Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Med Sci Sports Exerc ; 55(7): 1172-1183, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36878020

ABSTRACT

PURPOSE: The aim of this study was to investigate the function and mechanisms of ELABELA (ELA) in the aerobic exercise-induced antiapoptosis and angiogenesis of ischemic heart. METHODS: The myocardial infarction (MI) model of Sprague-Dawley rat was established by the ligation of the left anterior descending coronary artery. MI rats underwent 5 wk of Fc-ELA-21 subcutaneous injection and aerobic exercise training using a motorized rodent treadmill. Heart function was evaluated by hemodynamic measures. Cardiac pathological remodeling was evaluated by Masson's staining and the calculation of left ventricular weight index. Cell proliferation, angiogenesis, and Yes-associated protein (YAP) translocation were observed by immunofluorescence staining. Cell apoptosis was analyzed by TUNEL. Cell culture and treatment were used to elucidate the molecular mechanism of ELA. Protein expression was detected by Western blotting. Angiogenesis was observed by tubule formation test. One-way or two-way ANOVA and Student's t -test were used for statistical analysis. RESULTS: Aerobic exercise stimulated the endogenous ELA expression. Exercise and Fc-ELA-21 intervention significantly activated APJ-Akt-mTOR-P70S6K signaling pathway, kept more cardiomyocytes alive, and increased angiogenesis, so as to inhibit the cardiac pathological remodeling and improved the heart function of MI rats. Fc-ELA-32 also had the cellular and functional cardioprotective activities in vivo . In vitro , ELA-14 peptide regulated the phosphorylation and nucleoplasmic translocation of YAP and activated the APJ-Akt signaling pathway so as to increase the proliferation of H9C2 cells. Moreover, the antiapoptosis and the tubule formation of HUVECs were also enhanced by ELA-14, whereas the inhibition of Akt activity weakened such effects. CONCLUSIONS: ELA is a potential therapeutic member that plays a key role through APJ-Akt/YAP signaling axis in aerobic exercise-induced cardioprotection of MI rats.


Subject(s)
Myocardial Infarction , Proto-Oncogene Proteins c-akt , Rats , Animals , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Myocardial Infarction/prevention & control , Signal Transduction , Myocytes, Cardiac/metabolism
2.
Mol Biotechnol ; 65(3): 394-400, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35960440

ABSTRACT

Apelin receptor (APJ) ligands elabela (ELA) and apelin have divergent distributions and function differently in vitro and in vivo. Whether differences exist in their capacity of recruitment of ß-arrestins (ARRBs) to APJ remains unknown. The aim of the current study was to investigate the different effects of ELA and apelin on the interaction between APJ and ARRBs in live cells by NanoBiT®. NanoBiT® system is a new technology for studying protein-protein interaction in real-time in live cells, based on the emission of luminescence when two split components of NanoLuc luciferase, large Bit (LgBit) and small Bit (SmBit), complement each other to form an enzymatically active entity. We tagged the APJ and ARRBs with LgBit or SmBit and then evaluated their interactions in transiently transfected HEK293T cells, and determined the signal strength yielded as a result of the interaction. We also investigated the concentration-dependent response of the APJ-ARRB interaction in response to ELA and apelin. Finally, we assessed the effect of F13A, an APJ antagonist which is structurally very similar to apelin-13, on ELA- and apelin-mediated APJ-ARRB interactions. The NanoLuc® luciferase signal was highest in the pair of APJ-LgBit with SmBit-ARRB1 or SmBit-ARRB2. NanoLuc® luciferase signal increased in a concentration-dependent manner from 0.1 nM to 10 µM in response to ELA or apelin. Interestingly, ELA elicited weaker APJ-ARRB interaction signals than apelin. Pre-treatment with F13A potently reduced the APJ-ARRB interaction in response to both ELA and apelin. Our results demonstrated that both ELA and apelin promoted the interaction of APJ and ARRBs in a concentration-dependent manner, and ELA is less efficacious than apelin in inducing the recruitment of ARRBs to APJ, providing a biased functional aspect of ELA vs. apelin at the receptor signaling level. Additionally, ELA and apelin may share the same binding site(s) or pocket(s) at the APJ level.


