Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Exp Eye Res ; 244: 109919, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38729254

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly, which is primarily attributed to oxidative stress-induced damage to the retinal pigment epithelium (RPE). Human amniotic mesenchymal stem cells (hAMSC) were considered to be one of the most promising stem cells for clinical application due to their low immunogenicity, tissue repair ability, pluripotent potential and potent paracrine effects. The conditional medium (hAMSC-CM) and exosomes (hAMSC-exo) derived from hAMSC, as mediators of intercellular communication, play an important role in the treatment of retinal diseases, but their effect and mechanism on oxidative stress-induced retinal degeneration are not explored. Here, we reported that hAMSC-CM alleviated H2O2-induced ARPE-19 cell death through inhibiting mitochondrial-mediated apoptosis pathway in vitro. The overproduction of reactive oxygen species (ROS), alteration in mitochondrial morphology, loss of mitochondrial membrane potential and elevation of Bax/Bcl2 ratio in ARPE-19 cells under oxidative stress were efficiently reversed by hAMSC-CM. Moreover, it was found that hAMSC-CM protected cells against oxidative injury via PI3K/Akt/FoxO3 signaling. Intriguingly, exosome inhibitor GW4869 alleviated the inhibitory effect of hAMSC-CM on H2O2-induced decrease in cell viability of ARPE-19 cells. We further demonstrated that hAMSC-exo exerted the similar protective effect on ARPE-19 cells against oxidative damage as hAMSC-CM. Additionally, both hAMSC-CM and hAMSC-exo ameliorated sodium iodate-induced deterioration of RPE and retinal damage in vivo. These results first indicate that hAMSC-CM and hAMSC-exo protect RPE cells from oxidative damage by regulating PI3K/Akt/FoxO3 pathway, suggesting hAMSC-CM and hAMSC-exo will be a promising cell-free therapy for the treatment of AMD in the future.


Subject(s)
Amnion , Exosomes , Forkhead Box Protein O3 , Mesenchymal Stem Cells , Oxidative Stress , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Retinal Degeneration , Retinal Pigment Epithelium , Signal Transduction , Humans , Mesenchymal Stem Cells/metabolism , Exosomes/metabolism , Amnion/cytology , Culture Media, Conditioned/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/etiology , Forkhead Box Protein O3/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Apoptosis , Cells, Cultured , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial , Blotting, Western , Animals , Cell Survival , Hydrogen Peroxide/toxicity
2.
Can J Physiol Pharmacol ; 101(7): 369-381, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37192549

ABSTRACT

Obesity is a metabolic syndrome characterized by abnormal lipid deposition and energy imbalance. CD38 is a single-chain transmembrane glycoprotein widely expressed in a variety of cell types. The roles of skeletal muscle and brown fat in CD38 deficiency under HFD-induced obesity remain unknown. In this study, we established obesity model with HFD and examined the changes in metabolites with metabonomics. Our results showed that CD38 expression was increased in muscle and brown fat after HFD treatment. Moreover, the results of metabonomics showed that CD38 deficiency significantly altered the metabolites in energy metabolism, cofactor generation, and redox homeostasis. Furthermore, CD38 deficiency reduced the expressions of NADPH oxidase 2 and FASN in mRNA level. We found that the expressions of Sirt1, Sirt3, and PGC1α were upregulated in CD38-deficient muscle tissue. In brown fat, the Sirt1-3, cell death inducing DFFA-like effector A, ELOVL3, and Dio2 expressions were increased in CD38-deficient mice. Our results showed the uncoupling protein 1 expression was upregulated. And NAD+ supplementation increased the expression of Sirt1 and PGC1α after palmitic acid treatment. Taken together, our results demonstrated that the protection of CD38 deficiency on HFD-induced obesity was related to the inhibition of oxidative stress and increasing energy expenditure via activating NAD+/Sirtuins signaling pathways in muscle and brown fat.


Subject(s)
Adipose Tissue, Brown , NAD , Animals , Mice , Adipose Tissue, Brown/metabolism , Diet, High-Fat , Energy Metabolism , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , NAD/metabolism , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction , Sirtuin 1/metabolism
3.
Int J Mol Sci ; 24(21)2023 Nov 06.
Article in English | MEDLINE | ID: mdl-37958991

ABSTRACT

Diabetic cardiomyopathy is one of the diabetes mellitus-induced cardiovascular complications that can result in heart failure in severe cases, which is characterized by cardiomyocyte apoptosis, local inflammation, oxidative stress, and myocardial fibrosis. CD38, a main hydrolase of NAD+ in mammals, plays an important role in various cardiovascular diseases, according to our previous studies. However, the role of CD38 in diabetes-induced cardiomyopathy is still unknown. Here, we report that global deletion of the CD38 gene significantly prevented diabetic cardiomyopathy induced by high-fat diet plus streptozotocin (STZ) injection in CD38 knockout (CD38-KO) mice. We observed that CD38 expression was up-regulated, whereas the expression of Sirt3 was down-regulated in the hearts of diabetic mice. CD38 deficiency significantly promoted glucose metabolism and improved cardiac functions, exemplified by increased left ventricular ejection fraction and fractional shortening. In addition, we observed that CD38 deficiency markedly decreased diabetes or high glucose and palmitic acid (HG + PA)-induced pyroptosis and apoptosis in CD38 knockout hearts or cardiomyocytes, respectively. Furthermore, we found that the expression levels of Sirt3, mainly located in mitochondria, and its target gene FOXO3a were increased in CD38-deficient hearts and cardiomyocytes with CD38 knockdown under diabetic induction conditions. In conclusion, we demonstrated that CD38 deficiency protected mice from diabetes-induced diabetic cardiomyopathy by reducing pyroptosis and apoptosis via activating NAD+/Sirt3/FOXO3a signaling pathways.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Sirtuin 3 , Animals , Mice , Apoptosis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Mammals/metabolism , Myocytes, Cardiac/metabolism , NAD/metabolism , Oxidative Stress , Pyroptosis , Sirtuin 3/metabolism , Stroke Volume , Ventricular Function, Left
4.
Ann Noninvasive Electrocardiol ; 27(2): e12918, 2022 03.
Article in English | MEDLINE | ID: mdl-34951713

