Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Blood Adv ; 8(11): 2933-2951, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38484189

ABSTRACT

ABSTRACT: Natural killer (NK) cells represent the cytotoxic member within the innate lymphoid cell (ILC) family that are important against viral infections and cancer. Although the NK cell emergence from hematopoietic stem and progenitor cells through multiple intermediate stages and the underlying regulatory gene network has been extensively studied in mice, this process is not well characterized in humans. Here, using a temporal in vitro model to reconstruct the developmental trajectory of NK lineage, we identified an ILC-restricted oligopotent stage 3a CD34-CD117+CD161+CD45RA+CD56- progenitor population, that exclusively gave rise to CD56-expressing ILCs in vitro. We also further investigated a previously nonappreciated heterogeneity within the CD56+CD94-NKp44+ subset, phenotypically equivalent to stage 3b population containing both group-1 ILC and RORγt+ ILC3 cells, that could be further separated based on their differential expression of DNAM-1 and CD161 receptors. We confirmed that DNAM-1hi S3b and CD161hiCD117hi ILC3 populations distinctively differed in their expression of effector molecules, cytokine secretion, and cytotoxic activity. Furthermore, analysis of lineage output using DNA-barcode tracing across these stages supported a close developmental relationship between S3b-NK and S4-NK (CD56+CD94+) cells, whereas distant to the ILC3 subset. Cross-referencing gene signatures of culture-derived NK cells and other noncytotoxic ILCs with publicly available data sets validated that these in vitro stages highly resemble transcriptional profiles of respective in vivo ILC counterparts. Finally, by integrating RNA velocity and gene network analysis through single-cell regulatory network inference and clustering we unravel a network of coordinated and highly dynamic regulons driving the cytotoxic NK cell program, as a guide map for future studies on NK cell regulation.


Subject(s)
Killer Cells, Natural , Single-Cell Analysis , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Single-Cell Analysis/methods , Cell Lineage , Immunity, Innate , Cell Differentiation
2.
Sci Rep ; 14(1): 3514, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38346965

ABSTRACT

The past decade has seen substantial innovation in clinical trials, including new trial formats, endpoints, and others. Also there have been regulatory changes, increasing competitive pressures and other external factors which impact clinical trials. In parallel, trial timelines have increased and success rates remain stubbornly low. This has led many observers to question whether clinical trials have become overly complex and if this complexity is always needed. Here we present a large-scale analysis of protocols and other data from over 16,000 trials. Using a machine learning algorithm, we automatically assessed key features of these trials, such as number of endpoints, number of inclusion-exclusion criteria and others. Using a regression analysis we combined these features into a new metric, the Trial Complexity Score, which correlates with overall clinical trial duration. Looking at this score across different clinical phases and therapeutic areas we see substantial increases over time, suggesting that clinical trials are indeed becoming more complex. We discuss drivers of increasing trial complexity, necessary or helpful ('good') complexity versus unnecessary ('bad') complexity, and we explore mechanisms of how sponsors of clinical trials can reduce trial complexity where appropriate.

4.
Anal Bioanal Chem ; 414(2): 1039-1048, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34676433

ABSTRACT

The rapid development of nanozymes for ultrasensitive detection of contaminate has resulted in considerable attention. Herein, a carboxyl- and aminopropyl-functionalized copper organophyllosilicate (Cu-CAP) was synthesized by a facile, one-pot sol-gel method. The bifunctional groups endow it with superior catalytic activity than that of natural enzyme. Besides, it possesses outstanding catalytic stability under harsh conditions such as high temperature, extremely high or low pH, and high salinity. Apart from laccase-mimetic activity, Cu-CAP also shows oxidation of the peroxidase substrate 3,3',5,5'-tetramethylbenzidine (TMB) to the blue-colored TMBox in the presence of H2O2, which is similar to natural horseradish peroxidase (HRP). Interestingly, this colorimetric system was suppressed by hydroquinone (HQ) specifically. Inspired by this, Cu-CAP was used to develop a highly sensitive and selective colorimetric method for the determination of HQ. This assay displayed an extremely low detection limit of 23 nM and was applied for the detection of HQ in environmental water with high accuracy. This approach offers a new route for the rational design of high performance nanozymes for environmental and biosensing applications.


