Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Adv Sci (Weinh) ; : e2404423, 2024 May 20.
Article En | MEDLINE | ID: mdl-38767186

Establishing an advanced ecosystem incorporating freshwater harvesting, plastic utilization, and clean fuel acquisition is profoundly significant. However, low-efficiency evaporation, single energy utilization, and catalyst leakage severely hinder sustainable development. Herein, a nanofiber-based mortise-and-tenon structural Janus aerogel (MTSJA) is strategically designed in the first attempt and supports Z-scheme catalysts. By harnessing of the upper hydrophilic layer with hydrophilic channels embedding into the hydrophobic bottom layer to achieve tailoring bottom wettability states. MTSJA is capable of a fully-floating function for lower heat loss, water supply, and high-efficiency solar-to-vapor conversion. Benefiting from the ultrasonic cavitation effect and high sensitivity of materials to mechanical forces, this is also the first demonstration of synergistic solar and ultrasound fields to power simultaneous evaporation desalination and waste plastics as reusable substrates generating fuel energy. The system enables persistent desalination with an exceptional evaporation rate of 3.1 kg m-2 h-1 and 82.3% efficiency (21 wt.% NaCl solution and 1 sun), and realizes H2, CO, and CH4 yields with 16.1, 9.5, and 3 µmol h-1 g-1, respectively. This strategy holds great potential for desalination and plastics value-added transformation toward clean energy and carbon neutrality.

2.
J Ethnopharmacol ; 327: 117975, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38432576

ETHNOPHARMACOLOGICAL RELEVANCE: Liver fibrosis (LF) is a common reversible consequence of chronic liver damage with limited therapeutic options. Yinchen Gongying decoction (YGD) composed of two homologous plants: (Artemisia capillaris Thunb, Taraxacum monochlamydeum Hand.-Mazz.), has a traditionally application as a medicinal diet for acute icteric hepatitis. However, its impact on LF and underlying mechanisms remain unclear. AIM OF THE STUDY: This study aims to assess the impact of YGD on a carbon tetrachloride (CCl4) induced liver fibrosis and elucidate its possible mechanisms. The study seeks to establish an experimental foundation for YGD as a candidate drug for hepatic fibrosis. MATERIALS AND METHODS: LC-MS/MS identified 11 blood-entry components in YGD, and network pharmacology predicted their involvement in the FoxO signaling pathway, insulin resistance, and PI3K-AKT signaling pathway. Using a CCl4-induced LF mouse model, YGD's protective effects were evaluated in comparison to a positive control and a normal group. The underlying mechanisms were explored through the assessments of hepatic stellate cells (HSCs) activation, fibrotic signaling, and inflammation. RESULTS: YGD treatment significantly improved liver function, enhanced liver morphology, and reduced liver collagen deposition in CCl4-induced LF mice. Mechanistically, YGD inhibited HSC activation, elevated MMPs/TIMP1 ratios, suppressed the FoxO1/TGF-ß1/Smad2/3 and YAP pathways, and exhibited anti-inflammatory and antioxidant effects. Notably, YGD improved the insulin signaling pathway. CONCLUSION: YGD mitigates LF in mice by modulating fibrotic and inflammatory pathways, enhancing antioxidant responses, and specifically inhibiting FoxO1/TGF-ß1/Smad2/3 and YAP signal pathways.


Artemisia , Drugs, Chinese Herbal , Phosphatidylinositol 3-Kinases , Transforming Growth Factor beta1 , Mice , Animals , Transforming Growth Factor beta1/metabolism , Chromatography, Liquid , Phosphatidylinositol 3-Kinases/metabolism , Hepatic Stellate Cells , Tandem Mass Spectrometry , Liver , Signal Transduction , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Carbon Tetrachloride/pharmacology
3.
Cell Mol Gastroenterol Hepatol ; 17(1): 41-58, 2024.
Article En | MEDLINE | ID: mdl-37678798