ABSTRACT

BACKGROUND: We compared the methods of electrocardiogram (ECG) and X-ray localization of the peripherally inserted central catheter (PICC) tip position, in order to find a more convenient, practical, and safe method. OBJECTIVE: To investigate the value of applying electrocardiographic localization of the PICC tip position in critically ill patients with advanced cancer in Hebei Province, China. METHOD: Enrolled 137 advanced cancers requiring PICC placement. The position of the catheter tip was localized with the bedside electrocardiogram in real time. Then, the localization was performed using a chest X-ray (the gold standard). The accuracy of electrocardiographic location was checked. RESULTS: Specific P waves were observed in 130 patients. No change in the P waves was observed for the remaining seven patients. The age of the latter group of patients was more advanced (87.29 [5.15] years), a significant difference to that of the 130 patients with specific P waves (71.58 [14.84] years) (t = -6.704, p < .001). Specific P waves not only involve ascendance in P waves but also ascendance in QRS waves. CONCLUSIONS: The use of an ECG to localize the PICC tip in critically ill patients with advanced cancer may replace the unnecessary use of chest X-rays. Specific P waves not only involve an increase in P waves but also an increase in QRS waves. If there is no change in the P wave, a chest X-ray film must be obtained. In elderly patients, because there is a possibility of catheter tip malposition, a comprehensive evaluation should be performed before surgery.


Subject(s)
Catheterization, Central Venous , Catheterization, Peripheral , Central Venous Catheters , Neoplasms , Aged , Catheterization, Central Venous/adverse effects , Catheterization, Central Venous/methods , Catheterization, Peripheral/adverse effects , Catheterization, Peripheral/methods , Child, Preschool , Critical Illness , Electrocardiography/methods , Humans , Neoplasms/diagnostic imaging , Neoplasms/etiology
5.
Can J Physiol Pharmacol ; 99(8): 803-811, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33356884

ABSTRACT

Inflammation-induced activation and dysfunction of endothelial cells play an important role in the pathology of multiple vascular diseases. Nicaraven, a potent hydroxyl radical scavenger, has recently been found to have anti-inflammatory roles; however, the mechanism of its action is not fully understood. Here we investigated the effects of Nicaraven on tumor necrosis factor α (TNFα) - induced inflammatory response in human umbilical vein endothelial cells and we explore the underlying mechanisms related to the nuclear factor-κB (NF-κB) signaling pathway. Our results showed that Nicaraven significantly reduced the reactive oxygen species production after TNFα stimulation. Nicaraven suppressed TNFα-induced mRNA expression of multiple adhesion molecules and pro-inflammatory cytokines, including vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), E-selectin, MCP-1, TNFα, interleukin-1ß (IL-1ß), IL-6, and IL-8. In addition, Nicaraven inhibited monocyte adhesion and reduced the protein levels of VCAM-1 and ICAM-1. Mechanistically, Nicaraven prevented TNFα-induced activation of NF-κB signaling pathway by suppressing the phosphorylation of NF-κB p65, IκBα, and IκB kinase (IKK)α/ß, stabilizing IκBα, and inhibiting the translocation of p65 from cytosol to nucleus. Finally, we showed that Nicaraven improved the functions of endothelial cells, seen as the upregulation of endothelial nitric oxide synthase and increased nitric oxide levels. Our findings indicated that Nicaraven effectively inhibits TNFα-induced endothelial activation and inflammatory response at least partly through inhibiting NF-κB signaling pathway.


Subject(s)
NF-kappa B , Human Umbilical Vein Endothelial Cells , Humans , Signal Transduction
6.
J Cell Mol Med ; 22(7): 3638-3651, 2018 07.
Article in English | MEDLINE | ID: mdl-29682889

