Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Sheng Li Xue Bao ; 74(4): 505-512, 2022 Aug 25.
Article in Zh | MEDLINE | ID: mdl-35993201

ABSTRACT

The purpose of the present study was to investigate the effect of glutamate scavenger oxaloacetate (OA) combined with CGS21680, an adenosine A2A receptor (A2AR) agonist, on acute traumatic brain injury (TBI), and to elucidate the underlying mechanisms. C57BL/6J mice were subjected to moderate-level TBI by controlled cortical impact, and then were treated with OA, CGS21680, or OA combined with CGS21680 at acute stage of TBI. At 24 h post TBI, neurological severity score, brain water content, glutamate concentration in cerebrospinal fluid (CSF), mRNA and protein levels of IL-1ß and TNF-α, mRNA level and activity of glutamate oxaloacetate aminotransferase (GOT), and ATP level of brain tissue were detected. The results showed that neurological deficit, brain water content, glutamate concentration in CSF, and the inflammatory cytokine IL-1ß and TNF-α production were exacerbated in CGS21680 treated mice. Administrating OA suppressed the rise of both glutamate concentration in CSF and brain water content, and elevated the ATP level of cerebral tissue. More interestingly, neurological deficit, brain edema, glutamate concentration, IL-1ß and TNF-α levels were ameliorated significantly in mice treated with OA combined with CGS21680. The combined treatment exhibited better therapeutic effects than single OA treatment. We also observed that GOT activity was enhanced in single CGS21680 treatment group, and both the GOT mRNA level and GOT activity were up-regulated in early-stage combined treatment group. These results suggest that A2AR can improve the efficiency of GOT and potentiate the ability of OA to metabolize glutamate. This may be the mechanism that A2AR activation in combination group augmented the neuroprotective effect of OA rather than aggravated the brain damages. Taken together, the present study provides a new insight for the clinical treatment of TBI with A2AR agonists and OA.


Subject(s)
Adenosine A2 Receptor Agonists , Brain Injuries, Traumatic , Neuroprotective Agents , Oxaloacetic Acid , Receptor, Adenosine A2A , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Agonists/therapeutic use , Adenosine Triphosphate , Animals , Brain Injuries/drug therapy , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Glutamic Acid , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxaloacetic Acid/pharmacology , Oxaloacetic Acid/therapeutic use , RNA, Messenger , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Tumor Necrosis Factor-alpha/genetics , Water
2.
J Neuroinflammation ; 18(1): 241, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34666797

ABSTRACT

BACKGROUND: Cognitive impairment in the late stage of traumatic brain injury (TBI) is associated with the NOD-, LRR and pyrin domain-containing protein 3 (NLRP3) inflammasome, which plays an important role in neuroinflammation. Although classical inflammatory pathways have been well-documented in the late stage of TBI (4-8 weeks post-injury), the mechanism by which the NLRP3 inflammasome impairs cognition is still unclear. METHODS: Mice lacking the gene encoding for NLRP3 (NLRP3-knockout mice) and their wild-type littermates were used in a controlled cortical impact model of TBI. Levels of NLRP3 inflammasome and inflammatory factors such as IL-1ß and HMGB1 were detected in post-injury hippocampal tissue, as well as long-term potentiation. Behaviors were assessed by T-maze test, novel object recognition, and nesting tests. Glycyrrhizin was used to antagonize HMGB1. Calcium imaging were performed on primary neuronal cultures. RESULTS: By using the NLRP3-knockout TBI model, we found that the continuous activation of the NLRP3 inflammasome and high mobility group box 1 (HMGB1) release were closely related to cognitive impairment. We also found that inhibition of HMGB1 improved LTP reduction and cognitive function by increasing the phosphorylation level of the NMDAR1 subunit at serine 896 while reducing NLRP3 inflammasome activation. CONCLUSION: NLRP3 inflammasome damages memory in the late stage of TBI primarily through HMGB1 upregulation and provides an explanation for the long-term progression of cognitive dysfunction.


