Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
ACS Omega ; 5(45): 29127-29139, 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33225144

ABSTRACT

In enterocytes, protein RS1 (RSC1A1) mediates an increase of glucose absorption after ingestion of glucose-rich food via upregulation of Na+-d-glucose cotransporter SGLT1 in the brush-border membrane (BBM). Whereas RS1 decelerates the exocytotic pathway of vesicles containing SGLT1 at low glucose levels between meals, RS1-mediated deceleration is relieved after ingestion of glucose-rich food. Regulation of SGLT1 is mediated by RS1 domain RS1-Reg, in which Gln-Ser-Pro (QSP) is effective. In contrast to QSP and RS1-Reg, Gln-Glu-Pro (QEP) and RS1-Reg with a serine to glutamate exchange in the QSP motif downregulate the abundance of SGLT1 in the BBM at high intracellular glucose concentrations by about 50%. We investigated whether oral application of QEP improves diabetes in db/db mice and affects the induction of diabetes in New Zealand obese (NZO) mice under glucolipotoxic conditions. After 6-day administration of drinking water containing 5 mM QEP to db/db mice, fasting glucose was decreased, increase of blood glucose in the oral glucose tolerance test was blunted, and insulin sensitivity was increased. When QEP was added for several days to a high fat/high carbohydrate diet that induced diabetes in NZO mice, the increase of random plasma glucose was prevented, accompanied by lower plasma insulin levels. QEP is considered a lead compound for development of new antidiabetic drugs with more rapid cellular uptake. In contrast to SGLT1 inhibitors, QEP-based drugs may be applied in combination with insulin for the treatment of type 1 and type 2 diabetes, decreasing the required insulin amount, and thereby may reduce the risk of hypoglycemia.

2.
Diabetes ; 66(1): 25-35, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27999105

ABSTRACT

Hepatic DPP4 expression is elevated in subjects with ectopic fat accumulation in the liver. However, whether increased dipeptidyl peptidase 4 (DPP4) is involved in the pathogenesis or is rather a consequence of metabolic disease is not known. We therefore studied the transcriptional regulation of hepatic Dpp4 in young mice prone to diet-induced obesity. Already at 6 weeks of age, expression of hepatic Dpp4 was increased in mice with high weight gain, independent of liver fat content. In the same animals, methylation of four intronic CpG sites was decreased, amplifying glucose-induced transcription of hepatic Dpp4 In older mice, hepatic triglyceride content was increased only in animals with elevated Dpp4 expression. Expression and release of DPP4 were markedly higher in the liver compared with adipose depots. Analysis of human liver biopsy specimens revealed a correlation of DPP4 expression and DNA methylation to stages of hepatosteatosis and nonalcoholic steatohepatitis. In summary, our results indicate a crucial role of the liver in participation to systemic DPP4 levels. Furthermore, the data show that glucose-induced expression of Dpp4 in the liver is facilitated by demethylation of the Dpp4 gene early in life. This might contribute to early deteriorations in hepatic function, which in turn result in metabolic disease such as hepatosteatosis later in life.


Subject(s)
Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Fatty Liver/metabolism , Liver/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , CpG Islands/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Gene Expression Regulation , Glucose/metabolism , Hepatocytes/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/metabolism
3.
Hum Mol Genet ; 25(12): 2588-2599, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27126637

ABSTRACT

Obesity and ectopic fat disposition are risk factors for metabolic disease. Recent data indicate that IGFBP2 expression in liver is epigenetically inhibited during hepatic steatosis. The aim of this study was to investigate if epigenetic de-regulation of hepatic Igfbp2 occurs already early in life and is associated with increased risk for diet-induced obesity (DIO) during adolescence. Male C57BL/6J mice received a high-fat diet. After 3 weeks on this diet (age of 6 weeks), DIO-susceptible (responder, Resp) and DIO-resistant (non-responder, nResp) mice were identified by early weight gain. At the age of 6 weeks, Resp mice exhibited elevated blood glucose (p < 0.05), plasma insulin (p < 0.01), HOMA-IR and leptin/adiponectin ratio, whereas liver triglycerides were identical but significantly increased (p < 0.01) in Resp mice at 20 weeks of age. Igfbp2 expression was reduced in young Resp compared with nResp mice (p < 0.01), an effect that correlated with elevated DNA methylation of intronic CpG2605 (p < 0.01). The epigenetic inhibition of Igfbp2 was stable over time and preceded DIO and hepatosteatosis in adult mice. In vitro studies demonstrated that selective methylation of CpG2605 significantly reduced reporter activity by ∼85%, indicating that Igfbp2 expression is modulated by methylation. In human whole blood cells, methylation of IGFBP2 at the homologous CpG site was increased in obese men with impaired glucose tolerance. In conclusion, our data show that increased methylation of hepatic Igfbp2 during infancy predicts the development of fatty liver later in life and is linked to deterioration of glucose metabolism.