BACKGROUND & AIMS: The O-class of the forkhead transcription factor FoxO1 is a crucial factor mediating insulin→PI3K→Akt signaling and governs diverse cellular processes. However, the role of hepatocyte FoxO1 in liver fibrosis has not been well-established. In his study, we investigated the role of hepatocyte FoxO1 in liver fibrosis and uncovered the underlying mechanisms. METHODS: Liver fibrosis was established by carbon tetrachloride (CCL4) administration and compared between liver-specific deletion of FoxO1 deletion (F1KO) and control (CNTR) mice. Using genetic and bioinformatic strategies in vitro and in vivo, the role of hepatic FoxO1 in liver fibrosis and associated mechanisms was established. RESULTS: Increased FoxO1 expression and FoxO1 signaling activation were observed in CCL4-induced fibrosis. Hepatic FoxO1 deletion largely attenuated CCL4-induced liver injury and fibrosis compared with CNTR mice. F1KO mice showed ameliorated CCL4-induced hepatic inflammation and decreased TGF-ß1 mRNA and protein levels compared with those of CNTR mice. In primary hepatocytes, FoxO1 deficiency reduced TGF-ß1 expression and secretion. Conditioned medium (CM) collected from wild-type hepatocytes treated with CCL4 activated human HSC cell line (LX-2); such effect was attenuated by FoxO1 deletion in primary hepatocytes or neutralization of TGF-ß1 in the CM using TGF-ß1 antibody. Hepatic FoxO1 overexpression in CNTR mice promoted CCL4-induced HSC activation; such effect was blocked in L-TGF-ß1KO mice. CONCLUSIONS: Hepatic FoxO1 mediates CCL4-inducled liver fibrosis via upregulating hepatocyte TGF-ß1 expression, stimulating hepatic inflammation and TGF-ß1-mediated HSC activation. Hepatic FoxO1 may be a therapeutic target for prevention and treatment of liver fibrosis.


Hepatic Stellate Cells , Transforming Growth Factor beta1 , Animals , Humans , Mice , Hepatic Stellate Cells/pathology , Hepatocytes/metabolism , Inflammation/pathology , Liver Cirrhosis/genetics , Transforming Growth Factor beta1/metabolism
4.
J Endocrinol ; 249(2): 149-161, 2021 04 27.
Article En | MEDLINE | ID: mdl-33760755

Yes-associated protein (YAP), as a co-activator of transcription factors, is a downstream protein in the Hippo signaling pathway with important functions in cell proliferation, apoptosis, invasion and migration. YAP also plays a key role in the development of CCl4-induced liver fibrosis. However, the mechanism of YAP during hepatic fibrosis progression and reversion is still unclear. Mild liver fibrosis was developed after 4 months of high-fat diet (HFD) stimulation, and we found that the YAP signaling pathway was activated. Here, we aim to reveal whether specific knockout of Yap gene in the liver can improve liver fibrosis induced by insulin resistance (IR) stimulated by HFD, and further explain its specific mechanism. We found that liver-specific Yap gene knockout improved IR-induced liver fibrosis and liver dysfunction, and this mechanism is related to the inhibition of the insulin signal pathway at the FoxO1 level. These findings provide a new insight, and Yap is expected to be a new target to reverse the early stage of liver fibrosis induced by IR.


Adaptor Proteins, Signal Transducing/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Diet, High-Fat/adverse effects , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/drug effects , Glucose/metabolism , Glucose Tolerance Test , HEK293 Cells , Hepatocytes/drug effects , Humans , Insulin Resistance , Mice , Mice, Knockout , Signal Transduction , YAP-Signaling Proteins
5.
J Ethnopharmacol ; 263: 113227, 2020 Dec 05.
Article En | MEDLINE | ID: mdl-32783983

ETHNOPHARMACOLOGICAL RELEVANCE: Baihe Wuyao decoction (BWD), a prescription of Traditional Chinese Medicines, composed of Lilium brownii var. viridulum Baker.(Lilii Bulbus) and Lindera aggregata (Sims) Kosterm. (Linderae Radix), has been used to treat epigastric pain and superficial gastritis for hundreds of years in China. Recently, some compounds obtained from Lilii Bulbus and Linderae Radix had active effects of hepatic protection or liver fibrosis alleviation. Thus, we aim to evaluate the effects of BWD on treatment of chronic liver injury and liver fibrosis induced by carbon tetrachloride (CCl4) and to elucidate the possible molecular mechanism. MATERIALS AND METHODS: Mice were treated with BWD (low, medium and high dose), diammonium glycyrrhizinate or vehicle by oral gavage once daily, simultaneously intraperitoneal injected with a single dose of CCl4 (1 µl/g body weight) twice a week for consecutive 6 weeks. Next, all mice were sacrificed after fasted 12 h, and serums and liver tissues were harvested for analysis. The hepatic injury was detected by serum biomarker assay, including aspartate aminotransferase (AST) and alanine aminotransferase (ALT). The hepatic histology and collagen were illustrated by hematoxylin-eosin staining and Sirius red staining respectively. The antioxidant capacity of liver tissues was evaluated by the contents of superoxide dismutase (SOD) and malondialdehyde (MDA) in liver homogenization. The mRNA gene or protein expressions related to fibrosis, oxidative stress and inflammation molecules were performed by real-time quantitative PCR (RT-PCR) or Western-blot. RESULTS: BWD exhibited a good hepatic protection with ameliorating liver histological changes, decreasing serum AST and ALT contents, and reducing hepatic fibrosis with stimulation ECMs (such as Collagen1 and Collagen3) degradation. BWD inhibited hepatic stellate cells (HSCs) activation, promoted matrix metalloproteinase-2 (MMP2), MMP9, and MMP12 while suppressing tissue inhibitors of matrix metalloproteinase-1 (TIMP1) expression, and blocked traditional fibrosis TGF-ß1/Smad2/3 signal pathway. Moreover, BWD exhibited anti-inflammation effect proved by the reduction of liver Interleukin-1ß (IL-1ß), TNF-α, IL-11 mRNA levels and promoted anti-oxidation effects determined by inhibition of liver MDA and iNOS levels while promoting liver SOD and Mn-SOD. CONCLUSION: BWD ameliorates CCl4-induced CLI and liver fibrosis which is correlated to its blocking TGF-ß1/Smad2/3 signaling, anti-inflammation, and anti-oxidation effects. BWD, as a small traditional prescription, is a promising treatment for CLI and liver fibrosis through multiple pharmacological targets.


