Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Diabetes ; 2023 Dec 27.
Article in English | MEDLINE | ID: mdl-38149757

ABSTRACT

AIMS: Bariatric surgery results in rapid recovery of glucose control in subjects with type 2 diabetes mellitus. However, the underlying mechanisms are still largely unknown. The present study aims to clarify how bariatric surgery modifies pancreatic cell subgroup differentiation and transformation in the single-cell RNA level. METHODS: Male, 8-week-old Zucker diabetic fatty (ZDF) rats with obesity and diabetes phenotypes were randomized into sleeve gastrectomy (Sleeve, n = 9), Roux-en-Y gastric bypass (RYGB, n = 9), and Sham (n = 7) groups. Two weeks after surgery, the pancreas specimen was further analyzed using single-cell RNA-sequencing technique. RESULTS: Two weeks after surgery, compared to the Sham group, the metabolic parameters including fasting plasma glucose, plasma insulin, and oral glucose tolerance test values were dramatically improved after RYGB and Sleeve procedures (p < .05) as predicted. In addition, RYGB and Sleeve groups increased the proportion of pancreatic ß cells and reduced the ratio of α cells. Two multiple hormone-expressing cells were identified, the Gcg+/Ppy + and Ins+/Gcg+/Ppy + cells. The pancreatic Ins+/Gcg+/Ppy + cells were defined for the first time, and further investigation indicates similarities with α and ß cells, with unique gene expression patterns, which implies that pancreatic cell transdifferentiation occurs following bariatric surgery. CONCLUSIONS: For the first time, using the single-cell transcriptome map of ZDF rats, we reported a comprehensive characterization of the heterogeneity and differentiation of pancreatic endocrinal cells after bariatric surgery, which may contribute to the underlying mechanisms. Further studies will be needed to elucidate these results.

2.
Front Physiol ; 13: 836918, 2022.
Article in English | MEDLINE | ID: mdl-35464075

ABSTRACT

Metabolic and bariatric surgery (MBS) can generate a drastic shift of coding and noncoding RNA expression patterns in the gastrointestinal system, which triggers organ function remodeling and may induce type 2 diabetes (T2D) remission. Our previous studies have demonstrated that the altered expression profiles of duodenal and jejunal long noncoding RNAs (lncRNAs) after the duodenal-jejunal bypass (DJB), an investigational procedure and research tool of MBS, can improve glycemic control by modulating the entero-pancreatic axis and gut-brain axis, respectively. As an indiscerptible part of the intestine, the ileal lncRNA expression signatures after DJB and the critical pathways associated with postoperative correction of the impaired metabolism need to be investigated too. High-fat diet-induced diabetic mice were randomly assigned into two groups receiving either DJB or sham surgery. Compared to the sham group, 1,425 dysregulated ileal lncRNAs and 552 co-expressed mRNAs were identified in the DJB group. Bioinformatics analysis of the differently expressed mRNAs and predicted target genes or transcriptional factors indicated that the dysregulated ileal lncRNAs were associated with lipid and amino acid metabolism-related pathways. Moreover, a series of lncRNAs and their potential target mRNAs, especially NONMMUT040618, Pxmp4, Pnpla3, and Car5a, were identified on the pathway. In conclusion, DJB can induce remarkable alteration of ileal lncRNA and mRNA expression. The role of the ileum in DJB tends to re-establish the energy homeostasis by regulating the lipid and amino acid metabolism.

3.
Obes Surg ; 31(3): 1361-1368, 2021 03.
Article in English | MEDLINE | ID: mdl-33405187

ABSTRACT

BACKGROUND: We designed a novel malabsorptive procedure named as jejunal-ileal loop bipartition (JILB), in which a jejunal-ileal loop is created to reduce the effective length of food chyme passage in the small bowel, but without exclusion of any segment of the intestine. This study is to investigate the feasibility and efficacy of JILB on weight loss and glycemic control in obese diabetic mouse model. METHODS: High-fat diet-induced C57BL/6 mice with typical obese and diabetic phenotypes were randomly divided into two groups according to the surgical procedure performed, including JILB (n = 8) and sham group (n = 8). Age-matched naïve C57BL/6 mice fed with rodent chow diet were adopted as normal controls. Body weight, food intake, fasting plasma glucose (FPG), fasting plasma insulin (FPI), and oral glucose tolerance test (OGTT) were measured in vivo before and 2, 4, and 8 weeks after surgery. Plasma glucagon-like peptide 1 (GLP-1) was assayed before and 15 min after oral glucose challenge at the 8th week postoperatively. RESULTS: Comparing to the sham animals, JILB group consumed similar amount of food, but had lower body weight after surgery (P < 0.01). It led to significant lower FPG (p < 0.05) and improved glucose tolerance with lower FPI (p < 0.001). And GLP-1 secretion at 15 min after oral glucose challenge was higher than shams (P < 0.05). No intestinal obstruction was identified. CONCLUSIONS: JILB is potentially a metabolic and bariatric procedure that leads to effective weight loss and diabetes remission in obese diabetic subjects.


Subject(s)
Diabetes Mellitus, Type 2 , Obesity, Morbid , Animals , Blood Glucose , Diabetes Mellitus, Type 2/surgery , Gastrectomy , Insulin , Mice , Mice, Inbred C57BL , Obesity, Morbid/surgery , Rodentia
4.
Database (Oxford) ; 20192019 01 01.
Article in English | MEDLINE | ID: mdl-31089686

ABSTRACT

Gastrointestinal (GI) cancer is common, characterized by high mortality, and includes oesophagus, gastric, liver, bile duct, pancreas, rectal and colon cancers. The insufficient specificity and sensitivity of biomarkers is still a key clinical hindrance for GI cancer diagnosis and successful treatment. The emergence of `precision medicine', `basket trial' and `field cancerization' concepts calls for an urgent need and importance for the understanding of how organ system cancers occur at the molecular levels. Knowledge from both the literature and data available in public databases is informative in elucidating the molecular alterations underlying GI cancer. Currently, most available cancer databases have not offered a comprehensive discovery of gene-disease associations, molecular alterations and clinical information by integrated text mining and data mining in GI cancer. We develop GIDB, a panoptic knowledge database that attempts to automate the curation of molecular signatures using natural language processing approaches and multidimensional analyses. GIDB covers information on 8730 genes with both literature and data supporting evidence, 248 miRNAs, 58 lncRNAs, 320 copy number variations, 49 fusion genes and 2381 semantic networks. It presents a comprehensive database, not only in parallelizing supporting evidence and data integration for signatures associated with GI cancer but also in providing the timeline feature of major molecular discoveries. It highlights the most comprehensive overview, research hotspots and the development of historical knowledge of genes in GI cancer. Furthermore, GIDB characterizes genomic abnormalities in multilevel analysis, including simple somatic mutations, gene expression, DNA methylation and prognosis. GIDB offers a user-friendly interface and two customizable online tools (Heatmap and Network) for experimental researchers and clinicians to explore data and help them shorten the learning curve and broaden the scope of knowledge. More importantly, GIDB is an ongoing research project that will continue to be updated and improve the automated method for reducing manual work.


Subject(s)
Biomarkers, Tumor , Data Curation , Data Mining , Gastrointestinal Neoplasms , Natural Language Processing , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Humans , Precision Medicine
5.
Int J Mol Med ; 43(4): 1657-1668, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816423

ABSTRACT

Interleukin­22 (IL­22) has both pro­inflammatory and anti­inflammatory properties in a number tissues depending on the environment. Epithelial cells usually have a rapid turnover and are fueled by tissue stem cells. However, the question of whether IL­22 regulates tissue homeostasis through the modulation of stem cells remains unanswered. In this study, we investigated the role of IL­22 in the homeostasis of intestinal epithelial cells (IECs) during inflammation through a 3D organoid culture system. qPCR was performed to detect the changes in important gene transcriptions, and immunohistochemistry and western blot analysis were carried out to determine protein expression. As a result, we found that the expression of IL­22 was synchronously altered with the damage of the intestine. IL­22 treatment promoted cell proliferation and suppressed the cell differentiation of intestinal organoids. Surprisingly, IL­22 also led to self­renewal defects of intestinal stem cells (ISCs), thereby eventually resulting in the death of organoids. In examining the underlying mechanisms, we found that IL­22 activated signal transducer and activator of transcription 3 (Stat3) phosphorylation and suppressed the Wnt and Notch signaling pathways. Importantly, Wnt3a treatment attenuated the organoid defects caused by IL­22, which consolidated the importance of Wnt pathway at the downstream of IL­22. Collectively, the findings of this study indicate that IL­22 regulates the homeostasis of the intestinal epithelium and is critical for the regeneration of the intestine during inflammation. Thus, the data of this study may provide a potential strategy and a basis for the treatment of diseases of intestinal inflammation in clinical practice.


Subject(s)
Homeostasis , Inflammation/metabolism , Inflammation/pathology , Interleukins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Animals , Cell Self Renewal , Epithelial Cells/metabolism , Male , Mice, Inbred C57BL , Organoids/metabolism , Phosphorylation , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , Interleukin-22
6.
Obes Surg ; 29(4): 1174-1184, 2019 04.
Article in English | MEDLINE | ID: mdl-30610678

ABSTRACT

BACKGROUND: Single-anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI-S) has launched a huge challenge to classic Roux-en-Y gastric bypass (RYGB). Our objective was to compare diabetes remission and micronutrient deficiency in a mildly obese diabetic rat model undergoing SADI-S versus RYGB. METHODS: Thirty adult male mildly obese diabetic rats were randomly assigned to sham (S), SADI-S, and RYGB groups. Body weight, food intake, fasting plasma glucose (FPG), oral glucose tolerance test (OGTT), plasma insulin, GLP-1, and ghrelin levels were measured at indicated time points. Meanwhile, insulin sensitivity and pancreatic ß cell function were assessed during OGTT. Finally, plasma micronutrient evaluation and islet ß cell mass analysis were performed after all animals were sacrificed. RESULTS: As compared to sham, the SADI-S and RYGB groups achieved almost equivalent efficacy in caloric restriction and FPG control without excessive weight loss. During OGTT, the SADI-S and RYGB groups also provided comparable effects on glycemic excursion, insulin sensitivity, and ß cell function; however, only rats in the RYGB group showed significant changes in gut hormones, whereas the three groups were found to exhibit no significant difference in ß cell mass. In addition, only vitamin E in the RYGB group was deficient as compared with the SADI-S and S groups. CONCLUSION: In mildly obese diabetic rat, SADI-S and RYGB procedures have comparable efficacy in diabetes remission and risk of micronutrient deficiency. These data show that each of the surgery accomplishes diabetes improvements through both overlapping and distinct mechanisms requiring further investigation.


Subject(s)
Bariatric Surgery/methods , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 2/surgery , Micronutrients/deficiency , Obesity/surgery , Anastomosis, Surgical/adverse effects , Anastomosis, Surgical/methods , Animals , Bariatric Surgery/adverse effects , Blood Glucose/analysis , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Duodenum/surgery , Eating/physiology , Gastrectomy/methods , Gastric Bypass/adverse effects , Gastric Bypass/methods , Gastrointestinal Hormones/blood , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Ileum/surgery , Insulin Resistance/physiology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Male , Obesity/pathology , Obesity/physiopathology , Random Allocation , Rats , Rats, Wistar , Remission Induction , Weight Loss/physiology
7.
Front Neurosci ; 13: 1403, 2019.
Article in English | MEDLINE | ID: mdl-32009884

ABSTRACT

We have developed a low-cost electromagnetic vibrotactile stimulator that uses the magnetic field of an MR scanner as a permanent magnet to power a vibrating motor. A simple variable current power supply is controlled by software using a USB data acquisition controller. In our study, the function of our novel stimulator was verified in a vibration frequency discrimination working memory task, in which various ranges of frequencies and amplitudes are delivered in MRI scanner. Furthermore, our functional MRI study revealed activations of the primary and secondary somatosensory cortices during the perception of tactile stimulation. Therefore, the new designed electromagnetic vibrotactile stimulator is capable of generating various frequencies of tactile stimuli and represents a powerful and useful tool for studying somatosensory functions with functional MRI.

8.
Exp Cell Res ; 372(1): 61-72, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30236513

ABSTRACT

Thioredoxin 2 (Trx2), as a member of the thioredoxin system in mitochondria, is involved in controlling mitochondrial redox state. However, the role of Trx2 in cardiac biology is not fully understood. In the present study, the expression of Trx2 is silenced in quiescent neonatal rat ventricular cardiomyocytes (NRVCs) and mitochondrial respiratory function and cardiomyocyte hypertrophy are assessed. The results show that Trx2 depletion does not induce significant cytotoxicity in quiescent NRVCs. Remarkably, Trx2 depletion results in cardiomyocyte hypertrophy as determined by increased cell size and protein synthesis. Furthermore, Trx2 depletion inhibits AMPK activity and AMPK activator reversed cellular hypertrophy. Trx2 depletion enhances mitochondrial ROS generation without impact on cellular ROS level. Trx2 depletion has no effect on mitochondrial biogenesis. Specifically, Trx2 depletion increases mitochondrial respiration flux and total ATP concentration under quiescent conditions. To decipher the relationship between ROS generation, mitochondrial respiration flux, and AMPK signaling, mitochondrial metabolism and ROS was specifically inhibited, and the results show that AMPK inactivation and hypertrophic response in Trx2-silenced cells is reversed by respiration blockers but not ROS scavenger. In conclusion, these results show that beyond mitochondrial ROS scavenging, Trx2 controls mitochondrial respiratory function in quiescent cardiomyocytes and is implicated in cardiomyocyte hypertrophy via AMPK signaling.


Subject(s)
AMP-Activated Protein Kinases/genetics , Adenosine Triphosphate/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Thioredoxins/genetics , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Size , Gene Expression Regulation , Heart Ventricles/cytology , Heart Ventricles/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Oligomycins/pharmacology , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Ribonucleotides/pharmacology , Rotenone/pharmacology , Signal Transduction , Thioredoxins/antagonists & inhibitors , Thioredoxins/metabolism
9.
Surg Obes Relat Dis ; 14(6): 821-832, 2018 06.
Article in English | MEDLINE | ID: mdl-29631984

ABSTRACT

BACKGROUND: Metabolic and bariatric surgery is effective in ameliorating type 2 diabetes, although its underlying mechanisms are largely unknown. Our previous study indicated that the distinctly expressed duodenal long noncoding RNAs (lncRNAs) induced by the duodenal-jejunal bypass (DJB) might play a role in improving glycemic control via the enteropancreatic axis. Therefore, the physiologic role of the jejunum in metabolic regulation after DJB requires investigation. OBJECTIVES: To investigate the alterations in the jejunal Roux limb lncRNA expression signatures after DJB and analyze the functional pathways associated with metabolic improvement on a genome-wide scale in high-fat diet-induced diabetic mice. SETTING: University medical center. METHODS: Diabetic mice induced by high-fat diet were randomly assigned into 2 groups undergoing either DJB or sham surgery. The lncRNA and messenger (m)RNA expression profiles of the Roux limb segment of the jejunum in both groups were investigated using microarray. To identify the functional characteristics of the distinctly expressed lncRNAs, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis were conducted. The lncRNA-mRNA and lncRNA-transcription factor interaction networks were constructed based on Pearson correlation analysis. RESULTS: Compared with the sham group, 827 dysregulated (fold change ≥2.0) jejunal lncRNAs were identified in the DJB group. Both Kyoto Encyclopedia of Genes and Genomes pathway and gene ontology enrichment analysis revealed that 601 lncRNA-co-expressed mRNAs (fold change ≥2.0) were associated with neuromodulation-related pathways or biological processes, including serotonergic, glutamatergic, and dopaminergic synapses. In addition, hormonal regulation-related pathways, especially steroid biosynthesis, were also enriched. The results were further confirmed by bioinformatic analysis of target genes or transcription factors predicted on the basis of dysregulated jejunal lncRNAs. Furthermore, the NONMMUT023781 lncRNA may simultaneously target the Adcy8 mRNA both in cis and in trans and participate in neuromodulation and hormonal regulation. CONCLUSION: Alterations of jejunal Roux limb lncRNA and mRNA expression profiles trigger both neuromodulation and endocrine-related pathways, which play a critical role in type 2 diabetes remission after metabolic and bariatric surgery via the gut-brain axis. NONMMTU023781 and Adcy8 were identified as potential targets, which warrant further research.


Subject(s)
Bariatric Surgery , Diabetes Mellitus, Type 2/surgery , Duodenum/surgery , Jejunum/surgery , RNA, Long Noncoding/metabolism , Animals , Blood Glucose/metabolism , Brain/physiology , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Down-Regulation/physiology , Duodenum/metabolism , Gene Ontology , Genome/physiology , Jejunum/metabolism , Jejunum/physiology , Mice, Inbred C57BL , Microarray Analysis/methods , Neurotransmitter Agents/physiology , Postoperative Period , RNA, Messenger/metabolism , Random Allocation , Transcription Factors/physiology
10.
Biochem Biophys Res Commun ; 495(1): 693-699, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29117537

ABSTRACT

Hypoglycemia is a major barrier to achieving stable metabolic control in patients with diabetes which is a serious clinical concern. With progression of diabetes, the ability of pancreatic α-cells which respond to hypoglycemia becomes impaired; However, it is not clear whether the dysfunctional responses of α-cells during hypoglycemia are related with oxidative stress. In the present study, we investigated whether epigallocatechin-3-gallate (EGCG) has antioxidant potential on pancreatic alpha TC1-6 (αTC1-6) cell lines and protect the normal function of α-cells from H2O2 induced oxidative stress. ROS production, cell viability, glucagon secretion, and cell apoptosis were assessed. EGCG reduced ROS production and cell apoptosis, while restored cell viability and glucagon secretion within a particular concentration range. Moreover, EGCG activated Akt signaling and inhibited P38 as well as JNK mitogen-activated protein kinase (MAPK) pathway. Taken together, EGCG prevented αTC1-6 cells from H2O2 induced oxidative stress, restored dysfunction of glucagon secretion and inhibited cell apoptosis via the activation of Akt signaling and suppression of P38 and JNK pathway. These results provide rationale for combining the conventional anti-hyperglycemia therapy and antioxidant therapy in order to avert hypoglycemia in clinical treatment of diabetes.


Subject(s)
Antioxidants/administration & dosage , Catechin/analogs & derivatives , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Catechin/administration & dosage , Cell Line , Dose-Response Relationship, Drug , Hydrogen Peroxide/administration & dosage , Mice , Oxidative Stress/drug effects
11.
Cutan Ocul Toxicol ; 37(3): 228-232, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29237295

ABSTRACT

PURPOSE: To investigate the effect of microRNA-16 on hypoxia-induced VEGF expression of ARPE-19 cells. METHODS: ARPE-19 cells were cultivated under normoxia and hypoxia state. At 0 h, 12 h, 24 h, and 48 h after cultivation, the supernate of the culture medium was separated to test the VEGF secretion by ELISA, and the cells were purified to measure the expression of VEGF mRNA and microRNA-16 by qRT-PCR; microRNA-16 mimic was then transfected into ARPE-19 cells by the Hiperfect transfection reagent, a liposome transfection system. Scramble group and the non-transfected group were set as the controls. VEGF secretion and the level of VEGF mRNA were measured in these three groups. RESULTS: The VEGF secretion of the hypoxia ARPE cells was significantly higher than the initial state (p < 0.01). Compared with the normoxia cells, the VEGF secretion of the hypoxia cell was significantly increased (p < 0.01), and the division of VEGF secretion between these two groups increased by time. VEGF mRNA of the hypoxia ARPE cell was significantly higher than the initial state (p < 0.01). Compared with the normoxia cells, VEGF mRNA of the hypoxia cells was significantly increased (p < 0.01), and the division of VEGF mRNA between these two groups increased by time. After cultivating under hypoxia, the expression of microRNA-16 in ARPE-19 cells decreased significantly compared with the normal group, and the division of these two groups augmented by time. MicroRNA-16 was successfully transfected into ARPE-19 cells by the Hiperfect transfection system. Twenty four hours and 48 h after the transfection, the VEGF secretion of the miR-16 transfected cell was decreased significantly (p < 0.01) compared with the scramble and the non-transfected group under hypoxia, while VEGF mRNA level had no significant difference among these three groups. CONCLUSIONS: Hypoxia can increase the expression of VEGF mRNA and the secretion of VEGF protein of ARPE-19 cells. At the same time, microRNA-16 expression can be down-regulated by hypoxia. Transfection of microRNA-16 exogenously can down-regulate the VEGF protein secretion but cannot affect the expression of VEGF mRNA.


Subject(s)
Cell Hypoxia , MicroRNAs/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Line , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , MicroRNAs/antagonists & inhibitors , RNA, Messenger/metabolism , Retinal Pigment Epithelium/cytology , Transfection , Vascular Endothelial Growth Factor A/metabolism
12.
Curr Chem Genom Transl Med ; 11: 41-49, 2017.
Article in English | MEDLINE | ID: mdl-29238655

ABSTRACT

Type 2 diabetes mellitus (T2DM), a complex metabolic disorder typically accompanying weight gain, is associated with progressive ß-cell failure and insulin resistance. Bariatric surgery ameliorates glucose tolerance and provides a near-perfect treatment. Duodenal-jejunal bypass (DJB) is an experimental procedure and has been studied in several rat models, but its influence in db/db mice, a transgenic model of T2DM, remains unclear. To investigate the effectiveness of DJB in db/db mice, we performed the surgery and evaluated metabolism improvement. Results showed that mice in DJB group weighed remarkably less than sham group two weeks after surgery. Compared to the preoperative level, postoperative fasting blood glucose (FBG) was dramatically reduced. Statistical analysis revealed that changes in body weight and FBG were significantly correlated. Besides, DJB surgery altered plasma insulin level with approximate 40% reduction. Thus, for the first time we proved that DJB can achieve rapid therapeutic effect in transgenic db/db mice with severe T2DM as well as obesity. In addition, decreased insulin level reflected better insulin sensitivity induced by DJB. In conclusion, our study demonstrates that DJB surgery may be a potentially effective way to treat obesity-associated T2DM.

13.
Curr Chem Genom Transl Med ; 11: 31-40, 2017.
Article in English | MEDLINE | ID: mdl-28868238

ABSTRACT

OBJECTIVE: Glioblastomas multiforme (GBM) is the most malignant brain cancer, which presented vast genomic variation with complicated pathologic mechanism. METHOD: MicroRNA is a delicate post-transcriptional tuner of gene expression in the organisms by targeting and regulating protein coding genes. MiR-9 was reported as a significant biomarker for GBM patient prognosis and a key factor in regulation of GBM cancer stem cells. To explore the effect of miR-9 on GBM cell growth, we over expressed miR-9 in U87 and U251 cells. The cell viability decreased and apoptosis increased after miR-9 overexpression in these cells. To identify the target of miR-9, we scanned miR-9 binding site in the 3'UTRs region of expression SMC1A (structural maintenance of chromosomes 1A) genes and designed a fluorescent reporter assay to measure miR-9 binding to this region. Our results revealed that miR-9 binds to the 3'sUTR region of SMC1A and down-regulated SMC1A expression. RESULT: Our results indicated that miR-9 was a potential therapeutic target for GBM through triggering apoptosis of cancer cells.

14.
Surg Obes Relat Dis ; 13(7): 1212-1226, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28366671

ABSTRACT

BACKGROUND: The duodenum plays a role in the mechanism of type 2 diabetes remission after bariatric surgery. Roux-en-Y gastric bypass (RYGB) may change gene expression in the duodenum and metabolism. Long noncoding RNAs (lncRNAs) constitute a novel class of RNAs that regulate gene expression. Little is known about how duodenal lncRNAs respond to RYGB. Logically, studies on the changes of duodenal lncRNAs potentially can lead to an understanding of the mechanisms of bariatric surgery, as well as discovery of antidiabetic drug targets and biomarkers predicting postoperative outcome. OBJECTIVES: To investigate the expression signature of duodenal lncRNAs associated with glycemic improvement by duodenal-jejunal bypass (DJB), a component of RYGB, on a genome-wide scale in high-fat diet-induced diabetic mice. SETTING: University medical center. METHODS: High fat diet-induced diabetic mice were randomized into 2 groups receiving either the DJB or a sham procedure. Microarray was applied to screen the differentially expressed lncRNAs and messenger RNAs (mRNAs) in the duodenum between the DJB and sham groups, and the result was validated by quantitative real-time polymerase chain reaction in another cohort of animals. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to predict the potential lncRNA functions. Based on Pearson correlation analysis, the lncRNA-mRNA and lncRNA-transcription factor (TF) interaction networks were constructed to identify and rank core regulatory lncRNAs and transcription factors. RESULTS: A total of 301 lncRNAs, including 232 that were upregulated and 69 downregulated (fold change≥2.0), were differentially expressed in the duodenum between the DJB and sham groups. GO enrichment indicated that these lncRNA-coexpressed mRNAs were correlated with biological processes including cell proliferation, digestion, and catabolic and biosynthetic processes. KEGG pathway analysis revealed that in addition to the digestion and absorption signaling pathways, pancreatic secretion- and inflammatory process-related signaling pathways were mostly enriched in the DJB group. In addition, the lncRNA-mRNA interaction network combined with GO and KEGG pathway analysis suggested that as a top-ranked gene, NONMMUG021726 may play an important role in the mechanism of type 2 diabetes remission after DJB. CONCLUSION: DJB leads to drastic changes in lncRNA and mRNA expressions in the duodenum. The majority of top-ranked lncRNAs and mRNAs have roles in pancreatic secretion and inflammatory processes, implying that bypass of the duodenum may initiate insulin secretion and attenuate inflammation. In addition, modulators of such lncRNAs, most likely NONMMUG021726, have potential to become therapeutic targets or biomarkers for prediction of the outcomes of bariatric surgery.


Subject(s)
Diabetes Mellitus, Type 2/surgery , Diet, High-Fat , Duodenum/metabolism , Gastric Bypass , RNA, Long Noncoding/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Down-Regulation/physiology , Epistasis, Genetic , Fasting/blood , Gene Expression , Male , Mice, Inbred C57BL , Microarray Analysis , Random Allocation , Up-Regulation/physiology
15.
Cell Biol Toxicol ; 33(5): 457-466, 2017 10.
Article in English | MEDLINE | ID: mdl-28247283

ABSTRACT

Dysregulated glucagon secretion is a hallmark of type 2 diabetes (T2D). To date, few effective therapeutic agents target on deranged glucagon secretion. Family with sequence similarity 3 member D (FAM3D) is a novel gut-derived cytokine-like protein, and its secretion timing is contrary to that of glucagon. However, the roles of FAM3D in metabolic disorder and its biological functions are largely unknown. In the present study, we investigated whether FAM3D modulates glucagon production in mouse pancreatic alpha TC1 clone 6 (αTC1-6) cells. Glucagon secretion, prohormone convertase 2 (PC2) activity, and mitogen-activated protein kinase (MAPK) pathway were assessed. Exogenous FAM3D inhibited glucagon secretion, PC2 activity, as well as extracellular-regulated protein kinase 1/2 (ERK1/2) signaling and induced MAPK phosphatase 1 (MKP1) expression. Moreover, knockdown of MKP1 and inhibition of ERK1/2 abolished and potentiated the inhibitory effect of FAM3D on glucagon secretion, respectively. Taken together, FAM3D inhibits glucagon secretion via MKP1-dependent suppression of ERK1/2 signaling. These results provide rationale for developing the therapeutic potential of FAM3D for dysregulated glucagon secretion and T2D.


Subject(s)
Cytokines/pharmacology , Dual Specificity Phosphatase 1/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , MAP Kinase Signaling System/drug effects , Animals , Cell Line , Cytokines/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Enzyme Induction/drug effects , Glucagon/antagonists & inhibitors , Mice , Mitogen-Activated Protein Kinases/metabolism , Pancreas/cytology , Pancreas/drug effects , Pancreas/enzymology , Pancreas/metabolism , Proprotein Convertase 2/antagonists & inhibitors , Proprotein Convertase 2/biosynthesis , Proprotein Convertase 2/metabolism , Receptors, Formyl Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...