Subject(s)
Apelin Receptors , Humans , Apelin/metabolism , Apelin Receptors/metabolism , beta-Arrestins/metabolism , Binding Sites , HEK293 Cells
3.
Peptides ; 147: 170682, 2022 01.
Article in English | MEDLINE | ID: mdl-34742787

ABSTRACT

INTRODUCTION: Renal ischemia/reperfusion injury (IRI) is the most common cause of acute kidney injury (AKI), and patients with AKI have a high rate of mortality. Apelin is a therapeutic candidate for treatment of IRI and Elabela (ELA) is a recently discovered hormone that also activates the apelin receptor (APJ). We examined the use of ELA as a preventive treatment for IRI using in vitro and in vivo models. METHODS: Male mice were subjected to renal IRI, with or without administration of a stabilized form of ELA (Fc-ELA-21) for 4 days. Renal tubular lesions were measured using H&E staining, reactive oxygen species (ROS) were measured using a dihydroethidium stain assay, and renal cell apoptosis was measured using the TUNEL assay and flow cytometry. Immortalized human proximal tubular epithelial (HK-2) cells were pretreated with or without LY294002 and/or ELA-32, maintained at normoxic or hypoxic conditions, and then returned to normal culture conditions to mimic IRI. Cell apoptosis was determined using the TUNEL assay and cell proliferation was determined using the MTT assay. The levels of Akt, p-Akt, ERK1/2, p- ERK1/2, Bcl-2, Bax, caspase-3 and cleaved caspase-3 were measured using western blotting. RESULTS: Fc-ELA-21 administration reduced renal tissue damage, ROS production, and apoptosis in mice that had renal IRI. ELA-32 reduced HK-2 cell apoptosis and restored the proliferation of cells subjected to IRI. Akt phosphorylation had a role in the anti-apoptotic effect of ELA. CONCLUSION: This study of in vitro and in vivo models of IRI indicated that the preventive and anti-apoptotic effects of ELA were mediated via the PI3K/Akt signaling pathway.


Subject(s)
Acute Kidney Injury/drug therapy , Apoptosis/drug effects , Peptide Hormones/pharmacology , Reperfusion Injury/drug therapy , Acute Kidney Injury/pathology , Animals , Apoptosis/physiology , Cell Line , Cell Proliferation/drug effects , Half-Life , Humans , Kidney Tubules/cytology , Male , Mice, Inbred C57BL , Peptide Hormones/chemistry , Peptide Hormones/metabolism , Peptide Hormones/pharmacokinetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
4.
Adipocyte ; 10(1): 524-531, 2021 12.
Article in English | MEDLINE | ID: mdl-34670478

ABSTRACT

Peroxisome proliferator-activated receptor-γ (PPARγ) is the master regulator of adipogenesis, but knowledge about how PPARγ is regulated at the protein level is very limited. We aimed to identify PPARγ-interacting proteins which modulate PPARγ's protein levels and transactivating activities in human adipocytes. We expressed Flag-tagged PPARγ in human preadipocytes as bait to capture PPARγ-associated proteins, followed by mass spectroscopy and proteomics analysis, which identified serine/threonine kinase 38 (STK38) as a major PPARγ-associated protein. Protein pulldown studies confirmed this protein-protein interaction in transfected cells, and reporter assays demonstrated that STK38 enhanced PPARγ's transactivating activities without requiring STK38's kinase activity. In cell-based assays, STK38 increased PPARγ protein stability, extending PPARγ's half-life from ~1.08 to 1.95 h. Notably, in human preadipocytes, the overexpression of STK38 enhanced adipogenesis, whereas knockdown impaired the process in a PPARγ-dependent manner. Thus, we discovered that STK38 is a novel PPARγ-cofactor promoting adipogenesis, likely through stabilization of PPARγ.


Subject(s)
Adipogenesis , PPAR gamma , Protein Serine-Threonine Kinases , Adipocytes/metabolism , Humans , PPAR gamma/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics
5.
J Sport Health Sci ; 10(5): 594-603, 2021 09.
Article in English | MEDLINE | ID: mdl-33246164

