Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Pest Manag Sci ; 80(6): 2668-2678, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38411319

ABSTRACT

BACKGROUND: The Angoumois grain moth, Sitotroga cerealella, is a destructive pest of maize, wheat, and rice, causing economic losses and threatening food security. This study aimed to develop and characterize microcapsules of mesoporous silica nanospheres (MSN) and cyclodextrin-modified mesoporous silica nanospheres (CDMSN) containing two aldehydes, nonanal and decanal, found in plant essential oils, to assess their attractiveness to S. cerealella populations. RESULTS: Microcapsules with 2:1 ratio of nonanal and decanal exhibited an average encapsulation efficiency of 39.82% for MSN loaded with nonanal and decanal (MSN-ND) and 46.10% for CDMSN loaded with nonanal and decanal (CDMSN-ND). They have an elliptical shape with particle sizes of 115 nm for MSN and 175 nm for CDMSN. Gas chromatography-mass spectrometry analysis revealed in vitro release of nonanal in MSN at 96.24% and decanal at 96.42% by the 36th day. CDMSN showed releases of 93.83% for nonanal and 93.74% for decanal by the 50th day. CDMSN-ND attracted adult S. cerealella for 43 days, while MSN-ND remained effective for 29 days. In mass trapping assays in simulated grain warehouse, both MSN-ND and CDMSN-ND trapped over 50% of the adult population within 7 days, significantly reducing grain infestation rates below 10% by inhibiting F1 adult emergence. At temperatures ranging from 20 °C to 35 °C, both microcapsules exhibited significant and effective attraction rates for S. cerealella. Stored wheat seeds treated with CDMSN and CDMSN-ND over 1 year showed no significant differences in key germination parameters. CONCLUSION: Microencapsulated nonanal and decanal offer a promising, sustainable approach for controlling S. cerealella infestation in stored grains, contributing to global food security. © 2024 Society of Chemical Industry.


Subject(s)
Capsules , Cyclodextrins , Edible Grain , Silicon Dioxide , Silicon Dioxide/chemistry , Animals , Cyclodextrins/chemistry , Edible Grain/chemistry , Aldehydes/chemistry , Moths/growth & development , Insect Control/methods , Food Storage
2.
Cancer Manag Res ; 15: 1141-1153, 2023.
Article in English | MEDLINE | ID: mdl-37842130

ABSTRACT

Purpose: Immune checkpoint inhibitors (ICIs) have been developed for clinical application and proven effective for non-small cell lung cancer (NSCLC). Blockade of the programmed cell death 1 (PD-1) protein can partially reinvigorate circulating exhausted-phenotype CD8+ T cells (Tex cells) in preclinical models, however the clinical implication in anti-PD-1-based immunotherapy in NSCLC is unknown. Methods: Serum specimens were obtained before and during treatment from 145 patients with NSCLC patients who received anti-PD-1 treatment and their prognoses were followed-up. Indicators such as cell subpopulations, T cell invigoration were detected by clinical laboratory testing. Survival curves were estimated by the Kaplan-Meier method, Cox regression analysis was used to identify factors associated with prognoses of NSCLC patients. Results: The expressions of Ki-67 in PD-1+/CD8+ T cells in most NSCLC patients (97 of 145 cases) increased after treatment. The responding Ki-67+/CD8+ T cell population was mainly CD45RAlo CD27hi, containing cells with high expression of CTLA-4, PD-1, and 2B4 and low expression of NKG2-D (P < 0.0001). The maximum fold change of Ki-67+/PD-1+/CD8+T cells in treatment cycles and the tumor burden determined by imaging may be associated with survival. Patients with higher Ki-67 expression on PD-1+CD8+ T-cells (pretreatment) had statistically significant increased progression-free survival (PFS). A Ki-67 expression to tumor burden ratio greater than 0.6 at the 1st cycle of anti-PD-1 immunotherapy was associated with improvement of PFS and overall survival (P < 0.05). Conclusion: Activation of circulating Tex cells before or during therapy related to tumor burden may be associated with clinical efficacy of anti-PD-1 immune therapy in NSCLC.