Subject(s)
Copper/chemistry , Hydroquinones/analysis , Nanostructures/chemistry , Silicates/chemistry , Colorimetry/methods , Kinetics , Limit of Detection , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
5.
Bioresour Technol ; 322: 124560, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33371957

ABSTRACT

The development of synergetic biogeocomplices for biodegradation of recalcitrant organic pollutants is an urgently needed to achieve the environmental sustainability. The biogeosorbent based on the analcime-bearing rock immobilized Chlorella vulgaris f. globosa was developed to remove phenol from polluted waterbodies. The microalgae biofilm formation on the ABR resulted in 1.6 × 104 cells/mm2. Stress testing showed that low temperatures up to -30 °C did not adversely affect the cell viability, the dehydrogenase activity of the biogeosorbent exposed was 5.1 mg of formazan/mL. Under phenol-stress conditions, aggregation of suspended cells was observed. The biogeosorbent was more stress resistant than the microalgal suspension, and also reduced the time of exposure and had no secondary waste in comparison with the ABR. After having been treated, phenol removal was found to increase from 70 to 72% for MA, from 27 to 93% for ABR, from 82 to 93% for the biogeosorbent.


Subject(s)
Chlorella vulgaris , Microalgae , Biodegradation, Environmental , Phenol , Phenols
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 241: 118627, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-32623299

ABSTRACT

The luminescence intensity ratio method, exploiting the temperature-dependent luminescence of the thermally coupled energy levels, is regarded as a very promising approach for optical temperature measurement at the cellular level. In this study, it was found that bare NaYF4:Yb3+/Tm3+ nanoparticles cannot be used as a cellular thermosensor in principle because of their tendency to aggregate, which significantly affects the luminescent properties of the complex, introducing uncertainty in the intensity ratio measurement. NaYF4:Yb3+/Tm3+ up-conversion nanoparticles, coated with polyethylene glycol (PEG) and carboxyl groups (COOH), on the other hand, proved to be promising candidates for the role of thermosensors. For the first time the temperature sensitivity of the NaYF4:Yb3+/Tm3+@PEG@COOH thermosensor was calculated in water and in biotissues. It was found that the sensitivity of the thermosensor increased by 1.3 times during the transition from water to egg white and urine - from 1.17% × K-1 to 1.58% × K-1. This effect is associated with the chemical composition of the studied media. The results obtained suggest that using upconversion nanocomplexes as primary thermosensors is still difficult.


Subject(s)
Nanocomposites , Ytterbium , Luminescence , Temperature , Yttrium
7.
Diab Vasc Dis Res ; 15(4): 302-313, 2018 07.
Article in English | MEDLINE | ID: mdl-29499628

ABSTRACT

AIMS: Despite vast clinical experience linking diabetes and atherosclerosis, the molecular mechanisms leading to accelerated vascular damage are still unclear. Here, we investigated the effects of nuclear factor of activated T-cells inhibition on plaque burden in a novel mouse model of type 2 diabetes that better replicates human disease. METHODS & RESULTS: IGF-II/LDLR-/-ApoB100/100 mice were generated by crossbreeding low-density lipoprotein receptor-deficient mice that synthesize only apolipoprotein B100 (LDLR-/-ApoB100/100) with transgenic mice overexpressing insulin-like growth factor-II in pancreatic ß cells. Mice have mild hyperglycaemia and hyperinsulinaemia and develop complex atherosclerotic lesions. In vivo treatment with the nuclear factor of activated T-cells blocker A-285222 for 4 weeks reduced atherosclerotic plaque area and degree of stenosis in the brachiocephalic artery of IGF-II/LDLR-/-ApoB100/100 mice, as assessed non-invasively using ultrasound biomicroscopy prior and after treatment, and histologically after termination. Treatment had no impact on plaque composition (i.e. muscle, collagen, macrophages). The reduced plaque area could not be explained by effects of A-285222 on plasma glucose, insulin or lipids. Inhibition of nuclear factor of activated T-cells was associated with increased expression of atheroprotective NOX4 and of the anti-oxidant enzyme catalase in aortic vascular smooth muscle cells. CONCLUSION: Targeting the nuclear factor of activated T-cells signalling pathway may be an attractive approach for the treatment of diabetic macrovascular complications.


Subject(s)
Apolipoproteins B/deficiency , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Insulin-Like Growth Factor II/deficiency , NFATC Transcription Factors/antagonists & inhibitors , Plaque, Atherosclerotic , Pyrazoles/pharmacology , Receptors, LDL/deficiency , Animals , Apolipoprotein B-100 , Apolipoproteins B/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Catalase/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Insulin-Like Growth Factor II/genetics , Male , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/metabolism , NFATC Transcription Factors/metabolism , Oxidative Stress/drug effects , Phenotype , Receptors, LDL/genetics , Signal Transduction
8.
Kidney Int ; 89(2): 342-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26806835

ABSTRACT

Diabetic kidney disease is the leading cause of end-stage renal disease. Genetic factors have been suggested to contribute to its susceptibility. However, results from genetic studies are disappointing possibly because the role of glucose in diabetic kidney disease predisposed by epigenetic mechanisms has not been taken into account. Since thioredoxin-interacting protein (TXNIP) has been shown to play an important role in the pathogenesis of diabetic kidney disease, we tested whether glucose could induce expression of TXNIP in the kidney by epigenetic mechanisms. In kidneys from diabetic Sur1-E1506K(+/+) mice, hyperglycemia-induced Txnip expression was associated with stimulation of activating histone marks H3K9ac, H3K4me3, and H3K4me1, as well as decrease in the repressive histone mark H3K27me3 at the promoter region of the gene. Glucose also coordinated changes in histone marks and TXNIP gene expression in mouse SV40 MES13 mesangial cells and the normal human mesangial cell line NHMC. The involvement of histone acetylation in glucose-stimulated TXNIP expression was confirmed by reversing or enhancing acetylation using the histone acetyltransferase p300 inhibitor C646 or the histone deacetylase inhibitor trichostatin A. Thus, glucose is a potent inducer of histone modifications, which could drive expression of proinflammatory genes and thereby predispose to diabetic kidney disease.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/etiology , Epigenesis, Genetic , Mesangial Cells/metabolism , Thioredoxins/metabolism , Animals , Carrier Proteins/genetics , Cells, Cultured , Histone Code , Humans , Hyperglycemia/metabolism , Mice, Transgenic , Thioredoxins/genetics , Up-Regulation
9.
Diabetes ; 65(1): 239-54, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26395740

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone with extrapancreatic effects beyond glycemic control. Here we demonstrate unexpected effects of GIP signaling in the vasculature. GIP induces the expression of the proatherogenic cytokine osteopontin (OPN) in mouse arteries via local release of endothelin-1 and activation of CREB. Infusion of GIP increases plasma OPN concentrations in healthy individuals. Plasma endothelin-1 and OPN concentrations are positively correlated in patients with critical limb ischemia. Fasting GIP concentrations are higher in individuals with a history of cardiovascular disease (myocardial infarction, stroke) when compared with control subjects. GIP receptor (GIPR) and OPN mRNA levels are higher in carotid endarterectomies from patients with symptoms (stroke, transient ischemic attacks, amaurosis fugax) than in asymptomatic patients, and expression associates with parameters that are characteristic of unstable and inflammatory plaques (increased lipid accumulation, macrophage infiltration, and reduced smooth muscle cell content). While GIPR expression is predominantly endothelial in healthy arteries from humans, mice, rats, and pigs, remarkable upregulation is observed in endothelial and smooth muscle cells upon culture conditions, yielding a "vascular disease-like" phenotype. Moreover, the common variant rs10423928 in the GIPR gene is associated with increased risk of stroke in patients with type 2 diabetes.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Endothelial Cells/metabolism , Endothelin-1/genetics , Gastric Inhibitory Polypeptide/metabolism , Myocytes, Smooth Muscle/metabolism , Osteopontin/genetics , RNA, Messenger/metabolism , Receptors, Gastrointestinal Hormone/genetics , Aged , Aged, 80 and over , Animals , Aorta/cytology , Blotting, Western , Cardiovascular Diseases/genetics , Carotid Arteries/cytology , Case-Control Studies , Coronary Vessels/cytology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelin-1/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microvessels/cytology , Middle Aged , Osteopontin/metabolism , Peripheral Arterial Disease/metabolism , Plaque, Atherosclerotic/metabolism , Polymorphism, Single Nucleotide , Rats , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Stroke/complications , Stroke/genetics , Stroke/metabolism , Sus scrofa , Swine
10.
J Diabetes Res ; 2015: 428473, 2015.
Article in English | MEDLINE | ID: mdl-25918731

ABSTRACT

The pathogenesis of diabetic retinopathy (DR) remains unclear but hyperglycemia is an established risk factor. Endothelial dysfunction and changes in Ca2+ signaling have been shown to precede the onset of DR. We recently demonstrated that high extracellular glucose activates the Ca(2+)/calcineurin-dependent transcription factor NFAT in cerebral arteries and aorta, promoting the expression of inflammatory markers. Here we show, using confocal immunofluorescence, that NFAT is expressed in the endothelium of retinal microvessels and is readily activated by high glucose. This was inhibited by the NFAT blocker A-285222 as well as by the ectonucleotidase apyrase, suggesting a mechanism involving the release of extracellular nucleotides. Acute hyperglycemia induced by an IP-GTT (intraperitoneal glucose tolerance test) resulted in increased NFATc3 nuclear accumulation and NFAT-dependent transcriptional activity in retinal vessels of NFAT-luciferase reporter mice. In both Akita (Ins2(+/-) ) and streptozotocin- (STZ-) induced diabetic mice, NFAT transcriptional activity was elevated in retinal vessels. In vivo inhibition of NFAT with A-285222 decreased the expression of OPN and ICAM-1 mRNA in retinal vessels, prevented a diabetes driven downregulation of anti-inflammatory IL-10 in retina, and abrogated the increased vascular permeability observed in diabetic mice. Results identify NFAT signaling as a putative target for treatment of microvascular complications in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Microvessels/metabolism , NFATC Transcription Factors/metabolism , Retinal Vein/metabolism , Animals , Aorta/metabolism , Calcium/metabolism , Diabetes Complications , Glucose Tolerance Test , Hyperglycemia/metabolism , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcirculation , Osteopontin/metabolism , Permeability , Pyrazoles/chemistry , Retinal Vessels/pathology , Signal Transduction
11.
J Mol Med (Berl) ; 93(5): 511-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25471481

ABSTRACT

UNLABELLED: ORAI and stromal interaction molecule (STIM) are store-operated channel molecules that play essential roles in human physiology through a coupling mechanism of internal Ca(2+) store to Ca(2+) influx. However, the roles of ORAI and STIM in vascular endothelial cells under diabetic conditions remain unknown. Here, we investigated expression and signalling pathways of ORAI and STIM regulated by high glucose or hyperglycaemia using in vitro cell models, in vivo diabetic mice and tissues from patients. We found that ORAI1-3 and STIM1-2 were ubiquitously expressed in human vasculatures. Their expression was upregulated by chronic treatment with high glucose (HG, 25 mM D-glucose), which was accompanied by enhanced store-operated Ca(2+) influx in vascular endothelial cells. The increased expression was also observed in the aortae from genetically modified Akita diabetic mice (C57BL/6-Ins2(Akita)/J) and streptozocin-induced diabetic mice, and aortae from diabetic patients. HG-induced upregulation of ORAI and STIM genes was prevented by the calcineurin inhibitor cyclosporin A and NFATc3 siRNA. Additionally, in vivo treatment with the nuclear factor of activated T cells (NFAT) inhibitor A-285222 prevented the gene upregulation in Akita mice. However, HG had no direct effects on ORAI1-3 currents and the channel activation process through cytosolic STIM1 movement in the cells co-expressing STIM1-EYFP/ORAIs. We concluded that upregulation of STIM/ORAI through Ca(2+)-calcineurin-NFAT pathway is a novel mechanism causing abnormal Ca(2+) homeostasis and endothelial dysfunction under hyperglycaemia. KEY MESSAGE: ORAI1-3 and STIM1-2 are ubiquitously expressed in vasculatures and upregulated by high glucose. Increased expression is confirmed in Akita (Ins2(Akita)/J) and STZ diabetic mice and patients. Upregulation mechanism is mediated by Ca(2+)/calcineurin/NFATc3 signalling. High glucose has no direct effects on ORAI1-3 channel activity and channel activation process.


Subject(s)
Calcineurin/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Glucose/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction , Animals , Calcium Channels/genetics , Cell Line , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Middle Aged , RNA, Messenger , Up-Regulation
12.
Diabetes ; 62(6): 2088-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23349498

ABSTRACT

Low-grade inflammation in obesity is associated with accumulation of the macrophage-derived cytokine osteopontin (OPN) in adipose tissue and induction of local as well as systemic insulin resistance. Since glucose-dependent insulinotropic polypeptide (GIP) is a strong stimulator of adipogenesis and may play a role in the development of obesity, we explored whether GIP directly would stimulate OPN expression in adipose tissue and thereby induce insulin resistance. GIP stimulated OPN protein expression in a dose-dependent fashion in rat primary adipocytes. The level of OPN mRNA was higher in adipose tissue of obese individuals (0.13 ± 0.04 vs. 0.04 ± 0.01, P < 0.05) and correlated inversely with measures of insulin sensitivity (r = -0.24, P = 0.001). A common variant of the GIP receptor (GIPR) (rs10423928) gene was associated with a lower amount of the exon 9-containing isoform required for transmembrane activity. Carriers of the A allele with a reduced receptor function showed lower adipose tissue OPN mRNA levels and better insulin sensitivity. Together, these data suggest a role for GIP not only as an incretin hormone but also as a trigger of inflammation and insulin resistance in adipose tissue. Carriers of the GIPR rs10423928 A allele showed protective properties via reduced GIP effects. Identification of this unprecedented link between GIP and OPN in adipose tissue might open new avenues for therapeutic interventions.


Subject(s)
Adipose Tissue/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Insulin Resistance/physiology , Adipose Tissue/drug effects , Adolescent , Adult , Aged , Alleles , Animals , Cells, Cultured , Female , Humans , In Vitro Techniques , Insulin Resistance/genetics , Male , Mice , Middle Aged , Osteopontin/genetics , Rats , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Young Adult
13.
Exp Diabetes Res ; 2012: 176162, 2012.
Article in English | MEDLINE | ID: mdl-23243415

ABSTRACT

BACKGROUND: Diabetes is associated with increased cardiovascular disease, but the underlying cellular and molecular mechanisms are poorly understood. One proposed mechanism is that diabetes aggravates atherosclerosis by enhancing plaque inflammation. The Akita mouse has recently been adopted as a relevant model for microvascular complications of diabetes. Here we investigate the development of atherosclerosis and inflammation in vessels of Akita mice on LDLr⁻/⁻ background. METHODS AND RESULTS: Akita-LDLr⁻/⁻ and LDLr⁻/⁻ mice were fed high-fat diet from 6 to 24 weeks of age. Blood glucose levels were higher in both male and female Akita-LDLr⁻/⁻ mice (137% and 70%, resp.). Male Akita-LDLr⁻/⁻ mice had markedly increased plasma cholesterol and triglyceride levels, a three-fold increase in atherosclerosis, and enhanced accumulation of macrophages and T-cells in plaques. In contrast, female Akita-LDLr⁻/⁻ mice demonstrated a modest 29% increase in plasma cholesterol and no significant increase in triglycerides, atherosclerosis, or inflammatory cells in lesions. Male Akita-LDLr⁻/⁻ mice had increased levels of plasma IL-1ß compared to nondiabetic mice, whereas no such difference was seen between female diabetic and nondiabetic mice. CONCLUSION: Akita-LDLr⁻/⁻ mice display considerable gender differences in the development of diabetic atherosclerosis. In addition, the increased atherosclerosis in male Akita-LDLr⁻/⁻ mice is associated with an increase in inflammatory cells in lesions.


Subject(s)
Aorta/immunology , Aortitis/etiology , Atherosclerosis/etiology , Diabetes Mellitus, Type 1/complications , Diabetic Angiopathies/etiology , Receptors, LDL/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Aortitis/blood , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Biomarkers/blood , Blood Glucose/metabolism , Cells, Cultured , Cholesterol/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetic Angiopathies/blood , Diabetic Angiopathies/genetics , Diabetic Angiopathies/immunology , Diabetic Angiopathies/pathology , Disease Models, Animal , Female , Inflammation Mediators/blood , Interleukin-1beta/blood , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/genetics , Sex Factors , T-Lymphocytes/immunology , Triglycerides/blood
14.
J Biol Chem ; 287(28): 23451-63, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22610379

ABSTRACT

Contraction stimulates Na(+),K(+)-ATPase and AMP-activated protein kinase (AMPK) activity in skeletal muscle. Whether AMPK activation affects Na(+),K(+)-ATPase activity in skeletal muscle remains to be determined. Short term stimulation of rat L6 myotubes with the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR), activates AMPK and promotes translocation of the Na(+),K(+)-ATPase α(1)-subunit to the plasma membrane and increases Na(+),K(+)-ATPase activity as assessed by ouabain-sensitive (86)Rb(+) uptake. Cyanide-induced artificial anoxia, as well as a direct AMPK activator (A-769662) also increase AMPK phosphorylation and Na(+),K(+)-ATPase activity. Thus, different stimuli that target AMPK concomitantly increase Na(+),K(+)-ATPase activity. The effect of AICAR on Na(+),K(+)-ATPase in L6 myotubes was attenuated by Compound C, an AMPK inhibitor, as well as siRNA-mediated AMPK silencing. The effects of AICAR on Na(+),K(+)-ATPase were completely abolished in cultured primary mouse muscle cells lacking AMPK α-subunits. AMPK stimulation leads to Na(+),K(+)-ATPase α(1)-subunit dephosphorylation at Ser(18), which may prevent endocytosis of the sodium pump. AICAR stimulation leads to methylation and dephosphorylation of the catalytic subunit of the protein phosphatase (PP) 2A in L6 myotubes. Moreover, AICAR-triggered dephosphorylation of the Na(+),K(+)-ATPase was prevented in L6 myotubes deficient in PP2A-specific protein phosphatase methylesterase-1 (PME-1), indicating a role for the PP2A·PME-1 complex in AMPK-mediated regulation of Na(+),K(+)-ATPase. Thus contrary to the common paradigm, we report AMPK-dependent activation of an energy-consuming ion pumping process. This activation may be a potential mechanism by which exercise and metabolic stress activate the sodium pump in skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biphenyl Compounds , Blotting, Western , Carboxylic Ester Hydrolases/metabolism , Cell Hypoxia , Cells, Cultured , Enzyme Activation/drug effects , Methylation/drug effects , Mice , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Pyrones/pharmacology , RNA Interference , Rats , Ribonucleotides/pharmacology , Thiophenes/pharmacology
15.
Cardiovasc Res ; 93(3): 414-23, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22116621

ABSTRACT

AIMS: Alternative transcription and splicing of the allograft inflammatory factor-1 (AIF-1) gene results in the expression of two different proteins: AIF-1 and interferon responsive transcript-1 (IRT-1).  Here, we explore the impact of AIF-1 and IRT-1 on vascular smooth muscle cell (VSMC) activation and neointima formation, the mechanisms underlying their alternative splicing, and associations of AIF-1 and IRT-1 mRNA with parameters defining human atherosclerotic plaque phenotype. METHODS AND RESULTS: Translation of AIF-1 and IRT-1 results in different products with contrasting cellular distribution and functions. Overexpression of AIF-1 stimulates migration and proliferation of human VSMCs, whereas IRT-1 exerts opposite effects. Adenoviral infection of angioplasty-injured rat carotid arteries with AdAIF-1 exacerbates intima hyperplasia, whereas infection with AdIRT-1 reduces neointima. Expression of these variants is modulated by changes in nuclear factor of activated T-cells (NFAT) activity.  Pharmacological inhibition of NFAT or targeting of NFATc3 with small interfering RNA (siRNA) lowers the AIF-1/IRT-1 ratio and favours an anti-proliferative outcome.  NFAT acts as a repressor on the IRT-1 transcriptional start site, which is also sensitive to interferon-γ stimulation. Expression of AIF-1 mRNA in human carotid plaques associates with less extracellular matrix and a more pro-inflammatory plaque and plasma profile, features that may predispose to plaque rupture. In contrast, expression of IRT-1 mRNA associates with a less aggressive phenotype and less VSMCs at the most stenotic region of the plaque. CONCLUSION: Inhibition of NFAT signalling, by shifting the AIF-1/IRT-1 ratio, may be an attractive target to regulate the VSMC response to injury and manipulate plaque stability in atherosclerosis.


Subject(s)
Alternative Splicing/physiology , Carotid Artery Diseases , Coronary Artery Disease , DNA-Binding Proteins/genetics , NFATC Transcription Factors/metabolism , Neointima , Angioplasty, Balloon, Coronary/adverse effects , Animals , Calcium-Binding Proteins , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Coronary Vessels/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Microfilament Proteins , Muscle, Smooth, Vascular/pathology , Myometrium/blood supply , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
16.
Diabetes ; 60(9): 2424-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21810601

ABSTRACT

OBJECTIVE: The incretin hormone GIP (glucose-dependent insulinotropic polypeptide) promotes pancreatic ß-cell function by potentiating insulin secretion and ß-cell proliferation. Recently, a combined analysis of several genome-wide association studies (Meta-analysis of Glucose and Insulin-Related Traits Consortium [MAGIC]) showed association to postprandial insulin at the GIP receptor (GIPR) locus. Here we explored mechanisms that could explain the protective effects of GIP on islet function. RESEARCH DESIGN AND METHODS: Associations of GIPR rs10423928 with metabolic and anthropometric phenotypes in both nondiabetic (N = 53,730) and type 2 diabetic individuals (N = 2,731) were explored by combining data from 11 studies. Insulin secretion was measured both in vivo in nondiabetic subjects and in vitro in islets from cadaver donors. Insulin secretion was also measured in response to exogenous GIP. The in vitro measurements included protein and gene expression as well as measurements of ß-cell viability and proliferation. RESULTS: The A allele of GIPR rs10423928 was associated with impaired glucose- and GIP-stimulated insulin secretion and a decrease in BMI, lean body mass, and waist circumference. The decrease in BMI almost completely neutralized the effect of impaired insulin secretion on risk of type 2 diabetes. Expression of GIPR mRNA was decreased in human islets from carriers of the A allele or patients with type 2 diabetes. GIP stimulated osteopontin (OPN) mRNA and protein expression. OPN expression was lower in carriers of the A allele. Both GIP and OPN prevented cytokine-induced reduction in cell viability (apoptosis). In addition, OPN stimulated cell proliferation in insulin-secreting cells. CONCLUSIONS: These findings support ß-cell proliferative and antiapoptotic roles for GIP in addition to its action as an incretin hormone. Identification of a link between GIP and OPN may shed new light on the role of GIP in preservation of functional ß-cell mass in humans.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/genetics , Islets of Langerhans/metabolism , Osteopontin/genetics , Alleles , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Genome-Wide Association Study , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Male , Osteopontin/metabolism
17.
Cell Metab ; 13(1): 80-91, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21195351

ABSTRACT

Type 2 diabetes (T2D) evolves when insulin secretion fails. Insulin release from the pancreatic ß cell is controlled by mitochondrial metabolism, which translates fluctuations in blood glucose into metabolic coupling signals. We identified a common variant (rs950994) in the human transcription factor B1 mitochondrial (TFB1M) gene associated with reduced insulin secretion, elevated postprandial glucose levels, and future risk of T2D. Because islet TFB1M mRNA levels were lower in carriers of the risk allele and correlated with insulin secretion, we examined mice heterozygous for Tfb1m deficiency. These mice displayed lower expression of TFB1M in islets and impaired mitochondrial function and released less insulin in response to glucose in vivo and in vitro. Reducing TFB1M mRNA and protein in clonal ß cells by RNA interference impaired complexes of the mitochondrial oxidative phosphorylation system. Consequently, nutrient-stimulated ATP generation was reduced, leading to perturbed insulin secretion. We conclude that a deficiency in TFB1M and impaired mitochondrial function contribute to the pathogenesis of T2D.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Mitochondrial Proteins/genetics , Transcription Factors/genetics , Animals , Blood Glucose , Cell Line , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression , Gene Silencing , Genetic Loci , Genetic Variation , Humans , Insulin/blood , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Quantitative Trait Loci , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism
18.
Biochem J ; 429(1): 205-13, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20415663

ABSTRACT

Tight coupling between cytosolic and mitochondrial metabolism is key for GSIS (glucose-stimulated insulin secretion). In the present study we examined the regulatory contribution of PDH (pyruvate dehydrogenase) kinase 1, a negative regulator of PDH, to metabolic coupling in 832/13 clonal beta-cells. Knockdown of PDH kinase 1 with siRNA (small interfering RNA) reduced its mRNA (>80%) and protein level (>40%) after 72 h. PDH activity, glucose-stimulated cellular oxygen consumption and pyruvate-stimulated mitochondrial oxygen consumption increased 1.7- (P<0.05), 1.6- (P<0.05) and 1.6-fold (P<0.05) respectively. Gas chromatography/MS revealed an altered metabolite profile upon silencing of PDH kinase 1, determined by increased levels of the tricarboxylic acid cycle intermediates malate, fumarate and alpha-ketoglutarate. These metabolic alterations were associated with exaggerated GSIS (5-fold compared with 3.1-fold in control cells; P<0.01). Insulin secretion, provoked by leucine and dimethylsuccinate, which feed into the tricarboxylic acid cycle bypassing PDH, was unaffected. The oxygen consumption and metabolic data strongly suggest that knockdown of PDH kinase 1 in beta-cells permits increased metabolic flux of glucose-derived carbons into the tricarboxylic acid cycle via PDH. Enhanced insulin secretion is probably caused by increased generation of tricarboxylic acid cycle-derived reducing equivalents for mitochondrial electron transport to generate ATP and/or stimulatory metabolic intermediates. On the basis of these findings, we suggest that PDH kinase 1 is an important regulator of PDH in clonal beta-cells and that PDH kinase 1 and PDH are important for efficient metabolic coupling. Maintaining low PDH kinase 1 expression/activity, keeping PDH in a dephosphorylated and active state, may be important for beta-cells to achieve the metabolic flux rates necessary for maximal GSIS.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/physiology , Cell Line , Clone Cells , Humans , Insulin Secretion , Insulin-Secreting Cells/enzymology , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxygen Consumption/physiology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology
19.
Exp Physiol ; 95(4): 541-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20028849

ABSTRACT

The dominant RN mutation in pigs results in excessive glycogen storage in skeletal muscle. The mutation is situated in the PRKAG3 gene, which encodes a muscle-specific isoform of the AMP-activated protein kinase (AMPK) gamma3 subunit. AMPK is an important regulator of carbohydrate and fat metabolism in mammalian cells. The aim of the present study was to examine the effect of exercise on glycogen synthesis signalling pathways in muscle and to study enzyme activities of importance in carbohydrate metabolism in pigs with or without the PRKAG3 mutation. Glycogen content, metabolic enzyme activities and expression or phosphorylation of signalling proteins were analysed in skeletal muscle specimens obtained at rest, after a single treadmill exercise bout and after 3 h recovery. The PRKAG3 mutation carriers had higher glycogen content, a tendency for lower expression of AMPK (P < 0.07) and higher hexokinase and phosphorylase activities, whereas citrate synthase, 3-hydroxyacyl-CoA dehydrogenase and glycogen synthase activities did not differ between genotypes. Carriers and non-carriers of the RN mutation showed a similar degradation of glycogen after exercise, whereas the rate of resynthesis was faster in the carriers. Acute exercise stimulated Akt phosphorylation on Ser(473) in both genotypes, and the effect was greater in the carriers than in the non-carriers. Acute exercise also stimulated phosphorylation of Akt substrate of 160 kDA and Glycogen synthase kinase 3 in the carriers and GSK3alpha in the non-carriers. In conclusion, the increased rate of glycogen synthesis following exercise in pigs carrying the PRKAG3 mutation correlates with an increased signalling response of Akt and its substrate, AS160, and a higher activity of hexokinase, indicating an increased glucose influx and phosphorylation of glucose, directed towards glycogen synthesis.


Subject(s)
AMP-Activated Protein Kinases/genetics , Glycogen/biosynthesis , Physical Conditioning, Animal/physiology , Animals , GTPase-Activating Proteins/metabolism , Glycogen/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hexokinase/metabolism , Muscle, Skeletal/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Swine
20.
Hum Mol Genet ; 18(20): 3795-804, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19602480

ABSTRACT

Common variants in the transcription factor 7-like 2 (TCF7L2) gene have been identified as the strongest genetic risk factors for type 2 diabetes (T2D). However, the mechanisms by which these non-coding variants increase risk for T2D are not well-established. We used 13 expression assays to survey mRNA expression of multiple TCF7L2 splicing forms in up to 380 samples from eight types of human tissue (pancreas, pancreatic islets, colon, liver, monocytes, skeletal muscle, subcutaneous adipose tissue and lymphoblastoid cell lines) and observed a tissue-specific pattern of alternative splicing. We tested whether the expression of TCF7L2 splicing forms was associated with single nucleotide polymorphisms (SNPs), rs7903146 and rs12255372, located within introns 3 and 4 of the gene and most strongly associated with T2D. Expression of two splicing forms was lower in pancreatic islets with increasing counts of T2D-associated alleles of the SNPs: a ubiquitous splicing form (P = 0.018 for rs7903146 and P = 0.020 for rs12255372) and a splicing form found in pancreatic islets, pancreas and colon but not in other tissues tested here (P = 0.009 for rs12255372 and P = 0.053 for rs7903146). Expression of this form in glucose-stimulated pancreatic islets correlated with expression of proinsulin (r(2) = 0.84-0.90, P < 0.00063). In summary, we identified a tissue-specific pattern of alternative splicing of TCF7L2. After adjustment for multiple tests, no association between expression of TCF7L2 in eight types of human tissue samples and T2D-associated genetic variants remained significant. Alternative splicing of TCF7L2 in pancreatic islets warrants future studies. GenBank Accession Numbers: FJ010164-FJ010174.


Subject(s)
Alternative Splicing , Diabetes Mellitus, Type 2/genetics , Organ Specificity , TCF Transcription Factors/genetics , Adolescent , Adult , Cell Line , Colon/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Gene Expression , Humans , Islets of Langerhans/metabolism , Liver/metabolism , Male , Middle Aged , Molecular Sequence Data , Muscle, Skeletal/metabolism , Pancreas/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...