Drugs, Chinese Herbal/therapeutic use , End Stage Liver Disease/drug therapy , Liver Cirrhosis/drug therapy , Smad2 Protein/antagonists & inhibitors , Smad3 Protein/antagonists & inhibitors , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/isolation & purification , Antioxidants/pharmacology , Antioxidants/therapeutic use , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacology , End Stage Liver Disease/chemically induced , End Stage Liver Disease/metabolism , Liliaceae , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , Signal Transduction/drug effects , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
6.
Phys Rev Lett ; 124(20): 206601, 2020 May 22.
Article En | MEDLINE | ID: mdl-32501055

A quadrupole topological insulator, being one higher-order topological insulator with nontrivial quadrupole quantization, has been intensely investigated very recently. However, the tight-binding model proposed for such emergent topological insulators demands both positive and negative hopping coefficients, which imposes an obstacle in practical realizations. Here, we introduce a feasible approach to design the sign of hopping in acoustics, and construct the first acoustic quadrupole topological insulator that stringently emulates the tight-binding model. The inherent hierarchy quadrupole topology has been experimentally confirmed by detecting the acoustic responses at the bulk, edge, and corner of the sample. Potential applications can be anticipated for the topologically robust in-gap states, such as acoustic sensing and energy trapping.

7.
Pak J Pharm Sci ; 33(2): 597-603, 2020 Mar.
Article En | MEDLINE | ID: mdl-32276903

Diabetic nephropathy is one the most serious diabetic microangiopathies, which is the main cause of mortality in diabetic patients. Our research investigated the protective effects of rutin on kidney of the type 1 diabetes mice induced by streptozotocin (STZ). The levels of kidney weight index (KWI), postprandial plasma glucose (PPG), creatinine (Cre), blood urine nitrogen (BUN), the activity of super oxide dismutase (SOD), malondialdehyde (MDA) and glutathione per oxidase (GSH-Px) were all measured. The histological morphology of kidney tissues was observed by hematoxylin-eosin (HE) staining, masson staining and electron microscope. The collagen I (COL-I) and transforming growth factor-ß1 (TGF-ß1) levels were estimated by immunohistochemistry, western blot and Real-Time PCR respectively. The results revealed that the levels of SOD and GSH-Px all increased, while the levels of KWI, PPG, Cre, BUN and MDA all decreased in diabetic mice after the rutin treatment for eight weeks. Moreover, the histological morphology of kidney tissues was also improved. Furthermore, the expression of COL-I and TGF-ß1 in kidney tissues increased significantly in the diabetic mice, which were antagonized by the rutin treatment. Together, the result suggested that rutin can improve kidney injury of the type 1 diabetic mice.


Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/prevention & control , Diabetic Nephropathies/prevention & control , Kidney/drug effects , Rutin/therapeutic use , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/metabolism , Kidney/metabolism , Male , Mice , Rutin/pharmacology , Streptozocin/toxicity
8.
J Adv Nurs ; 75(11): 3058-3067, 2019 Nov.
Article En | MEDLINE | ID: mdl-31241192

AIM: To evaluate the effectiveness of a mobile application-assisted nurse-led management model in childhood asthma. BACKGROUND: Studies have shown that a nurse-led asthma management model can improve asthma outcomes. However, the role of a mobile application-assisted nurse-led model in paediatric asthma management has not been studied well. DESIGN: A multi-centre randomized clinical trial. METHODS: The trial was conducted between March 2017-March 2018. A total of 152 children (6 to 11.9 years old) were enrolled, with 77 children in the experimental group and 75 in the control group. All children received nurse-led asthma management and other routine treatment measures, including inhaled corticosteroids. Meanwhile, a mobile application was used to manage asthma only for children in the experimental group. Primary outcome was frequency of asthma exacerbations. All outcomes were evaluated twice a month for 12 months. RESULTS: Compared with the pre-enrollment period, frequency of asthma exacerbations decreased in the post-enrollment period in the two groups, with a greater decrease in the experimental group. Compared with children in the control group, children in the experimental group had better secondary outcomes, such as improved adherence, higher Childhood Asthma Control Test scores, decreased respiratory tract infections, days of antibiotic use, days of school absence, parental work loss, and medical expenses. CONCLUSION: A mobile application-assisted nurse-led management model decreased asthma exacerbations and improved secondary outcomes in children with asthma. Further research is needed to verify its validity in larger population samples. IMPACT: Children with asthma benefited from a nurse-led asthma management model when combined with mobile application. This trial suggested that computer and Internet technologies should be incorporated into nurse-led asthma strategy in paediatric asthma management. TRIAL REGISTRATION: The current trial was registered online with the Chinese Clinical Trial Registry (registration number: ChiCTR1800016726).


Adrenal Cortex Hormones/therapeutic use , Asthma/drug therapy , Asthma/nursing , Mobile Applications , Nursing Care/methods , Reminder Systems , Telemedicine/methods , Child , Child, Preschool , China , Female , Humans , Male , Treatment Outcome
9.
Endocrinology ; 160(5): 1333-1347, 2019 05 01.
Article En | MEDLINE | ID: mdl-30951171

The transcription factor forkhead box O1 (FoxO1) is a key mediator in the insulin signaling pathway and controls multiple physiological functions, including hepatic glucose production (HGP) and pancreatic ß-cell function. We previously demonstrated that S256 in human FOXO1 (FOXO1-S256), equivalent to S253 in mouse FoxO1 (FoxO1-S253), is a key phosphorylation site mediating the effect of insulin as a target of protein kinase B on suppression of FOXO1 activity and expression of target genes responsible for gluconeogenesis. Here, we investigated the role of FoxO1-S253 phosphorylation in control of glucose homeostasis in vivo by generating global FoxO1-S253A/A knockin mice, in which FoxO1-S253 alleles were replaced with alanine (A substitution) blocking FoxO1-S253 phosphorylation. FoxO1-S253A/A mice displayed mild increases in feeding blood glucose and insulin levels but decreases in fasting blood glucose and glucagon concentrations, as well as a reduction in the ratio of pancreatic α-cells/ß-cells per islet. FoxO1-S253A/A mice exhibited a slight increase in energy expenditure but barely altered food intake and glucose uptake among tissues. Further analyses revealed that FoxO1-S253A/A enhances FoxO1 nuclear localization and promotes the effect of glucagon on HGP. We conclude that dephosphorylation of S253 in FoxO1 may reflect a molecular basis of pancreatic plasticity during the development of insulin resistance.


Forkhead Box Protein O1/metabolism , Glucose/metabolism , Homeostasis , Serine/metabolism , Alanine/genetics , Alanine/metabolism , Animals , Blood Glucose/metabolism , Forkhead Box Protein O1/genetics , Glucagon/blood , Insulin/blood , Insulin Resistance , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Serine/genetics
10.
Diabetes ; 67(11): 2167-2182, 2018 11.
Article En | MEDLINE | ID: mdl-30201683

Dysregulation of hepatic glucose production (HGP) serves as a major underlying mechanism for the pathogenesis of type 2 diabetes. The pancreatic hormone glucagon increases and insulin suppresses HGP, controlling blood glucose homeostasis. The forkhead transcription factor Foxo1 promotes HGP through increasing expression of genes encoding the rate-limiting enzymes responsible for gluconeogenesis. We previously established that insulin suppresses Foxo1 by Akt-mediated phosphorylation of Foxo1 at Ser256 in human hepatocytes. In this study, we found a novel Foxo1 regulatory mechanism by glucagon, which promotes Foxo1 nuclear translocation and stability via cAMP- and protein kinase A-dependent phosphorylation of Foxo1 at Ser276 Replacing Foxo1-S276 with alanine (A) or aspartate (D) to block or mimic phosphorylation, respectively, markedly regulates Foxo1 stability and nuclear localization in human hepatocytes. To establish in vivo function of Foxo1-Ser276 phosphorylation in glucose metabolism, we generated Foxo1-S273A and Foxo1-S273D knock-in (KI) mice. The KI mice displayed impaired blood glucose homeostasis, as well as the basal and glucagon-mediated HGP in hepatocytes. Thus, Foxo1-Ser276 is a new target site identified in the control of Foxo1 bioactivity and associated metabolic diseases.