ABSTRACT

PURPOSE: The aim of this study was to investigate the potential of dynamic resistance exercise to generate skeletal muscle-derived follistatin like-1 (FSTL1), which may induce cardioprotection in rats following myocardial infarction (MI) by inducing angiogenesis. METHODS: Male, adult Sprague-Dawley rats were randomly divided into 5 groups (n = 12 in each group): sham group (S), sedentary MI group (MI), MI + resistance exercise group (MR), MI + adeno-associated virus (AAV)-FSTL1 injection group (MA), and MI + AAV-FSTL1 injection + resistance exercise group (MAR). The AAV-FSTL1 vector was prepared by molecular biology methods and injected into the anterior tibialis muscle. The MI model was established by ligation of the left anterior descending coronary artery. Rats in the MR and MAR groups underwent 4 weeks of dynamic resistance exercise training using a weighted climbing-up ladder. Heart function was evaluated by hemodynamic measures. Collagen volume fraction of myocardium was observed and analyzed by Masson's staining. Human umbilical vein vessel endothelial cells culture and recombinant human FSTL1 protein or transforming growth factor-ß receptor 1 (TGFßR1) inhibitor treatment were used to elucidate the molecular signaling mechanism of FSTL1. Angiogenesis, cell proliferation, and disco interacting protein 2 homolog A (DIP2A) location were observed by immunofluorescence staining. The expression of FSTL1, DIP2A, and the activation of signaling pathways were detected by Western blotting. Angiogenesis of endothelial cells was observed by tubule experiment. One-way analysis of variance and Student's t test were used for statistical analysis. RESULTS: Resistance exercise stimulated the secretion of skeletal muscle FSTL1, which promoted myocardial angiogenesis, inhibited pathological remodeling, and protected cardiac function in MI rats. Exercise facilitated skeletal muscle FSTL1 to play a role in protecting the heart. Exogenous FSTL1 promoted the human umbilical vein vessel endothelial cells proliferation and up-regulated the expression of DIP2A, while TGFßR1 inhibitor intervention down-regulated the phosphorylation level of Smad2/3 and the expression of vascular endothelial growth factor-A, which was not conducive to angiogenesis. FSTL1 bound to the receptor, DIP2A, to regulate angiogenesis mainly through the Smad2/3 signaling pathway. FSTL1-DIP2A directly activated Smad2/3 and was not affected by TGFßR1. CONCLUSION: Dynamic resistance exercise stimulates the expression of skeletal muscle-derived FSTL1, which could supplement the insufficiency of cardiac FSTL1 and promote cardiac rehabilitation through the DIP2A-Smad2/3 signaling pathway in MI rats.


Subject(s)
Angiogenesis Inducing Agents , Follistatin-Related Proteins/pharmacology , Myocardial Infarction/therapy , Nuclear Proteins/metabolism , Physical Conditioning, Animal/methods , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Animals , Disease Models, Animal , Male , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
6.
Biosci Rep ; 40(9)2020 09 30.
Article in English | MEDLINE | ID: mdl-32808659

ABSTRACT

Endotoxemia-induced acute kidney injury (AKI) is a common clinical condition that lacks effective treatments. Elabela (ELA) is a recently discovered kidney peptide hormone, encoded by the gene apela, and has been reported to improve cardio-renal outcomes in sepsis. However, ELA is a small peptide and is largely unsuitable for clinical use because of its short in vivo half-life. In the present study, we evaluated the potential renoprotective effects of a long-acting constant fragment (Fc)-ELA fusion protein in liposaccharide (LPS)-induced AKI in mice. LPS administration in mice for 5 days greatly lowered the gene expression of apela and impaired kidney function, as evidenced by elevated serum creatinine and the ratio of urine protein to creatinine. In addition, renal inflammation and macrophage infiltration were apparent in LPS-challenged mice. Treatment with the Fc-ELA fusion protein partially restored apela expression and attenuated the kidney inflammation. Moreover, LPS treatment induced reactive oxygen species (ROS) production and apoptosis in kidney HK-2 cells as well as in the mouse kidney, which were mitigated by ELA or Fc-ELA treatment. Finally, we found that ELA promoted the survival of HK-2 cells treated with LPS, and this action was abolished by LY204002, a PI3K/Akt inhibitor. Collectively, we have demonstrated that the Fc-ELA fusion protein has significant renoprotective activities against LPS-induced AKI in mice.


Subject(s)
Acute Kidney Injury/drug therapy , Immunoglobulin Fc Fragments/genetics , Lipopolysaccharides/toxicity , Peptide Hormones/genetics , Recombinant Fusion Proteins/pharmacology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Animals , Apoptosis/drug effects , Cell Line , Female , Gene Expression/drug effects , Humans , Immunoglobulin Fc Fragments/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mice, Inbred C57BL , Peptide Hormones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics
7.
Acta Diabetol ; 57(6): 705-713, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32008161