ABSTRACT

We previously observed that disruption of FK506-binding protein 12.6 (FKBP12.6) gene resulted in cardiac hypertrophy in male mice. Studies showed that overexpression of FKBP12.6 attenuated thoracic aortic constriction (TAC)-induced cardiac hypertrophy in mice, whereas the adenovirus-mediated overexpression of FKBP12.6 induced hypertrophy and apoptosis in cultured neonatal cardiomyocytes, indicating that the role of FKBP12.6 in cardiac hypertrophy is still controversial. In this study, we aimed to investigate the roles and mechanisms of FKBP12.6 in angiotensin II (AngII)-induced cardiac hypertrophy using various transgenic mouse models in vivo and in vitro. FKBP12.6 knockout (FKBP12.6-/- ) mice and cardiac-specific FKBP12.6 overexpressing (FKBP12.6 TG) mice were infused with AngII (1500 ng/kg/min) for 14 days subcutaneously by implantation of an osmotic mini-pump. The results showed that FKBP12.6 deficiency aggravated AngII-induced cardiac hypertrophy, while cardiac-specific overexpression of FKBP12.6 prevented hearts from the hypertrophic response to AngII stimulation in mice. Consistent with the results in vivo, overexpression of FKBP12.6 in H9c2 cells significantly repressed the AngII-induced cardiomyocyte hypertrophy, seen as reductions in the cell sizes and the expressions of hypertrophic genes. Furthermore, we demonstrated that the protection of FKBP12.6 on AngII-induced cardiac hypertrophy was involved in reducing the concentration of intracellular Ca2+ ([Ca2+ ]i), in which the protein significantly inhibited the key Ca2+ /calmodulin-dependent signalling pathways such as calcineurin/cardiac form of nuclear factor of activated T cells 4 (NFATc4), calmodulin kinaseII (CaMKII)/MEF-2, AKT/Glycogen synthase kinase 3ß (GSK3ß)/NFATc4 and AKT/mTOR signalling pathways. Our study demonstrated that FKBP12.6 protects heart from AngII-induced cardiac hypertrophy through inhibiting Ca2+ /calmodulin-mediated signalling pathways.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Cardiomegaly/metabolism , Tacrolimus Binding Proteins/metabolism , Angiotensin II/metabolism , Angiotensin II/toxicity , Animals , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cell Line , Gene Expression , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tacrolimus Binding Proteins/genetics
7.
Cell Physiol Biochem ; 48(6): 2350-2363, 2018.
Article in English | MEDLINE | ID: mdl-30114710

ABSTRACT

BACKGROUND/AIMS: Previous studies showed that CD38 deficiency protected heart from ischemia/reperfusion injury and high fat diet (HFD)-induced obesity in mice. However, the role of CD38 in HFD-induced heart injury remains unclear. In the present study, we have investigated the effects and mechanisms of CD38 deficiency on HFD-induced heart injury. METHODS: The metabolites in heart from wild type (WT) and CD38 knockout (CD38-/-) mice were examined using metabolomics analysis. Cell viability, lactate hydrogenase (LDH) release, super oxide dismutase (SOD) activity, reactive oxygen species (ROS) production, triglyceride concentration and gene expression were examined by biochemical analysis and QPCR. RESULTS: Our results revealed that CD38 deficiency significantly elevated the intracellular glutathione (GSH) concentration and GSH/GSSG ratio, decreased the contents of free fatty acids and increased intracellular NAD+ level in heart from CD38-/- mice fed with HFD. In addition, in vitro knockdown of CD38 significantly attenuated OA-induced cellular injury, ROS production and lipid synthesis. Furthermore, the expression of mitochondrial deacetylase Sirt3 as well as its target genes FOXO3 and SOD2 were markedly upregulated in the H9C2 cell lines after OA stimulation. In contrast, the expressions of NOX2 and NOX4 were significantly decreased in the cells after OA stimulation. CONCLUSION: Our results demonstrated that CD38 deficiency protected heart from HFD-induced oxidative stress via activating Sirt3/FOXO3-mediated anti-oxidative stress pathway.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Diet, High-Fat , Forkhead Box Protein O3/metabolism , Membrane Glycoproteins/genetics , Oxidative Stress , Sirtuin 3/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , Animals , Cell Line , Glutathione/metabolism , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxide Dismutase/metabolism
8.
J Cell Mol Med ; 21(8): 1492-1502, 2017 08.
Article in English | MEDLINE | ID: mdl-28296029

ABSTRACT

Cardiac hypertrophy is an early hallmark during the clinical course of heart failure and regulated by various signalling pathways. Recently, we observed that mouse embryonic fibroblasts from CD38 knockout mice were significantly resistant to oxidative stress such as H2 O2 -induced injury and hypoxia/reoxygenation-induced injury. In addition, we also found that CD38 knockout mice protected heart from ischaemia reperfusion injury through activating SIRT1/FOXOs-mediated antioxidative stress pathway. However, the role of CD38 in cardiac hypertrophy is not explored. Here, we investigated the roles and mechanisms of CD38 in angiotensin II (Ang-II)-induced cardiac hypertrophy. Following 14 days of Ang-II infusion with osmotic mini-pumps, a comparable hypertension was generated in both of CD38 knockout and wild-type mice. However, the cardiac hypertrophy and fibrosis were much more severe in wild-type mice compared with CD38 knockout mice. Consistently, RNAi-induced knockdown of CD38 decreased the gene expressions of atrial natriuretic factor (ANF) and brain natriuretic peptide (BNP) and reactive oxygen species generation in Ang-II-stimulated H9c2 cells. In addition, the expression of SIRT3 was elevated in CD38 knockdown H9c2 cells, in which SIRT3 may further activate the FOXO3 antioxidant pathway. The intracellular Ca2+ release induced by Ang-II markedly decreased in CD38 knockdown H9c2 cells, which might be associated with the decrease of nuclear translocation of NFATc4 and inhibition of ERK/AKT phosphorylation. We concluded that CD38 plays an essential role in cardiac hypertrophy probably via inhibition of SIRT3 expression and activation of Ca2+ -NFAT signalling pathway. Thus, CD38 may be a novel target for treating cardiac hypertrophy.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Angiotensin II/pharmacology , Cardiomegaly/genetics , Membrane Glycoproteins/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/deficiency , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Calcium/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Line , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression Regulation , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuins/genetics , Sirtuins/metabolism
9.
Lipids Health Dis ; 16(1): 82, 2017 Apr 27.
Article in English | MEDLINE | ID: mdl-28449683