Subject(s)
DNA Methylation/genetics , Fatty Liver/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Obesity/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose , Diet, High-Fat , Fatty Liver/blood , Fatty Liver/pathology , Female , Gene Expression Regulation , Humans , Insulin/blood , Insulin Resistance/genetics , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Liver/metabolism , Liver/pathology , Male , Mice , Middle Aged , Obesity/blood , Obesity/pathology
4.
Diabetologia ; 58(3): 604-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25527001

ABSTRACT

AIMS/HYPOTHESIS: Oestrogens have previously been shown to exert beta cell protective, glucose-lowering effects in mouse models. Therefore, the recent development of a glucagon-like peptide-1 (GLP-1)-oestrogen conjugate, which targets oestrogen into cells expressing GLP-1 receptors, offers an opportunity for a cell-specific and enhanced beta cell protection by oestrogen. The purpose of this study was to compare the effects of GLP-1 and GLP-1-oestrogen during beta cell failure under glucolipotoxic conditions. METHODS: Male New Zealand obese (NZO) mice were treated with daily s.c. injections of GLP-1 and GLP-1-oestrogen, respectively. Subsequently, the effects on energy homeostasis and beta cell integrity were measured. In order to clarify the targeting of GLP-1-oestrogen, transcription analyses of oestrogen-responsive genes in distinct tissues as well as microarray analyses in pancreatic islets were performed. RESULTS: In contrast to GLP-1, GLP-1-oestrogen significantly decreased food intake resulting in a substantial weight reduction, preserved normoglycaemia, increased glucose tolerance and enhanced beta cell protection. Analysis of hypothalamic mRNA profiles revealed elevated expression of Pomc and Leprb. In livers from GLP-1-oestrogen-treated mice, expression of lipogenic genes was attenuated and hepatic triacylglycerol levels were decreased. In pancreatic islets, GLP-1-oestrogen altered the mRNA expression to a pattern that was similar to that of diabetes-resistant NZO females. However, conventional oestrogen-responsive genes were not different, indicating rather indirect protection of pancreatic beta cells. CONCLUSIONS/INTERPRETATION: GLP-1-oestrogen efficiently protects NZO mice against carbohydrate-induced beta cell failure by attenuation of hyperphagia. In this regard, targeted delivery of oestrogen to the hypothalamus by far exceeds the anorexigenic capacity of GLP-1 alone.


Subject(s)
Estrogens/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Hyperphagia/drug therapy , Hyperphagia/metabolism , Insulin-Secreting Cells/drug effects , Animals , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Obese , New Zealand
5.
J Mol Cell Cardiol ; 76: 208-17, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25173922

ABSTRACT

In the insulin resistant heart, energy fuel selection shifts away from glucose utilization towards almost complete dependence on long-chain fatty acids (LCFA). This shift results in excessive cardiac lipid accumulation and eventually heart failure. Lipid-induced cardiomyopathy may be averted by strategies that increase glucose uptake without elevating LCFA uptake. Protein kinase-D1 (PKD1) is involved in contraction-induced glucose, but not LCFA, uptake allowing to hypothesize that this kinase is an attractive target to treat lipid-induced cardiomyopathy. For this, cardiospecific constitutively active PKD1 overexpression (caPKD1)-mice were subjected to an insulin resistance-inducing high fat-diet for 20-weeks. Substrate utilization was assessed by microPET and cardiac function by echocardiography. Cardiomyocytes were isolated for measurement of substrate uptake, lipid accumulation and insulin sensitivity. Wild-type mice on a high fat-diet displayed increased basal myocellular LCFA uptake, increased lipid deposition, greatly impaired insulin signaling, and loss of insulin-stimulated glucose and LCFA uptake, which was associated with concentric hypertrophic remodeling. The caPKD1 mice on high-fat diet showed none of these characteristics, whereas on low-fat diet a shift towards cardiac glucose utilization in combination with hypertrophy and ventricular dilation was observed. In conclusion, these data suggest that PKD pathway activation may be an attractive therapeutic strategy to mitigate lipid accumulation, insulin resistance and maladaptive remodeling in the lipid-overloaded heart, but this requires further investigation.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Insulin Resistance , Protein Kinase C/metabolism , Animals , Diet, High-Fat/adverse effects , Female , Gene Expression , Heart Ventricles/metabolism , Heart Ventricles/pathology , Histone Deacetylases/metabolism , Lipid Metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Phosphorylation , Protein Kinase C/genetics , Protein Processing, Post-Translational
6.
Diabetes ; 63(12): 4230-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25053586