Forkhead Box Protein O1/metabolism , Glucagon/metabolism , Glucose/metabolism , Homeostasis/physiology , Signal Transduction/physiology , Animals , Gluconeogenesis/physiology , Glycogenolysis/physiology , Hepatocytes/metabolism , Mice , Mice, Transgenic , Phosphorylation
11.
J Mol Cell Cardiol ; 116: 165-174, 2018 03.
Article En | MEDLINE | ID: mdl-29452158

The slowly activating delayed rectifier K+ current (IKs) is one of the main repolarizing currents in the human heart. Evidence has shown that angiotensin II (Ang II) regulates IKs through the protein kinase C (PKC) pathway, but the related results are controversial. This study was designed to identify PKC isoenzymes involved in the regulation of IKs by Ang II and the underlying molecular mechanism. The whole-cell patch-clamp technique was used to record IKs in isolated guinea pig ventricular cardiomyocytes and in human embryonic kidney (HEK) 293 cells co-transfected with human KCNQ1/KCNE1 genes and Ang II type 1 receptor genes. Ang II inhibited IKs in a concentration-dependent manner in native cardiomyocytes. A broad PKC inhibitor Gö6983 (not inhibiting PKCε) and a selective cPKC inhibitor Gö6976 did not affect the inhibitory action of Ang II. In contrast, the inhibition was significantly attenuated by PKCε-selective peptide inhibitor εV1-2. However, direct activation of PKC by phorbol 12-myristate 13-acetate (PMA) increased the cloned human IKs in HEK293 cells. Similarly, the cPKC peptide activator significantly enhanced the current. In contrast, the PKCε peptide activator inhibited the current. Further evidence showed that PKCε knockdown by siRNA antagonized the Ang II-induced inhibition on KCNQ1/KCNE1 current, whereas knockdown of cPKCs (PKCα and PKCß) attenuated the potentiation of the current by PMA. Moreover, deletion of four putative phosphorylation sites in the C-terminus of KCNQ1 abolished the action of PMA. Mutation of two putative phosphorylation sites in the N-terminus of KCNQ1 and one site in KCNE1 (S102) blocked the inhibition of Ang II. Our results demonstrate that PKCε isoenzyme mediates the inhibitory action of Ang II on IKs and by phosphorylating distinct sites in KCNQ1/KCNE1, cPKC and PKCε isoenzymes produce the contrary regulatory effects on the channel. These findings have provided new insight into the molecular mechanism underlying the modulation of the KCNQ1/KCNE1 channel.


Angiotensin II/pharmacology , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Kinase C-epsilon/metabolism , Animals , Enzyme Activation/drug effects , Guinea Pigs , HEK293 Cells , Heart Ventricles/metabolism , Humans , Isoenzymes/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Tetradecanoylphorbol Acetate/pharmacology
12.
J Mol Cell Cardiol ; 85: 155-67, 2015 Aug.
Article En | MEDLINE | ID: mdl-26055448

Coxsackievirus B3 (CVB3) is the major pathogen of human viral myocarditis. CVB3 has been found to manipulate and modify the cellular lipid metabolism for viral replication. The cellular AMP-activated protein kinase (AMPK) is a key regulator of multiple metabolic pathways, including lipid metabolism. Here we explore the potential roles AMPK plays in CVB3 infection. We found that AMPK is activated by the viral replication during CVB3 infection in Hela cells and primary myocardial cells. RNA interference mediated inhibition of AMPK could increase the CVB3 replication in cells, indicating that AMPK contributed to restricting the viral replication. Next, we showed that CVB3 replication could be inhibited by several different pharmacological AMPK activators including metformin, A769662 and AICAR. And the constitutively active AMPK mutant (CA-AMPK) could also inhibit the CVB3 replication. Furthermore, we found that CVB3 infection increased the cellular lipid levels and showed that the AMPK agonist AICAR both restricted CVB3 replication and reduced lipid accumulation through inhibiting the lipid synthesis associated gene expression. We further found that CVB3 infection would also induce AMPK activated in vivo. The AMPK agonist metformin, which has been widely used in diabetes therapy, could decrease the viral replication and further protect the mice from myocardial histological and functional changes in CVB3 induced myocarditis, and improve the survival rate of infected mice. Lastly, it was demonstrated that the AICAR-mediated restriction of viral replication could be rescued partially by exogenous palmitate, the first product of fatty acid biosynthesis, demonstrating that AMPK activation restricted CVB3 infection through its inhibition of lipid synthesis. Taken together, these data in the present study suggest a model in which AMPK is activated by CVB3 infection and restricts viral replication by inhibiting the cellular lipid accumulation, and inform a potential novel therapeutic strategy for CVB3-associated diseases.