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease is one of the most common liver diseases in the world and is a typical hepatic manifestation of metabolic syndrome which is characterized with lipid accumulation in liver. Nicotinamide phosphoribosyltransferase (NAMPT) has been recently identified as an enzyme involved in nicotinamide adenine dinucleotide (NAD+) biosynthesis and plays an important role in cellular metabolism in variety of organs in mammals. The aim of this study was to investigate the effects of NAMPT on high fat diet-induced hepatic steatosis. METHODS: Hepatic steatosis model was induced by high fat diet (HFD) in C57BL/6 mice in vivo. HepG2 and Hep1-6 hepatocytes were transfected with NAMPT vector plasmid or treated with NAMPT inhibitor FK866 and then incubated with oleic acid. Lipids accumulation was examined by HE staining or oil red staining. Quantitative RT-PCR and Western blot were used to measure expressions of the genes involved in lipogenic synthesis. RESULTS: FK866 significantly promoted liver steatosis in the mice fed with HFD and hepatic lipid accumulation in vitro, accompanied by the increases of the expressions of lipogenic genes such as sterol regulatory element-binding protein 1 (SREBP1) and fatty acid synthase (FASN). Nicotinamide mononucleotide (NMN) and NAD+ significantly rescued the actions of FK866 in vitro. In contrast, overexpression of NAMPT in HepG2 and Hep1-6 hepatocytes ameliorated hepatic lipid accumulation. In addition, FK866 decreased the protein levels of Sirt1 and phospho-AMPKα in liver of the HFD fed mice. Furthermore, Resveratrol, a Sirt1 activator, significantly reduced lipogenic gene expressions, while EX-527, a Sirt1 specific inhibitor, had the opposite effects. CONCLUSION: Our results demonstrated that inhibition of NAMPT aggravated the HFD- or oleic acid-induced hepatic steatosis through suppressing Sirt1-mediated signaling pathway. On the one hand, the inhibition of NAMPT reduced the production of NAD+ through inhibiting the NAD+ salvage pathway, resulting in the decrease of Sirt1 activity, and then attenuated the deacetylation of SREBP1 in which the inhibition of SREBP1 activity promoted the expressions of FASN and ACC. On the other hand, the reduced Sirt1 activity alleviated the activation of AMPKα to further enhance SREBP1 activities.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cytokines/genetics , Liver/enzymology , Nicotinamide Phosphoribosyltransferase/genetics , Non-alcoholic Fatty Liver Disease/genetics , Sirtuin 1/genetics , Sterol Regulatory Element Binding Protein 1/genetics , AMP-Activated Protein Kinases/metabolism , Acrylamides/pharmacology , Animals , Carbazoles/pharmacology , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Diet, High-Fat/adverse effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , NAD/pharmacology , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Oleic Acid/pharmacology , Piperidines/pharmacology , Resveratrol , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Stilbenes/pharmacology
10.
Int Immunopharmacol ; 129: 111660, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38350357

ABSTRACT

BACKGROUND: Human amniotic mesenchymal stem cells (hAMSCs) derived from amniotic membrane have multilineage differentiation, immunosuppressive, and anti-inflammation which makes them suitable for the treatment of various diseases. OBJECTIVE: This study aimed to explore the therapeutic effect and molecular mechanism of hAMSCs in ventricular remodeling (VR). METHODS: hAMSCs were characterized by a series of experiments such as flow cytometric analysis, immunofluorescence, differentiative induction and tumorigenicity. Mouse VR model was induced by isoproterenol (ISO) peritoneally, and the therapeutic effects and the potential mechanisms of hAMSCs transplantation were evaluated by echocardiography, carboxy fluorescein diacetate succinimidyl ester (CFSE) labeled cell tracing, histochemistry, qRT-PCR and western blot analysis. The co-culturing experiments were carried out for further exploring the mechanisms of hAMSCs-derived conditioned medium (CM) on macrophage polarization and fibroblast fibrosis in vitro. RESULTS: hAMSCs transplantation significantly alleviated ISO-induced VR including cardiac hypertrophy and fibrosis with the improvements of cardiac functions. CFSE labeled hAMSCs kept an undifferentiated state in heart, indicating that hAMSCs-mediated the improvement of ISO-induced VR might be related to their paracrine effects. hAMSCs markedly inhibited ISO-induced inflammation and fibrosis, seen as the increase of M2 macrophage infiltration and the expressions of CD206 and IL-10, and the decreases of CD86, iNOS, COL3 and αSMA expressions in heart, suggesting that hAMSCs transplantation promoted the polarization of M2 macrophages and inhibited the polarization of M1 macrophages. Mechanically, hAMSCs-derived CM significantly increased the expressions of CD206, IL-10, Arg-1 and reduced the expressions of iNOS and IL-6 in RAW264.7 macrophages in vitro. Interestingly, RAW264.7-CM remarkably promoted the expressions of anti-inflammatory factors such as IL-10, IDO, and COX2 in hAMSCs. Furthermore, the CM derived from hAMSCs pretreated with RAW264.7-CM markedly inhibited the expressions of fibrogenesis genes such as αSMA and COL3 in 3T3 cells. CONCLUSION: Our results demonstrated that hAMSCs effectively alleviated ISO-induced cardiac hypertrophy and fibrosis, and improved the cardiac functions in mice, and the underlying mechanisms might be related to inhibiting the inflammation and fibrosis during the ventricular remodeling through promoting the polarization of CD206hiIL-10hi macrophages in heart tissues. Our study strongly suggested that by taking the advantages of the potent immunosuppressive and anti-inflammatory effects, hAMSCs may provide an alternative therapeutic approach for prevention and treatment of VR clinically.