ABSTRACT

Type 2 diabetes in humans and in obese mice is polygenic. In recent genome-wide association studies, genetic markers explaining a small portion of the genetic contribution to the disease were discovered. However, functional evidence linking these genes with the pathogenesis of diabetes is scarce. We performed RNA sequencing-based transcriptomics of islets from two obese mouse strains, a diabetes-susceptible (NZO) and a diabetes-resistant (B6-ob/ob) mouse, after a short glucose challenge and compared these results with human data. Alignment of 2,328 differentially expressed genes to 106 human diabetes candidate genes revealed an overlap of 20 genes, including TCF7L2, IGFBP2, CDKN2A, CDKN2B, GRB10, and PRC1. The data provide a functional validation of human diabetes candidate genes, including those involved in regulating islet cell recovery and proliferation, and identify additional candidates that could be involved in human ß-cell failure.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Islets of Langerhans/metabolism , Obesity/genetics , Transcriptome/genetics , Animals , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Diabetes Mellitus, Type 2/complications , GRB10 Adaptor Protein/genetics , Gene Expression Profiling , Genes, p16 , Genetic Predisposition to Disease , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Mice , Mice, Inbred Strains , Obesity/complications , Transcription Factor 7-Like 2 Protein/genetics
7.
Am J Physiol Endocrinol Metab ; 307(2): E225-36, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24895286

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) in cardiomyocytes induces translocation of glucose transporter GLUT4 and long-chain fatty acid (LCFA) transporter CD36 from endosomal stores to the sarcolemma to enhance glucose and LCFA uptake, respectively. Ca(2+)/calmodulin-activated kinase kinase-ß (CaMKKß) has been positioned directly upstream of AMPK. However, it is unknown whether acute increases in [Ca(2+)]i stimulate translocation of GLUT4 and CD36 and uptake of glucose and LCFA or whether Ca(2+) signaling converges with AMPK signaling to exert these actions. Therefore, we studied the interplay between Ca(2+) and AMPK signaling in regulation of cardiomyocyte substrate uptake. Exposure of primary cardiomyocytes to inhibitors or activators of Ca(2+) signaling affected neither AMPK-Thr(172) phosphorylation nor basal and AMPK-mediated glucose and LCFA uptake. Despite their lack of an effect on substrate uptake, Ca(2+) signaling activators induced GLUT4 and CD36 translocation. In contrast, AMPK activators stimulated GLUT4/CD36 translocation as well as glucose/LCFA uptake. When cardiomyocytes were cotreated with Ca(2+) signaling and AMPK activators, Ca(2+) signaling activators further enhanced AMPK-induced glucose/LCFA uptake. In conclusion, Ca(2+) signaling shows no involvement in AMPK-induced GLUT4/CD36 translocation and substrate uptake but elicits transporter translocation via a separate pathway requiring CaMKKß/CaMKs. Ca(2+)-induced transporter translocation by itself appears to be ineffective to increase substrate uptake but requires additional AMPK activation to effectuate transporter translocation into increased substrate uptake. Ca(2+)-induced transporter translocation might be crucial under excessive cardiac stress conditions that require supraphysiological energy demands. Alternatively, Ca(2+) signaling might prepare the heart for substrate uptake during physiological contraction by inducing transporter translocation.


Subject(s)
CD36 Antigens/metabolism , Calcium Signaling/physiology , Fatty Acids/metabolism , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , Animals , Calcimycin/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Myocytes, Cardiac/drug effects , Protein Transport/drug effects , Rats , Rats, Inbred Lew , Sarcolemma/drug effects , Thapsigargin/pharmacology
8.
Exp Mol Pathol ; 96(3): 339-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24607416

ABSTRACT

AIM: Cardiac troponin I (cTnI) and T (cTnT) are the most important biomarkers in the diagnosis of acute myocardial infarction (AMI). Nevertheless, they can be elevated in the absence of AMI. It is unclear if such elevations represent irreversible cardiomyocyte-damage or leakage from viable cardiomyocytes. Our objective is to evaluate whether cTn is released from viable cardiomyocytes in response to ischemia and to identify differences in the release of cTn and its molecular forms. METHODS AND RESULTS: HL-1 cardiomyocytes (mouse) were subjected to ischemia (modeled by anoxia with glucose deprivation). The total contents and molecular forms of cTn were determined in culture media and cell lysates. Cell viability was assessed from the release of lactate dehydrogenase (LDH). Before the release of LDH, the intracellular cTn content in ischemic cells decreased significantly compared to control (52% for cTnI; 23% for cTnT) and was not matched by a cTn increase in the medium. cTnI decreased more rapidly than cTnT, resulting in an intracellular cTnT/cTnI ratio of 25.5 after 24 h of ischemia. Western blots revealed changes in the relative amounts of fragmented cTnI and cTnT in ischemic cells. CONCLUSIONS: HL-1 cardiomyocytes subjected to simulated ischemia released cTnI and cTnT only in combination with the release of LDH. We find no evidence of cTn release from viable cardiomyocytes, but did observe a significant decrease in cTn content, before the onset of cell death. Intracellular decrease of cTn in viable cardiomyocytes can have important consequences for the interpretation of cTn values in clinical practice.