Adenylate Kinase/metabolism , Enterovirus B, Human/physiology , Lipid Metabolism , Virus Replication , Animals , Enzyme Activation , HeLa Cells , Humans , Male , Mice, Inbred BALB C , Mice, Obese
13.
Circ Heart Fail ; 8(1): 198-208, 2015 Jan.
Article En | MEDLINE | ID: mdl-25477432

BACKGROUND: Heart failure is a leading cause of morbidity and mortality in the USA and is closely associated with diabetes mellitus. The molecular link between diabetes mellitus and heart failure is incompletely understood. We recently demonstrated that insulin receptor substrates 1, 2 (IRS1, 2) are key components of insulin signaling and loss of IRS1 and IRS2 mediates insulin resistance, resulting in metabolic dysregulation and heart failure, which is associated with downstream Akt inactivation and in turn activation of the forkhead transcription factor Foxo1. METHODS AND RESULTS: To determine the role of Foxo1 in control of heart failure in insulin resistance and diabetes mellitus, we generated mice lacking Foxo1 gene specifically in the heart. Mice lacking both IRS1 and IRS2 in adult hearts exhibited severe heart failure and a remarkable increase in the ß-isoform of myosin heavy chain (ß-MHC) gene expression, whereas deletion of cardiac Foxo1 gene largely prevented the heart failure and resulted in a decrease in ß-MHC expression. The effect of Foxo1 deficiency on rescuing cardiac dysfunction was also observed in db/db mice and high-fat diet mice. Using cultures of primary ventricular cardiomyocytes, we found that Foxo1 interacts with the promoter region of ß-MHC and stimulates gene expression, mediating an effect of insulin that suppresses ß-MHC expression. CONCLUSIONS: Our study suggests that Foxo1 has important roles in promoting diabetic cardiomyopathy and controls ß-MHC expression in the development of cardiac dysfunction. Targeting Foxo1 and its regulation will provide novel strategies in preventing metabolic and myocardial dysfunction and influencing MHC plasticity in diabetes mellitus.


Diabetic Cardiomyopathies/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Insulin Resistance/genetics , Myosin Heavy Chains/genetics , RNA/genetics , Ventricular Function, Left/physiology , Animals , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Forkhead Box Protein O1 , Immunoprecipitation , Mice , Mice, Inbred C57BL , Microscopy, Electron , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Myosin Heavy Chains/biosynthesis , Real-Time Polymerase Chain Reaction
14.
Hypertension ; 64(5): 1131-40, 2014 Nov.
Article En | MEDLINE | ID: mdl-25069665

The renin-angiotensin system is a major determinant of blood pressure regulation. It consists of a cascade of enzymatic reactions involving 3 components: angiotensinogen, renin, and angiotensin-converting enzyme, which generate angiotensin II as a biologically active product. Angiotensinogen is largely produced in the liver, acting as a major determinant of the circulating renin-angiotensin system, which exerts acute hemodynamic effects on blood pressure regulation. How the expression of angiotensinogen is regulated is not completely understood. Here, we hypothesize that angiotensinogen is regulated by forkhead transcription factor forkhead box class O1 (Foxo1), an insulin-suppressed transcription factor, and thereby controls blood pressure in mice. We generated liver-specific Foxo1 knockout mice, which exhibited a reduction in plasma angiotensinogen and angiotensin II levels and a significant decrease in blood pressure. Using hepatocyte cultures, we demonstrated that overexpression of Foxo1 increased angiotensinogen expression, whereas hepatocytes lacking Foxo1 demonstrated a reduction of angiotensinogen gene expression and partially impaired insulin inhibition on angiotensinogen gene expression. Furthermore, mouse angiotensinogen prompter analysis demonstrated that the angiotensinogen promoter region contains a functional Foxo1-binding site, which is responsible for both Foxo1 stimulation and insulin suppression on the promoter activity. Together, these data demonstrate that Foxo1 regulates hepatic angiotensinogen gene expression and controls plasma angiotensinogen and angiotensin II levels, modulating blood pressure control in mice.


Angiotensinogen/physiology , Blood Pressure/physiology , Forkhead Transcription Factors/physiology , Hemodynamics/physiology , Liver/physiology , Angiotensin II/drug effects , Angiotensin II/genetics , Angiotensin II/physiology , Angiotensinogen/drug effects , Angiotensinogen/genetics , Animals , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/physiology , Insulin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Renin-Angiotensin System/physiology
15.
Am J Physiol Heart Circ Physiol ; 306(2): H233-42, 2014 Jan 15.
Article En | MEDLINE | ID: mdl-24213609