ABSTRACT

AIMS: Type 2 diabetes mellitus (T2DM) is now very prevalent in China. Due to the lower rate of controlled diabetes in China compared to that in developed countries, there is a higher incidence of serious cardiovascular complications, especially acute coronary syndrome (ACS). The aim of this study was to establish a potent risk predictive model in the economically disadvantaged northwest region of China, which could predict the probability of new-onset ACS in patients with T2DM. METHODS: Of 456 patients with T2DM admitted to the First Affiliated Hospital of Xi'an Jiaotong University from January 2018 to January 2019 and included in this study, 270 had no ACS, while 186 had newly diagnosed ACS. Overall, 32 demographic characteristics and serum biomarkers of the study patients were analysed. The least absolute shrinkage and selection operator regression was used to select variables, while the multivariate logistic regression was used to establish the predictive model that was presented using a nomogram. The area under the receiver operating characteristics curve (AUC) was used to evaluate the discriminatory capacity of the model. A calibration plot and Hosmer-Lemeshow test were used for the calibration of the predictive model, while the decision curve analysis (DCA) was used to evaluate its clinical validity. RESULTS: After random sampling, 319 and 137 T2DM patients were included in the training and validation sets, respectively. The predictive model included age, body mass index, diabetes duration, systolic blood pressure (SBP), diastolic blood pressure (DBP), low-density lipoprotein cholesterol, serum uric acid, lipoprotein(a), hypertension history and alcohol drinking status as predictors. The AUC of the predictive model and that of the internal validation set was 0.830 [95% confidence interval (CI) 0.786-0.874] and 0.827 (95% CI 0.756-0.899), respectively. The predictive model showed very good fitting degree, and DCA demonstrated a clinically effective predictive model. CONCLUSIONS: A potent risk predictive model was established, which is of great value for the secondary prevention of diabetes. Weight loss, lowering of SBP and blood uric acid levels and appropriate control for DBP may significantly reduce the risk of new-onset ACS in T2DM patients in Northwest China.


Subject(s)
Acute Coronary Syndrome/diagnosis , Acute Coronary Syndrome/etiology , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/diagnosis , Models, Statistical , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/epidemiology , Aged , Biomarkers/blood , Blood Pressure/physiology , Body Mass Index , China/epidemiology , Cholesterol, LDL/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetic Angiopathies/blood , Diabetic Angiopathies/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Predictive Value of Tests , Prevalence , Prognosis , Risk Factors , Uric Acid/blood
8.
JACC Clin Electrophysiol ; 5(7): 854-862, 2019 07.
Article in English | MEDLINE | ID: mdl-31320015

ABSTRACT

OBJECTIVES: This study tested the hypothesis that a biphasic defibrillation waveform with an ascending first phase (ASC) causes less myocardial damage by pathology and injury current than a standard biphasic truncated exponential (BTE) waveform in a swine model. BACKGROUND: Although lifesaving, defibrillation shocks have significant iatrogenic effects that reduce their benefit for patient survival. METHODS: An ASC waveform with an 8-ms linear ramp followed by an additional positive 0.5-ms decaying portion with amplitudes of 20 J (ASC 20J) and 25 J (ASC 25J) was used. The control was a 25-J BTE conventional waveform (BTE 25J) RESULTS: The ASC 20J and ASC 25J shocks were both successful in 6 of 6 pigs, but the BTE 25J was successful in only 6 of 14 pigs (p < 0.05). Post-shock ST-segment elevation (injury current) in the right ventricular electrode was significantly greater with BTE 25J than with ASC 20J and ASC 25J. With a blinded pathology reading, hemorrhage, inflammation, thrombi, and necrosis 24 h post-shock were significantly greater with BTE 25J than with ASC 20J and ASC 25J. Troponin levels were also markedly lower at 3, 4, 5, and 6 h post-shock. CONCLUSIONS: Defibrillation shocks cause electrophysiological, histological, and biochemical signs of myocardial damage and necrosis. These signs of damage are markedly less for an ASC waveform than for a conventional BTE waveform.


Subject(s)
Defibrillators , Electric Countershock , Heart Ventricles , Myocardium/pathology , Animals , Defibrillators/adverse effects , Defibrillators/standards , Disease Models, Animal , Electric Countershock/adverse effects , Electric Countershock/methods , Electric Countershock/standards , Electrocardiography , Electrodes , Female , Heart Ventricles/injuries , Heart Ventricles/physiopathology , Male , Necrosis/etiology , Swine , Troponin C/blood
9.
Int J Cardiol ; 292: 180-187, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31101543

ABSTRACT

Activation of the apelin receptor, or APJ, by apelin is considered a therapeutic avenue for cardiovascular disease, including heart failure. Recently, a novel endogenous ligand for APJ named Elabela (ELA) has been discovered and is known to possess anti-heart failure activity in animal models. However, the short in vivo half-life of ELA constrains its clinical potential. To extend its half-life in vivo, we attempted to make IgG-Fc-ELA fusion proteins. We found that Fc-ELA-32 fusion proteins are cleaved during protein production, whereas Fc-ELA-21 fusion proteins are expressed intact, so we focused our studies on the latter. The Fc-ELA-21 fusion protein retained its functionality in vitro and had a half-life of approximately 44 h in circulation in mice after subcutaneous injection. Daily injection of the fusion protein in MI rats for 4 weeks significantly mitigated heart dysfunction with respect to hemodynamics. At the cellular and tissue levels, treatment of Fc-ELA-21 fusion protein significantly increased angiogenesis, promoted cardiomyocyte proliferation and reduced apoptosis and heart fibrosis near the infarct area. In comparison, ELA-21 had a half-life of 13 min and showed no significant cardioprotective activities. These data suggest that Fc-ELA-21 may be a potential therapeutic for heart failure.


Subject(s)
Myocardial Infarction/blood , Myocardial Infarction/drug therapy , Peptide Hormones/blood , Peptide Hormones/therapeutic use , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/therapeutic use , Amino Acid Sequence , Animals , HEK293 Cells , Half-Life , Humans , Male , Peptide Hormones/administration & dosage , Peptide Hormones/genetics , Rats , Rats, Sprague-Dawley
10.
Sci Rep ; 8(1): 17586, 2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30487532

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

11.
Sci Rep ; 8(1): 11428, 2018 07 30.
Article in English | MEDLINE | ID: mdl-30061611

ABSTRACT

Apelin is a peptide hormone with anti-oxidative and anti-inflammatory activities and is proposed to be a potential therapeutic for many disease conditions, including sepsis. However, short in vivo half-life of the apelin peptide would limit its potential clinical applications. This study aims to investigate the effects of Fc-apelin, a novel long-acting apelin fusion protein, on lipopolysaccharide (LPS)-induced liver injury. Liver injury was induced by systemic injection of LPS in mice. Hepatoprotective activities of Fc-apelin against inflammation were evaluated in LPS mice and/or hepatoma Huh-7 cells with respect to serum ALT, apoptosis, oxidative stress, macrophage infiltration and gene expression. We found that LPS induced systemic inflammation and liver damage. Co-administration of Fc-apelin significantly attenuated serum ALT elevation, diminished LPS-induced apoptosis and ROS production in the liver and in Huh-7 cells, mitigated hepatic macrophage infiltration, and reduced TNFα and IL-6 gene expression. Collectively, Fc-apelin fusion protein exerts protective effects against LPS-induced liver damage and may serve as a potential therapeutic for endotoxin-induced liver injury.


Subject(s)
Apelin/pharmacology , Liver/injuries , Receptors, Fc/metabolism , Recombinant Fusion Proteins/pharmacology , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Interleukin-6/blood , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Diabetes Metab Res Rev ; 34(5): e2997, 2018 07.
Article in English | MEDLINE | ID: mdl-29577579

ABSTRACT

BACKGROUND: Apelin is a peptide ligand of the G-protein-coupled receptor APJ and exhibits anti-diabetes and anti-heart failure activities. However, short serum half-life of the apelin peptide limits its potential clinical applications. This study aimed to develop a long-acting apelin analog. METHODS: To extend apelin's in vivo half-life, we made a recombinant protein by fusing the IgG Fc fragment to apelin-13 (Fc-apelin-13), conducted pharmacokinetics studies in mice, and determined in vitro biological activities in suppressing cyclic adenosine monophosphate and activating extracellular signal-regulated kinase signalling by reporter assays. We investigated the effects of Fc-apelin-13 on food intake, body weight, fasting blood glucose and insulin levels, glucose tolerance test, hepatic steatosis, and cardiac function and fibrosis by subcutaneous administration of Fc-apelin-13 in diet-induced obese mice for 4 weeks. RESULTS: The estimated half-life of Fc-apelin-13 in blood was approximately 33 hours. Reporter assays showed that Fc-apelin-13 was active in suppressing cyclic adenosine monophosphate response element and activating serum response element activities. Four weeks of Fc-apelin-13 treatment in obese mice did not affect food intake and body weight, but resulted in a significant improvement of glucose tolerance, and a decrease in hepatic steatosis and fibrosis, as well as in serum alanine transaminase levels. Moreover, cardiac stroke volume and output were increased and cardiac fibrosis was decreased in the treated mice. CONCLUSIONS: Fc-apelin-13 fusion protein has an extended in vivo half-life and exerts multiple benefits on obese mice with respect to the improvement of glucose disposal, amelioration of liver steatosis and heart fibrosis, and increase of cardiac output. Hence, Fc-apelin-13 is potentially a therapeutic for obesity-associated disease conditions.


Subject(s)
Diet/adverse effects , Fatty Liver/drug therapy , Heart Failure/drug therapy , Immunoglobulin Fc Fragments/chemistry , Intercellular Signaling Peptides and Proteins/administration & dosage , Obesity/complications , Recombinant Fusion Proteins/administration & dosage , Animals , Fatty Liver/etiology , Fatty Liver/pathology , Heart Failure/etiology , Heart Failure/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology
13.
PLoS One ; 13(1): e0190904, 2018.
Article in English | MEDLINE | ID: mdl-29315328

ABSTRACT

The skeletal muscle is the largest organ in the human body, depositing energy as protein/amino acids, which are degraded in catabolic conditions such as fasting. Alanine is synthesized and secreted from the skeletal muscle that is used as substrates of gluconeogenesis in the liver. During fasting, the expression of PGC-1α, a transcriptional coactivator of nuclear receptors, is increased in the liver and regulates gluconeogenesis. In the present study, we observed increased mRNA expression of PGC-1α and alanine aminotransferase 2 (ALT2) in the skeletal muscle during fasting. In C2C12 myoblast cells overexpressing PGC-1α, ALT2 expression was increased concomitant with an increased alanine level in the cells and medium. In addition, PGC-1α, along with nuclear receptor ERR, dose-dependently enhanced the ALT2 promoter activity in reporter assay using C2C12 cells. In the absence of glucose in the culture medium, mRNA levels of PGC-1α and ALT2 increased. Endogenous PGC-1α knockdown in C2C12 cells reduced ALT2 gene expression level, induced by the no-glucose medium. Taken together, in the skeletal muscle, PGC-1α activates ALT2 gene expression, and alanine production may play roles in adaptation to fasting.


Subject(s)
Alanine/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Alanine Transaminase/genetics , Animals , Cell Line , Fasting , Gene Expression Regulation , Liver/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Promoter Regions, Genetic
14.
Sci Rep ; 6: 32424, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27561749

ABSTRACT

Exercise training has been reported to ameliorate heart dysfunction in both humans and animals after myocardial infarction (MI), but the underlying mechanisms are poorly understood. Follistatin-like1 (FSTL1) is a cardioprotective factor against ischemic injury and is induced in cardiomyocytes and skeletal muscle in ischemic and hypoxic conditions. To test the hypothesis that FSTL1 may be a molecular link between exercise and improved heart function post MI, we subjected MI-rats, induced by left coronary artery ligation, to two modes of exercise: intermittent aerobic exercise (IAE) or mechanical vibration training (MVT), for four weeks and examined the relevance of FSTL1 to exercise-mediated cardiac effects. Exercise improved the functional performance, reduced fibrosis of MI-hearts and induced FSTL1 expression, the TGFß-Smad2/3 signaling and angiogenesis in myocardium. In gastrocnemius, exercise increased the cross-sectional area of myocytes and FSTL1 expression. Importantly, exercise increased circulating FSTL1 levels, which were positively correlated with the skeletal muscle FSTL1 expression and negatively correlated with heart fibrosis. Overall, the IAE was more effective than that of MVT in cardioprotection. Finally, exogenous FSTL1 administration directly improved angiogenesis as well as functionality of post-MI hearts. Taken together, we have demonstrated that FSTL1 is a potential mediator of exercise-induced cardioprotection in post-MI rats.


Subject(s)
Fibrosis/rehabilitation , Follistatin-Related Proteins/genetics , Myocardial Infarction/rehabilitation , Physical Conditioning, Animal , Animals , Fibrosis/genetics , Fibrosis/physiopathology , Fibrosis/therapy , Follistatin/metabolism , Follistatin-Related Proteins/metabolism , Humans , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats
15.
J Lipid Res ; 57(7): 1256-63, 2016 07.
Article in English | MEDLINE | ID: mdl-27178044

ABSTRACT

The glucocorticoid-induced leucine zipper (GILZ), a primary target of glucocorticoids, is expressed in human adipocytes, but its importance in adipocyte function is unknown. Because TNFα is increased in obese adipose tissue and antagonizes a number of glucocorticoid actions, we investigated the interplay of these pathways. GILZ knockdown increased and GILZ overexpression decreased interleukin-6 (IL-6) and leptin mRNA and protein secretion. GILZ knockdown increased the magnitude of the glucocorticoid effect on leptin secretion, but did not affect the glucocorticoid suppression of IL-6. Although GILZ silencing decreased adiponectin mRNA levels, it did not affect the amount of adiponectin secreted. GILZ negatively modulated pro-inflammatory signaling pathways, blocking basal and TNFα-stimulated (1 h) p65 nuclear factor κB nuclear translocation and transcriptional activity by binding to p65 in the cytoplasm. GILZ silencing increased basal ERK1/2 and JNK phosphorylation, and decreased MAPK phosphatase-1 protein levels. Longer term TNFα (4 h or 24 h) treatment decreased GILZ expression in human adipocytes. Furthermore, adipose tissue GILZ mRNA levels were reduced in proportion to the degree of obesity and expression of inflammatory markers. Overall, these results suggest that GILZ antagonizes the pro-inflammatory effects of TNFα in human adipocytes, and its downregulation in obesity may contribute to adipose inflammation and dysregulated adipokine production, and thereby systemic metabolism.


Subject(s)
Inflammation/genetics , Obesity/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism , Adipokines/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adult , Biopsy , Dual Specificity Phosphatase 1/metabolism , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Leptin/metabolism , MAP Kinase Signaling System/genetics , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Obesity/metabolism , Obesity/pathology , RNA, Messenger/biosynthesis , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcription Factors/biosynthesis , Tumor Necrosis Factor-alpha/genetics
16.
Diabetes Metab Res Rev ; 31(6): 562-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25865565

ABSTRACT

BACKGROUND: Alanine transaminase (ALT) plays an important role in gluconeogenesis by converting alanine into pyruvate for glucose production. Early studies have shown that ALT activities are upregulated in gluconeogenic conditions and may be implicated in the development of diabetes. ALT consists of two isoforms, ALT1 and ALT2, with distinctive subcellular and tissue distributions. Whether and how they are regulated are largely unknown. METHODS: By using Western blotting analysis, we measured hepatic ALT isoforms at the protein level in obese and diabetic animals and in Fao hepatoma cells treated with dexamethasone and insulin. In addition, we measured glucose output in Fao cells over-expressing ALT1 and ALT2. RESULTS: Both ALT isoforms in the liver were increased in diabetic Goto-Kakizaki rats and during fasting. However, in ob/ob mice, only ALT2, but not ALT1, protein levels were elevated, and the increase of ALT2 was correlated with that of ALT activity. We further demonstrated that, in vitro, both ALT1 and ALT2 were induced by glucocorticoid dexamethasone, but suppressed by insulin in Fao cells. Finally, we showed that the over-expression of ALT1 and ALT2 in Fao cells directly increased glucose output. CONCLUSIONS: We have shown the similarity and difference in the regulation of ALT isoforms in gluconeogenic conditions at the protein level, supporting that ALT isoenzymes play an important role in glucose metabolism and may be implicated the development of insulin resistance and diabetes.


Subject(s)
Alanine Transaminase/metabolism , Diabetes Mellitus, Type 2/enzymology , Enzyme Induction , Gluconeogenesis , Liver/enzymology , Obesity/enzymology , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/chemistry , Alanine Transaminase/genetics , Animals , Cell Line , Dexamethasone/pharmacology , Diabetes Mellitus, Type 2/metabolism , Enzyme Induction/drug effects , Enzyme Repression/drug effects , Glucocorticoids/pharmacology , Gluconeogenesis/drug effects , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
17.
Obesity (Silver Spring) ; 23(5): 1014-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25919922

ABSTRACT

OBJECTIVE: To determine whether super-activation of PPARγ can reprogram human myoblasts into brown-like adipocytes and to establish a new cell model for browning research. METHODS: To enhance the PPARγ signaling, M3, the transactivation domain of MyoD, was fused to PPARγ. PPARγ and M3-PPARγ-lentiviral vectors were used to convert human myoblasts into adipocytes. Brown adipocyte markers of the reprogrammed adipocytes were assessed by qPCR and protein analyses. White adipocytes differentiated from subcutaneous stromal vascular cells and perithyroid brown fat tissues were used as references. RESULTS: In transient transfections, M3-PPARγ had a stronger constitutive activity than PPARγ by reporter assay. Although the transduction of either PPARγ or M3-PPARγ induced adipogenesis in myoblasts, M3-PPARγ drastically induced the brown adipocyte markers of UCP1, CIDEA, and PRDM16 by 1,050, 2.4, and 5.0 fold, respectively and increased mitochondria contents by 4 fold, compared to PPARγ. CONCLUSIONS: Super-activation of PPARγ can effectively convert human myoblasts into brown-like adipocytes and a new approach to derive brown-like adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , PPAR gamma/metabolism , Adipogenesis/physiology , Cell Differentiation/physiology , Humans , Mitochondria/metabolism , Myoblasts/metabolism , Stromal Cells/metabolism , Transcription Factors/metabolism
18.
J Inherit Metab Dis ; 38(5): 941-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25758935

ABSTRACT

Intellectual disability is genetically heterogeneous, and it is likely that many of the responsible genes have not yet been identified. We describe three siblings with isolated, severe developmental encephalopathy. After extensive uninformative genetic and metabolic testing, whole exome sequencing identified a homozygous novel variant in glutamic pyruvate transaminase 2 (GPT2) or alanine transaminase 2 (ALT2), c.459 C > G p.Ser153Arg that segregated with developmental encephalopathy in the family. This variant was predicted to be damaging by all in silico prediction algorithms. GPT2 is the gene encoding ALT2 which is responsible for the reversible transamination of alanine and 2-oxoglutarate to form pyruvate and glutamate. GPT2 is expressed in brain and is in the pathway to generate glutamate, an excitatory neurotransmitter. Functional assays of recombinant wild-type and mutant ALT2 proteins demonstrated the p.Ser153Arg mutation resulted in a severe loss of enzymatic function. We suggest that recessively inherited loss of function GPT2 mutations are a novel cause of intellectual disability.


Subject(s)
Brain Diseases/genetics , Intellectual Disability/genetics , Mutation, Missense , Transaminases/genetics , Adolescent , Alanine Transaminase/genetics , Brain Diseases/congenital , Child, Preschool , Consanguinity , Female , Humans , Male , Pedigree , Siblings
19.
Sci Rep ; 5: 8170, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25639753

ABSTRACT

Elabela (ELA) or Toddler is a recently discovered hormone which is required for normal development of heart and vasculature through activation of apelin receptor (APJ), a G protein-coupled receptor (GPCR), in zebrafish. The present study explores whether the ELA-APJ signaling pathway is functional in the mammalian system. Using reverse-transcription PCR, we found that ELA is restrictedly expressed in human pluripotent stem cells and adult kidney whereas APJ is more widely expressed. We next studied ELA-APJ signaling pathway in reconstituted mammalian cell systems. Addition of ELA to HEK293 cells over-expressing GFP-AJP fusion protein resulted in rapid internalization of the fusion receptor. In Chinese hamster ovarian (CHO) cells over-expressing human APJ, ELA suppresses cAMP production with EC50 of 11.1 nM, stimulates ERK1/2 phosphorylation with EC50 of 14.3 nM and weakly induces intracellular calcium mobilization. Finally, we tested ELA biological function in human umbilical vascular endothelial cells and showed that ELA induces angiogenesis and relaxes mouse aortic blood vessel in a dose-dependent manner through a mechanism different from apelin. Collectively, we demonstrate that the ELA-AJP signaling pathways are functional in mammalian systems, indicating that ELA likely serves as a hormone regulating the circulation system in adulthood as well as in embryonic development.


Subject(s)
Peptide Hormones/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Apelin Receptors , Blood Vessels/physiology , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Endocytosis , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic , Peptide Hormones/genetics , Phosphorylation , Receptors, G-Protein-Coupled/genetics
20.
J Biomol Screen ; 19(8): 1164-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907126

ABSTRACT

Niemann-Pick disease type C (NPC) is a rare neurodegenerative disorder caused by recessive mutations in the NPC1 or NPC2 gene that result in lysosomal accumulation of unesterified cholesterol in patient cells. Patient fibroblasts have been used for evaluation of compound efficacy, although neuronal degeneration is the hallmark of NPC disease. Here, we report the application of human NPC1 neural stem cells as a cell-based disease model to evaluate nine compounds that have been reported to be efficacious in the NPC1 fibroblasts and mouse models. These cells are differentiated from NPC1 induced pluripotent stem cells and exhibit a phenotype of lysosomal cholesterol accumulation. Treatment of these cells with hydroxypropyl-ß-cyclodextrin, methyl-ß-cyclodextrin, and δ-tocopherol significantly ameliorated the lysosomal cholesterol accumulation. Combined treatment with cyclodextrin and δ-tocopherol shows an additive or synergistic effect that otherwise requires 10-fold higher concentration of cyclodextrin alone. In addition, we found that hydroxypropyl-ß-cyclodextrin is much more potent and efficacious in the NPC1 neural stem cells compared to the NPC1 fibroblasts. Miglustat, suberoylanilide hydroxamic acid, curcumin, lovastatin, pravastatin, and rapamycin did not, however, have significant effects in these cells. The results demonstrate that patient-derived NPC1 neural stem cells can be used as a model system for evaluation of drug efficacy and study of disease pathogenesis.


Subject(s)
Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/pathology , Niemann-Pick Disease, Type C/pathology , Cell Differentiation , Cells, Cultured , Cholesterol/metabolism , Cyclodextrins/pharmacology , Drug Synergism , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Neural Stem Cells/pathology , Neurons/drug effects , Neurons/pathology , Niemann-Pick Disease, Type C/drug therapy , Tocopherols/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...