Subject(s)
Cell Death/physiology , Myocardial Infarction/diagnosis , Myocytes, Cardiac/metabolism , Troponin I/metabolism , Troponin T/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Ischemia/pathology , L-Lactate Dehydrogenase/metabolism , Mice , Myocytes, Cardiac/pathology
9.
Article in English | MEDLINE | ID: mdl-22580174

ABSTRACT

The fatty acid transporter and scavenger receptor CD36 is increasingly being implicated in the pathogenesis of insulin resistance and its progression towards type 2 diabetes and associated cardiovascular complications. The redistribution of CD36 from intracellular stores to the plasma membrane is one of the earliest changes occurring in the heart during diet induced obesity and insulin resistance. This elicits an increased rate of fatty acid uptake and enhanced incorporation into triacylglycerol stores and lipid intermediates to subsequently interfere with insulin-induced GLUT4 recruitment (i.e., insulin resistance). In the present paper we discuss the potential of CD36 to serve as a target to rectify abnormal myocardial fatty acid uptake rates in cardiac lipotoxic diseases. Two approaches are described: (i) immunochemical inhibition of CD36 present at the sarcolemma and (ii) interference with the subcellular recycling of CD36. Using in vitro model systems of high-fat diet induced insulin resistance, the results indicate the feasibility of using CD36 as a target for adaptation of cardiac metabolic substrate utilization. In conclusion, CD36 deserves further attention as a promising therapeutic target to redirect fatty acid fluxes in the body.


Subject(s)
CD36 Antigens/metabolism , Diabetic Cardiomyopathies/prevention & control , Insulin Resistance , Lipotropic Agents/therapeutic use , Membrane Transport Modulators/therapeutic use , Molecular Targeted Therapy , Myocardium/metabolism , Animals , Biological Transport/drug effects , CD36 Antigens/chemistry , Diabetic Cardiomyopathies/metabolism , Heart/drug effects , Humans , Lipid Metabolism/drug effects , Lipotropic Agents/pharmacology , Membrane Transport Modulators/pharmacology
10.
J Biol Chem ; 287(44): 37530-9, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22936810

ABSTRACT

Cardiac glucose utilization is regulated by reversible translocation of the glucose transporter GLUT4 from intracellular stores to the plasma membrane. During the onset of diet-induced insulin resistance, elevated lipid levels in the circulation interfere with insulin-stimulated GLUT4 translocation, leading to impaired glucose utilization. Recently, we identified vesicle-associated membrane protein (VAMP) 2 and 3 to be required for insulin- and contraction-stimulated GLUT4 translocation, respectively, in cardiomyocytes. Here, we investigated whether overexpression of VAMP2 and/or VAMP3 could protect insulin-stimulated GLUT4 translocation under conditions of insulin resistance. HL-1 atrial cardiomyocytes transiently overexpressing either VAMP2 or VAMP3 were cultured for 16 h with elevated concentrations of palmitate and insulin. Upon subsequent acute stimulation with insulin, we measured GLUT4 translocation, plasmalemmal presence of the fatty acid transporter CD36, and myocellular lipid accumulation. Overexpression of VAMP3, but not VAMP2, completely prevented lipid-induced inhibition of insulin-stimulated GLUT4 translocation. Furthermore, the plasmalemmal presence of CD36 and intracellular lipid levels remained normal in cells overexpressing VAMP3. However, insulin signaling was not retained, indicating an effect of VAMP3 overexpression downstream of PKB/Akt. Furthermore, we revealed that endogenous VAMP3 is bound by the contraction-activated protein kinase D (PKD), and contraction and VAMP3 overexpression protect insulin-stimulated GLUT4 translocation via a common mechanism. These observations indicate that PKD activates GLUT4 translocation via a VAMP3-dependent trafficking step, which pathway might be valuable to rescue constrained glucose utilization in the insulin-resistant heart.