The related transcriptional enhancer factor-1 (RTEF-1) increases gene transcription of hypoxia-inducible factor 1α (HIF-1α) and enhances angiogenesis in endothelium. Both hypoxia and inflammatory factor TNF-α regulate gene expression of HIF-1α, but how RTEF-1 and TNF-α coordinately regulate HIF-1α gene transcription is unclear. Here, we found that RTEF-1 interacts with p65 subunit of NF-κB, a primary mediator of TNF-α. RTEF-1 increased HIF-1α promoter activity, whereas expression of p65 subunit inhibited the stimulatory effect. By contrast, knockdown of p65 markedly enhanced RTEF-1 stimulation on the HIF-1α promoter activity (7-fold). A physical interaction between RTEF-1 and p65 was confirmed by coimmunoprecipitation experiments in cells and glutathione S-transferase (GST)-pull-down assays. A computational analysis of RTEF-1 crystal structures revealed that a conserved surface of RTEF-1 potentially interacts with p65 via four amino acid residues located at T347, Y349, R351, and Y352. We performed site-directed mutagenesis and GST-pull-down assays and demonstrated that Tyr352 (Y352) in RTEF-1 is a key site for the formation of RTEF-1 and p65-NF-κB complex. An alanine mutation at Y352 of RTEF-1 disrupted the interaction of RTEF-1 with p65. Moreover, expression of RTEF-1 decreased TNF-α-induced HIF-1α promoter activity, IL-1ß, and IL-6 mRNA levels in cells; however, the effect of RTEF-1 was largely lost when Y352 was mutated to alanine. These results indicate that RTEF-1 interacts with p65-NF-κB through Y352 and that they antagonize each other for HIF-1α transcriptional activation, suggesting a novel mechanism by which RTEF-1 regulates gene expression, linking hypoxia to inflammation.


DNA-Binding Proteins/metabolism , Molecular Docking Simulation , Muscle Proteins/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Sequence Data , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutagenesis, Site-Directed , Mutation, Missense , Promoter Regions, Genetic , Protein Binding , TEA Domain Transcription Factors , Transcription Factor RelA/chemistry , Transcription Factor RelA/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic
16.
Diabetes ; 62(11): 3887-900, 2013 Nov.
Article En | MEDLINE | ID: mdl-24159000

Cardiac failure is a major cause of death in patients with type 2 diabetes, but the molecular mechanism that links diabetes to heart failure remains unclear. Insulin resistance is a hallmark of type 2 diabetes, and insulin receptor substrates 1 and 2 (IRS1 and IRS2) are the major insulin-signaling components regulating cellular metabolism and survival. To determine the role of IRS1 and IRS2 in the heart and examine whether hyperinsulinemia causes myocardial insulin resistance and cellular dysfunction via IRS1 and IRS2, we generated heart-specific IRS1 and IRS2 gene double-knockout (H-DKO) mice and liver-specific IRS1 and IRS2 double-knockout (L-DKO) mice. H-DKO mice had reduced ventricular mass; developed cardiac apoptosis, fibrosis, and failure; and showed diminished Akt→forkhead box class O-1 signaling that was accompanied by impaired cardiac metabolic gene expression and reduced ATP content. L-DKO mice had decreased cardiac IRS1 and IRS2 proteins and exhibited features of heart failure, with impaired cardiac energy metabolism gene expression and activation of p38α mitogen-activated protein kinase (p38). Using neonatal rat ventricular cardiomyocytes, we further found that chronic insulin exposure reduced IRS1 and IRS2 proteins and prevented insulin action through activation of p38, revealing a fundamental mechanism of cardiac dysfunction during insulin resistance and type 2 diabetes.


Heart Failure/etiology , Hyperinsulinism/physiopathology , Insulin Receptor Substrate Proteins/deficiency , Insulin Resistance/physiology , Mitogen-Activated Protein Kinase 14/physiology , Signal Transduction/drug effects , Animals , Energy Metabolism/genetics , Insulin/physiology , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats
17.
Endocrinology ; 153(2): 631-46, 2012 Feb.
Article En | MEDLINE | ID: mdl-22147007

Dysregulation of blood glucose and triglycerides are the major characteristics of type 2 diabetes mellitus. We sought to identify the mechanisms regulating blood glucose and lipid homeostasis. Cell-based studies established that the Foxo forkhead transcription factors Forkhead box O (Foxo)-1, Foxo3, and Foxo4 are inactivated by insulin via a phosphatidylinositol 3-kinase/Akt-dependent pathway, but the role of Foxo transcription factors in the liver in regulating nutrient metabolism is incompletely understood. In this study, we used the Cre/LoxP genetic approach to delete the Foxo1, Foxo3, and Foxo4 genes individually or a combination of two or all in the liver of lean or db/db mice and assessed the role of Foxo inactivation in regulating glucose and lipid homeostasis in vivo. In the lean mice or db/db mice, hepatic deletion of Foxo1, rather than Foxo3 or Foxo4, caused a modest reduction in blood glucose concentrations and barely affected lipid homeostasis. Combined deletion of Foxo1 and Foxo3 decreased blood glucose levels, elevated serum triglyceride and cholesterol concentrations, and increased hepatic lipid secretion and caused hepatosteatosis. Analysis of the liver transcripts established a prominent role of Foxo1 in regulating gene expression of gluconeogenic enzymes and Foxo3 in the expression of lipogenic enzymes. Our findings indicate that Foxo1 and Foxo3 inactivation serves as a potential mechanism by which insulin reduces hepatic glucose production and increases hepatic lipid synthesis and secretion in healthy and diabetic states.