Subject(s)
Fluoresceins , Interleukin-10 , Mesenchymal Stem Cells , Succinimides , Mice , Humans , Animals , Interleukin-10/pharmacology , Amnion , Isoproterenol , Ventricular Remodeling , Macrophages , Inflammation/chemically induced , Inflammation/therapy , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Fibrosis , Cardiomegaly
11.
Int Immunopharmacol ; 124(Pt B): 110875, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37742368

ABSTRACT

BACKGROUND: Extensions of mesenchymal stem cells (MSCs) in vitro may lead to the loss of their biological functions. However, hypoxic culturation has been shown to enhance the proliferation, survival, and immunomodulatory capacity of MSCs. OBJECTIVE: We aimed to investigate the effects of long-term hypoxic cultivation on the properties of human umbilical cord-derived MSCs (hUCMSCs) and the therapeutic effects of their extracellular vesicles (EVs) in allergic rhinitis (AR). METHODS: Proliferation, senescence, telomerase activity and multipotent properties of hUCMSCs were analyzed under long-term culturation of hypoxia (1%) or normoxia (21%), and the therapeutic effects of their conditional medium (CM) and EVs were evaluated in OVA-induced AR mice. Effects of hypoxia-EVs (Hy-EVs) or normoxia-EVs (No-EVs) on human monocyte-derived dendritic cells (DCs) were investigated, and the possible mechanisms of Hy-EVs in induction of immunotolerance were further explored. RESULTS: Long-term hypoxia significantly promoted the proliferation, inhibited cell senescence, maintained the multipotent status of hUCMSCs. Hy-CM and Hy-EVs showed better therapeutic effects in AR mice compared to No-EVs, seen as improvement of AR-related behaviors such as rubbing and sneezing, and attenuation of inflammation in nasal tissues. In addition, Hy-EVs significantly reduced the expressions of HLA-DR, CD80, CD40, and CD83 induced by OVA plus LPS in DCs, inhibiting the maturation of DCs. Furthermore, we observed that VEGF was remarkably enriched in Hy-EVs, but not in No-EVs, and the inhibition of DCs maturation was markedly neutralized by VEGF antibodies, suggesting that VEGF derived from Hy-EVs was responsible for the inhibition of DCs maturation. CONCLUSION: Our results demonstrated that long-term hypoxia significantly promoted the proliferation, inhibited cell senescence, maintained the multipotent status of hUCMSCs, and hypoxia treated hUCMSCs-derived EVs enhanced their therapeutic effects in AR mice through VEGF-mediated inhibition of DCs maturation.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Rhinitis, Allergic , Humans , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , Mesenchymal Stem Cells/metabolism , Rhinitis, Allergic/therapy , Rhinitis, Allergic/metabolism , Hypoxia/therapy , Hypoxia/metabolism , Dendritic Cells/metabolism , Extracellular Vesicles/metabolism
12.
Phys Chem Chem Phys ; 14(15): 5297-306, 2012 Apr 21.
Article in English | MEDLINE | ID: mdl-22402404

ABSTRACT

Second-order nonlinear optical properties of a tetrathiafulvalene (TTF) derivative of spiropyran have been studied based on density functional theory (DFT) combined with the finite field (FF) calculations. Our DFT-FF calculations confirm a switching behavior of the static first hyperpolarizability caused by the redox and photochromic reaction. The photochromic reaction generates spiropyran-merocyanine conversion by reversible cleavage of the C-O bond, which is relative to the close- and open-ring forms 1-c and 1-o. The open-ring form 1-o displays the large static first hyperpolarizability relative to its close-ring form 1-c according to our DFT-FF calculations with three functionals. The electronic structure analysis and spin unrestricted calculations show that the redox processes significantly affect the geometrical structure of the TTF unit, and thus enhance the static first hyperpolarizabilities. The one-electron-oxidized species having good planar structure of the TTF unit are ~30 and ~200 times as large as that of the neutral compounds 1-c and 1-o, respectively. But the difference in the static first hyperpolarizability between one- and two-electron-oxidized states of spiropyran species is not substantial according to our DFT-FF calculations, and the spiropyran-merocyanine conversion of two-electron-oxidized species does not largely affect their static first hyperpolarizability. On the basis of the large change in the static first hyperpolarizability, our DFT-FF calculations support a five-state switching of the static first hyperpolarizability based on the redox and photoisomerization.