Subject(s)
Glucose Transporter Type 4/metabolism , Insulin Resistance , Myocytes, Cardiac/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , Animals , CD36 Antigens/metabolism , Cell Line , Dietary Fats/pharmacology , Gene Expression , Heart Diseases/metabolism , Heart Diseases/pathology , Insulin/pharmacology , Insulin/physiology , Lipid Metabolism , Male , Mice , Myocardial Contraction , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Palmitates/pharmacology , Protein Kinase C/metabolism , Protein Transport , Rats , Rats, Inbred Lew , Signal Transduction , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 3/genetics
11.
Am J Physiol Heart Circ Physiol ; 303(3): H323-31, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22636676

ABSTRACT

Cardiac myosin-binding protein C (cMyBP-C) is involved in the regulation of cardiac myofilament contraction. Recent evidence showed that protein kinase D (PKD) is one of the kinases that phosphorylate cMyBP-C. However, the mechanism by which PKD-induced cMyBP-C phosphorylation affects cardiac contractile responses is not known. Using immunoprecipitation, we showed that, in contracting cardiomyocytes, PKD binds to cMyBP-C and phosphorylates it at Ser(315). The effect of PKD-mediated phosphorylation of cMyBP-C on cardiac myofilament function was investigated in permeabilized ventricular myocytes, isolated from wild-type (WT) and from cMyBP-C knockout (KO) mice, incubated in the presence of full-length active PKD. In WT myocytes, PKD increased both myofilament Ca(2+) sensitivity (pCa(50)) and maximal Ca(2+)-activated tension of contraction (T(max)). In cMyBP-C KO skinned myocytes, PKD increased pCa(50) but did not alter T(max). This suggests that cMyBP-C is not involved in PKD-mediated sensitization of myofilaments to Ca(2+) but is essential for PKD-induced increase in T(max). Furthermore, the phosphorylation of both PKD-Ser(916) and cMyBP-C-Ser(315) was contraction frequency-dependent, suggesting that PKD-mediated cMyBP-C phosphorylation is operational primarily during periods of increased contractile activity. Thus, during high contraction frequency, PKD facilitates contraction of cardiomyocytes by increasing Ca(2+) sensitivity and by an increased T(max) through phosphorylation of cMyBP-C.


Subject(s)
Carrier Proteins/metabolism , Excitation Contraction Coupling , Myocardial Contraction , Myocytes, Cardiac/enzymology , Protein Kinase C/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Carrier Proteins/genetics , Electric Stimulation , Excitation Contraction Coupling/drug effects , Immunoprecipitation , Male , Mice , Mice, Knockout , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myofibrils/enzymology , Phosphorylation , Protein Binding , Rats , Rats, Inbred Lew , Serine
12.
J Biol Chem ; 287(8): 5871-81, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22158620

ABSTRACT

Increased contraction enhances substrate uptake into cardiomyocytes via translocation of the glucose transporter GLUT4 and the long chain fatty acid (LCFA) transporter CD36 from intracellular stores to the sarcolemma. Additionally, contraction activates the signaling enzymes AMP-activated protein kinase (AMPK) and protein kinase D1 (PKD1). Although AMPK has been implicated in contraction-induced GLUT4 and CD36 translocation in cardiomyocytes, the precise role of PKD1 in these processes is not known. To study this, we triggered contractions in cardiomyocytes by electric field stimulation (EFS). First, the role of PKD1 in GLUT4 and CD36 translocation was defined. In PKD1 siRNA-treated cardiomyocytes as well as cardiomyocytes from PKD1 knock-out mice, EFS-induced translocation of GLUT4, but not CD36, was abolished. In AMPK siRNA-treated cardiomyocytes and cardiomyocytes from AMPKα2 knock-out mice, both GLUT4 and CD36 translocation were abrogated. Hence, unlike AMPK, PKD1 is selectively involved in glucose uptake. Second, we analyzed upstream factors in PKD1 activation. Cardiomyocyte contractions enhanced reactive oxygen species (ROS) production. Using ROS scavengers, we found that PKD1 signaling and glucose uptake are more sensitive to changes in intracellular ROS than AMPK signaling or LCFA uptake. Furthermore, silencing of death-activated protein kinase (DAPK) abrogated EFS-induced GLUT4 but not CD36 translocation. Finally, possible links between PKD1 and AMPK signaling were investigated. PKD1 silencing did not affect AMPK activation. Reciprocally, AMPK silencing did not alter PKD1 activation. In conclusion, we present a novel contraction-induced ROS-DAPK-PKD1 pathway in cardiomyocytes. This pathway is activated separately from AMPK and mediates GLUT4 translocation/glucose uptake, but not CD36 translocation/LCFA uptake.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Muscle Contraction , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , CD36 Antigens/metabolism , Cell Line , Female , Gene Knockdown Techniques , Glucose Transporter Type 4/metabolism , Male , Mice , Myocytes, Cardiac/cytology , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Transport , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Article in English | MEDLINE | ID: mdl-21571515