Forkhead Transcription Factors/metabolism , Hyperlipidemias/metabolism , Hypoglycemia/metabolism , Liver/metabolism , Animals , Blood Glucose/metabolism , Cell Cycle Proteins , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Gluconeogenesis/physiology , Glucose/metabolism , Homeostasis , Lipid Metabolism/physiology , Mice , Mice, Inbred NOD , Mice, Knockout
18.
Acta Pharmacol Sin ; 29(11): 1313-8, 2008 Nov.
Article En | MEDLINE | ID: mdl-18954525

AIM: To determine whether different Na+/K+-ATPase signal transduction pathways have positive inotropic effects on normal ventricular myocytes (NC) and failing ventricular myocytes (FC), and are involved in an increase of [Ca2+]i induced by strophanthidin (Str). METHODS: A guinea pig model of congestive heart failure was made by constricting descending aorta. The left ventricular myocytes were enzymatically isolated. The effects of 25 micromol/L Str with different signal-transducing inhibitors on contractility and the calcium transient of NC or FC from guinea pigs were simultaneously assessed and compared with those in the 25 micromol/L Str-only group by a video-based, motion-edge detection system. RESULTS: Str at 1, 10, and 25 micromol/L in NC and Str at 0.1, 1, 10, and 25 micromol/L) in FC elevated the calcium transient amplitude and increased the positive inotropic effects in a concentration-dependent manner, respectively. At the same concentration, the effects of Str were more potent in FC than in NC. In FC, both the mitogen-activated protein kinase (MAPK) and reactive oxygen species (ROS) signal transduction pathway of Na+/K+-ATPase were involved in the increase of the calcium transient induced by Str, but only activation of the MAPK pathway increased the calcium transient in NC. However, only the ROS pathway was involved in positive inotropic effects both in NC and FC. CONCLUSION: The present study suggests that Na+/K+-ATPase signaling pathways involved in the inotropic effects of Str in NC and FC are consistent, and Na+/K+-ATPase signaling pathways involved in the increase of [Ca2+]i by Str in NC and FC are different.


Calcium/pharmacology , Myocytes, Cardiac/metabolism , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/physiology , Strophanthidin/pharmacology , Animals , Guinea Pigs , Heart Failure/drug therapy , Heart Failure/physiopathology , Heart Ventricles/cytology , Heart Ventricles/drug effects , In Vitro Techniques , Male , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Signal Transduction/drug effects
19.
Yao Xue Xue Bao ; 43(3): 259-66, 2008 Mar.
Article En | MEDLINE | ID: mdl-18630261

Effect of strophanthidin (Str) on intracellular calcium concentration ([Ca2+]i) was investigated on isolated ventricular myocytes of guinea pig. Single ventricular myocytes were obtained by enzymatic dissociation technique. Fluorescent signal of [Ca2+]i was detected with confocal microscopy after incubation of cardiomycytes in Tyrode' s solution with Fluo3-AM. The result showed that Str increased [Ca2+]i in a concentration-dependent manner. The ventricular myocytes began to round-up into a contracture state once the peak level of [Ca2+]i was achieved in the presence of Str (10 micromol L(- 1)), but remained no change in the presence of Str (1 and 100 nmol L(-1)). Tetrodotoxin (TTX), nisodipine, and high concentration of extracellular Ca2+ changed the response of cardiomycytes to Str (1 and 100 nmol L(-1)) , but had no obvious effects on the action of Str (10 micromol L(-1)). The elevation of [Ca2+]i caused by Str at all of the detected concentrations was partially antagonized by rynodine (10 micromol L(-1)) or the removal of Ca2+ from Tyrode's solution. In Na+, K+ -free Tyrode' s solution, the response of cardiomycytes in [Ca2+]i elevation to Str (10 micromol L(-1)) was attenuated, while remained no change to Str (1 and 100 nmol L(-1)). TTX, nisodipine, and high concentration of extracellular Ca2+ changed the response of cardiomycytes to Str at all of the detected concentrations in Na+, K+ -free Tyrode's solution. The study suggests that the elevation of [Ca2+]i by Str at the low (nomomolar) concentrations is partially mediated by the extracellular calcium influx through Ca2+ channel or a "slip mode conductance" of TTX sensitive Na+ channel. While the effect of Str at high (micromolar) concentrations was mainly due to the inhibition of Na+, K+ -ATPase. Directly triggering the release of intracellular Ca2+ from sarcoplasmic reticulum (SR) by Str may be also involved in the mechanism of [Ca2+]i elevation.


Calcium Channels/metabolism , Calcium/metabolism , Sarcolemma/pathology , Sarcoplasmic Reticulum/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Strophanthidin/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Aequorin/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Fura-2/pharmacology , Fura-2/supply & distribution , Guinea Pigs , Myocardium/pathology , Nifedipine/pharmacology , Ryanodine/pharmacology , Sarcolemma/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology
...