13.
Stem Cell Res Ther ; 13(1): 224, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35659360

ABSTRACT

BACKGROUND: Liver fibrosis is an outcome of restoring process in chronic liver injury. Human amniotic mesenchymal stem cells (hAMSCs) derived from amniotic membrane have multilineage differentiation, immunosuppressive, and anti-inflammatory potential which makes them suitable for treating liver fibrosis. This study aimed to explore the effect and mechanism of hAMSCs on liver fibrosis. METHODS: hAMSCs were transplanted into carbon tetrachloride (CCl4)-induced liver fibrosis mice via tail vein, and the effects of hAMSCs on hepatic fibrosis were assessed. The effects of hAMSCs and hAMSCs conditional medium (CM) on the activation of hepatic stellate cells (HSCs) were investigated in vivo and in vitro. Antibody array assay was used to identify the cytokines secreted by hAMSCs that may inhibit the activation of HSCs. Finally, the underlying mechanisms were explored by assessing IGF-1R/PI3K/AKT and GSK3ß/ß-catenin signaling pathways in the activated HSCs (LX-2) with hAMSCs and hAMSCs transfected with corresponding siRNAs. RESULTS: Our results showed that hAMSCs possessed the characterizations of mesenchymal stem cells. hAMSCs significantly reduced liver fibrosis and improved liver function in mice by inhibiting HSCs activation in vivo. Both hAMSCs and hAMSC-CM remarkably inhibited the collagen deposition and activation of LX-2 cells in vitro. Antibody array assay showed that insulin-like growth factor binding protein-3 (IGFBP-3), Dickkopf-3 (DKK-3), and Dickkopf-1 (DKK-1) were highly expressed in the co-culture group and hAMSC-CM group compared with LX-2 group. Western blot assay demonstrated that IGFBP-3, DKK-3, and DKK-1 derived from hAMSCs inhibit LX-2 cell activation through blocking canonical Wnt signaling pathway. CONCLUSIONS: Our results demonstrated that IGFBP-3, Dkk3, and DKK-1 secreted by hAMSCs attenuated liver fibrosis in mice through inhibiting HSCs activation via depression of Wnt/ß-catenin signaling pathway, suggesting that hAMSCs or hAMSC-CM provides an alternative therapeutic approach for the treatment of liver fibrosis.


Subject(s)
Mesenchymal Stem Cells , Wnt Signaling Pathway , Amnion , Animals , Hepatic Stellate Cells/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/therapy , Mesenchymal Stem Cells/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism
14.
Signal Transduct Target Ther ; 7(1): 148, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35513381

ABSTRACT

Endothelial activation plays an essential role in the pathogenesis of sepsis-induced acute lung injury, however, the detailed regulatory mechanisms remain largely unknown. Here, we reported that TRIM47, an E3 ubiquitin ligase of the tripartite motif-containing protein family, was highly expressed in vascular endothelial cells. TRIM47-deficient mice were effectively resistant to lipopolysaccharide (LPS)-induced acute lung injury and death by attenuating pulmonary inflammation. TRIM47 was upregulated during TNFα-induced endothelial activation in vitro. Knockdown of TRIM47 in endothelial cells inhibited the transcription of multiple pro-inflammatory cytokines, reduced monocyte adhesion and the expression of adhesion molecules, and suppressed the secretion of IL-1ß and IL-6 in endothelial cells. By contrast, overexpression of TRIM47 promoted inflammatory response and monocyte adhesion upon TNFα stimulation. In addition, TRIM47 was able to activate the NF-κB and MAPK signaling pathways during endothelial activation. Furthermore, our experiments revealed that TRIM47 resulted in endothelial activation by promoting the K63-linked ubiquitination of TRAF2, a key component of the TNFα signaling pathway. Taken together, our studies demonstrated that TRIM47 as a novel activator of endothelial cells, promoted LPS-induced pulmonary inflammation and acute lung injury through potentiating the K63-linked ubiquitination of TRAF2, which in turn activates NF-κB and MAPK signaling pathways to trigger an inflammatory response in endothelial cells.


Subject(s)
Acute Lung Injury , Pneumonia , Animals , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Endothelial Cells/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , NF-kappa B/genetics , NF-kappa B/metabolism , Pneumonia/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
15.
Front Oncol ; 12: 853935, 2022.
Article in English | MEDLINE | ID: mdl-35402260

ABSTRACT

Tripartite motif containing 65 (TRIM65) is an E3 ubiquitin ligase that has been implicated in a variety of cellular processes as well as tumor progression, but its biological role and the underlying mechanism in cervical cancer is unclear. Here, we reported that TRIM65 expression in human cervical cancer tissues was significantly higher than that in the adjacent normal cervical tissues, and TRIM65 knockdown enhanced autophagic flux and cell apoptosis, but not cell cycle, to dramatically inhibit the proliferation and migration of cervical cancer cells. Furthermore, our experiments showed that TRIM65 exhibited oncogenic activities via directly targeting p53, a tumor suppressor and a common upsteam regulator between autophagy and apoptosis, promoting ubiquitination and proteasomal degradation of p53. Taken together, our studies demonstrated that TRIM65 knockdown promotes cervical cancer cell death through enhancing autophagy and apoptosis, suggesting that TRIM65 may be a potential therapeutic target for cervical cancer clinically.

