucleus for chromatin assembly. But, the role of IPO4 in cancer, especially in gastric cancer (GC), has not been fully understood. We aim to determine the expression and function of IPO4 in GC. MATERIALS AND METHODS: Bioinformatics analysis was used to study the association of IPO4 and GC using GEO data and the Kaplan-Meier plotter. The quantitative real-time polymerase chain reaction and Western blot analysis were used to determine the IPO4 level in GC cells and tissues. Small interfering RNAs (siRNAs) were used to knockdown endogenous IPO4 expression in GC cells. Cell counting kit-8 (CCK-8), colony formation and transwell assays were used to examine the effect of IPO4 on cell proliferation and migration. RESULTS: IPO4 mRNA is overexpressed in GC tissues using bioinformatics analysis of three groups' transcriptome data, and high level of IPO4 is negatively correlated with poor long-term survival using the Kaplan-Meier plotter analysis. Western blot analysis further shows that IPO4 protein levels are also overexpressed in GC tissues and a number of GC cell lines. Endogenous IPO4 level can be inhibited by specific siRNA effectively. Importantly, CCK-8, colony formation, and transwell assays demonstrate that IPO4 knockdown by siRNA impairs GC cell proliferation and migration. CONCLUSIONS: Our data suggest that IPO4 contributes to GC progression and poor prognosis, and may function as a driving force in GC progression.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Membrane Transport Proteins/genetics , Stomach Neoplasms/metabolism , Adult , Aged , Cell Line, Tumor , Cell Proliferation , Computational Biology , Female , Gene Expression Profiling , Humans , Male , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/physiology , Middle Aged , Stomach Neoplasms/genetics , Stomach Neoplasms/physiopathology
12.
Medicine (Baltimore) ; 96(43): e8359, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29069020

ABSTRACT

This study is conducted to investigate the involvement of T-helper (Th) cells and regulatory T cells in epithelial ovarian cancer (EOC).The percentages of Th22, Th17, Th1, and regulatory T cells in the peripheral blood of EOC patients, benign ovarian epithelial neoplasm (BOEN) patients, and healthy control (HC) were examined by flow cytometry. Enzyme-linked immunosorbent assay was used to determine serum levels of interleukin (IL)-22, IL-17, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α).Th22 and Th17 were significantly increased in EOC patients. The plasma concentrations of IL-22 and TNF-α were significantly elevated in EOC patients compared with BOEN patients and HC. In EOC patients, there was an increased trend of Th22, IL-22, and TNF-α in stage III-IV patients compared with stage I-II patients. A positive correlation was seen among Th22, Th17, and Th1 cells in EOC patients. Similarly, positive correlations were detected between Th22 cells and IL-22 or TNF-α and between Th1 cells and interferon-γ (IFN-γ) in EOC patients. Besides, no significant difference was found in Th1 cells and regulatory T cells among EOC and BOEN patients and HC.There is a higher circulating frequency of Th22, Th17 cells, IL-22, and TNF-α concentration in EOC patients, which may conjointly participate in the pathogenesis and growth of EOC.


Subject(s)
Cytokines/blood , Neoplasms, Glandular and Epithelial/blood , Ovarian Neoplasms/blood , T-Lymphocytes, Helper-Inducer , T-Lymphocytes, Regulatory , Adult , Aged , Carcinoma, Ovarian Epithelial , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lymphocyte Count , Middle Aged
13.
Sci Rep ; 7(1): 11802, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28924212

ABSTRACT

Triple-negative breast cancer (TNBC) refers to one aggressive histological subtype of breast cancer with high heterogeneity and poor prognosis after standard therapy. Lack of clearly established molecular mechanism driving TNBC progression makes personalized therapy more difficult. Thus, identification of genetic variants associated with TNBC prognosis will show clinic significance for individualized treatments. Our study is aimed to evaluate the prognostic value of the genome wide association study (GWAS)-identified CHST9 rs1436904 and AQP4 rs527616 genetic variants in our established early-stage TNBC sample database. Cox regression was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). CHST9 rs1436904G allele was significantly associated with decreased disease-free survival time (DFS) (8.5 months shorter in GG genotype carriers compared to TT genotype carriers, HR = 1.70, 95% CI = 1.03-2.81, P = 0.038). Stratified analyses showed an increased risk of cancer progression in CHST9 rs1436904G allele carriers harboring larger tumor (tumor size > 2 cm), without lymph-node metastasis, being premenopausal at diagnosis or with vascular invasion (P = 0.032, 0.017, 0.008 or 0.003). Our findings demonstrate that the GWAS-identified 18q11.2 CHST9 rs1436904 polymorphism significantly contributes to prognosis of early-stage TNBC, suggesting its clinical potential in the screening of high-risk TNBC patients for recurrence and the possibility of patient-tailored therapeutic decisions.


Subject(s)
Genotype , Neoplasm Recurrence, Local , Polymorphism, Genetic , Sulfotransferases/genetics , Triple Negative Breast Neoplasms , Adult , Disease-Free Survival , Female , Genome-Wide Association Study , Humans , Middle Aged , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Survival Rate , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality
14.
Breast Cancer Res Treat ; 166(1): 109-116, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28744749

ABSTRACT

PURPOSE: As a subtype of breast cancer, triple-negative breast cancer (TNBC) shows poor prognosis and high heterogeneity. Precise identification of TNBC subgroups relevant to clinical prognosis is crucial in the design and administration of individualized treatments. This study aimed to evaluate the prognostic value of the functional BRCA1 rs799917 genetic variant in TNBC. METHODS: Associations between the rs799917 polymorphism and progression risk were investigated after genotyping 370 TNBC patients. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated by Cox regression.  RESULTS: We found that the rs799917T allele was associated with a significantly increased risk of disease progression and shortened progression-free survival time (PFS) (P = 0.001 for log-rank test). Notably, TNBC patients with the rs799917 CC genotype showed about 22 months prolonged PFS compared to the TT genotype after radiotherapy (HR 4.44, 95% CI 1.98-9.93; P = 2.9 × 10-4). Additionally, in overweight patients, the mean PFS of the rs799917TT genotype was 10 months shorter than that of the CC genotype (HR 3.57, 95% CI 1.46-8.73, P = 0.005). CONCLUSIONS: Our findings demonstrate that the functional BRCA1 genetic variant contributes to prognosis of TNBC. Our study also highlights the clinical potential of this polymorphism in the screening of high-risk TNBC patients for recurrence and the possibility of patient-tailored decisions especially during radiotherapy.


Subject(s)
BRCA1 Protein/genetics , Genetic Variation , Open Reading Frames , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Alleles , BRCA1 Protein/metabolism , Biomarkers, Tumor , Female , Genetic Predisposition to Disease , Genotype , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Polymorphism, Single Nucleotide , Prognosis , Proportional Hazards Models , Treatment Outcome , Triple Negative Breast Neoplasms/radiotherapy
15.
Biomed Pharmacother ; 89: 689-695, 2017 May.
Article in English | MEDLINE | ID: mdl-28267672

ABSTRACT

PURPOSE: There is a compelling need for prolonged local anesthetic that would be used for analgesia with a single administration. However, due to the low molecular weight of local anesthetics (LA) (lidocaine, bupivacaine, procaine, dibucaine, etc), they present fast systemic absorption. METHODS: The aim of the present study was to develop and evaluate bupivacaine lipid-polymer hybrid nanoparticles (BVC LPNs), and compared with BVC loaded PLGA nanoparticles (BVC NPs). Their morphology, particle size, zeta potential and drug loading capacity were evaluated. In vitro release study, stability and cytotoxicity were studied. In vivo evaluation of anesthetic effects was performed on animal models. RESULTS: A facile nanoprecipitation and self-assembly method was optimized to obtain BVC LPNs, composed of PLGA, lecithin and DSPE-PEG2000, of ∼175nm particle size. Compared to BVC NPs, BVC LPNs exhibited prolonged in vitro release in phosphate-buffered saline (pH=7.4). Further, BVC LPNs displayed enhanced in vitro stability in 10% FBS and lower cytotoxicity (the concentration of BVC ranging from 1.0µM to 20µM). In addition, BVC LPNs exhibited significantly prolonged analgesic duration. CONCLUSION: These results demonstrate that the LPNs could function as promising drug delivery system for overcoming the drawbacks of poor stability and rapid drug leakage, and prolonging the anesthetic effect with slight toxicity.


Subject(s)
Anesthetics, Local/administration & dosage , Anesthetics, Local/pharmacology , Bupivacaine/administration & dosage , Bupivacaine/pharmacology , Anesthetics, Local/chemistry , Animals , Bupivacaine/chemistry , Cell Survival/drug effects , Cells, Cultured , Delayed-Action Preparations , Drug Delivery Systems , Drug Stability , Lactic Acid/chemistry , Lipids/chemistry , Male , Mice , Mice, Inbred BALB C , Nanoparticles , Pain Measurement/drug effects , Particle Size , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/chemistry , Solubility
16.
Sci Rep ; 6: 27013, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27245877

ABSTRACT

Metallopeptidase 13 (MMP13), a well-known and highly regulated zinc-dependent MMP collagenase, plays a crucial part in development and progression of esophageal squamous cell carcinoma (ESCC). Therefore, we examined associations between ESCC susceptibility and four haplotype-tagging single nucleotide polymorphisms (htSNPs) using a two stage case-control strategy. Odds ratios (OR) and 95% confidence intervals (95% CI) were computed by logistic regression model. After analyzing 1588 ESCC patients and frequency-matched 1600 unaffected controls, we found that MMP13 rs2252070 G > A genetic polymorphism is significantly associated with ESCC risk in Chinese Han populations (GA: OR = 0.63, 95% CI = 0.54-0.74, P = 1.7 × 10(-6), AA: OR = 0.73, 95% CI = 0.66-0.81, P = 1.8 × 10(-6)). Interestingly, the rs2252070 G-to-A change was shown to diminish a Sp1-binding site in ESCC cells. Reporter gene assays indicated that the rs2252070 A allele locating in a potential MMP13 promoter has low promoter activities. After measuring MMP13 gene expression in sixty-six pairs of esophageal cancer and normal tissues, we observed that the rs2252070 A protective allele carriers showed decreased oncogene MMP13 expression. Results of these analyses underline the support of the notion that MMP13 might function as a key oncogene in esophageal carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Matrix Metalloproteinase 13/genetics , Polymorphism, Single Nucleotide , Sp1 Transcription Factor/genetics , Adult , Alleles , Asian People , Base Sequence , Binding Sites , Carcinoma, Squamous Cell/ethnology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Esophageal Neoplasms/ethnology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Gene Frequency , Haplotypes , Humans , Male , Matrix Metalloproteinase 13/metabolism , Middle Aged , Odds Ratio , Promoter Regions, Genetic , Risk Factors , Sp1 Transcription Factor/metabolism
17.
PLoS One ; 9(11): e110863, 2014.
Article in English | MEDLINE | ID: mdl-25365256

ABSTRACT

Recent genome-wide association studies (GWAS) have identified eleven leukocyte telomere length (LTL)-related single nucleotide polymorphisms (SNPs). Since LTL has been associated with risk of many malignancies, LTL-related SNPs may contribute to cancer susceptibility. To test this hypothesis in hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC), we genotyped these eleven LTL-related SNPs in a case-control set including 1186 HBV-related HCC cases, 508 chronic HBV carriers and 1308 healthy controls at the discovery stage. The associations of HCC risk with these SNPs were further confirmed in an independent case-control set. We found that 1p34.2 rs621559 and 14q21 rs398652 were significantly associated with HBV-related HCC risk (both P<0.005 after Bonferroni corrections). There was no significant difference of either rs621559 or rs398652 genotypes between chronic HBV carriers and healthy controls, demonstrating that the association was not due to predisposition to HBV infection. In the pooled analyses (1806 HBV-related HCC cases and 1954 controls), we observed a decreased HCC risk, 0.72-times, associated with the 1p34.2 rs621559 AA genotype compared to the GG genotype (P = 1.6×10(-6)). Additionally, there was an increased HCC risk, 1.27-fold, associated with the rs398652 GG genotype (P = 3.3×10(-6)). A statistical joint effect between the rs621559 GG and rs398652 GG genotypes may exist in elevating risk of HBV-related HCC. We show, for the first time, that rs398652 and rs621559 might be marker genetic variants for risk of HBV-related HCC in the Chinese population.


Subject(s)
Alleles , Carcinoma, Hepatocellular/etiology , Hepatitis B virus , Hepatitis B/complications , Hepatitis B/genetics , Leukocytes , Liver Neoplasms/etiology , Telomere/genetics , Adult , Aged , Case-Control Studies , Female , Gene Frequency , Genetic Variation , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk , Risk Factors
18.
Oncotarget ; 5(15): 6178-90, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25115392

ABSTRACT

FBXO31, a subunit of the SCF ubiquitin ligase, played a crucial role in neuronal development, DNA damage response and tumorigenesis. Here, we investigated the expression and prognosis value of FBXO31 in human primary gastric cancer (GC) samples. Meanwhile, the biological role and the regulation mechanism of FBXO31 were evaluated. We found that FBXO31 mRNA and protein was decreased dramatically in the GC tissue compared with the adjacent non-cancerous tissues. FBXO31 expression was significantly associated with tumor size, tumor infiltration, clinical grade and patients' prognosis. FBXO31 overexpression significantly decreased colony formation and induced a G1-phase arrest and inhibited the expression of CyclinD1 protein in GC cells. Further evidence was obtained from knockdown of FBXO31. Ectopic expression of FBXO31 dramatically inhibited xenograft tumor growth in nude mice. miR-20a and miR-17 mimics inhibited, whereas the inhibitor of miR-20a and miR-17 increased, the expression of FBXO31, respectively. miR-20a and miR-17 directly bind to the 3'-UTR of FBXO31. The level of miR-20a and miR-17 in GC tissue was significantly higher than that in surrounding normal mucosa. Moreover, a highly significant negative correlation between miR-20a (miR-17) and FBXO31 was observed in these GC samples. Therefore, effective therapy targeting the miR-20a (miR-17)-FBXO31-CyclinD1 pathway may help control GC progression.


Subject(s)
F-Box Proteins/biosynthesis , MicroRNAs/metabolism , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/biosynthesis , Animals , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Down-Regulation , F-Box Proteins/genetics , Female , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
19.
J Cell Mol Med ; 18(4): 610-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24447545

ABSTRACT

Runt-related transcription factor 3 (RUNX3) is a putative tumour suppressor via regulating the expression of a series of target genes. Clinical studies demonstrated that loss of RUNX3 expression is associated with gastric cancer progression and poor prognosis, but the underlying mechanism is not entirely clear. Accumulating evidence shows that the epithelial-mesenchymal transition (EMT) plays an important role in cancer relapse and metastasis. Therefore, we addressed whether RUNX3 has a role in the EMT in gastric cancer. Knockdown of RUNX3 promoted cell invasion and increased the protein expression of the mesenchymal marker vimentin in human gastric cancer cells. Overexpression of RUNX3 suppressed cell invasion and decreased the protein expression of vimentin in the cells and inhibited gastric cancer cells colonization in nude mice. Furthermore, overexpression of RUNX3 increased the expression of microRNA-30a (miR-30a), and miR-30a directly targeted the 3' untranslated region of vimentin and decreased its protein level. miR-30a inhibitor abrogated RUNX3-mediated inhibition of cell invasion and downregulation of vimentin. Thus, RUNX3 suppressed gastric cancer cell invasion and vimentin expression by activating miR-30a. In gastric cancer patients, levels of RUNX3 were positively correlated with miR-30a and negatively associated with the levels of vimentin. Collectively, our data suggest a novel molecular mechanism for the tumour suppressor activity of RUNX3. Effective therapy targeting the RUNX3 pathway may help control gastric cancer cell invasion and metastasis by inhibiting the EMT.


Subject(s)
Core Binding Factor Alpha 3 Subunit/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Vimentin/biosynthesis , Animals , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...