3.
Cells ; 12(4)2023 02 20.
Article in English | MEDLINE | ID: mdl-36831336

ABSTRACT

Diallyl trisulfide (DAT) is a biologically active component of garlic essential oil and exhibits multi-targeted activity against many organisms. The current study tested the capacity of DAT to decrease the male fertility of Sitotroga cerealella. The effects on testis morphology, sperm number, motility, and lipid homeostasis were observed in adult males fumigated with DAT at a dose of 0.01 µL/L in air. The results indicated that the DAT significantly decreased the dimorphic sperm number. Meanwhile, the ultrastructural analysis of the sperm showed that the DAT caused malformed and aberrant structures of mitochondrial derivatives of dimorphic sperm. Additionally, the lipid homeostasis and ATP contents in the male adults were significantly decreased after treatment. Moreover, the total sperm motility was reduced, while the wave-propagation velocity, amplitude, frequency, and wavelength were significantly decreased compared with the controls. Overall, this study reported, for the first time, that DAT impairs energy metabolism, inhibits dimorphic spermatogenesis, and decreases sperm motility, while these abnormalities in sperm lead to adult-male infertility.


Subject(s)
Garlic , Moths , Oils, Volatile , Male , Animals , Garlic/chemistry , Sperm Motility , Seeds , Spermatogenesis , Antioxidants/pharmacology , Fertility , Homeostasis
4.
Transl Cancer Res ; 9(12): 7540-7549, 2020 Dec.
Article in English | MEDLINE | ID: mdl-35117354

ABSTRACT

BACKGROUND: Gastric cancer is a highly heterogeneous disease and its traditional histopathological classification is difficult to meet clinical needs. Oxaliplatin is an antitumor drug with high efficiency and low toxicity. Therefore, the insensitivity or secondary drug resistance of oxaliplatin to gastric cancer is vital for tumor progression. The aim of this study was to investigate the sensitivity of gastric cancer cells to oxaliplatin after ARID1A (AT-rich interactive domain1A gene) gene silencing. METHODS: MGC-803 and AGS cells were selected as gastric cancer cells for study. ARID1A protein and mRNA expression was detected by Western blot and quantitative reverse-transcription PCR (qRT-PCR). The short hairpin RNA (shRNA) fragment of ARID1A gene silencing was constructed and introduced into gastric cancer cells. The cell proliferation activity was calculated using CCK8 and the IC50 was calculated. The flow cytometry was used to detect the cell cycle and apoptosis rate. The ability of cell invasion was detected by transwell method. Cells were treated with different concentrations of oxaliplatin. RESULTS: The proliferation of gastric cancer cells was promoted by ARID1A gene silencing (P<0.01), the quantity of cells in S phase increased (P<0.05), and the invasive ability increased (P<0.05). After treatment with oxaliplatin at different concentrations, ARID1A gene silencing reduced the inhibition rate of oxaliplatin on gastric cancer cells and apoptosis rate (P<0.05), and increased IC 50 (P<0.01). CONCLUSIONS: ARID1A gene silencing, a factor promoting proliferation of gastric cancer cells, would reduce the sensitivity of gastric cancer cells to oxaliplatin, which can provide a basis for the exploration of targeted drugs for individualized treatment of gastric cancer.

5.
Nutr Metab (Lond) ; 16: 88, 2019.
Article in English | MEDLINE | ID: mdl-31889968

ABSTRACT

BACKGROUND: Obesity is a global epidemic disease that increases the risk of metabolic syndrome. However, therapeutic drugs for obesity are still scarce. In recent years, peptides have been identified as new biological regulators. RIFV (R-I-F-V-P-I-K-G-R-P-A-P), a novel active peptide from our peptide database. METHODS: We performed oil red O staining and triglyceride measurement to analyze the influence of RIFV on white preadipocytes differentiation. Then the effects of RIFV on cell proliferation, apoptosis and cell cycle were determined by using CCK-8 assay and flow cytometry. The mRNA and protein levels of adipogenesis-related genes were respectively detected by qRT-PCR and western blot. Rescue experiment was conducted to confirm whether RIFV could regulate adipocytes differentiation via targeting C/EBP-ß. Finally, the luciferase reporter gene assay was performed to verify the regulation of RIFV on C/EBP-ß gene. RESULTS: RIFV was revealed to inhibit the differentiation of human white adipocytes without affecting their proliferation. Additionally, RIFV could also suppress the differentiation of mouse primary white preadipocytes isolated from inguinal fat tissues. Furthermore, RIFV may have an inhibitory effect on adipogenesis by inhibiting the regulation of the adipogenic gene C/EBP-ß. CONCLUSIONS: Our results indicated that RIFV may be a novel essential regulator of adipocyte differentiation and represents a therapeutic strategy for obesity and related complications.

6.
Biochim Biophys Acta Gene Regul Mech ; 1861(2): 125-132, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29391195

ABSTRACT

The novel obesity-associated protein Phosphotyrosine Interaction Domain containing 1 (PID1) inhibits insulin-PI3K/Akt signaling pathway and insulin-stimulated glucose uptake in vitro. In this study, we generated fat tissue-specific aP2-PID1 transgenic (aP2-PID1tg) mice and PID1 knockout (PID1-/-) mice to explore how PID1 affects glucose metabolism in vivo. We observed insulin resistance and impaired insulin-PI3K/Akt signaling in aP2-PID1tg mice. Consistent with these data, the PID1-/- mice displayed improved glucose tolerance and insulin sensitivity under chow diet, with increased Akt phosphorylation in white adipose tissue (WAT). We further demonstrated that PID1 could interact with low density lipoprotein receptor-related protein 1 (LRP1) but not the insulin receptor (IR) in adipocytes, and its overexpression could lead to decreased GLUT4 level. Our results thus indentify PID1 as a critical regulator of glucose metabolism in adipocytes.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Glucose/metabolism , Homeostasis , 3T3-L1 Cells , Adipose Tissue, White/metabolism , Animals , Carrier Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Glucose Transporter Type 4/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism
7.
Chin J Cancer ; 36(1): 97, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273089

ABSTRACT

BACKGROUND: Metastatic colorectal cancer (mCRC) patients with progressive disease after all available standard therapies need new medication for further treatment. Famitinib is a small-molecule multikinase inhibitor, with promising anticancer activities. This multicenter, randomized, double-blinded, placebo-controlled, phase II clinical trial was designed to evaluate the safety and efficacy of famitinib in mCRC. METHODS: Famitinib or placebo was administered orally once daily. The primary endpoint was progression-free survival (PFS). Secondary endpoints included objective response rate (ORR), disease control rate (DCR), overall survival (OS), quality-of-life (QoL), and safety. RESULTS: Between July 18, 2012 and Jan 22, 2014, a total of 167 patients were screened, and 154 patients were randomized in a 2:1 ratio to receive either famitinib (n = 99) or placebo (n = 55). The median PFS was 2.8 and 1.5 months in the famitinib and placebo groups (hazard ratio = 0.60, 95% confidence interval = 0.41-0.86, P = 0.004). The DCR was 59.8% and 31.4% (P = 0.002) and the ORR was 2.2% and 0.0% (P = 0.540) in the famitinib and placebo groups, respectively. The most frequent grade 3-4 adverse events were hypertension (11.1%), hand-foot syndrome (10.1%), thrombocytopenia (10.1%), and neutropenia (9.1%). Serious adverse events occurred in 11 (11.1%) patients in the famitinib group and 5 (9.1%) in the placebo group (P = 0.788). The median OS of the famitinib and placebo groups was 7.4 and 7.2 months (P = 0.657). CONCLUSION: Famitinib prolonged PFS in refractory mCRC patients with acceptable tolerability. Trial registration This study was registered on ClinicalTrials.gov (NCT01762293) and was orally presented in the 2015 ASCO-Gastrointestinal Symposium.


Subject(s)
Colorectal Neoplasms/drug therapy , Indoles/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrroles/administration & dosage , Administration, Oral , Adult , Aged , Double-Blind Method , Drug Administration Schedule , Female , Humans , Indoles/adverse effects , Male , Neoplasm Metastasis , Protein Kinase Inhibitors/adverse effects , Pyrroles/adverse effects , Survival Analysis , Treatment Outcome
8.
Int J Clin Exp Med ; 8(5): 7728-36, 2015.
Article in English | MEDLINE | ID: mdl-26221323

ABSTRACT

BACKGROUND AND AIMS: It remains a challenge to prevent the local recurrence or distant metastasis of gastric carcinoma after D2 gastrectomy. Cytokine-induced killer (CIK) cells have shown promising activity against solid tumors in vitro and in vivo. We investigated the effect of adjuvant chemotherapy combined with autologous CIK therapy after D2 gastrectomy compared with adjuvant chemotherapy alone after D2 gastrectomy in patients with stage II-III gastric cancer. METHODS: From January 2009 to December 2011, 226 patients with stage II-III gastric cancer who had had curative D2 gastrectomy were enrolled. Eighty-nine patients (CIK group) received adjuvant chemotherapy combining with autologous CIK therapy and 137 patients (control group) received adjuvant chemotherapy alone. Disease-free survival (DFS) and overall survival (OS) were evaluated. RESULTS: Patients in the CIK group had longer DFS and OS than patients in the control group (DFS 41.0 months vs. 32.0 months, OS 45.0 months vs. 44.0 months, by log-rank test P = 0.006 and P = 0.028, respectively). In subgroup analysis, no significant differences in DFS and OS were observed between the two groups for the patients with stages II disease. Patients with stage III disease in the CIK group had longer median DFS and OS than patients in the control group (P = 0.031 and P = 0.038, respectively). In multivariate analysis, the stage and the interaction of stage and CIK therapy were independent prognostic factors for DFS and OS. CONCLUSIONS: The data suggest that adjuvant chemotherapy combined with autologous CIK therapy can improve prognosis for gastric carcinoma patients after D2 gastrectomy, especially for the patients with stage III disease.

9.
Mol Genet Genomics ; 290(5): 1659-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25773316

ABSTRACT

Brown adipose tissue (BAT) functions to dissipate energy in response to cold exposure or overfeeding. Counteracting obesity has been extensively considered as a promising target. Long noncoding RNAs (lncRNAs) are an important class of pervasive genes involved in a variety of biological functions. However, the potential biological functions of lncRNAs during mouse brown fat cell differentiation have not been fully understood. Here, we performed lncRNA and mRNA expression profile analysis using microarray technology and identified 1064 lncRNAs with differential expression (fold change| ≥4, p ≤ 0.01) on day 0 and day 8 during differentiation. Furthermore, candidate lncRNAs were characterized by comprehensive examination of their genomic context, gene ontology (GO) enrichment of their associated protein-coding genes and pathway analysis. We identified three lncRNAs (Gm15051, Tmem189 and Cebpd) associated with their flanking coding genes (Hoxa1, C/EBPß and C/EBPδ), which participated in adipose commitment. Collectively, our findings indicated lncRNAs are involved in mouse BAT development and provide potential targets for obesity therapy.


Subject(s)
Adipocytes/cytology , Adipose Tissue, Brown/cytology , Cell Differentiation/genetics , RNA, Long Noncoding/physiology , Transcriptome , Animals , Gene Expression Profiling , Mice , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
10.
Mol Biol Rep ; 42(5): 927-35, 2015 May.
Article in English | MEDLINE | ID: mdl-25421647

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs involved in the regulation of gene expression. MiR-1908 is a recently identified miRNA that is highly expressed in human adipocytes. However, it is not known what role of miR-1908 is involved in the regulation of human adipocytes. In this study, we demonstrate that the level of miR-1908 increases during the adipogenesis of human multipotent adipose-derived stem (hMADS) cells and human preadipocytes-visceral. Overexpression of miR-1908 in hMADS cells inhibited adipogenic differentiation and increased cell proliferation, suggesting that miR-1908 is involved in the regulation of adipocyte cell differentiation and metabolism, and, thus, may have an effect on human obesity.


Subject(s)
Adipocytes/physiology , Adipogenesis/physiology , MicroRNAs/physiology , Adipogenesis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation , Humans , MicroRNAs/genetics
11.
Mol Med Rep ; 10(2): 761-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920049

ABSTRACT

Murine P19 embryonal carcinoma cells are multipotent cells that can differentiate into cardiomyocytes when treated with dimethyl sulfoxide. This experimental model provides an invaluable tool to study different aspects of cardiac differentiation, such as the function of cardiac­specific transcription factors and signaling pathways, and the regulation of contractile protein expression. The role of mitochondria during cardiac differentiation is unclear. In this context, we have examined the mitochondrial-related changes in undifferentiated and differentiated P19 cells. We observed that mitochondrial DNA content sharply decreased in P19 cell aggregates compared to undifferentiated cells, accompanied by decreased levels of adenosine triphosphate (ATP) and reactive oxygen species (ROS). Following the aggregation stage, the mitochondrial DNA content reached its highest level on day 7 of the differentiation process, with the intracellular ROS level showing a trend to increase, similar to cellular ATP production. In conclusion, our study on differentiating P19 embryonal carcinoma cells provides new insights into the role of mitochondria in the differentiation of P19 stem cells into beating cardiomyocytes.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Adenosine Triphosphate/metabolism , Animals , Cell Differentiation/drug effects , DNA, Mitochondrial/metabolism , Dimethyl Sulfoxide/pharmacology , Mice , Mitochondria/genetics , Mitochondria/ultrastructure , Reactive Oxygen Species/metabolism
12.
Mol Cell Endocrinol ; 393(1-2): 65-74, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24931160

ABSTRACT

Visceral obesity is an independent risk factor for metabolic syndrome, and abnormal fat accumulation is linked to increases in the number and size of adipocytes. MiR-146b was a miRNA highly expressed in mature adipocytes while very lowly expressed in human mesenchymal stem cells (hMSCs) and human visceral preadipocytes (vHPA). In this paper, we mainly focused on the roles of miR-146b in adipogenesis. We found miR-146b could inhibit the proliferation of visceral preadipocytes and promote their differentiation. MiR-146b in human visceral adipocytes inhibited the expression of KLF7, a member of the Kruppel-like transcription factors, as demonstrated by a firefly luciferase reporter assay, indicating that KLF7 is a direct target of the endogenous miR-146b. MiR-146b expression was significantly altered in visceral and subcutaneous adipose tissues in human overweight and obese subjects, and in the epididymal fat tissues and brown fat tissues of diet-induced obese mice. Our data indicates that miR-146b may be a new therapeutic target against human visceral obesity and metabolic dysfunction.


Subject(s)
Adipocytes/pathology , Adipogenesis/genetics , Cell Differentiation , Gene Expression Regulation , MicroRNAs/metabolism , Obesity/genetics , Animals , Blotting, Western , Cell Cycle/genetics , Cell Proliferation , Humans , Mice , Mice, Obese , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction
13.
Cell Biochem Biophys ; 70(1): 667-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24771405

ABSTRACT

LYR motif-containing 1 (LYRM1) was recently discovered to be involved in adipose tissue homeostasis and obesity-associated insulin resistance. We previously demonstrated that LYRM1 overexpression might contribute to insulin resistance and mitochondrial dysfunction. Additionally, knockdown of LYRM1 enhanced insulin sensitivity and mitochondrial function in 3T3-L1 adipocytes. We investigated whether knockdown of LYRM1 in 3T3-L1 adipocytes could rescue insulin resistance and mitochondrial dysfunction induced by the cyanide p-trifluoromethoxyphenyl-hydrazone (FCCP), a mitochondrion uncoupler, to further ascertain the mechanism by which LYRM1 is involved in obesity-associated insulin resistance. Incubation of 3T3-L1 adipocytes with 1 µM FCCP for 12 h decreased insulin-stimulated glucose uptake, reduced intracellular ATP synthesis, increased intracellular reactive oxygen species (ROS) production, impaired insulin-stimulated Glucose transporter type 4 (GLUT4) translocation, and diminished insulin-stimulated tyrosine phosphorylation of Insulin receptor substrate-1 (IRS-1) and serine phosphorylation of Protein Kinase B (Akt). Knockdown of LYRM1 restored insulin-stimulated glucose uptake, rescued intracellular ATP synthesis, reduced intracellular ROS production, restored insulin-stimulated GLUT4 translocation, and rescued insulin-stimulated tyrosine phosphorylation of IRS-1 and serine phosphorylation of Akt in FCCP-treated 3T3-L1 adipocytes. This study indicates that FCCP-induced mitochondrial dysfunction and insulin resistance are ameliorated by knockdown of LYRM1.


Subject(s)
Adipocytes/cytology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Gene Knockdown Techniques , Insulin Resistance/genetics , Mitochondria/drug effects , 3T3-L1 Cells , Adenosine Triphosphate/biosynthesis , Animals , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proton Ionophores/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
14.
Cell Biochem Biophys ; 70(2): 771-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24771406

ABSTRACT

Obesity has become a global public health problem associated with complications including type 2 diabetes, cardiovascular disease, and several cancers. Adipocyte differentiation (adipogenesis) plays an important role in obesity and energy homeostasis. Adipose tissue secretes multiple cytokines and adipokines which can cause the complications of obesity, especially insulin resistance. TNF-α, IL-6, leptin, and resistin have been identified as the main regulators of obesity and insulin activity. miR-378 is highly induced during adipogenesis and has been reported to be positively regulated in adipogenesis. In the current study, matured human adipocytes were treated with TNF-α, IL-6, leptin, or resistin on the 15th day after the induction of human pre-adipocyte differentiation. We demonstrated that TNF-α, IL-6, and leptin upregulated miR-378 expression indicating that miR-378 probably is a novel mediator in the development of insulin resistance related to obesity.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Interleukin-6/pharmacology , Leptin/pharmacology , MicroRNAs/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Adipocytes/cytology , Adipocytes/metabolism , Humans
15.
Cell Biochem Biophys ; 68(2): 283-90, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23801157

ABSTRACT

During the development of obesity, adipose tissue releases a host of different adipokines and inflammatory cytokines, such as leptin, resistin, tumor necrosis factor α (TNF-α), Interleukin-6 (IL-6), and adiponectin, which mediate insulin resistance. Recently, some microRNAs (miRNAs) regulated by adiponectin were identified as novel targets for controlling adipose tissue inflammation. Therefore, the relationship between adipokines and miRNA is worth studying. MiR-335 is an adipogenesis-related miRNA and implicated in both fatty acid metabolism and lipogenesis. In this study, we focused on the association of miR-335 and adipokines, and examined the expression trend of miR-335 during human adipocyte differentiation. Our results showed that miR-335 is significantly upregulated with treatment of leptin, resistin, TNF-α, and IL-6 in human mature adipocytes, and its expression elevated in the process of adipocyte differentiation. Interestingly, the transcriptional regulation of miR-335 by these adipokines seems independent of its host gene (mesoderm-specific transcript homolog, MEST). Thus, we cloned and identified potential promoter of miR-335 within the intron of MEST. As a result, a fragment about 600-bp length upstream sequences of miR-335 had apparent transcription activity. These findings indicated a novel role for miR-335 in adipose tissue inflammation, and miR-335 might play an important role in the process of obesity complications via its own transcription mechanism.


Subject(s)
Adipose Tissue/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , Adipogenesis , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Cells, Cultured , HEK293 Cells , Humans , Interleukin-6/pharmacology , Leptin/pharmacology , Obesity/metabolism , Obesity/pathology , Promoter Regions, Genetic , Resistin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
16.
Cell Physiol Biochem ; 34(6): 1983-97, 2014.
Article in English | MEDLINE | ID: mdl-25562147

ABSTRACT

BACKGROUND/AIM: Emerging evidence suggests that microRNA (miRNA) mediated gene regulation influences the maintenance of metabolic homeostasis, particularly the states of obesity and insulin resistance, thereby providing a potential link between miRNAs and nonalcoholic fatty liver disease (NAFLD). METHODS: Sprague-Dawley rats fed a high-fat diet (HFD) were used to establish a rat model of NAFLD. The miRNA expression profile of liver tissues was evaluated using Illumina HiSeq deep sequencing. Selected miRNAs were then validated by real-time PCR at both 4- and 12-week time points. Furthermore, the expression levels of these miRNAs were assessed in HepG2 cells and human hepatocytes treated with free fatty acids (FFAs) and proinflammatory factors (tumour necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). RESULTS: Our results showed that consumption of a HFD for 4 weeks caused simple steatosis, which progressed to steatohepatitis at 12 weeks. miRNA deep sequencing analysis identified 44 known up-regulated miRNAs (fold change >1.5) and 12 down-regulated miRNAs (fold change <0.5). Among the abnormally expressed miRNAs, miR-200a, miR-200b, miR-200c, miR-146a, miR-146b and miR-152 were up-regulated both in vitro and vivo. Interestingly, the expression levels of these six miRNAs were increased in HepG2 cells and human hepatocytes after treatment with FFAs and proinflammatory factors. CONCLUSION: These findings suggest a critical role for miRNAs in the pathogenesis of NAFLD.


Subject(s)
Gene Expression Regulation , MicroRNAs/biosynthesis , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Diet, High-Fat , Fatty Acids, Nonesterified/administration & dosage , Hep G2 Cells , Hepatocytes/drug effects , High-Throughput Nucleotide Sequencing , Humans , Insulin Resistance/genetics , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/pathology , Rats
17.
Mol Biol Rep ; 40(11): 6469-76, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24052233

ABSTRACT

Our data in the previous report demonstrated that C10orf116 (AFRO) is an adipocyte lineage-specific nuclear factor that can modulate the master adipogenesis transcription factors early during differentiation. However, more precise functional properties of this gene need to be clarified and await further investigation. Therefore, in this study, we performed an expression profile of cellular MicroRNAs (miRNAs) in the C10orf116 overexpression 3T3-L1 adipocytes and performed target prediction and functional enrichment of the differentially expressed miRNAs. Our study identified 34 miRNAs up-regulated in the 3T3-L1 adipocytes stably overexpressing C10orf116, whereas 43 miRNAs up-regulated in the control cells. The target genes of differentially expressed miRNAs were found to be involved in multiple signalling pathways, such as Wnt, TGF-beta, MAPK, Jak-STAT, insulin signalling pathway, et al. Our data provided novel information for the identification of C10orf116.


Subject(s)
Adipocytes/metabolism , Gene Expression , MicroRNAs/genetics , Nuclear Proteins/genetics , Transcriptome , 3T3-L1 Cells , Adipogenesis/genetics , Animals , Gene Expression Regulation , Lipid Metabolism/genetics , Mice , Reproducibility of Results , Transfection
18.
Mol Med Rep ; 8(3): 806-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23846528

ABSTRACT

Fatty acid binding protein 3 (FABP3, also termed heart-type fatty acid binding protein) is a member of the intracellular lipid-binding protein family that may be essential in fatty acid transport, cell growth, cellular signaling and gene transcription. Previously, we demonstrated that FABP3 was involved in apoptosis-associated congenital cardiac malformations; however, its mechanism of regulation remains unclear. Apoptosis has increasingly been considered to be important in cardiac development. In the present study, a zebrafish model was used to investigate the involvement of FABP3­morpholino (MO)-induced apoptosis and mitochondrial dysfunction in cardiac development. During the early stages of cardiac development, injection of FABP3­MO into zebrafish resulted in significant impairment in cardiac development and promoted the rate of apoptosis which was correlated with significant dysfunction of the mitochondria. For example, the ATP content was markedly decreased at 24 and 48 h post-fertilization (pf), reactive oxygen species production was significantly enhanced at 24 and 48 h pf and the mitochondrial DNA copy number was reduced at 24, 48 and 72 h pf. Additionally, Nkx2.5 expression was upregulated in FABP3-MO zebrafish, and Wnt signaling molecules (Wnt1, Wnt5 and Wnt11) were also highly expressed in FABP3-MO zebrafish at 24, 48 and 72 h pf. In conclusion, the results indicated that FABP3 knockdown exhibited significant toxic effects on cardiac development and mitochondrial function, which may be responsible for the knockdown of FABP3-induced apoptosis. Apoptosis was one of the mechanisms underlying this effect, and was correlated with the activation of Wnt signaling. These studies identified FABP3 as a candidate gene underlying the etiology of congenital heart defects.


Subject(s)
Apoptosis/drug effects , Fatty Acid-Binding Proteins/antagonists & inhibitors , Mitochondria/metabolism , Morpholinos/toxicity , Zebrafish Proteins/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , DNA Copy Number Variations/drug effects , DNA, Mitochondrial/metabolism , Embryo, Nonmammalian/metabolism , Fatty Acid Binding Protein 3 , Fatty Acid-Binding Proteins/metabolism , Heart/drug effects , Heart/growth & development , Homeobox Protein Nkx-2.5 , Larva/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Morpholinos/chemistry , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/metabolism
19.
Mol Med Rep ; 7(5): 1477-81, 2013 May.
Article in English | MEDLINE | ID: mdl-23467766

ABSTRACT

Data from our previous study demonstrated that C10orf116 is an adipocyte lineage-specific nuclear factor, which regulates master adipogenesis transcription factors during early differentiation. However, the precise functional properties of this gene have yet to be identified and further investigation is required. In the present study, we report the effects of C10orf116 expression on cell proliferation and apoptosis in vitro and observed that the overexpression of C10orf116 stimulates proliferation and inhibits apoptosis in preadipocytes. Furthermore, we investigated the effects of C10orf116 on glucose uptake and demonstrated that the ectopic expression of C10orf116 significantly increases insulin-stimulated glucose uptake in adipocytes by increasing glucose transporter type 4 (GLUT4) expression levels. Collectively, these data further support the hypothesis that C10orf116 is important in regulating glucose transport in adipocytes as well as the number of preadipocytes. The results of the present study may also provide insights into the complex mechanisms involved in the development of obesity.


Subject(s)
Apoptosis , Glucose/metabolism , Nuclear Proteins/metabolism , 3T3-L1 Cells , Animals , Apoptosis/drug effects , Benzimidazoles/metabolism , Biological Transport/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Mice
20.
Cell Biochem Biophys ; 66(3): 489-97, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23274913

ABSTRACT

NYGGF4, also known as phosphotyrosine interaction domain containing 1(PID1), is a recently discovered gene which is involved in obesity-related insulin resistance (IR) and mitochondrial dysfunction. We aimed to further elucidate the effects and mechanisms underlying NYGGF4-induced IR by investigating the effect of overexpressing mitochondrial transcription factor A (TFAM), which is essential for mitochondrial DNA transcription and replication, on NYGGF4-induced IR and mitochondrial abnormalities in 3T3-L1 adipocytes. Overexpression of TFAM increased the mitochondrial copy number and ATP content in both control 3T3-L1 adipocytes and NYGGF4-overexpressing adipocytes. Reactive oxygen species (ROS) production was enhanced in NYGGF4-overexpressing adipocytes and reduced in TFAM-overexpressing adipocytes; co-overexpression of TFAM significantly attenuated ROS production in NYGGF4-overexpressing adipocytes. However, overexpression of TFAM did not affect the mitochondrial transmembrane potential (ΔΨm) in control 3T3-L1 adipocytes or NYGGF4-overexpressing adipocytes. In addition, co-overexpression of TFAM-enhanced insulin-stimulated glucose uptake by increasing Glucose transporter type 4 (GLUT4) translocation to the PM in NYGGF4-overexpressing adipocytes. Overexpression of NYGGF4 significantly inhibited tyrosine phosphorylation of Insulin receptor substrate 1 (IRS-1) and serine phosphorylation of Akt, whereas overexpression of TFAM strongly induced phosphorylation of IRS-1 and Akt in NYGGF4-overexpressing adipocytes. This study demonstrates that NYGGF4 plays a role in IR by impairing mitochondrial function, and that overexpression of TFAM can restore mitochondrial function to normal levels in NYGGF4-overexpressing adipocytes via activation of the IRS-1/PI3K/Akt signaling pathway.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Insulin Resistance , Mitochondria/pathology , Mitochondrial Proteins/genetics , Transcription Factors/genetics , 3T3-L1 Cells , Adenosine Triphosphate/metabolism , Adipocytes/drug effects , Animals , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Insulin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Size/drug effects , Phosphorylation/drug effects , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...