16.
J Ultrasound Med ; 30(11): 1467-74, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22039019

ABSTRACT

OBJECTIVES: The purpose of this study was to compare the use of sonography and non-contrast-enhanced magnetic resonance imaging (MRI) for detection and staging of glottic carcinoma. METHODS: In this study, 30 consecutive patients with glottic squamous cell carcinoma underwent high-frequency sonography and non-contrast-enhanced MRI before surgical treatment. Tumor detectability and imaging findings were evaluated independently and then compared with the surgical and postoperative pathologic findings. RESULTS: Sonography showed a trend toward higher detectability than MRI in early (stage T1 and T2) glottic carcinoma (94.1% [16 of 17 patients] for sonography versus 76.5% [13 of 17 patients] for MRI; P = .335). With regard to T staging, the overall accuracy rates were 80.0% (24 of 30) for sonography and 76.7% (23 of 30) for MRI; the difference was not statistically significant (P > .99). Non-contrast-enhanced MRI was superior to sonography in showing tumor involvement in the retrolaryngeal extent of glottic carcinoma. CONCLUSIONS: Sonography is suggested as a noninvasive complementary modality for detection and initial staging of glottic carcinoma.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Laryngeal Neoplasms/diagnostic imaging , Laryngeal Neoplasms/pathology , Magnetic Resonance Imaging/methods , Adult , Aged , Aged, 80 and over , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Neoplasm Staging , Ultrasonography
17.
RSC Adv ; 11(20): 11786-11792, 2021 Mar 23.
Article in English | MEDLINE | ID: mdl-35423764

ABSTRACT

New carbon-loaded nickel chalcogenide electrode materials (NiS2/GO and NiSe2/rGO) have been synthesized through an easy-to-operate process: NiSe2 was obtained based on NiS2 hollow spheres, and was successfully synthesized with l-cysteine assistance under the hydrothermal method at 120 °C. GO of different mass fraction was added together with l-cysteine. The electrochemical performance of NiS2/GO and NiSe2/rGO has been greatly improved because the formation of a carbon-loaded layer effectively increased the specific surface area and reduced the charge transport resistance. Compared with pure NiS2 and NiSe2, NiS2/GO and NiSe2/rGO presented much better specific capacitance (1020 F g-1 and 722 F g-1 respectively at a current density of 1 A g-1) and more superior rate capability (when the current density was raised to 5 A g-1 the specific capacitance remained at 569 F g-1 and 302 F g-1). This work highlights the advantages of nickel compounds through a very simple experimental method, and contributes to providing a good reference for preparation of superior supercapacitor materials with high performance.

18.
Stem Cell Res Ther ; 12(1): 501, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34507619

ABSTRACT

BACKGROUND: Hyperpigmentation of skin is caused by an imbalance between the melanosome/melanin synthesis in melanocytes and the melanosome/melanin degradation in keratinocytes. Although studies showed that stem cells play a role in hypopigmentation, the underlying mechanisms are far not elucidated. Human amniotic stem cells (hASCs) including human amniotic mesenchymal stem cells (hAMSCs) and human amniotic epithelial stem cells (hAESCs) were considered to be a promising cell source for stem cells-based therapy of many diseases clinically due to their pluripotent potential, no tumorigenesis and immunogenicity, no ethical issues, and potent paracrine effects. Here, we reported that both hASCs and their conditional medium (CM) had a potent anti-hyperpigmentation in skin in vivo and in vitro. METHODS: hAESCs and hAMSCs were identified by RT-PCR, flow cytometric analysis and immunofluorescence. Effects of hASCs and hASC-CM on pigmentation were evaluated in B16F10 cells stimulated with α-melanocyte-stimulating hormone (α-MSH), and mouse ears or human skin substitutes treated with ultraviolet radiation B (UVB). Expressions of the key proteins related with melanogenesis and autophagic flux were detected by western blot in B16F10 cells for further exploring the effects and the underlying mechanisms of hAESC-CM and hAMSC-CM on melanogenesis and melanosome degradation. The hAMSCs exosomes-derived miRNAs were determined by sequencing. RT-PCR, western blot, melanin content analysis and luciferase activity assay were used to determine the hypopigmentation of miR-181a-5p and miR-199a. RESULTS: In our study, we observed that both hASCs and their CM significantly alleviated the α-MSH in B16F10 cells or UVB-induced hyperpigmentation in mouse ears or human skin substitutes by suppressing melanin synthesis and promoting melanosome degradation in vivo and in vitro. Furthermore, we demonstrated that miR-181a-5p and miR-199a derived from hASCs exosomes remarkably inhibited melanogenesis by suppressing MITF (microphthalmia-associated transcription factor) which is a master regulator for governing melanogenesis and promoting melanosome degradation through activating autophagy, respectively. CONCLUSIONS: Our studies provided strong evidence that the conditional medium and exosomes derived from hAMSCs inhibit skin hyperpigmentation by suppressing melanogenesis and promoting melanosome degradation, indicating that the hASCs exosomes or their released microRNAs might be as reagents for cell-free therapy in hyperpigmented disorders clinically.