ABSTRACT

In response to a chronic high plasma concentration of long-chain fatty acids (FAs), the heart is forced to increase the uptake of FA at the cost of glucose. This switch in metabolic substrate uptake is accompanied by an increased presence of the FA transporter CD36 at the cardiac plasma membrane and over time results in the development of cardiac insulin resistance and ultimately diabetic cardiomyopathy. FA can interact with peroxisome proliferator-activated receptors (PPARs), which induce upregulation of the expression of enzymes necessary for their disposal through mitochondrial ß-oxidation, but also stimulate FA uptake. This then leads to a further increase in FA concentration in the cytoplasm of cardiomyocytes. These metabolic changes are supposed to play an important role in the development of cardiomyopathy. Although the onset of this pathology is an increased FA utilization by the heart, the subsequent lipid overload results in an increased production of reactive oxygen species (ROS) and accumulation of lipid intermediates such as diacylglycerols (DAG) and ceramide. These compounds have a profound impact on signaling pathways, in particular insulin signaling. Over time the metabolic changes will introduce structural changes that affect cardiac contractile characteristics. The present mini-review will focus on the lipid-induced changes that link metabolic perturbation, characteristic for type 2 diabetes, with cardiac remodeling and dysfunction.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Dietary Fats/adverse effects , Fatty Acids/metabolism , Myocardium/metabolism , Animals , CD36 Antigens/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Ceramides/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Diglycerides/metabolism , Humans , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/pathology , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptors/metabolism , Reactive Oxygen Species/metabolism
14.
Cell Mol Life Sci ; 68(15): 2525-38, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21547502

ABSTRACT

Cardiomyocytes use glucose as well as fatty acids for ATP production. These substrates are transported into the cell by glucose transporter 4 (GLUT4) and the fatty acid transporter CD36. Besides being located at the sarcolemma, GLUT4 and CD36 are stored in intracellular compartments. Raised plasma insulin concentrations and increased cardiac work will stimulate GLUT4 as well as CD36 to translocate to the sarcolemma. As so far studied, signaling pathways that regulate GLUT4 translocation similarly affect CD36 translocation. During the development of insulin resistance and type 2 diabetes, CD36 becomes permanently localized at the sarcolemma, whereas GLUT4 internalizes. This juxtaposed positioning of GLUT4 and CD36 is important for aberrant substrate uptake in the diabetic heart: chronically increased fatty acid uptake at the expense of glucose. To explain the differences in subcellular localization of GLUT4 and CD36 in type 2 diabetes, recent research has focused on the role of proteins involved in trafficking of cargo between subcellular compartments. Several of these proteins appear to be similarly involved in both GLUT4 and CD36 translocation. Others, however, have different roles in either GLUT4 or CD36 translocation. These trafficking components, which are differently involved in GLUT4 or CD36 translocation, may be considered novel targets for the development of therapies to restore the imbalanced substrate utilization that occurs in obesity, insulin resistance and diabetic cardiomyopathy.


Subject(s)
CD36 Antigens/metabolism , Glucose Transporter Type 4/metabolism , Myocytes, Cardiac/metabolism , Animals , Fatty Acids/metabolism , Glucose/metabolism , Humans , Intracellular Space/metabolism , Models, Biological , Protein Transport
15.
Am J Physiol Cell Physiol ; 298(6): C1549-59, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20375272

ABSTRACT

Insulin and contraction stimulate both cardiac glucose and long-chain fatty acid (LCFA) uptake via translocation of the substrate transporters GLUT4 and CD36, respectively, from intracellular compartments to the sarcolemma. Little is known about the role of vesicular trafficking elements in insulin- and contraction-stimulated glucose and LCFA uptake in the heart, especially whether certain trafficking elements are specifically involved in GLUT4 versus CD36 translocation. Therefore, we studied the role of coat proteins, actin- and microtubule-filaments and endosomal pH on glucose and LCFA uptake into primary cardiomyocytes under basal conditions and during stimulation with insulin or oligomycin (contraction-like AMP-activated protein kinase activator). Inhibition of coat protein targeting to Golgi/endosomes decreased insulin/oligomycin-stimulated glucose (-42%/-51%) and LCFA (-39%/-68%) uptake. Actin disruption decreased insulin/oligomycin-stimulated glucose uptake (-41%/-75%), while not affecting LCFA uptake. Microtubule disruption did not affect substrate uptake under any condition. Endosomal alkalinization increased basal sarcolemmal CD36 (2-fold), but not GLUT4, content, and concomitantly decreased basal intracellular membrane GLUT4 and CD36 content (-60% and -62%, respectively), indicating successful CD36 translocation and incomplete GLUT4 translocation. Additionally, endosomal alkalinization elevated basal LCFA uptake (1.4-fold) in a nonadditive manner to insulin/oligomycin, and decreased insulin/oligomycin-stimulated glucose uptake (-32%/-68%). In conclusion, 1) CD36 translocation, just like GLUT4 translocation, is a vesicle-mediated process depending on coat proteins, and 2) GLUT4 and CD36 trafficking are differentially dependent on endosomal pH and actin filaments. The latter conclusion suggests novel strategies to alter cardiac substrate preference as part of metabolic modulation therapy.


Subject(s)
Actin Cytoskeleton/metabolism , CD36 Antigens/metabolism , Deoxyglucose/metabolism , Endosomes/metabolism , Glucose Transporter Type 4/metabolism , Myocytes, Cardiac/metabolism , Palmitic Acid/metabolism , AMP-Activated Protein Kinases/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Biological Transport , Brefeldin A/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Coatomer Protein/metabolism , Colchicine/pharmacology , Endosomes/drug effects , Endosomes/ultrastructure , Enzyme Activation , Enzyme Activators/pharmacology , Hydrogen-Ion Concentration , Insulin/metabolism , Male , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Oligomycins/pharmacology , Protein Transport , Rats , Rats, Inbred Lew , Thiazolidines/pharmacology , Tubulin Modulators/pharmacology
16.
Article in English | MEDLINE | ID: mdl-20206486

ABSTRACT

Transport of long-chain fatty acids across the cell membrane has long been thought to occur by passive diffusion. However, in recent years there has been a fundamental shift in understanding, and it is now generally recognized that fatty acids cross the cell membrane via a protein-mediated mechanism. Membrane-associated fatty acid-binding proteins ('fatty acid transporters') not only facilitate but also regulate cellular fatty acid uptake, for instance through their inducible rapid (and reversible) translocation from intracellular storage pools to the cell membrane. A number of fatty acid transporters have been identified, including CD36, plasma membrane-associated fatty acid-binding protein (FABP(pm)), and a family of fatty acid transport proteins (FATP1-6). Fatty acid transporters are also implicated in metabolic disease, such as insulin resistance and type-2 diabetes. In this report we briefly review current understanding of the mechanism of transmembrane fatty acid transport, and the function of fatty acid transporters in healthy cardiac and skeletal muscle, and in insulin resistance/type-2 diabetes. Fatty acid transporters hold promise as a future target to rectify lipid fluxes in the body and regain metabolic homeostasis.


Subject(s)
Cell Membrane/metabolism , Fatty Acid Transport Proteins/physiology , Fatty Acids/pharmacokinetics , Muscles/metabolism , Animals , Biological Transport/physiology , CD36 Antigens/physiology , Fatty Acid-Binding Proteins/physiology , Glucose Transporter Type 4/physiology , Humans , Mice , Models, Biological , Rats
17.
FEBS Lett ; 583(13): 2294-300, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19527715

ABSTRACT

Insulin and muscle contraction increase fatty acid transport into muscle by inducing the translocation of FAT/CD36. We examined (a) whether these effects are additive, and (b) whether other fatty acid transporters (FABPpm, FATP1, FATP4, and FATP6) are also induced to translocate. Insulin and muscle contraction increased glucose transport and plasmalemmal GLUT4 independently and additively (positive control). Palmitate transport was also stimulated independently and additively by insulin and by muscle contraction. Insulin and muscle contraction increased plasmalemmal FAT/CD36, FABPpm, FATP1, and FATP4, but not FATP6. Only FAT/CD36 and FATP1 were stimulated in an additive manner by insulin and by muscle contraction.


Subject(s)
CD36 Antigens/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Muscle Contraction/physiology , Animals , Aspartate Aminotransferase, Mitochondrial/metabolism , Biological Transport , Glucose Transporter Type 4/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects
18.
J Endocrinol ; 201(2): 199-209, 2009 May.
Article in English | MEDLINE | ID: mdl-19273501

ABSTRACT

Insulin stimulates cardiac long-chain fatty acid (LCFA) and glucose uptake via translocation of human homolog of rat fatty acid translocase (CD36) and GLUT4 respectively, from intracellular membrane compartments to the sarcolemma, a process dependent on the activation of phosphatidylinositol-3 kinase. To identify downstream kinases of insulin signaling involved in translocation of CD36 and GLUT4 in the heart, we tested i) which cardiac protein kinase C (PKC) isoforms (alpha, delta, epsilon or zeta) are activated by insulin, and ii) whether PKC isoform-specific inhibition affects insulin-stimulated substrate uptake in the heart. Insulin-stimulated LCFA and glucose uptake were completely blunted by inhibition of PKC-zeta, but not by inhibition of conventional or novel PKCs. Concomitantly, translocation of CD36 and GLUT4 to the sarcolemma was completely blunted upon inhibition of PKC-zeta. However, insulin, in contrast to the diacylglycerol-analog phorbol-12-myristate-13-acetate (PMA), did not induce membrane-attachment of the conventional and novel PKCs-alpha, -delta, and -epsilon. PKC-zeta was already entirely membrane-bound in non-stimulated cells, and neither insulin nor PMA treatment had any effect on the subcellular localization of PKC-zeta. Furthermore, insulin treatment did not change phosphorylation of PKC-alpha, -delta, and -zeta or enzymatic activity of PKC-zeta towards a PKC-zeta substrate peptide. It is concluded that PKC-zeta, but not any other PKC isoform, is necessary for insulin-induced translocation of GLUT4 and CD36. However, PKC-zeta is already fully active under basal conditions and not further activated by insulin, indicating that its role in insulin-stimulated uptake of both glucose and LCFA is permissive rather than regulatory.


Subject(s)
CD36 Antigens/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Myocytes, Cardiac/drug effects , Protein Kinase C/physiology , Animals , Cells, Cultured , Enzyme Activation/drug effects , Fatty Acids/metabolism , Male , Models, Biological , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Rats , Rats, Inbred Lew , Tetradecanoylphorbol Acetate/pharmacology
19.
Biochem J ; 419(2): 447-55, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19138173

ABSTRACT

Although CPT-I (carnitine palmitoyltransferase-I) is generally regarded to present a major rate-controlling site in mitochondrial beta-oxidation, it is incompletely understood whether CPT-I is rate-limiting in the overall LCFA (long-chain fatty acid) flux in the heart. Another important site of regulation of the LCFA flux in the heart is trans-sarcolemmal LCFA transport facilitated by CD36 and FABPpm (plasma membrane fatty acid-binding protein). Therefore, we explored to what extent a chronic pharmacological blockade of the LCFA flux at the level of mitochondrial entry of LCFA-CoA would affect sarcolemmal LCFA uptake. Rats were injected daily with saline or etomoxir, a specific CPT-I inhibitor, for 8 days at 20 mg/kg of body mass. Etomoxir-treated rats displayed a 44% reduced cardiac CPT-I activity. Sarcolemmal contents of CD36 and FABPpm, as well as the LCFA transport capacity, were not altered in the hearts of etomoxir-treated versus control rats. Furthermore, rates of LCFA uptake and oxidation, and glucose uptake by cardiac myocytes from etomoxir-treated rats were not different from control rats, neither under basal nor under acutely induced maximal metabolic demands. Finally, hearts from etomoxir-treated rats did not display triacylglycerol accumulation. Therefore CPT-I appears not to present a major rate-controlling site in total cardiac LCFA flux. It is likely that sarcolemmal LCFA entry rather than mitochondrial LCFA-CoA entry is a promising target for normalizing LCFA flux in cardiac metabolic diseases.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Epoxy Compounds/pharmacology , Fatty Acids/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Biological Transport/drug effects , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Male , Oxidation-Reduction/drug effects , Rats , Triglycerides/metabolism
20.
Cardiovasc Res ; 79(2): 249-58, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18469026

ABSTRACT

Circulating long-chain fatty acids (LCFA) and glucose are the main sources for energy production in the heart. In the healthy heart the ratio of glucose and LCFA oxidation is sensitively balanced and chronic alterations in this substrate mix are closely associated with cardiac dysfunction. While it has been accepted for several years that cardiac glucose uptake is mediated by facilitated transport, i.e. by means of the glucose transport proteins GLUT1 and GLUT4, only in the last few years it has become clear that proteins with high-affinity binding sites to LCFA, referred to as LCFA transporters, are responsible for bulk LCFA uptake. Similar to the GLUTs, the LCFA transporters CD36 and FABP(pm) can be recruited from an intracellular storage compartment to the sarcolemma to increase the rate of substrate uptake. Permanent relocation of LCFA transporters, mainly CD36, from intracellular stores to the sarcolemma is accompanied by accumulation of lipids and lipid metabolites in the heart. As a consequence, insulin signalling and glucose utilization are impaired, leading to decreased contractile activity of the heart. These observations underline the particular role and interplay of substrate carriers for glucose and LCFA in modulating cardiac metabolism, and the development of heart failure. The signalling and trafficking pathways and subcellular machinery regulating translocation of glucose and LCFA transporters are beginning to be unravelled. More knowledge on substrate transporter recycling, especially the similarities and differences between glucose and LCFA transporters, is expected to enable novel therapies aimed at changing the subcellular distribution of glucose and LCFA transporters, thereby manipulating the substrate preference of the diseased heart to help restore cardiac function.


Subject(s)
Fatty Acid Transport Proteins/metabolism , Glucose/metabolism , Heart Diseases/metabolism , Sarcolemma/metabolism , Animals , Biological Transport/physiology , Disease Models, Animal , Humans , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...