Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Drug Des Devel Ther ; 18: 1349-1368, 2024.
Article in English | MEDLINE | ID: mdl-38681208

ABSTRACT

Background: Sepsis is recognized as a multiorgan and systemic damage caused by dysregulated host response to infection. Its acute systemic inflammatory response highly resembles that of lipopolysaccharide (LPS)-induced endotoxemia. Propofol and dexmedetomidine are two commonly used sedatives for mechanical ventilation in critically ill patients and have been reported to alleviate cognitive impairment in many diseases. In this study, we aimed to explore and compare the effects of propofol and dexmedetomidine on the encephalopathy induced by endotoxemia and to investigate whether ferroptosis is involved, finally providing experimental evidence for multi-drug combination in septic sedation. Methods: A total of 218 C57BL/6J male mice (20-25 g, 6-8 weeks) were used. Morris water maze (MWM) tests were performed to evaluate whether propofol and dexmedetomidine attenuated LPS-induced cognitive deficits. Brain injury was evaluated using Nissl and Fluoro-Jade C (FJC) staining. Neuroinflammation was assessed by dihydroethidium (DHE) and DCFH-DA staining and by measuring the levels of three cytokines. The number of Iba1+ and GFAP+ cells was used to detect the activation of microglia and astrocytes. To explore the involvement of ferroptosis, the levels of ptgs2 and chac1; the content of iron, malondialdehyde (MDA), and glutathione (GSH); and the expression of ferroptosis-related proteins were investigated. Conclusion: The single use of propofol and dexmedetomidine mitigated LPS-induced cognitive impairment, while the combination showed poor performance. In alleviating endotoxemic neural loss and degeneration, the united sedative group exhibited the most potent capability. Both propofol and dexmedetomidine inhibited neuroinflammation, while propofol's effect was slightly weaker. All sedative groups reduced the neural apoptosis, inhibited the activation of microglia and astrocytes, and relieved neurologic ferroptosis. The combined group was most prominent in combating genetic and biochemical alterations of ferroptosis. Fpn1 may be at the core of endotoxemia-related ferroptosis activation.


Subject(s)
Dexmedetomidine , Endotoxemia , Ferroptosis , Lipopolysaccharides , Mice, Inbred C57BL , Propofol , Dexmedetomidine/pharmacology , Animals , Propofol/pharmacology , Ferroptosis/drug effects , Mice , Male , Endotoxemia/drug therapy , Endotoxemia/metabolism , Endotoxemia/chemically induced , Lipopolysaccharides/pharmacology , Dose-Response Relationship, Drug , Brain Diseases/drug therapy , Brain Diseases/metabolism , Brain Diseases/pathology , Hypnotics and Sedatives/pharmacology
2.
Nano Lett ; 24(4): 1415-1422, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38232178

ABSTRACT

Charge and spin are two intrinsic attributes of carriers governing almost all of the physical processes and operation principles in materials. Here, we demonstrate the manipulation of electronic and spin states in designed Co-quantum dot/WS2 (Co-QDs/WS2) heterostructures by employing a metal-dielectric composite substrate and via scanning tunneling microscope. By repeatedly scanning under a unipolar bias, switching the bias polarity, or applying a pulse through nonmagnetic or magnetic tips, the Co-QDs morphologies exhibit a regular and reproducible transformation between bright and dark dots. First-principles calculations reveal that these tunable characters are attributed to the variation of density of states and the transition of magnetic anisotropy energy induced by carrier accumulation. It also suggests that the metal-dielectric composite substrate is successful in creating the interfacial potential for carrier accumulation and realizes the electrically controllable modulations. These results will promote the exploration of electron-matter interactions in quantum systems and provide an innovative way to facilitate the development of spintronics.

3.
BMC Anesthesiol ; 23(1): 299, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37670239

ABSTRACT

BACKGROUND: Postoperative cognitive dysfunction (POCD) has been reported as a significant complication in elderly patients. Various methods have been proposed for reducing the incidence and severity of POCD. Intravenous lidocaine administration has been reported in the literature to reduce POCD, but the effect of lidocaine remains controversial. METHODS: We screened Medline, Embase, Cochrane Library, and China National Knowledge Infrastructure (up to April 2022) databases following a search strategy for intravenous lidocaine on POCD. We also screened related bibliographies on lidocaine for POCD. Ten articles comprising 1517 patients were selected and analyzed. We divided the postoperative follow-up period as follows: short term (<30 days), medium term (30-90 days), and long term (>90 days). OUTCOMES: We found that lidocaine could attenuate the overall incidence of POCD, especially in the short term. There were no differences between lidocaine and placebo on the overall severity of POCD. CONCLUSION: Lidocaine administered intravenously could attenuate the overall incidence of POCD and its severity in the short term.


Subject(s)
Postoperative Cognitive Complications , Aged , Humans , Administration, Intravenous , China , Databases, Factual , Lidocaine
5.
J Bioenerg Biomembr ; 55(2): 103-114, 2023 04.
Article in English | MEDLINE | ID: mdl-37046136

ABSTRACT

Endothelial dysfunction is a key early link in the pathogenesis of atherosclerosis, and the accumulation of senescent vascular endothelial cells causes endothelial dysfunction. Phosphoenolpyruvate (PEP), which is a high-energy glycolytic intermediate, protects against ischemia-reperfusion injury in isolated rat lung, heart, and liver tissue by quickly providing ATP. However, it was reported that serum PEP concentrations are 13-fold higher in healthy elderly compare to the young. Unlike that of other cell types, the energy required for the physiological function of endothelial cells is mainly derived from glycolysis. Recently, it is unclear whether circulating accumulation of PEP affects endothelial cell function. In this study, we found for the first time that 50-250 µM of PEP significantly promoted THP-1 monocyte adhesion to human umbilical vein endothelial cells (HUVECs) through increased expression of vascular endothelial adhesion factor 1 (VCAM1) and intercellular adhesion factor 1 (ICAM1) in HUVECs. Meanwhile, 50-250 µM of PEP decreased the expression of endothelial nitric oxide synthase (eNOS) and cellular level of nitric oxide (NO) in HUVECs. Moreover, PEP increased levels of ROS, enhanced the numbers of SA-ß-Gal-positive cells and upregulated the expression of cell cycle inhibitors such as p21, p16 and the phosphorylation level of p53 on Ser15, and the expression of proinflammatory factors including TNF-α, IL-1ß, IL-6, IL-8, IL-18 and MCP-1 in HUVECs. Furthermore, PEP increased both oxygen consumption rate (OCR) and glycolysis rate, and was accompanied by reduced NAD+/NADH ratios and enhanced phosphorylation levels of AMPKα (Thr172), p38 MAPK (T180/Y182) and NF-κB p65 (Ser536) in HUVECs. Notably, PEP had no significant effect on hepG2 cells. In conclusion, these results demonstrated that PEP induced dysfunction and senescence in vascular endothelial cells through stimulation of metabolic reprogramming.


Subject(s)
Cellular Senescence , Signal Transduction , Rats , Animals , Humans , Aged , Cells, Cultured , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology
6.
Eur J Pharmacol ; 944: 175584, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36781043

ABSTRACT

Apigenin is a natural flavonoid which is widely found in vegetables and fruits. However, the mechanism of apigenin in oxidative stress-induced myocardial injury has not been fully elucidated. We established an isoproterenol (Iso)-induced myocardial injury mouse model and a hypoxia/reoxygenation (H/R)-induced H9c2 cell injury model, followed by pretreatment with apigenin to explore its protective effects. Apigenin can significantly alleviate isoproterenol-induced oxidative stress, cell apoptosis and myocardial remodeling in vivo. Apigenin pretreatment can also significantly improve cardiomyocyte morphology, decrease H/R induced oxidative stress, and attenuate cell apoptosis and inflammation in vitro. Further mechanism study revealed that apigenin treatment reversed isoprenaline and H/R-induced decrease of Sirtuin1 (SIRT1). Molecular docking results proved that apigenin can form hydrogen bond with 230 Glu, a key site of SIRT1 activation, indicating that apigenin is an agonist of SIRT1. Moreover, SIRT1 knockdown by siRNA significantly reversed the protective effect of apigenin in H/R-induced myocardial injury. In conclusion, apigenin protects cardiomyocyte function from oxidative stress-induced myocardial injury by modulating SIRT1 signaling pathway, which provides a new potential therapeutic natural compound for the clinical treatment of cardiovascular diseases.


Subject(s)
Apigenin , Sirtuin 1 , Animals , Mice , Apigenin/pharmacology , Apoptosis , Hypoxia/metabolism , Isoproterenol/pharmacology , Molecular Docking Simulation , Myocytes, Cardiac , Oxidative Stress , Signal Transduction , Sirtuin 1/metabolism
7.
J Pharm Pharmacol ; 75(2): 253-263, 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36179123

ABSTRACT

OBJECTIVES: To investigate the function and regulatory mechanisms of delphinidin in the treatment of hepatocellular carcinoma. METHODS: HepG2 and HuH-7 cells were treated with different concentrations of delphinidin. Cell viability was analysed by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. The cell autophagy and autophagic flux were analysed by LC3b-green fluorescent protein (GFP)-Adv and LC3b-GFP-monomeric red fluorescent protein-Adv transfected HepG2 and HuH-7 cells, respectively. Cell apoptosis was analysed by Hoechst33342 staining, terminal deoxynucleotidyl transferase dUTP nick end labeling staining and DNA laddering. Cell autophagy, apoptosis and survival related protein expressions were detected by Western blotting. KEY FINDINGS: After treatment with different concentrations of delphinidin, the cell survival rate was significantly decreased. Delphinidin could block the autophagic flux, resulting in a significant increase in autophagosomes, and led to an increase in cell apoptosis. The combined application of delphinidin and cisplatin could promote the antitumour effect and reduce the dose of cisplatin in tumour cells. Further mechanism studies reveal that delphinidin could inhibit the multidrug resistance gene 1 (MDR1) and the tumour-promoting transcription cofactor DEAD-box helicase 17 (DDX17) expression in tumour cells. Overexpression of DDX17 could reverse delphinidin's antitumor function in tumour cells. CONCLUSIONS: Delphinidin has a strong anti-tumour effect by inducing tumour cell autophagic flux blockage and apoptosis by inhibiting of both MDR1 and DDX17 expression.


Subject(s)
Cisplatin , Liver Neoplasms , Humans , Cisplatin/pharmacology , Genes, MDR , Apoptosis , Autophagy , Cell Line, Tumor , DEAD-box RNA Helicases/pharmacology
8.
Signal Transduct Target Ther ; 7(1): 391, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36522308

ABSTRACT

Aging is a gradual and irreversible pathophysiological process. It presents with declines in tissue and cell functions and significant increases in the risks of various aging-related diseases, including neurodegenerative diseases, cardiovascular diseases, metabolic diseases, musculoskeletal diseases, and immune system diseases. Although the development of modern medicine has promoted human health and greatly extended life expectancy, with the aging of society, a variety of chronic diseases have gradually become the most important causes of disability and death in elderly individuals. Current research on aging focuses on elucidating how various endogenous and exogenous stresses (such as genomic instability, telomere dysfunction, epigenetic alterations, loss of proteostasis, compromise of autophagy, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, deregulated nutrient sensing) participate in the regulation of aging. Furthermore, thorough research on the pathogenesis of aging to identify interventions that promote health and longevity (such as caloric restriction, microbiota transplantation, and nutritional intervention) and clinical treatment methods for aging-related diseases (depletion of senescent cells, stem cell therapy, antioxidative and anti-inflammatory treatments, and hormone replacement therapy) could decrease the incidence and development of aging-related diseases and in turn promote healthy aging and longevity.


Subject(s)
Health Promotion , Neurodegenerative Diseases , Humans , Aged , Aging/metabolism , Cellular Senescence/genetics , Genomic Instability , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy
9.
Eur J Radiol ; 157: 110566, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36274361

ABSTRACT

OBJECTIVES: To explore the ability of conventional MRI and histogram analysis of apparent diffusion coefficient (ADC) for differentiating between malignant and benign palatal lesions. MATERIALS AND METHODS: A retrospective analysis was performed on MRI images of 86 patients with palatal lesions confirmed by histopathology between January 2015 and December 2018. Each lesion was evaluated based on the conventional MRI characteristics, including size, location, morphology, inner texture, enhancement, capsule, bone destruction, and nerve invasion. ADC histogram analysis was fitted to a bimodal Gaussian distribution mixture curve. Nine histogram analysis parameters were extracted from ADC maps and were compared between malignant and benign palatal lesions. Statistical analysis was performed to assess the differential performance of each parameter individually and combined. RESULTS: On conventional MRI, the capsule structure and nerve invasion were useful characteristics for the differential diagnosis of palatal lesions. Histogram analysis showed significant differences between the groups in terms of mean ADC values of the lower curve and overall curve of the bimodal histogram, as well as ADC10 and ADC50. The optimal diagnostic threshold of ADC value was ADC50 = 1.17 × 10-3 mm2/s (area under curve [AUC] = 0.934, sensitivity = 93.0 %, specificity = 86.2 %). The combination of conventional MRI and ADC values yielded the best predictive performance, with an AUC of 0.949, and an increase in sensitivity from 80.7 % to 96.5 % and specificity from 58.6 % to 82.8 %, compared to conventional MRI. CONCLUSIONS: Histogram analysis of DWI combined with conventional MRI allows accurate differentiation between benign and malignant palatal lesions.


Subject(s)
Diffusion Magnetic Resonance Imaging , Magnetic Resonance Imaging , Humans , Retrospective Studies , Diffusion Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/methods , Area Under Curve , Diagnosis, Differential , Palate , Sensitivity and Specificity , ROC Curve
10.
Front Oncol ; 12: 943032, 2022.
Article in English | MEDLINE | ID: mdl-35992805

ABSTRACT

DEAD-box (DDX)5 and DDX17, which belong to the DEAD-box RNA helicase family, are nuclear and cytoplasmic shuttle proteins. These proteins are expressed in most tissues and cells and participate in the regulation of normal physiological functions; their abnormal expression is closely related to tumorigenesis and tumor progression. DDX5/DDX17 participate in almost all processes of RNA metabolism, such as the alternative splicing of mRNA, biogenesis of microRNAs (miRNAs) and ribosomes, degradation of mRNA, interaction with long noncoding RNAs (lncRNAs) and coregulation of transcriptional activity. Moreover, different posttranslational modifications, such as phosphorylation, acetylation, ubiquitination, and sumoylation, endow DDX5/DDX17 with different functions in tumorigenesis and tumor progression. Indeed, DDX5 and DDX17 also interact with multiple key tumor-promoting molecules and participate in tumorigenesis and tumor progression signaling pathways. When DDX5/DDX17 expression or their posttranslational modification is dysregulated, the normal cellular signaling network collapses, leading to many pathological states, including tumorigenesis and tumor development. This review mainly discusses the molecular structure features and biological functions of DDX5/DDX17 and their effects on tumorigenesis and tumor progression, as well as their potential clinical application for tumor treatment.

11.
Int J Biol Sci ; 18(8): 3107-3121, 2022.
Article in English | MEDLINE | ID: mdl-35637957

ABSTRACT

Glucocorticoids are essential participants in the regulation of lipid metabolism. On a tissue-specific level, glucocorticoid signal is controlled by 11ß-Hydroxysteroid dehydrogenase 1 (11ß-HSD1). Up-regulation of 11ß-HSD1 expression during non-alcoholic fatty liver disease (NAFLD) has been previously shown, while 11ß-HSD1 inhibition has been shown to reduce hepatic lipids in NAFLD, but the underlying mechanisms remain unclear. Here, in this study, we created in vitro cell culture and in vivo transgenic hepatocyte-specific 11ß-HSD1 mouse models of NAFLD to determine the regulatory mechanisms of 11ß-HSD1 during lipid metabolism dysfunction. We found that 11ß-HSD1 overexpression activated glucocorticoid receptors and promoted their nuclear translocation, and then stimulating gp78. The induction of gp78 sharply reduced expression of Insig2, but not Insig1, which led to up-regulation of lipogenesis regulatory proteins including SREBP1, FAS, SCD1, and ACC1. Our results suggested that overexpression of 11ß-HSD1 induced lipid accumulation, at least partially through the GR/gp78/Insig2/SREBP1 pathway, which may serve as a potential diagnostic and therapeutic target for treatment of NAFLD.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Non-alcoholic Fatty Liver Disease , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Glucocorticoids , Humans , Lipids , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/genetics
12.
Biochem Biophys Res Commun ; 612: 169-175, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35533489

ABSTRACT

Hepatic lipid accumulation is an initiation factor in fatty liver disease, and promoting a reduction in hepatic lipid accumulation is an important treatment strategy. DEAD box RNA helicase 17 (DDX17) is a member of the DEAD-box family and a molecular chaperone. Previous studies have demonstrated that DDX17 is a transcriptional coregulator of tumorigenesis, inflammation, and macrophage cholesterol efflux. The liver is the main site for lipid metabolism, and metabolic (dysfunction)-associated fatty liver disease (MAFLD) is one of the most common chronic liver diseases. However, the impact of DDX17 on hepatic lipid accumulation has not been verified. In this study, we found, for the first time, that oleic acid/palmitic acid (OA/PA)-induced lipid accumulation was largely abrogated by DDX17 overexpression in both HepG2 (a human hepatocellular carcinoma line) and Hep1-6 (a murine hepatocellular carcinoma line) cells, and this effect was due to a marked reduction in cellular triglyceride (TG) content. Moreover, the overexpression of DDX17 was accompanied by a significant decrease in the expression of genes involved in de novo fatty acid synthesis (FAS, ACC, and SCD-1) in both HepG2 and Hep1-6 cells. In conclusion, DDX17 protected against OA/PA-induced lipid accumulation in hepatocytes through de novo lipogenesis inhibition.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Lipid Metabolism , Lipogenesis , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Oleic Acid/metabolism , Oleic Acid/pharmacology , Palmitic Acid/metabolism , Palmitic Acid/pharmacology
13.
Oxid Med Cell Longev ; 2022: 5184135, 2022.
Article in English | MEDLINE | ID: mdl-35186188

ABSTRACT

OBJECTIVE: To investigate the protective effects and regulatory mechanism of miR-488-3p on doxorubicin-induced cardiotoxicity. METHODS: The C57BL/6 mice and primary cardiomyocytes were used to construct doxorubicin-induced cardiomyocyte injury models in vivo and in vitro. The levels of miR-488-3p and its downstream target genes were analyzed by quantitative real-time PCR. Mouse cardiac function, cell survival, cellular injury-related proteins, and the apoptosis level of cardiomyocytes were analyzed by echocardiography, MTT analysis, Western blotting, and DNA laddering separately. RESULTS: Cardiomyocyte injury caused by a variety of stimuli can lead to the reduction of miR-488-3p level, especially when stimulated with doxorubicin. Doxorubicin led to significant decrease in cardiac function, cell autophagic flux blockage, and apoptosis in vivo and in vitro. The expression of miR-488-3p's target gene, CyclinG1, increased remarkably in the doxorubicin-treated neonatal mouse cardiomyocytes. Overexpression of miR-488-3p inhibited CyclinG1 expression, increased cardiomyocyte viability, and attenuated doxorubicin-induced cardiomyocyte autophagic flux blockage and apoptosis. CONCLUSIONS: miR-488-3p is one of the important protective miRNAs in doxorubicin-induced cardiotoxicity by inhibiting the expression of CyclinG1, which provides insight into the possible clinical application of miR-488-3p/CyclinG1 as therapeutic targets in doxorubicin-induced cardiovascular diseases.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Cyclin G1/antagonists & inhibitors , Doxorubicin/adverse effects , MicroRNAs/metabolism , Myocytes, Cardiac/drug effects , Animals , Humans , Male , Mice , Rats
14.
FEBS Lett ; 596(4): 510-525, 2022 02.
Article in English | MEDLINE | ID: mdl-35043979

ABSTRACT

Lysophosphatidylcholine (LPC), the active metabolite of palmitate, triggers hepatocyte death by activating endoplasmic reticulum stress and JNK signalling-mediated lipoapoptosis. However, LPC-induced cytotoxicity in hepatocytes is not well understood. Here, we found for the first time that LPC-induced cell rounding occurred prior to apoptosis. LPC-induced rounding of cells reduced both cell-extracellular matrix (ECM) adhesion and cell-cell junctions, which promoted detachment-induced apoptosis (defined as anoikis) in hepatocytes. Further study revealed that LPC altered cellular morphology and cell adhesion by inhibiting integrin and cadherin signalling-mediated microfilament polymerization. We also found that ECM supplementation and microfilament cytoskeletal stabilization inhibited LPC-induced hepatocyte death by attenuating anoikis. Our data indicate a novel cytotoxic process and signalling pathway induced by LPC.


Subject(s)
Anoikis/drug effects , Cadherins/genetics , Cell Adhesion/drug effects , Integrins/genetics , Intercellular Junctions/drug effects , Lysophosphatidylcholines/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Anoikis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cadherins/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Integrins/metabolism , Intercellular Junctions/metabolism , Intercellular Junctions/ultrastructure , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Vinculin/genetics , Vinculin/metabolism
15.
Eur Radiol ; 32(1): 254-261, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34255162

ABSTRACT

OBJECTIVES: To compare the correlation of depth of invasion (DOI) measured on multiple magnetic resonance imaging (MRI) sequences and pathological DOI, in order to determine the optimal MRI sequence for measurement. METHODS: A total of 122 oral tongue squamous cell carcinoma (OTSCC) patients were retrospectively analyzed, who had received preoperative MRI and surgical resection. DOIs measured on fat-suppressed T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI), dynamic enhanced-T1 high-resolution insotropic volume examination (e-THRIVE), and contrast-enhanced fat-suppressed T1WI (CE-T1WI) were respectively compared to those measured in pathologic specimens. The cutoff value of the best correlated MRI sequence was determined, and the T staging accuracy of MRI-derived DOI was evaluated. RESULTS: DOI derived from e-THRIVE showed the best correlation (r = 0.936, p < 0.001) with pathological DOI. The area under the curve values of MRI-derived DOI distinguishing T1 stage from T2 stage and distinguishing T2 stage from T3 stage were 0.969 and 0.974, respectively. The T staging criteria of MRI-derived DOI were 6.2 mm and 11.4 mm, with a staging accuracy of 86.9% compared to pathological DOI criteria of 5 mm and 10 mm. CONCLUSION: E-THRIVE was the optimal MR sequence to measure the MR-derived DOI, and DOI derived from e-THRIVE could serve as a potential cut-off value as a clinical T staging indicator of OTSCC. KEY POINTS: • Multiparametric MRI helps radiologists to assess the neoplasm invasion in patients with oral tongue squamous cell carcinoma. • Retrospective study indicated that measurement was most accurate on enhanced-T1 high-resolution insotropic volume examination dynamic contrast enhancement images. • T staging of oral tongue squamous cell carcinoma was accurate according to the dynamic contrast enhancement MRI-derived depth of invasion.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Multiparametric Magnetic Resonance Imaging , Tongue Neoplasms , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Humans , Magnetic Resonance Imaging , Neoplasm Staging , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck , Tongue Neoplasms/diagnostic imaging , Tongue Neoplasms/pathology
16.
J Mol Endocrinol ; 68(1): 35-49, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34723832

ABSTRACT

miR-23a-3p and miR-23b-3p are members of the miR-23~27~24-2 superfamily. The role of miR-23a/b-3p in regulating hepatic lipid accumulation is still unknown. Here, we found that increased miR-23a-3p and miR-23b-3p levels were accompanied by an increase in the protein levels of the sterol regulatory element-binding protein-1 (SREBP-1) and fatty acid synthase (FAS) in the steatotic livers of mice fed a high-fat diet and leptin receptor-deficient type 2 diabetic mice (db/db). Importantly, overexpression of miR-23a/b-3p in Hep1-6 cells elevated the intracellular triglyceride level and upregulated the expression of Srebp-1c and Fas. Taken together, these results suggested that miR-23a/b-3p enhanced mRNA stability by binding the 5'-UTR of Srebp-1c and Fas mRNA, thereby promoting triglyceride accumulation in hepatocytes.


Subject(s)
Fatty Acid Synthase, Type I/genetics , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , MicroRNAs/genetics , Sterol Regulatory Element Binding Protein 1/genetics , 5' Untranslated Regions , Animals , Diet, High-Fat , Disease Susceptibility , Fatty Acid Synthase, Type I/metabolism , Hepatocytes/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , RNA Interference , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
17.
Cell Prolif ; 54(12): e13140, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34647385

ABSTRACT

OBJECTIVE: In this study, we aim to explore the role of bone marrow macrophage-derived exosomes in hepatic insulin resistance, investigate the substance in exosomes that regulates hepatic insulin signalling pathways, reveal the specific molecular mechanisms involved in hepatic insulin resistance and further explore the role of exosomes in type 2 diabetes. MATERIALS AND METHODS: High-fat diet (HFD)-fed mice were used as obesity-induced hepatic insulin resistance model, exosomes were isolated from BMMs which were extracted from HFD-fed mice by ultracentrifugation. Exosomes were analysed the spectral changes of microRNA expression using a microRNA array. The activation of the insulin signalling pathway and the level of glycogenesis were examined in hepatocytes after transfected with miR-143-5p mimics. Luciferase assay and western blot were used to assess the target of miR-143-5p. RESULTS: BMMs from HFD-fed mice were polarized towards M1, and miR-143-5p was significantly upregulated in exosomes of BMMs from HFD-fed mice. Overexpression of miR-143-5p in Hep1-6 cells led to decreased phosphorylation of AKT and GSK and glycogen synthesis. Dual-luciferase reporter assay and western blot demonstrated that mitogen-activated protein kinase phosphatase-5 (Mkp5, also known as Dusp10) was the target gene of miR-143-5p. Moreover, the overexpression of MKP5 could rescue the insulin resistance induced by transfection miR-143-5p mimics in Hep1-6. CONCLUSION: Bone marrow macrophage-derived exosomal miR-143-5p induces insulin resistance in hepatocytes through repressing MKP5.


Subject(s)
Bone Marrow Cells/metabolism , Dual-Specificity Phosphatases/biosynthesis , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Insulin Resistance , Macrophages/metabolism , MicroRNAs/metabolism , Animals , Diet, High-Fat , Exosomes , Mice
18.
Exp Ther Med ; 22(5): 1248, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34539844

ABSTRACT

Sepsis-induced myocardial dysfunction is a leading cause of the high mortality rates associated with sepsis. The aim of the present study was to investigate the effect of butorphanol on sepsis-induced cardiomyocyte dysfunction. Lipopolysaccharide (LPS) was used to induce H9C2 cardiomyocytes to establish an in vitro sepsis model. The effect of butorphanol on the viability of LPS-induced H9C2 cells was analyzed using a Cell Counting Kit-8 assay. The levels of tumor necrosis factor-α, interleukin (IL)-1ß and IL-6 were detected using ELISA. Western blotting was used to analyze the expression levels of inflammation-and apoptosis-related proteins. Cell apoptosis was measured using a TUNEL assay. The expression levels of κ-opioid receptor (KOR) were analyzed using reverse transcription-quantitative PCR analysis and western blotting. Following LPS induction, the levels of inflammatory cytokines and proapoptotic proteins were found to be upregulated in H9C2 cells, while butorphanol treatment downregulated these levels. The expression levels of KOR were also upregulated following butorphanol treatment in LPS-induced H9C2 cells. Addition of the KOR inhibitor, nor-binaltorphimine, alleviated the inhibitory effects of butorphanol on inflammation and apoptosis in LPS-induced H9C2 cells. In conclusion, the findings of the present study provided evidence indicating that butorphanol may alleviate LPS-induced inflammation and apoptosis in cardiomyocytes by upregulating KOR expression, which may provide a novel insight into the potential therapeutic effects of butorphanol and its underlying mechanism of action.

19.
Oxid Med Cell Longev ; 2021: 5550498, 2021.
Article in English | MEDLINE | ID: mdl-34221233

ABSTRACT

Lipotoxicity-induced apoptosis, also referred to as lipoapoptosis, is one of the important initial factors promoting the progression from hepatosteatosis to nonalcoholic steatohepatitis (NASH). Saturated free fatty acids (SFAs), which are increased significantly in NASH, are directly hepatotoxic which induce hepatocyte lipoapoptosis. Previously, we reported that protein phosphatase 4 (PP4) was a novel regulator of hepatic insulin resistance and lipid metabolism, but its role in hepatic lipoapoptosis remains unexplored. In this study, we found out that PP4 was upregulated in the livers of western diet-fed-induced NASH mice and SFA-treated murine primary hepatocytes and HepG2 cells. In addition, we found for the first time that suppression of PP4 decreased SFA-induced JNK activation and expression of key modulators of hepatocyte lipoapoptosis including p53-upregulated modulator of apoptosis (PUMA) and Bcl-2-interacting mediator (Bim) and reduced hepatocyte lipoapoptosis level as well both in vitro and in vivo. Further study revealed that PP4 induced JNK activation and lipoapoptosis-related protein expression by regulating the RAC1/MLK3 pathway instead of the PERK/CHOP pathway. The effects of palmitate-treated and PP4-induced lipoapoptosis pathway activation were largely abolished by RAC1 inhibition. Moreover, we identified that PP4 interacted with RAC1 and regulated GTPase activity of RAC1. In conclusion, these results demonstrated that PP4 was a novel regulator of hepatocyte lipoapoptosis and mediated hepatocyte lipoapoptosis by regulating the RAC1/MLK3/JNK signaling pathway. Our finding provided new insights into the mechanisms of this process.


Subject(s)
Hepatocytes/metabolism , Kallikreins/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Neuropeptides/metabolism , Phosphoprotein Phosphatases/metabolism , Prostate-Specific Antigen/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Hepatocytes/cytology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinase Kinase 11
20.
J Immunol Res ; 2021: 1815098, 2021.
Article in English | MEDLINE | ID: mdl-34307691

ABSTRACT

Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.


Subject(s)
Adiponectin/metabolism , Myocardial Reperfusion Injury/immunology , Myocytes, Cardiac/pathology , Animals , Animals, Newborn , Apoptosis/immunology , Cell Hypoxia/immunology , Cell Survival , Cells, Cultured , Culture Media/metabolism , Disease Models, Animal , Female , Humans , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/immunology , Necroptosis/immunology , Oxidative Stress/immunology , Pregnancy , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...