Subject(s)
Hyperpigmentation , MicroRNAs , Animals , Humans , Melanocytes , Melanosomes , Mice , MicroRNAs/genetics , Stem Cells , Ultraviolet Rays
19.
Stem Cell Res Ther ; 12(1): 364, 2021 06 26.
Article in English | MEDLINE | ID: mdl-34174964

ABSTRACT

BACKGROUND: Obesity is a metabolic disorder syndrome characterized by excessive fat accumulation that is related to many diseases. Human amniotic mesenchymal stem cells (hAMSCs) have a great potential for cell-based therapy due to their characteristics such as pluripotency, low immunogenicity, no tumorigenicity, potent paracrine effects, and no ethical concern. Recently, we observed that both hAMSCs and their conditioned medium (hAMSCs-CM) efficiently repaired skin injury, inhibited hepatocellular carcinoma, and alleviated high-fat diet (HFD)-induced diabetes. However, the effects and the underlying mechanisms of hAMSCs-CM on high-fat diet (HFD)-induced obesity were not explored. METHODS: The characteristics of hAMSCs were confirmed by flow cytometry, RT-PCR, and immunofluorescence. Obese mice were induced by administrating HFD for 15 weeks and simultaneously, the mice were intraperitoneally injected with hAMSCs-CM weekly to evaluate the effects of hAMSCs-CM on HFD-induced obesity. GTT and ITT assays were used to assess the effects of hAMSCs-CM on HFD-induced glucose tolerance and insulin resistance. The lipid accumulation and adipocytes hypertrophy in mouse adipose tissues were determined by histological staining, in which the alterations of blood lipid, liver, and kidney function were also examined. The role of hAMSCs-CM in energy homeostasis was monitored by examining the oxygen consumption (VO2), carbon dioxide production (VCO2), and food and water intake in mice. Furthermore, the expressions of the genes related to glucose metabolism, fatty acid ß oxidation, thermogenesis, adipogenesis, and inflammation were determined by western blot analysis, RT-PCR, and immunofluorescence staining. The roles of hAMSCs-CM in adipogenesis and M1/M2 macrophage polarization were investigated with 3T3-L1 preadipocytes or RAW264.7 cells in vitro. RESULTS: hAMSCs-CM significantly restrained HFD-induced obesity in mice by inhibiting adipogenesis and lipogenesis, promoting energy expenditure, and reducing inflammation. The underlying mechanisms of the anti-obesity of hAMSCs-CM might be involved in inhibiting PPARγ and C/EBPα-mediated lipid synthesis and adipogenesis, promoting GLUT4-mediated glucose metabolism, elevating UCP1/PPARα/PGC1α-regulated energy expenditure, and enhancing STAT3-ARG1-mediated M2-type macrophage polarization. CONCLUSION: Our studies demonstrated that hAMSCs significantly alleviated HFD-induced obesity through their paracrine effects. Obviously, our results open up an attractive therapeutic modality for the prevention and treatment of obesity and other metabolic disorders clinically. The cytokines, exosomes, or micro-vesicles secreted from hAMSCs significantly inhibited HFD-induced obesity in mice by inhibiting lipid production and adipogenesis, promoting energy consumption, and reducing inflammation.


Subject(s)
Diet, High-Fat , Mesenchymal Stem Cells , 3T3-L1 Cells , Adipogenesis , Animals , Culture Media, Conditioned/pharmacology , Diet, High-Fat/adverse effects , Humans , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/therapy
20.
Int J Biol Sci ; 17(15): 4305-4315, 2021.
Article in English | MEDLINE | ID: mdl-34803499

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in hepatocytes. CD38 was initially identified as a lymphocyte surface antigen and then has been found to exist in a variety of cell types. Our previous studies showed that CD38-/- mice were resistant to high-fat diet (HFD)-induced obesity. However, the role and mechanism of CD38 in HFD-induced NAFLD is still unclear. Here, we reported that CD38-/- mice significantly alleviated HFD-induced hepatic steatosis. HFD or oleic acid (OA) remarkably increased the mRNA and protein expressions of CD38 in mouse hepatic tissues and primary hepatocytes or hepatic cell lines in vitro and in vivo, suggesting that CD38 might play a role in HFD-induced hepatic steatosis. We observed that CD38 deficiency markedly decreased HFD- or OA-induced the lipid accumulation and oxidative stress in CD38-/- livers or primary hepatocytes, respectively. In contrast, overexpression of CD38 in Hep1-6 cells aggravated OA-induced lipid accumulation and oxidative stress. Furthermore, CD38 deficiency markedly inhibited HFD- or OA-induced the expressions of NOX4, and increased the expression of PPARα, CPT1, ACOX1 and SOD2 in liver tissue and hepatocytes from CD38-/- mice, indicating that CD38 deficiency-mediated the enhancement of fatty acid oxidation and the inhibition of oxidative stress contributed to protecting NAFLD. More importantly, Ex527 (Sirt1 inhibitor) and 3-TYP (Sirt3 inhibitor) significantly enhanced OA-induced lipid accumulation and oxidative stress in CD38-/- primary hepatocytes, suggesting that the anti-lipid accumulation of CD38 deficiency might be dependent on NAD/Sirtuins-mediated enhancement of FAA ß-oxidation and suppression of oxidative stress in hepatocytes. In conclusion, we demonstrated that CD38 deficiency protected mice from HFD-induced NAFLD by reducing lipid accumulation and suppressing oxidative stress via activating NAD/Sirtuins signaling pathways.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Diet, High-Fat/adverse effects , Lipid Metabolism/drug effects , Membrane Glycoproteins/metabolism , NAD/metabolism , Non-alcoholic Fatty Liver Disease/chemically induced , Sirtuins/metabolism , ADP-ribosyl Cyclase 1/genetics , Animals , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , NAD/genetics , Non-alcoholic Fatty Liver Disease/genetics , Oxidative Stress , Signal Transduction , Sirtuins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL