Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38920294

ABSTRACT

BACKGROUND AND OBJECTIVES: The primary objective of this study was to evaluate admission serum Anion Gap (AG) as a predictor of all-cause mortality in critically ill patients with cirrhosis. MATERIALS AND METHODS: A total of 3,084 cirrhotic patients were included and randomly divided into training and validation cohorts (n = 2,159 and 925, respectively). Patients were categorized into high and normal AG groups based on their AG values. Cox regression and Kaplan-Meier survival analysis were used to assess the relationships between AG levels and outcomes. RESULTS: Both cohorts showed strong parameter similarity (P > 0.05). High AG were associated with significantly lower survival probabilities. Cox models confirmed elevated AG as a risk factor, even after adjusting for covariates (HR: 1.920, 1.793, and 1.764 for 30-day, 60-day, and hospital mortality, respectively). Subgroup analyses, especially regarding chronic kidney disease, revealed complex interactions. Serum AG displayed predictive power comparable to established scoring systems. CONCLUSION: Elevated AG at admission is a valuable predictor of poor outcomes and increased mortality risk in critically ill cirrhotic patients. Serum AG can serve as an easily accessible tool for risk assessment and prognosis evaluation in this population.

2.
Med Oncol ; 40(11): 319, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37796396

ABSTRACT

Gastric cancer represents a significant global health concern, necessitating the exploration of novel therapeutic options. Diosmetin, a natural flavonoid derived from citrus and vegetables, has demonstrated promising anti-tumor activity against various tumor cells. However, the potential anticancer effect of diosmetin in gastric cancer and its underlying mechanism have yet to be elucidated. In this study, we aimed to investigate the impact of diosmetin on cell proliferation, migration, cell cycle progression and apoptosis in human gastric cancer HGC-27 cells. Our findings revealed that diosmetin effectively suppressed cell proliferation, induced G2/M phase cell cycle arrest, and triggered cell apoptosis. Mechanistically, diosmetin downregulated the expression of antiapoptotic proteins Bcl-2 and Bcl-xL, while upregulated the level of proapoptotic proteins such as Bax, cleaved PARP and cleaved caspase-3. Additionally, diosmetin inhibited Akt and FoxO1 phosphorylation, while activated the MAPK signaling pathway. Notably, pretreatment of IGF-1, an Akt activator, attenuated the diosmetin-induced apoptosis. Furthermore, pretreatment with SP600125, a JNK inhibitor, significantly reduced the protein level of LC3B, while promoted the expression of cleaved caspase-3 and cleaved PARP. Collectively, our results suggest that diosmetin holds promise as an effective therapeutic agent against gastric cancer by inducing apoptosis through inhibition of the Akt/FoxO1 pathway and promoting protective autophagy via the MAPK/JNK signaling pathway.


Subject(s)
Stomach Neoplasms , Humans , Apoptosis , Autophagy , Caspase 3 , Flavonoids/pharmacology , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases , Poly(ADP-ribose) Polymerase Inhibitors , Proto-Oncogene Proteins c-akt , Stomach Neoplasms/drug therapy , Cell Line, Tumor
3.
J Alzheimers Dis ; 90(3): 1153-1162, 2022.
Article in English | MEDLINE | ID: mdl-36214002

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) is important in the etiology of Alzheimer's disease (AD). Removal of Aß from the brain is a major strategy for the prevention and treatment of AD. OBJECTIVE: To clarify whether Aß42 can be cleared by intestinal excretion and whether the gut microbiota (GM) can affect the excretory clearance of Aß42 in the peripheral blood and intestines. METHODS: Male 8-month-old C57BL6 mice were maintained on either normal chow or received broad-spectrum antibiotics in their drinking water for one week. Sterile saline, fluorescein isothiocyanate (FITC), or FITC-Aß42 (fluorescein isothiocyanate-labeled amyloid-ß42 peptides) was injected 1 h before sampling. Related changes of Aß42 before and after injection were evaluated. RESULTS: FITC-Aß42 was injected into mice through the tail vein and could later be detected in feces. Furthermore, the fecal concentrations of FITC-Aß42 were higher in mice that had been fed antibiotics to alter their GM than in normal mice. However, the FITC-Aß42 concentrations in blood showed the opposite pattern. CONCLUSION: Aß42 can be excreted into the intestinal lumen and is regulated by the GM.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Animals , Mice , Male , Gastrointestinal Microbiome/physiology , Fluorescein-5-isothiocyanate , Mice, Transgenic , Mice, Inbred C57BL , Disease Models, Animal , Amyloid beta-Peptides/metabolism , Alzheimer Disease/therapy , Gastrointestinal Tract/metabolism , Brain/metabolism , Anti-Bacterial Agents
4.
Therap Adv Gastroenterol ; 14: 17562848211035670, 2021.
Article in English | MEDLINE | ID: mdl-34394726

ABSTRACT

BACKGROUND AND AIMS: Currently, there are no definitive therapies for coronavirus disease 2019 (COVID-19). Gut microbial dysbiosis has been proved to be associated with COVID-19 severity and probiotics is an adjunctive therapy for COIVD-19. However, the potential benefit of probiotics in COVID-19 has not been studied. We aimed to assess the relationship of probiotics use with clinical outcomes in patients with COVID-19. METHODS: We conducted a propensity-score matched retrospective cohort study of adult patients with COVID-19. Eligible patients received either probiotics plus standard care (probiotics group) or standard care alone (non-probiotics group). The primary outcome was the clinical improvement rate, which was compared among propensity-score matched groups and in the unmatched cohort. Secondary outcomes included the duration of viral shedding, fever, and hospital stay. RESULTS: Among the propensity-score matched groups, probiotics use was related to clinical improvement rates (log-rank p = 0.028). This relationship was driven primarily by a shorter (days) time to clinical improvement [difference, -3 (-4 to -1), p = 0.022], reduction in duration of fever [-1.0 (-2.0 to 0.0), p = 0.025], viral shedding [-3 (-6 to -1), p < 0.001], and hospital stay [-3 (-5 to -1), p = 0.009]. Using the Cox model with time-varying exposure, use of probiotics remained independently related to better clinical improvement rate in the unmatched cohort. CONCLUSION: Our study suggested that probiotics use was related to improved clinical outcomes in patients with COVID-19. Further studies are required to validate the effect of probiotics in combating the COVID-19 pandemic.

5.
Oncol Lett ; 21(5): 407, 2021 May.
Article in English | MEDLINE | ID: mdl-33841568

ABSTRACT

Increased microRNA (miR)-32 expression in colorectal cancer (CRC) tissues enhances CRC cell proliferation, migration, invasion and attenuates CRC cell apoptosis by repressing the expression of phosphatase and tensin homolog (PTEN). Forkhead box K1 (FOXK1) was identified as a potential interacting transcription factor using DNA pull-down assays and mass spectrometry. The present study aimed to elucidate the role of FOXK1 in regulating miR-32 expression in CRC. The expressions of FOXK1, miR-32, transmembrane protein 245 gene (TMEM245) and PTEN were compared between CRC and normal colonic tissues. Levels of miR-32, TMEM245, PTEN and the proliferation and apoptosis of CRC cells were studied using FOXK1-overexpression or knockdown, or by simultaneously interfering with FOXK1 and miR-32 expression. Direct FOXK1 binding to the miR-32 promoter was verified using chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays. The results showed elevated FOXK1, miR-32 and TMEM245 expression, and significantly decreased PTEN expression in CRC, compared with normal colonic tissues. Correlations between the expressions of TMEM245 and miR-32, FOXK1 and miR-32, and FOXK1 and TMEM245 were positive and significant. FOXK1-knockdown led to decreased miR-32 and TMEM245 expression and increased PTEN expression, whereas FOXK1-overexpression had the opposite effect. Overexpressed FOXK1 promoted the malignancy of CRC cells in vitro by stimulating proliferation and reducing apoptosis; whereas FOXK1-depletion suppressed such malignancy and a miR-32 inhibitor partially reversed the effects of FOXK1. The results of ChIP and dual-luciferase reporter assays indicated that FOXK1 directly binds to the promoter of TMEM245/miR-32. Thus, the FOXK1-miR-32-PTEN signaling axis may play a crucial role in the pathogenesis and development of CRC.

6.
Med Sci Monit ; 27: e928478, 2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33686049

ABSTRACT

BACKGROUND Bifidobacterium is a potentially effective and safe treatment for patients with inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease. However, information on the influence of B. bifidum on gut microbial diversity of treated and pretreated IBD patients is limited. MATERIAL AND METHODS Our study investigated therapeutic and preventive effects of B. bifidum ATCC 29521 on C57BL/6 mice with dextran sulfate sodium (DSS)-induced acute colitis via 16S ribosomal ribonucleic acid (rRNA) gene sequencing. RESULTS Treatment and pretreatment of mice with B. bifidum ATCC 29521 significantly alleviated the severity of acute colitis on the basis of clinical and pathologic indicators. 16S rRNA gene sequencing showed that administration of B. bifidum shifted composition of the gut microbiome in mice with DSS-induced colitis in both treated and pretreated groups. Mice pretreated with B. bifidum ATCC 29521 for 21 days exhibited a significant increase in diversity of the gut microbiome. Principal coordinate analysis showed that gut microbiota structure was shaped by different treatments and time points. On the basis of linear discriminant analysis of effect size, the abundance of the genus Escherichia-Shigella, belonging to the family Enterobacteriaceae, was reduced in the B. bifidum-treated group, indicating that pathogens were inhibited by the B. bifidum treatment. Furthermore, the genera Intestinimonas and Bacteroides were significantly associated with the B. bifidum-pretreated group. CONCLUSIONS 16S rRNA gene sequencing showed that pretreatment with B. bifidum ATCC 29521 reduced intestinal inflammation and altered the gut microbiota to favor the genera Intestinimonas and Bacteroides.


Subject(s)
Bifidobacterium bifidum/metabolism , Colitis/drug therapy , Gastrointestinal Microbiome/drug effects , Animals , Bacteria/genetics , Colitis/microbiology , Colitis, Ulcerative/genetics , Colon/pathology , Dextran Sulfate/adverse effects , Dextran Sulfate/pharmacology , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Inflammatory Bowel Diseases/pathology , Mice , Mice, Inbred C57BL , Probiotics/therapeutic use , RNA, Ribosomal, 16S/genetics
7.
Oncol Lett ; 20(3): 2411-2419, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32782558

ABSTRACT

Colorectal cancer (CRC) is a common human malignant tumor, and the fourth most common cause of cancer-associated mortality in China. However, the pathogenesis of CRC is not yet fully understood. The present study aimed to investigate the expression and clinical significance of microRNA (miR)-126 and insulin receptor substrate-1 (IRS-1), as well as the role of miR-126 in the prognosis of patients with CRC. A total of 86 colorectal tissue specimens, including 40 CRC and adjacent normal tissue, 26 colorectal adenoma tissue and 20 normal colorectal tissue samples, were collected for the present study. Reverse transcription-quantitative PCR analysis was performed to determine miR-126 and IRS-1 mRNA expression levels, while western blotting and immunohistochemistry (IHC) analyses were performed to determine IRS-1 protein expression levels. The correlation between miR-126 and IRS-1 expression, as well as the association between altered miR-126 and IRS-1 expression levels and clinicopathological characteristics, and the overall survival time of patients with CRC were assessed. The results demonstrated that miR-126 expression was significantly downregulated, while IRS-1 protein expression was upregulated in CRC tissues compared with that in adjacent normal tissues, colorectal adenoma tissues and normal colorectal tissues, respectively. IHC analysis exhibited strong positive staining of IRS-1 protein in CRC tissues, while absent or weak staining of IRS-1 protein was detected in adjacent normal tissues, colorectal adenoma tissues and normal colorectal tissues. miR-126 expression was inversely correlated with IRS-1 protein expression in CRC tissues (r=-0.420; P<0.05). Furthermore, downregulated miR-126 expression was associated with advanced clinicopathological characteristics of the disease and a shorter overall survival time in patients with CRC. Taken together, the results of the present study suggest that miR-126 downregulation may be a candidate molecular marker predictive of poor prognosis of patients with CRC.

8.
Biosci Rep ; 40(8)2020 08 28.
Article in English | MEDLINE | ID: mdl-32700743

ABSTRACT

Colon cancer is one of the major causes of cancer-related deaths worldwide. Long non-coding RNA (lncRNA) LINC01123 has been suggested to act as an oncogene in non-small cell lung cancer and a prognostic signature in head and neck squamous cell carcinoma. However, its role in colon cancer remains obscure. From TCGA database, LINC01123 was observed to be up-regulated in colon adenocarcinoma (COAD). Subsequently, the up-regulated LINC01123 was also detected in colon cancer cells. Functionally, LINC01123 could enhance cell proliferation, migration, invasion and angiogenesis. Moreover, the chemoresistance of colon cancer cells was verified to be promoted by LINC01123. Afterward, LINC01123 was found to bind with Ago2 and miR-34c-5p. Besides, miR-34c-5p was confirmed to inhibit the cellular process and chemoresistance of colon cancer cells. Then, VEGFA was disclosed to coexist with LINC01123 and miR-34c-5p in RNA-induced silencing complex. And TCGA database suggested that its expression was correlated with different stages of COAD. Moreover, it was uncovered that VEGFA could bind with miR-34c-5p and its expression positively correlated with LINC01123 expression. Finally, LINC01123 was proofed to regulate colon cancer progression and cells chemoresistance via VEGFA. In conclusion, LINC01123/miR-34c-5p/VEGFA axis promotes colon cancer malignancy and cells chemoresistance.


Subject(s)
Adenocarcinoma/metabolism , Cell Proliferation , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm , RNA, Long Noncoding/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Movement , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Databases, Factual , Gene Expression Regulation , HCT116 Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Neovascularization, Pathologic , RNA, Long Noncoding/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Exp Cell Res ; 395(1): 112170, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32682783

ABSTRACT

Colorectal cancer is the second leading cause of cancer mortality worldwide with poor prognosis and high recurrence. Aberrant Wnt/ß-catenin signaling promotes oncogenesis by transcriptional activation of c-Myc and its downstream signals. EDAR is characterized as an important effector of canonical Wnt signaling in developing skin appendages, but the interplay between EDAR and Wnt signaling in tumorigenesis and progression remains to be elucidated. In this study, we revealed that EDAR expression is prevalently elevated in colorectal cancer tissues compared with normal tissues. Further analysis suggests there is a strict correlation between EDAR expression and colorectal cancer progression. EDAR silencing by shRNA in colorectal cancer cells showed proliferative suppression via retarding cell cycle at G1 phase. Xenograft mice transplanted with shEDAR-transduced tumor cells significantly alleviated tumor burden in comparison with control mice. Furthermore, downregulation of EDAR was accompanied by reduction of ß-catenin, c-Myc and other G1 cell cycle regulators, while ß-catenin agonist restored the expression of these proteins and overrode the proliferative block induced by EDAR knockdown. These findings indicate that EDAR functions as a component of Wnt/ß-catenin signaling pathway, and is a potential modulator in colorectal carcinogenesis.


Subject(s)
Cell Proliferation/physiology , Colonic Neoplasms , Colorectal Neoplasms/pathology , Neoplasm Recurrence, Local/metabolism , Receptors, Ectodysplasin/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neoplasm Recurrence, Local/genetics , Receptors, Ectodysplasin/genetics , Wnt Signaling Pathway/genetics
10.
Sci Rep ; 9(1): 7764, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31123322

ABSTRACT

Early identification of severe acute pancreatitis (SAP) is critical for clinical decision-making. The apolipoprotein B-to-apolipoprotein A1 ratio (ApoB/A1 ratio) reflects the balance between pro-inflammation and anti-inflammation in vivo. This study investigated the association between serum ApoB/A1 ratio at admission and acute pancreatitis (AP) severity. A total of 375 patients with first attack of AP were retrospectively recruited from January 2014 to December 2017. The severity of AP was assessed at admission based on the 2012 revised Atlanta Classification. Serum lipids levels were tested on the first 24 h of hospitalization, of which the correlations with clinical features or scoring systems were also measured. The ApoB/A1 ratio markedly increased across disease severity of AP. The ApoB/A1 ratio, expressed as both quartile and continuous variables, was significantly associated with a high risk of SAP, even after adjustment for other conventional SAP risk factors. The ApoB/A1 ratio positively correlated with the revised 2012 Atlanta Classification, Ranson score, Bedside Index for Severity in AP score, Modified Computed Tomography Severity Index score, and Acute Physiology and Chronic Health Evaluation II score for AP severity. The optimal cut-off value of ApoB/A1 ratio for detecting SAP was 0.88, with a sensitivity of 83.08% and a specificity of 69.03%. Serum ApoB/A1 ratio at admission is closely correlated with disease severity in patients with AP and can serve as a reliable indicator for SAP in clinical setting.


Subject(s)
Apolipoprotein A-I/analysis , Apolipoproteins B/analysis , Pancreatitis/metabolism , Adult , Aged , Apolipoprotein A-I/blood , Apolipoproteins B/blood , Biomarkers/blood , Female , Humans , Male , Middle Aged , Pancreatitis/blood , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Sensitivity and Specificity , Severity of Illness Index
11.
Exp Ther Med ; 17(4): 3209-3214, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30936995

ABSTRACT

MicroRNA-32 (miR-32) is upregulated in colorectal cancer (CRC) tissues; its overexpression leads to increased cell proliferation, migration and invasion, as well as reduced apoptosis of CRC cells, at least partly by inhibiting the target gene phosphatase and tensin homolog. However, the mechanisms of its upregulation have remained elusive. In the present study, the effects of methylation and acetylation on the expression of miR-32 were investigated. The promoter methylation status of miR-32 in the CRC cell lines HT-29 and HCT-116 and the normal colonic epithelial cell line NCM460 was investigated by bisulfate sequencing polymerase chain reaction (BSP). The potential role of methylation and histone acetylation in the regulation of miR-32 expression in CRC cells was investigated using the demethylation reagent 5-aza-2'-deoxycytidine (5-Aza-dC), the histone deacetylase inhibitor trichostatin A (TSA) and transfection of DNA methyltransferase 1 (DNMT1) overexpression plasmid. BSP revealed that CpG sites in the miR-32 promoter region of CRC and normal colonic epithelial cells were all hypomethylated, with methylation rates of 0.12, 1.14 and 0.64% in HCT-116, HT-29 and NCM460 cells, respectively. Treatment with 5-Aza-dC and/or TSA and transfection with DNMT1 plasmid did not significantly alter the expression of miR-32. Therefore, the present results suggest that methylation and histone acetylation do not affect miR-32 expression in CRC cells.

12.
Oncol Lett ; 17(4): 3743-3750, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30881496

ABSTRACT

The pathogenesis of colorectal cancer (CRC) is poorly understood. MicroRNA (miR)-32 upregulation in CRC tissues was previously reported, where it increased the proliferation, migration and invasion, and reduced apoptosis of CRC cells by inhibiting the expression of phosphatase and tensin homolog (PTEN). However, the mechanism underlying miR-32 upregulation remains unknown. miR-32 is an intronic miRNA located within intron 14 of the transmembrane protein 245 gene (TMEM245). The present study aimed to elucidate the biological pathways underlying miR-32 regulation in CRC. A truncated promoter containing the 5'-flanking region of TMEM245/miR-32 gene was constructed. The promoter region was analyzed by dual luciferase reporter assay in CRC cells. DNA pull-down assay and mass spectrometry (MS) were used to identify proteins binding to the core promoter. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and transcription factor (TF) analyses were used to identify the binding proteins. The -320 to -1 bp fragment of the 5'-flanking region exhibited the highest luciferase activity. The regions spanning -606 to -320 bp exhibited a significant decrease in luciferase activity, compared with the -320 to -1 bp fragment. DNA pull-down assay and MS revealed 403 potential miR-32 promoter binding proteins. GO and KEGG pathway analysis indicated that these proteins were involved in numerous physiological and biochemical processes, including 'structural molecule activity', 'RNA binding', 'small molecule metabolic process' and 'biogenesis'. Furthermore, TF analysis revealed 10 potential interacting TFs, including SMAD family member 1 (SMAD1), signal transducer and activator of transcription 1 (STAT1) and forkhead box K1 (Foxk1). These results suggested that the core promoter region may be located within-320 to -1 bp of the 5'-flanking region of TMEM245/miR-32 gene, while the region from -606 to -320 bp may harbor repressive regulatory elements. The TFs SMAD1, STAT1 and Foxk1 may be involved in the transcriptional regulation of miR-32.

13.
Korean J Parasitol ; 56(4): 325-334, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30196664

ABSTRACT

Toxoplasma gondii is an apicomplexan zoonotic protozoan parasite that infects most species of warm-blooded animals, including humans. The heavy incidence and severe or lethal damage caused by T. gondii infection clearly indicate a need for the development of an effective vaccine. T. gondii GRA8 is a member of the dense granules protein family and is used as a marker of acute infection. In the present study, we evaluated the protective immunity induced by DNA vaccination based on a recombinant eukaryotic plasmid, pDsRed2-GRA8, against acute toxoplasmosis in mice. BALB/c mice were intramuscularly immunized with the pDsRed2-GRA8 plasmid and then challenged by infection with the highly virulent GFP-RH strain of T. gondii. The specific immune responses and protective efficacy against T. gondii of this vaccine were analyzed by measuring cytokine and serum antibody titers, splenocyte proliferation assays, and the survival times of mice after challenge. Our results showed that mice immunized with pDsRed2-GRA8 demonstrated specific humoral and cellular responses, induced higher IgG antibody titers with predominant IgG2a production; increased levels of IL-10, IL-12 (p70), IFN-γ, TNF-α, and splenocyte proliferation; and prolonged survival times compared to those of control mice. The present study showed that DNA immunization with pDsRed2-GRA8 induced humoral and cellular immune responses, and all immunized mice showed greater Th1-type immune responses and longer survival times than those of control mice. These results indicated that T. gondii GRA8 DNA immunization induces a partial protective effect against acute toxoplasmosis.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Toxoplasmosis/prevention & control , Vaccines, DNA/immunology , Acute Disease , Animals , Antibodies, Protozoan/blood , Cell Proliferation , Cytokines/blood , Disease Models, Animal , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G/blood , Mice, Inbred BALB C , Spleen/cytology , Spleen/immunology
14.
Oncotarget ; 8(1): 705-721, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27893428

ABSTRACT

Increasing evidence suggests that miRNAs are widely dysregulated in ulcerative colitis (UC), potentially affecting UC pathogenesis, diagnosis, and therapy. microRNA (miR) -206 has been reported to be upregulated in UC; however, its function and role in UC remain unknown. Here, we elucidate the function of miR-206 in the pathogenesis of UC. In patients with active-UC, miR-206 and adenosine A3 receptor (A3AR) levels were significantly upregulated and downregulated, respectively, and were inversely correlated. A3AR was expressed in the colon mucosa (particularly in colon epithelial-cell membranes). In HT-29 cells, miR-206 downregulated A3AR mRNA/protein expression by directly targeting the A3AR 3'-UTR; miR-206 overexpression and knockdown respectively increased and decreased TNF-α-induced nuclear NF-κB/p65, p-IκB-α, IKKα, p-IKKα and IL-8/IL-1ß secretion. However, A3AR-siRNA reversed the miR-206 inhibitory effect. Furthermore, miR-206 increased dextran sodium sulphate-induced colitis severity (i.e., increased bodyweight loss, DAI score, colon shrinkage, and MPO activity), which was partially ameliorated by miR-206-antagomir treatment. miR-206-agomir treatment potently suppressed A3AR expression and increased NF-κB signalling and downstream cytokine (TNF-α/IL-8/IL-1ß) expression in the mouse colon, in contrast to miR-206-antagomir administration. Taken together, our results demonstrated that miR-206 has a proinflammatory role in UC by downregulating A3AR expression and activating NF-κB signalling.


Subject(s)
Colitis, Ulcerative/genetics , Gene Expression Regulation , MicroRNAs/genetics , RNA Interference , Receptor, Adenosine A3/genetics , 3' Untranslated Regions , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Silencing , HT29 Cells , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , NF-kappa B/metabolism , Protein Transport , Receptor, Adenosine A3/metabolism , Severity of Illness Index , Signal Transduction
15.
Sci Rep ; 6: 30824, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27476546

ABSTRACT

MicroRNAs (miRNAs) act as important post-transcriptional regulators of gene expression by targeting the 3'-untranslated region of their target genes. Altered expression of miR-16 is reported in human ulcerative colitis (UC), but its role in the development of the disease remains unclear. Adenosine through adenosine A2a receptor (A2aAR) could inhibit nuclear factor-kappaB (NF-κB) signaling pathway in inflammation. Here we identified overexpression of miR-16 and down-regulation of A2aAR in the colonic mucosa of active UC patients. We demonstrated that miR-16 negatively regulated the expression of the A2aAR at the post-transcriptional level. Furthermore, transfection of miR-16 mimics promoted nuclear translocation of NF-κB p65 protein and expression of pro-inflammatory cytokines, IFN-γ and IL-8 in colonic epithelial cells. Treatment with miR-16 inhibitor could reverse these effects in cells. The A2aAR-mediated effects of miR-16 on the activation of the NF-κB signaling pathway were confirmed by the A2aAR knockdown assay. Our results suggest that miR-16 regulated the immune and inflammatory responses, at least in part, by suppressing the expression of the A2aAR to control the activation of the NF-κB signaling pathway.


Subject(s)
Colitis, Ulcerative/pathology , Gene Expression Regulation , MicroRNAs/genetics , NF-kappa B/metabolism , RNA, Messenger/metabolism , Receptor, Adenosine A2A/metabolism , 3' Untranslated Regions , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Cytokines/metabolism , HT29 Cells , Humans , Intestinal Mucosa/metabolism , NF-kappa B/genetics , RNA, Messenger/genetics , Receptor, Adenosine A2A/genetics , Signal Transduction
16.
Surg Endosc ; 30(8): 3357-61, 2016 08.
Article in English | MEDLINE | ID: mdl-26497947

ABSTRACT

BACKGROUND AND AIM: Laparoscopic-assisted surgery (LAC) is an alternative to open surgery for gastrointestinal stromal tumors (GISTs). Endoscopic full-thickness resection (EFTR), a recently developed procedure, is increasingly used to resect GISTs originated from the muscularis propria. In this retrospective study, we aimed to compare EFTR with LAC as minimally invasive treatments for GISTs, especially those with a diameter <2 cm, originating from the muscularis propria. Moreover, we evaluated the clinical efficacy, safety, and feasibility of EFTR for GISTs. METHODS: The study included 68 patients with GISTs originating from the muscularis propria (35 patients who underwent EFTR, and 33 who underwent LAC) who were treated at the Affiliated Hospital of Guangdong Medical University (Zhanjiang, China) between January 2011 and December 2013. The therapeutic outcomes of EFTR and LAC were reviewed retrospectively. RESULTS: In the EFTR group, the mean tumor size was 13 ± 5 mm, the mean procedure time was 91 ± 63 min, and the complete resection rate was 100 %. There were 35 "artificial" perforations and four cases of intraoperative bleeding; all complications were successfully managed endoscopically without emergency surgery. In the LAC group, the mean tumor size was 16 ± 4 mm, the mean operation time was 155 ± 37 min, and complications included three wound infections and one anastomotic leakage. CONCLUSIONS: EFTR was associated with a lower complication rate than LAC, with favorable en bloc and sufficient tumor tissue for histological diagnosis. EFTR seems to be an efficacious, relatively safe, and minimally invasive treatment for GISTs and could replace LAC surgical resection in cases where the tumor is smaller than 2 cm in diameter.


Subject(s)
Endoscopy, Gastrointestinal , Gastrointestinal Neoplasms/surgery , Gastrointestinal Stromal Tumors/surgery , Laparoscopy , Blood Loss, Surgical , Female , Humans , Length of Stay , Male , Middle Aged , Operative Time , Postoperative Complications , Retrospective Studies
17.
Immunol Lett ; 170: 88-94, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26349055

ABSTRACT

The receptor for advanced glycation end products (RAGE) and its proinflammatory ligands are critically implicated in the pathological progression of ulcerative colitis (UC). Functional polymorphisms in the regulatory elements and/or ligand-binding regions of the RAGE gene affect the expression and function of RAGE and thus may increase susceptibility to UC. In this study, a total of 266 unrelated UC patients and 247 control subjects were analyzed for 3 RAGE single nucleotide polymorphisms (SNPs) (-429 T/C, -374 T/A, and G82S) using an improved small-amplicon high resolution melting curve (HRM) analysis assay. Serum levels of soluble RAGE (sRAGE) were determined by a double sandwich ELISA system. The genotypes, alleles and haplotypes were analyzed and compared between UC patients and control subjects. Three pairs of genotyping primers for three RAGE polymorphism loci (-429 T/C, -374 T/A, and G82S) were developed based on HRM. Significant differences in the allele distribution of the G82S polymorphism was found among UC cases and controls from a Chinese population. Carriers of the RAGE G82S variant genotype were at higher risk of UC (OR=2.594, 95% CI: 1.778-3.784, P<0.001) than homozygous wild-type individuals. Further analyses revealed that the 82 (GS+SS) variant genotype was associated with patients who have extended UC (OR=1.924, 95% CI: 1.163-3.181, P=0.010), and a family history of inflammatory bowel disease (IBD) (OR=1.923, 95% CI: 1.049-3.521, P=0.032). The polymorphisms -374 T/A and -429 T/C did not demonstrate any association with UC, but an association was found between the -374(TA+AA) variant genotypes and the serum sRAGE level (P=0.002). Moreover, haplotypes T/A/A and T/A/G showed significantly different frequencies between UC patients and controls (OR=3.337, 95% CI: 1.892-6.091, P=0.026; OR=0.530, 95% CI: 0.351-0.801, P=0.002). The present study developed novel primers based on HRM to provide preliminary evidence in a Chinese population that the RAGE polymorphism is involved in genetic susceptibility to UC and that the 82(GS+SS) genotype of G82S is a risk factor for UC. Furthermore, RAGE polymorphisms may be related to the location of UC as well as a family history of IBD in a Chinese population.


Subject(s)
Asian People/genetics , Colitis, Ulcerative/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Receptor for Advanced Glycation End Products/genetics , Adult , Alleles , Biomarkers , Case-Control Studies , China , Colitis, Ulcerative/blood , Female , Gene Frequency , Genotype , Haplotypes , Humans , Male , Middle Aged , Odds Ratio , Receptor for Advanced Glycation End Products/blood , Risk
18.
DNA Cell Biol ; 34(7): 470-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25974152

ABSTRACT

Hepatic fibrosis, which results from chronic liver disease, currently lacks effective treatment. MicroRNAs, a group of small noncoding RNA molecules, have been observed to play an essential role in liver diseases, including hepatic fibrosis. In this study, we described the regulation of nuclear factor kappa B (NF-κB) inhibitor alpha (IκBα) and its possible signaling pathway by miR-126 in human hepatic stellate cell (HSC) line LX-2. The 3'-untranslated region (3'-UTR) of IκBα combined with miR-126 was analyzed by using a dual-luciferase reporter assay. Furthermore, the effects of miR-126 on IκBα mRNA and protein and NF-κB protein expression were assessed by real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and western blot analysis in the human HSC LX-2 cell line transfected with miR-126 mimic or inhibitor. Moreover, to understand the molecular mechanism of miR-126 in promoting liver fibrosis through NF-κB signaling pathway, the NF-κB downstream signaling factors expression such as transforming growth factor (TGF)-ß1 and collagen I mRNA were detected by real-time qRT-PCR. We identified that IκBα is a potential target gene of miR-126, by directly targeting its 3'-UTR. Endogenous miR-126 and exogenous miR-126 mimic inhibited IκBα expression. Moreover, overexpression of miR-126 reduced total and the cytoplasm IκBα protein expression and increased total and cytoblast NF-κB protein expression of LX-2. Conversely, knockdown of miR-126 could inhibit NF-κB activation by upregulation of IκBα protein expression. Further, miR-126 promoted TNF-a-induced TGF-ß1 and collagen I mRNA expression in LX-2 cells. miR-126 may play an important role in hepatic fibrosis by downregulating the expression of IκBα partly through the NF-κB signaling pathway.


Subject(s)
Hepatic Stellate Cells/metabolism , MicroRNAs/genetics , NF-kappa B/metabolism , 3' Untranslated Regions , Base Sequence , Binding Sites , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Gene Expression , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , MicroRNAs/metabolism , RNA Interference , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation
19.
Sci Rep ; 5: 9047, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25762375

ABSTRACT

The role of the adenosine A3 receptor (A3AR) in experimental colitis is controversial. The A3AR agonist N(6)-(3-iodobenzyl)adenosine-5'-N-methyluronamide (IB-MECA) has been shown to have a clinical benefit, although studies in A3AR-deficient mice suggest a pro-inflammatory role. However, there are no studies on the effect of 2-Cl-IB-MECA and the molecular mechanism of action of A3AR in murine colitis models in vivo. Is it the same as that observed in vitro? The interaction between 2-CL-IB-MECA and A3AR in a murine colitis model and the signaling pathways associated with this interaction remain unclear. Here we demonstrate a role for the NF-κB signaling pathway and its effect on modifying the activity of proinflammatory factors in A3AR-mediated biological processes. Our results demonstrated that A3AR activation possessed marked effects on experimental colitis through the NF-κB signaling pathway.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Colitis/metabolism , NF-kappa B/metabolism , Receptor, Adenosine A3/metabolism , Signal Transduction/drug effects , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/administration & dosage , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Inflammation Mediators/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Peroxidase/metabolism , Receptor, Adenosine A3/genetics
20.
Mediators Inflamm ; 2014: 818251, 2014.
Article in English | MEDLINE | ID: mdl-24864134

ABSTRACT

To investigate the expression of adenosine A3 receptor (A3AR) in human colonic epithelial cells and the effects of A3AR activation on tumor necrosis factor alpha (TNF-α-) induced inflammation in order to determine its mechanism of action in human colonic epithelial cells, human colonic epithelial cells (HT-29 cells) were treated with different concentrations of 2-Cl-IB-MECA prior to TNF-α stimulation, followed by analysis of NF-κB signaling pathway activation and downstream IL-8 and IL-1ß production. A3AR mRNA and protein were expressed in HT-29 cells and not altered by changes in TNF-α or 2-Cl-IB-MECA. Pretreatment with 2-Cl-IB-MECA prior to stimulation with TNF-α attenuated NF-κB p65 nuclear translocation as p65 protein decreased in the nucleus of cells and increased in the cytoplasm, inhibited the degradation of IκB-α, and reduced phosphorylated-IκB-α level significantly, compared to TNF-α-only-treated groups. Furthermore, 2-Cl-IB-MECA significantly decreased TNF-α-stimulated IL-8 and IL-1ß mRNA expression and secretion, compared to the TNF-α-only treated group. These results confirm that A3AR is expressed in human colonic epithelial cells and demonstrate that its activation has an anti-inflammatory effect, through the inhibition of NF-κB signaling pathway, which leads to inhibition of downstream IL-8 and IL-1ß expression. Therefore, A3AR activation may be a potential treatment for gut inflammatory diseases such as inflammatory bowel disease.


Subject(s)
Inflammation/metabolism , Receptor, Adenosine A3/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Colon/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HT29 Cells , Humans , Inflammation/chemically induced , Interleukin-1beta/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Receptor, Adenosine A3/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...