Subject(s)
Brain Injuries, Traumatic/metabolism , Cognitive Dysfunction/metabolism , HMGB1 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Brain Injuries, Traumatic/pathology , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Coculture Techniques , Cognitive Dysfunction/pathology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques
3.
Tumour Biol ; 36(5): 3903-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25577248

ABSTRACT

Cystatin SN (cystatin 1, CST1) is a member of the cystatin superfamily that inhibits the proteolytic activity of cysteine proteases. CST1 is a tumor biomarker that provides useful information for the diagnosis of esophageal, gastric, and colorectal carcinomas. However, the significance of CST1 in pancreatic cancer is unknown. The aim of this study was to assess whether CST1 is a potential biomarker for early diagnosis of malignant pancreatic neoplasms. Microarray analysis of mRNA extracted from pancreatic cancer and pancreatic normal tissues was performed. Bioinformatics revealed that CST1 was one of the highest expressed genes on the array in pancreatic cancer, compared with normal tissue. In addition, the upregulation of CST1 in pancreatic cancer and several pancreatic cancer cell lines was confirmed using real-time PCR (RT-PCR), immunohistochemistry, and Western blotting. Next, CST1 knockdown using siRNA reduced the expression of the proliferation-related proteins p-AKT and PCNA significantly, as well as colony formation and xenograft development in vitro. Consistent with this, CST1 mRNA overexpression was correlated closely with malignancy-associated proteins such as PCNA, cyclin D1, cyclin A2, and cyclin E in pancreatic cancer cell lines. In conclusion, our data suggest that CST1 might contribute to the proliferation of pancreatic cancer cells and could be a potential biomarker for the early detection of pancreatic cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinogenesis , Early Detection of Cancer , Pancreatic Neoplasms/genetics , Salivary Cystatins/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Microarray Analysis , Middle Aged , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , RNA, Messenger/biosynthesis , Salivary Cystatins/genetics , Xenograft Model Antitumor Assays
4.
J Trace Elem Med Biol ; 74: 127061, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35987182

ABSTRACT

BACKGROUND: Numerous studies have investigated the relationship between serum copper/zinc ratio and lung cancer. However, the results are inconsistent. Therefore, we evaluated the association between copper/zinc ratio and lung cancer. MATERIALS AND METHODS: Observational studies reporting serum copper/zinc ratio in lung cancer patients and controls were identified from PubMed, Web of Science, EMBASE, CNKI and Wanfang databases online before December 2021. Summary standard mean difference (SMD) and the corresponding 95 % confidence interval (95 % CI) were applied to compare the serum serum copper/zinc ratio between lung cancer patients and controls using a random-effects model. RESULTS: Thirty-nine articles including 3598 lung cancer patients, 1402 benign lung diseases, and 3314 healthy controls were included in this study. The pooled results showed that the lung cancer patients had significantly higher serum copper/zinc ratio than healthy controls [SMD (95 % CI): 1.62 (1.31, 1.93)] and patients with benign lung diseases [SMD (95 % CI): 0.60 (0.36, 0.84)]. The results were robust according to sensitivity analysis. Meanwhile, consistent results were obtained both in European populations and Asian populations. Moreover, serum copper/zinc ratio was significant higher in patients with advanced stage of lung cancer than that in patients with early stage of lung cancer. CONCLUSION: The results showed that elevated serum copper/zinc ratio might be associated with increased risk of lung cancer.


Subject(s)
Lung Neoplasms , Zinc , Asian People , Copper , Humans
5.
Front Public Health ; 10: 999095, 2022.
Article in English | MEDLINE | ID: mdl-36203661

ABSTRACT

Fluorescent lamp manufacturing workers have been extensively exposed to mercury (Hg). Our aim was to assess their health risks using several approved occupational health risk assessment methods, and to find out which method was more suitable for identification of occupational health risks. Work locations, and air and urine samples were collected from 530 exposed workers in Zhejiang, China. Based on the calculated exposure doses, health risks and risk ratios (RRs) as health risk indices, were evaluated using: Environmental Protection Agency (EPA), Australian, Romanian, Singaporean, International Council on Mining and Metals (ICMM), and Control of Substances Hazardous to Health (COSHH) methods. Among the workers, 86.0% had higher Hg levels than the Chinese occupational exposure limits of 0.02 mg/m3, and 16.7% urine samples were higher than the biological exposure limits of 35.0 µg/g·creatinine. Among workers at the injection, etc. locations, their average RRs, evaluated by the EPA, COSHH and Singaporean methods were 0.97, 0.76, and 0.60, respectively, and were significantly higher than the ICMM (0.39), Australian (0.30) and Romanian (0.29) methods. The RRs from the Singaporean method showed significant correlations with the urinary Hg levels (P < 0.01). In conclusion, the Singaporean method was more appropriate than the others for health risk evaluation because the excessive risks were significantly associated with urinary Hg levels among the workers.


Subject(s)
Mercury , Occupational Exposure , Occupational Health , Australia/epidemiology , Creatinine , Humans , Mercury/urine , Occupational Exposure/adverse effects , Occupational Exposure/analysis
6.
Neurosci Lett ; 769: 136431, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34974110

ABSTRACT

NLRP3 inflammasome plays a crucial role in the innate immune system. Our group previously reported that the microglial adenosine 2A receptor (A2AR) regulates canonical neuroinflammation, which is affected by the glutamate concentration. However, the regulatory effect of A2AR on NLRP3 inflammasome and the effects of glutamate concentration remain unknown. Therefore, we aimed to investigate the regulatory effect of microglial A2AR on NLRP3 inflammasome assembly and activation as well as the effects of glutamate concentration on the inflammasome assembly and activation. Experiments were conducted on magnetically sorted primary microglia from P14 mice. The results showed that pharmacological A2AR activation ameliorated NLRP3 activation under no or low glutamate concentrations, but this effect was reversed by high glutamate concentrations. Moreover, the mRNA levels of NLRP3 inflammasome-related genes were not affected by A2AR activation or the glutamate concentration. We further demonstrated that A2AR activation inhibited the interaction between NLRP3 and caspase 1 under no or low glutamate concentrations while promoting their interaction under high glutamate concentrations. The oligomerization of ASC also showed a similar trend. In conclusion, our findings proved that the high glutamate concentration could reverse the inhibition of A2AR on NLRP3 inflammasome activation by modulating its assembly, which provides new insights into the regulatory effect of A2AR on neuroinflammation under different pathological conditions.


Subject(s)
Glutamic Acid/metabolism , Inflammasomes/metabolism , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Cells, Cultured , Glutamic Acid/pharmacology , Mice , Microglia/drug effects , Protein Multimerization
7.
Neurochem Int ; 149: 105145, 2021 10.
Article in English | MEDLINE | ID: mdl-34324942

ABSTRACT

The heteromeric complexes of adenosine 2A receptor (A2AR) and N-methyl-D-aspartate receptor (NMDAR) have recently been confirmed in cell experiments, while its in situ detection at the subcellular level of brain tissue has not yet been achieved. Proximity Ligation Assay (PLA) enables the detection of low-abundance proteins and their interactions at the cellular level with high specificity and sensitivity, while Transmission electron microscope (TEM) is an excellent tool for observing subcellular structures. To develop a highly efficient and reproducible technique for in situ detection of protein interactions at subcellular levels, in this study, we modified the standard PLA sample preparation method to make the samples suitable for analysis by transmission electron microscopy. Using this technique, we successfully detected the heteromers of A2AR and NMDAR1, the essential subunit of NMDA receptor on the hippocampal synaptic structure in mice. Our results show that the distribution of this heteromer is different in different hippocampal subregions. This technique holds the potential for being a reliable method to detect protein interactions at the subcellular level and unravel their unknown functions.


Subject(s)
Hippocampus/ultrastructure , Microscopy, Electron, Transmission/methods , Receptor, Adenosine A2A/ultrastructure , Receptors, N-Methyl-D-Aspartate/ultrastructure , Synapses/ultrastructure , Animals , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding/physiology , Receptor, Adenosine A2A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
8.
Cell Prolif ; 54(2): e12971, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33349993

ABSTRACT

OBJECTIVES: The present study clarified the role and signalling pathway of Ski in regulating proliferation and apoptosis in fibroblasts under high-glucose (HG) conditions. MATERIALS AND METHODS: The proliferation and apoptosis of rat primary fibroblasts were assessed using EdU incorporation and TUNEL assays. The protein and phosphorylation levels of the corresponding factors were measured using immunofluorescence staining and Western blotting. Immunoprecipitation was used to determine the interactions between Ski and FoxO1 or Ski and HDAC1. The Ski protein was overexpressed via recombinant adenovirus transfection, and FoxO1 and HDAC1 were knocked down using targeted small-interfering RNA. RESULTS: The present study found that HG inhibited fibroblast proliferation, increased apoptosis and reduced Ski levels in rat primary fibroblasts. Conversely, increasing Ski protein levels alleviated HG-induced proliferation inhibition and apoptosis promotion. Increasing Ski protein levels also increased Ski binding to FoxO1 to decrease FoxO1 acetylation, and interfering with FoxO1 caused loss of the regulatory effect of Ski in fibroblasts under HG. Increasing Ski protein levels decreased FoxO1 acetylation via HDAC1-mediated deacetylation. CONCLUSIONS: Therefore, these findings confirmed for the first time that Ski regulated fibroblast proliferation and apoptosis under HG conditions via the FoxO1 pathway.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Glucose/pharmacology , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Acetylation/drug effects , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Male , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Phosphorylation/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Smad2 Protein , Smad3 Protein/metabolism
9.
Ann Palliat Med ; 10(7): 7173-7183, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34154359

ABSTRACT

BACKGROUND: The pathogenesis of gastric cardiac polyps is not yet clear, and there is little research on their clinical and histopathologic characteristics and correlation with gastroesophageal reflux. Early detection and treatment of premalignant lesions in this area can prevent the development of cancer. We aimed to evaluate the clinical and histopathologic characteristics and risk factors of gastric cardiac polyps to improve the current understanding of the disease. METHODS: Patients diagnosed with gastric cardiac polyps at the Third Affiliated Hospital of Sun Yat-Sen University between January 1, 2010, and December 31, 2019, were sought for the study. The exclusion criteria were missing clinical data, insufficient pathological reports, gastric malignancy, or a previous history of gastroduodenal surgery. Ultimately, 140 patients were included in the case group, while 140 patients diagnosed with chronic superficial gastritis from the same 10-year period were identified randomly and selected as a control group. The exclusion criteria for this group were the same as those for the case group. Patients in both groups were matched in age and gender to ensure comparability between the two groups. We evaluated and compared the demographic and clinical data and endoscopic impressions of each group and analyzed the endoscopic, histologic features of gastric cardiac polyps. RESULTS: Gastroesophageal reflux was significantly associated with a higher risk of gastric cardiac polyps after adjustment for other covariates [adjusted odds ratio (OR) =2.809; 95% confidence interval (CI): 1.178-6.697; P=0.020]. Most gastric polyps were single (97.9%), sessile (92.8%), and small polyps with a diameter less than 1 cm (88.6%). Most were located in the gastroesophageal junction region (65.0%) with a smooth surface (56.4%) or surface congestion (30.0%). Hyperplastic (inflammatory) polyps (88.0%) were the most common pathological type and comprised gastric-type foveolar epithelium, squamous epithelium, or admixture of the two epithelia, with a minority showing intestinal metaplasia, mild, or moderate epithelial dysplasia. CONCLUSIONS: Gastroesophageal reflux was associated with a significantly higher incidence of gastric cardiac polyps with a 2.8-fold increased risk. Most gastric cardiac polyps were found to be benign lesions and had a favorable prognosis in the clinic despite their malignant potential.


Subject(s)
Gastroesophageal Reflux , Stomach Neoplasms , Case-Control Studies , Esophagogastric Junction , Gastroesophageal Reflux/complications , Humans , Metaplasia , Stomach Neoplasms/etiology
10.
Gastroenterol Rep (Oxf) ; 9(2): 154-165, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34026223

ABSTRACT

BACKGROUND: Butyrate acts as a regulator in multiple inflammatory organ injuries. However, the role of butyrate in acute liver injury has not yet been fully explored. In the present study, we aimed to investigate the association between butyrate and lipopolysaccharide (LPS)-induced acute liver injury and the signaling pathways involved. METHODS: LPS-induced acute liver injury was induced by intraperitoneal injection of LPS (5 mg/kg) in G-protein-coupled receptor 43 (GPR43)-knockout (KO) and wild-type female C57BL/6 mice. Sodium butyrate (500mg/kg) was administered intraperitoneally 30 min prior to LPS exposure. Liver injury was detected by serum markers, tissue morphology, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). Pro-inflammatory-factor levels were detected by enzyme-linked immunosorbent assay and real-time polymerase chain reaction (RT-PCR). Cell models were first treated with sodium butyrate (4 µmol/mL), followed by LPS (1 µg/mL) half an hour later in GPR43 small interfering RNA (siRNA)-transfected or control RAW264.7 cells. Cell-inflammation status was evaluated through detecting pro-inflammatory-factor expression by RT-PCR and also through checking toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB)-element levels including TLR4, TRAF6, IKKß, IкBα, phospho-IкBα, p65, and phospho-p65 by Western blot. The interaction between GPR43 and ß-arrestin-2 was tested by co-immunoprecipitation. RESULTS: Sodium butyrate reversed the LPS-induced tissue-morphology changes and high levels of serum alanine aminotransferase, aspartate transaminase, myeloperoxidase, TUNEL, and pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-6. The ameliorating effect of sodium butyrate was weakened in GPR43-KO mice and GPR43 siRNA RAW264.7 cells, compared with those of GPR43-positive controls. Sodium butyrate downregulated some elements of the TLR4/NF-κB pathway, including phospho-IκBα and phospho-p65, in RAW264.7 cells. Increased interactions between GPR43 and ß-arrestin-2, and between ß-arrestin-2 and IкBα were observed. CONCLUSION: Sodium butyrate significantly attenuated LPS-induced liver injury by reducing the inflammatory response partially via the GPR43/ß-arrestin-2/NF-κB signaling pathway.

11.
Front Aging Neurosci ; 13: 721474, 2021.
Article in English | MEDLINE | ID: mdl-34539383

ABSTRACT

Tau hyperphosphorylation is a characteristic alteration present in a range of neurological conditions, such as traumatic brain injury (TBI) and neurodegenerative diseases. Treatments targeting high-mobility group box protein 1 (HMGB1) induce neuroprotective effects in these neuropathologic conditions. However, little is known about the interactions between hyperphosphorylated tau and HMGB1 in neuroinflammation. We established a model of TBI with controlled cortical impacts (CCIs) and a tau hyperphosphorylation model by injecting the virus encoding human P301S tau in mice, and immunofluorescence, western blotting analysis, and behavioral tests were performed to clarify the interaction between phosphorylated tau (p-tau) and HMGB1 levels. We demonstrated that p-tau and HMGB1 were elevated in the spatial memory-related brain regions in mice with TBI and tau-overexpression. Animals with tau-overexpression also had significantly increased nucleotide-binding oligomerization domain-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome activation, which manifested as increases in apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), activating caspase-1 and interleukin 1 beta (IL-1ß) levels. In addition, NLRP3-/- mice and the HMGB1 inhibitor, glycyrrhizin, were used to explore therapeutic strategies for diseases with p-tau overexpression. Compared with wild-type (WT) mice with tau-overexpression, downregulation of p-tau and HMGB1 was observed in NLRP3-/- mice, indicating that HMGB1 alterations were NLRP3-dependent. Moreover, treatment with glycyrrhizin at a late stage markedly reduced p-tau levels and improved performance in the Y- and T-mazes and the ability of tau-overexpressing mice to build nests, which revealed improvements in spatial memory and advanced hippocampal function. The findings identified that p-tau has a triggering role in the modulation of neuroinflammation and spatial memory in an NLRP3-dependent manner, and suggest that treatment with HMGB1 inhibitors may be a better therapeutic strategy for tauopathies.

12.
World J Clin Cases ; 8(6): 1116-1128, 2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32258082

ABSTRACT

BACKGROUND: Hepatoid carcinoma (HC) is an extremely rare neoplasm that is morphologically similar to hepatocellular carcinoma. HC has been described in various organs; however, HC of the pancreas is extremely rare. To our knowledge, only 38 cases have been reported. We present a case of HC of the pancreas in a 36-year-old male patient. CASE SUMMARY: A 36-year-old cachexic man with no significant past medical history was transferred to our hospital with a history of painless jaundice, elevated blood glucose and significant weight loss. Lab tests showed elevated serum transaminases, bilirubin and alpha-fetoprotein levels. Magnetic resonance imaging of the upper abdomen showed a diffusely enlarged pancreas, appearing "sausage-shaped". Magnetic resonance cholangiopancreatography showed upstream ductal dilation secondary to stricture of the main pancreatic duct and the common bile duct, which were not visible. Immunohistochemistry of biopsied tissue from a percutaneous pancreatic biopsy showed tumor cell positivity for HepPar1, polyclonal carcinoembryonic antigen and CK19, suggestive of HC of the pancreas. The characteristics of 39 patients with HC of the pancreas were reviewed. CONCLUSION: HC of the pancreas is more prevalent in males, and patients have a median age of 57 years. It is most commonly asymptomatic or presents as abdominal back pain, and the pancreatic tail is the most common location. At the time of diagnosis, liver metastasis is often present.

13.
Gastroenterol Rep (Oxf) ; 7(6): 434-443, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31857905

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) have been applied as biomarkers in many diseases. However, scarce biomarkers are available in single lncRNA differential expression associated with different clinical stages of liver cirrhosis (LC). The aim of the study is to identify some lncRNAs that can serve as non-invasive sensitive biomarkers for early diagnosis and grade of LC. METHODS: Blood lncRNA expression was evaluated in three independent cohorts with 305 participants including healthy controls, hepatitis B virus (HBV) carriers, and patients with chronic hepatitis B (CHB) or LC. First, candidate lncRNAs were screened by CapitalBiotech microarray to diagnose cirrhosis. Quantitative reverse-transcriptase polymerase chain reaction was then used to investigate the expression of selected lncRNAs in the whole group of cirrhosis and different Child-Pugh classes. Ultimately, the diagnostic accuracy of the promising biomarker was examined and validated via Mann-Whitney test and receiver-operating characteristics analysis. RESULTS: Lnc-TCL6 was identified as a sensitive biomarker for early diagnosis of LC (Child-Pugh A) compared with healthy controls (area under the ROC curve [AUC] = 0.636), HBV carriers (AUC = 0.671), and CHB patients (AUC = 0.672). Furthermore, lnc-TCL6 showed a favourable capacity in discriminating among different Child-Pugh classes (AUC: 0.711-0.837). Compared with healthy controls, HBV carriers, and CHB patients, the expression of lnc-TCL6 was obviously up-regulated in Child-Pugh A patients and, conversely, significantly down-regulated in Child-Pugh C patients. CONCLUSIONS: Lnc-TCL6 is a novel potential biomarker for early diagnosis of LC and is a possible predictor of disease progression.

14.
Iran J Immunol ; 15(2): 122-132, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29947341

ABSTRACT

BACKGROUND: Hemin is an important sterile component that induces a neuroinflammatory response after intracerebral hemorrhage, in which NLRP3 inflammasome activation has also proved to be involved. Although microglial activation acts as a key contributor in the neuroinflammatory response, the relationship between hemin and NLRP3 in microglia remains poorly understood. OBJECTIVE: To investigate whether or not hemin regulates microglia-mediated secondary injury through activating the NLRP3/caspase-1 signaling pathway in microglia. METHODS: In this study, N9 microglial cells were treated with hemin, and subsequently used to detect the production of caspase-1 p10 and NLRP3 inflammasome assembly. An ELISA was subsequently performed to measure the secretion of IL-1ß. RESULTS: It was found that the production of activated caspase-1 was dose- and time-dependent with regards to hemin. Moreover, hemin was observed to be capable of inducing the assembly of the NLRP3 inflammasome without any increase in IL-1ß. Similarly, the supernatant of hemin-treated primary microglial cells did not increase in IL-1ß secretion. Furthermore, hemin-induced NLRP3 inflammasome activation did not significantly affect pyroptosis. CONCLUSION: Hemin is a potential sterile danger signal molecule that can induce inflammasome activation without directly mediating inflammation damage on microglia.


Subject(s)
Hemin/metabolism , Inflammasomes/metabolism , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Apoptosis , Biomarkers , Caspase 1/metabolism , Cell Line , Cells, Cultured , Interleukin-1beta/biosynthesis , Mice , Protein Binding , Signal Transduction
15.
Int J Oncol ; 53(2): 781-790, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29845224

ABSTRACT

Cystatin SN (cystatin 1, CST1) is a member of the cystatin superfamily which inhibits the proteolytic activity of cysteine proteases. CST1 is a tumor biomarker that provides useful information for the diagnosis of esophageal, gastric and colorectal carcinomas. MicroRNAs (miRNAs or miRs) play an important role in tumor cell proliferation. However, the exact role of let­7d and CST1 in colon cancer remains unknown. The aim of this study was to assess whether let­7d inhibits colorectal carcinogenesis through the CST1/p65 pathway, and determine whether it may be used as a potential target for clinical therapy. Microarray analysis of mRNAs extracted from colon cancer and normal tissues was performed. The results of gene expression microanalysis revealed that CST1 expression was upregulated in colon cancer compared with normal tissues. In addition, the upregulation of CST1 expression and the downregulation of let­7d expression in patients with colon cancer and in several colorectal cancer cell lines were confirmed by reverse transcription-quantitative PCR (RT­qPCR), immunohistochemistry and western blot analysis. In addition, siRNA targeting CST1 (CST1­siRNA) and let­7d-mimics were used in the HCT116 cells, and the results revealed that CST1 and let­7d played a role in colorectal cancer cell proliferation. Let­7d inhibited colorectal carcinogenesis through the CST1/p65 pathway. Thus, the findings of the present study indicate that CST1 may be a potential target for the future clinical therapy of colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Profiling/methods , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis/methods , Salivary Cystatins/genetics , Signal Transduction , Aged , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Male , Middle Aged , Salivary Cystatins/metabolism , Transcription Factor RelA/genetics , Up-Regulation
16.
J Dig Dis ; 15(8): 425-34, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24814616

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the potential mechanism and signaling pathway involved in chemotherapy-induced intestinal mucosal injury (CIMI), which is a common physiopathological problem in patients with cancer. METHODS: For the in vivo experiment, mice received intraperitoneal injection of 5-fluorouracil (5-FU) at a dose of 75 mg/kg/day for 1, 3 or 5 days. Villus height and crypt depth of the small intestine, cell apoptosis and proliferation were then examined to determine the extent of CIMI. The expressions of Akt, p53, PUMA and p21 were evaluated both in vivo in mice models and in vitro in the IEC-6 and HCT116 cell lines. RESULTS: After 5-FU therapy both the intestinal villus height (275.93 µm vs 164.52 µm, P < 0.001) and crypt depth (64.13 µm vs 42.48 µm, P < 0.001) were decreased. The apoptotic index was greatly increased from 0.32% to 15.84% (P < 0.001) and proliferation was suppressed (63.58% vs 39.15%, P < 0.001). Additionally, p53 expression was significantly increased in the intestinal crypt along with the expressions of PUMA and p21. Western blot showed that the administration of 5-FU induced p53/PUMA-mediated apoptosis and upregulated p21 expression to suppress cell proliferation. CONCLUSION: Chemotherapy might mediate intestinal injury via p53/PUMA-mediated apoptotic signaling and the suppression of proliferation in response to p21.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Apoptosis Regulatory Proteins/biosynthesis , Fluorouracil/toxicity , Mucositis/chemically induced , Tumor Suppressor Proteins/biosynthesis , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Fluorouracil/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Mucositis/metabolism , Mucositis/pathology , Proto-Oncogene Proteins/biosynthesis , Signal Transduction/drug effects , Tumor Cells, Cultured
17.
World J Gastroenterol ; 19(30): 4925-34, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23946597

ABSTRACT

AIM: To investigate whether tumor necrosis factor-α (TNF-α) mediates ischemia-reperfusion (I/R)-induced intestinal mucosal injury through c-Jun N-terminal kinase (JNK) activation. METHODS: In this study, intestinal I/R was induced by 60-min occlusion of the superior mesenteric artery in rats followed by 60-min reperfusion, and the rats were pretreated with a TNF-α inhibitor, pentoxifylline, or the TNF-α antibody infliximab. After surgery, part of the intestine was collected for histological analysis. The mucosal layer was harvested for RNA and protein extraction, which were used for further real-time polymerase chain reaction, enzyme-linked immunosorbent assay and Western blotting analyses. The TNF-α expression, intestinal mucosal injury, cell apoptosis, activation of apoptotic protein and JNK signaling pathway were analyzed. RESULTS: I/R significantly enhanced expression of mucosal TNF-α at both the mRNA and protein levels, induced severe mucosal injury and cell apoptosis, activated caspase-9/caspase-3, and activated the JNK signaling pathway. Pretreatment with pentoxifylline markedly downregulated TNF-α at both the mRNA and protein levels, whereas infliximab pretreatment did not affect the expression of TNF-α induced by I/R. However, pretreatment with pentoxifylline or infliximab dramatically suppressed I/R-induced mucosal injury and cell apoptosis and significantly inhibited the activation of caspase-9/3 and JNK signaling. CONCLUSION: The results indicate there was a TNF-α-mediated JNK activation response to intestinal I/R injury.


Subject(s)
Ileum/immunology , Intestinal Mucosa/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Jejunum/immunology , Reperfusion Injury/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Disease Models, Animal , Enzyme Activation , Ileum/drug effects , Ileum/enzymology , Ileum/pathology , Infliximab , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Jejunum/drug effects , Jejunum/enzymology , Jejunum/pathology , Ligation , Male , Mesenteric Arteries/surgery , Mesenteric Vascular Occlusion/complications , Pentoxifylline/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/etiology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL