Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Channels (Austin) ; 11(1): 20-33, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27440385

ABSTRACT

Two-pore channels (TPC1-3) comprise a subfamily of the eukaryotic voltage-gated ion channels (VGICs) superfamily that are mainly expressed in acidic stores in plants and animals. TPCS are widespread across the animal kingdom, with primates, mice and rats lacking TPC3, and mainly act as Ca+ and Na+ channels, although it was also suggested that they could be permeable to other ions. Nowadays, TPCs have been related to the development of different diseases, including Parkinson´s disease, obesity or myocardial ischemia. Due to this, their study has raised the interest of the scientific community to try to understand their mechanism of action in order to be able to develop an efficient drug that could regulate TPCs activity. In this review, we will provide an updated view regarding TPCs structure, function and activation, as well as their role in different pathophysiological processes.


Subject(s)
Ion Channels , Animals , Humans , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/physiology , NADP/analogs & derivatives , NADP/physiology , Phosphatidylinositol Phosphates/physiology
2.
J Biosci ; 41(4): 643-658, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27966485

ABSTRACT

Two-pore channels (TPCs or TPCNs) are novel voltage-gated ion channels that have been postulated to act as Ca2+ and/or Na+ channels expressed exclusively in acidic organelles such as endosomes and lysosomes. TPCNs participate in the regulation of diverse biological processes and recently have been proposed to be involved in the pathophysiology of metabolic disorders such as obesity, fatty liver disease and type 2 diabetes mellitus. Due to the importance of these pathologies in the development of cardiovascular diseases, we aimed to study the possible role of two-pore channel 1 (TPCN1) in the regulation of cardiac metabolism. To explore the cardiac function of TPCN1, we developed proteomic approaches as 2-DE-MALDI-MS and LC-MALDI-MS in the cardiac left ventricle of TPCN1 KO and WT mice, and found alterations in several proteins implicated in glucose and fatty acid metabolism in TPCN1 KO vs. WT mice. The results confirmed the altered expression of HFABP, a key fatty acid transport protein, and of enolase and PGK1, the key enzymes in the glycolytic process. Finally, in vitro experiments performed in neonatal rat cardiomyocytes, in which TPCN1 was silenced using siRNAs, confirmed that the downregulation of TPCN1 gene expression increased 2-deoxy-D-[3H]-glucose uptake and GLUT4 mobilization into cell peripherals in cardiac cells. Our results are the first to suggest a potential role for TPCNs in cardiac metabolism regulation.


Subject(s)
Calcium Channels/genetics , Fatty Acid-Binding Proteins/biosynthesis , Glucose Transporter Type 4/biosynthesis , Phosphoglycerate Kinase/biosynthesis , Phosphopyruvate Hydratase/biosynthesis , Animals , Calcium/metabolism , Calcium Channels/biosynthesis , Cardiovascular Diseases/etiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Fatty Acid Binding Protein 3 , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Humans , Lipid Metabolism/genetics , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphoglycerate Kinase/genetics , Phosphopyruvate Hydratase/genetics , Proteomics , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
J Physiol ; 594(11): 3061-77, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26757341

ABSTRACT

KEY POINTS: Two-pore channels (TPCs) were identified as a novel family of endolysosome-targeted calcium release channels gated by nicotinic acid adenine dinucleotide phosphate, as also as intracellular Na(+) channels able to control endolysosomal fusion, a key process in autophagic flux. Autophagy, an evolutionarily ancient response to cellular stress, has been implicated in the pathogenesis of a wide range of cardiovascular pathologies, including heart failure. We report direct evidence indicating that TPCs are involved in regulating autophagy in cardiomyocytes, and that TPC knockout mice show alterations in the cardiac lysosomal system. TPC downregulation implies a decrease in the viability of cardiomyocytes under starvation conditions. In cardiac tissues from both humans and rats, TPC transcripts and protein levels were higher in females than in males, and correlated negatively with markers of autophagy. We conclude that the endolysosomal channels TPC1 and TPC2 are essential for appropriate basal and induced autophagic flux in cardiomyocytes, and also that they are differentially expressed in male and female hearts. ABSTRACT: Autophagy participates in physiological and pathological remodelling of the heart. The endolysosomal two-pore channels (TPCs), TPC1 and TPC2, have been implicated in the regulation of autophagy. The present study aimed to investigate the role of TPC1 and TPC2 in basal and induced cardiac autophagic activity. In cultured cardiomyocytes, starvation induced a significant increase in TPC1 and TPC2 transcripts and protein levels that paralleled the increase in autophagy identified by increased LC3-II and decreased p62 levels. Small interfering RNA depletion of TPC2 alone or together with TPC1 increased both LC3II and p62 levels under basal conditions and in response to serum starvation, suggesting that, under conditions of severe energy depletion (serum plus glucose starvation), changes in the autophagic flux (as assessed by use of bafilomycin A1) occurred either when TPC1 or TPC2 were downregulated. The knockdown of TPCs diminished cardiomyocyte viability under starvation and simulated ischaemia. Electron micrographs of hearts from TPC1/2 double knockout mice showed that cardiomyocytes contained large numbers of immature lysosomes with diameters significantly smaller than those of wild-type mice. In cardiac tissues from humans and rats, TPC1 and TPC2 transcripts and protein levels were higher in females than in males. Furthermore, transcript levels of TPCs correlated negatively with p62 levels in heart tissues. TPC1 and TPC2 are essential for appropriate basal and induced autophagic flux in cardiomyocytes (i.e. there is a negative effect on cell viability under stress conditions in their absence) and they are differentially expressed in male and female human and murine hearts, where they correlate with markers of autophagy.


Subject(s)
Autophagy/physiology , Calcium Channels/physiology , Lysosomes/physiology , Myocytes, Cardiac/physiology , Sex Characteristics , Aged , Animals , Animals, Newborn , Atrial Appendage/physiology , Cells, Cultured , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley
4.
Endocrine ; 52(1): 11-29, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26662184

ABSTRACT

Nesfatin-1 is a new energy-regulating peptide widely expressed at both central and peripheral tissues with pleiotropic effects. In the last years, the study of nesfatin-1 actions and its possible implication in the development of different diseases has created a great interest among the scientific community. In this review, we will summarize nesfatin-1 main functions, focusing on its cardiovascular implications.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Cardiovascular Physiological Phenomena/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Energy Metabolism/genetics , Energy Metabolism/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Adipose Tissue/metabolism , Animals , Humans , Nucleobindins
6.
Cell Physiol Biochem ; 37(1): 176-92, 2015.
Article in English | MEDLINE | ID: mdl-26303782

ABSTRACT

BACKGROUND: The adipokine chemerin has been associated with cardiovascular disease. We investigated the effects of chemerin on viability and intracellular signalling in murine cardiomyocytes, and the effects of insulin and TNF-α on cardiomyocyte chemerin production. METHODS: Hoechst dye vital staining and cell cycle analysis were used to analyse the viability of murine cardiac cells in culture. Western blot was used to explore the phosphorylation of AKT and caspase-9 activity in neonatal rat cardiomyocytes and HL-1 cells. Finally, RT-qPCR, ELISA and western blot were performed to examine chemerin and CMKLR1 expression after insulin and TNF-α treatment in cardiac cells. RESULTS: Chemerin treatment increased apoptosis, reduced phosphorylation of AKT at Thr308 and increased caspase-9 activity in murine cardiomyocytes. Insulin treatment lowered chemerin and CMKLR1 mRNA and protein levels, and the amount of chemerin in the cell media, while TNF-α treatment increased chemerin mRNA and protein levels but decreased expression of the CMKLR1 gene. CONCLUSION: Chemerin induces apoptosis, reduces AKT phosphorylation and increases the cleavage of caspase-9 in murine cardiomyocytes. The expression of chemerin is regulated by important metabolic (insulin) and inflammatory (TNF-α) mediators at cardiac level. Our results suggest that chemerin could play a role in the physiopathology of cardiac diseases.


Subject(s)
Adipokines/metabolism , Apoptosis/physiology , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Caspase 9/metabolism , Cells, Cultured , Insulin/metabolism , Mice , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Chemokine/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Endocrinology ; 154(12): 4757-67, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24064358

ABSTRACT

Nesfatin-1, a satiety-inducing peptide identified in hypothalamic regions that regulate energy balance, is an integral regulator of energy homeostasis and a putative glucose-dependent insulin coadjuvant. We investigated its production by human cardiomyocytes and its effects on glucose uptake, in the main cardiac glucose transporter GLUT-4 and in intracellular signaling. Quantitative RT-PCR, Western blots, confocal immunofluorescence microscopy, and ELISA of human and murine cardiomyocytes and/or cardiac tissue showed that cardiomyocytes can synthesize and secrete nesfatin-1. Confocal microscopy of cultured cardiomyocytes after GLUT-4 labeling showed that nesfatin-1 mobilizes this glucose transporter to cell peripherals. The rate of 2-deoxy-D-[(3)H]glucose incorporation demonstrated that nesfatin-1 induces glucose uptake by HL-1 cells and cultured cardiomyocytes. Nesfatin-1 induced dose- and time-dependent increases in the phosphorylation of ERK1/2, AKT, and AS160. In murine and human cardiac tissue, nesfatin-1 levels varied with diet and coronary health. In conclusion, human and murine cardiomyocytes can synthesize and secrete nesfatin-1, which is able to induce glucose uptake and the mobilization of the glucose transporter GLUT-4 in these cells. Nesfatin-1 cardiac levels are regulated by diet and coronary health.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Glucose Transporter Type 4/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Animal Feed/analysis , Animals , Calcium-Binding Proteins/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Diet , Dietary Fats/pharmacology , Female , Gene Expression Regulation/physiology , Glucose/metabolism , Glucose Transporter Type 4/genetics , Humans , Male , Mice , Nerve Tissue Proteins/genetics , Nucleobindins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
8.
PLoS One ; 7(6): e37505, 2012.
Article in English | MEDLINE | ID: mdl-22701570

ABSTRACT

BACKGROUND: Heart failure (HF) involves alterations in metabolism, but little is known about cardiomyopathy-(CM)-specific or diabetes-independent alterations in gene expression of proteins involved in fatty-acid (FA) uptake and oxidation or in calcium-(Ca(2+))-handling in the human heart. METHODS: RT-qPCR was used to quantify mRNA expression and immunoblotting to confirm protein expression in left-ventricular myocardium from patients with HF (n = 36) without diabetes mellitus of ischaemic (ICM, n = 16) or dilated (DCM, n = 20) cardiomyopathy aetiology, and non-diseased donors (CTL, n = 6). RESULTS: Significant increases in mRNA of genes regulating FA uptake (CD36) and intracellular transport (Heart-FA-Binding Protein (HFABP)) were observed in HF patients vs CTL. Significance was maintained in DCM and confirmed at protein level, but not in ICM. mRNA was higher in DCM than ICM for peroxisome-proliferator-activated-receptor-alpha (PPARA), PPAR-gamma coactivator-1-alpha (PGC1A) and CD36, and confirmed at the protein level for PPARA and CD36. Transcript and protein expression of Ca(2+)-handling genes (Two-Pore-Channel 1 (TPCN1), Two-Pore-Channel 2 (TPCN2), and Inositol 1,4,5-triphosphate Receptor type-1 (IP3R1)) increased in HF patients relative to CTL. Increases remained significant for TPCN2 in all groups but for TPCN1 only in DCM. There were correlations between FA metabolism and Ca(2+)-handling genes expression. In ICM there were six correlations, all distinct from those found in CTL. In DCM there were also six (all also different from those found in CTL): three were common to and three distinct from ICM. CONCLUSION: DCM-specific increases were found in expression of several genes that regulate FA metabolism, which might help in the design of aetiology-specific metabolic therapies in HF. Ca(2+)-handling genes TPCN1 and TPCN2 also showed increased expression in HF, while HF- and CM-specific positive correlations were found among several FA and Ca(2+)-handling genes.


Subject(s)
Calcium/metabolism , Fatty Acids/metabolism , Gene Expression Regulation/physiology , Heart Failure/physiopathology , Metabolic Networks and Pathways/genetics , Myocardium/metabolism , CD36 Antigens/metabolism , Calcium Channels/metabolism , Case-Control Studies , DNA Primers/genetics , Female , Gene Expression Regulation/genetics , Heart Failure/genetics , Heart Failure/metabolism , Heart Ventricles/cytology , Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Male , Middle Aged , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Transcription Factors/metabolism
9.
Biochem Pharmacol ; 82(5): 491-504, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21640714

ABSTRACT

PURPOSE: We investigated whether the direct renin inhibitor aliskiren can affect metabolism in cardiomyocytes from rat, mouse and human sources. METHODS AND RESULTS: At 10-50 µmol/L, aliskiren significantly increased medium-chain-fatty-acid uptake in primary-cultured neonatal-rat and HL-1 adult-mouse-derived cardiomyocytes (BODIPY-induced fluorescence intensity). The fatty-acid transporter CD-36 was correspondingly translocated to, but the glucose transporter Glut-4 away from, the sarcoplasmic reticulum/plasma membrane, in primary-cultured neonatal-rat (CD-36, Glut-4) and adult-human (CD-36) cardiomyocytes (confocal immunocytochemistry). Immunoblotting showed that aliskiren induced phosphorylation of ERK1/2 in cardiomyocytes from all three sources; responses were dose- and time-dependent, unaffected by renin treatment, and did not cause alterations in expression of (P)R or Igf2/M6P receptors. Microarray analysis of the complete genome of aliskiren-treated neonatal-rat cardiomyocytes, with RT-qPCR and immunoblot confirmation assays in rat and human primary cardiomyocytes, showed that aliskiren up-regulated mRNA and increased protein expression of several enzymes important in lipid and glucose metabolism and in cholesterol biosynthesis. Cardiomyocyte cell-cycle and viability were unaffected by aliskiren. CONCLUSIONS: Aliskiren can induce changes in fatty-acid and glucose uptake and expression of key enzymes of lipid and cholesterol metabolism, which are not associated with increased expression of (P)R or Igf2/M6P receptors, in cultured cardiomyocytes.


Subject(s)
Amides/pharmacology , Fatty Acids/metabolism , Fumarates/pharmacology , Lipid Metabolism/drug effects , Myocytes, Cardiac/drug effects , Renin/antagonists & inhibitors , Animals , CD36 Antigens/analysis , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Cholesterol/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose Transporter Type 4/analysis , Humans , Lauric Acids/metabolism , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Receptor, IGF Type 2/analysis
10.
Endocrinology ; 151(7): 3286-98, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410201

ABSTRACT

The current study aimed to compare the effects of the peptide hormone ghrelin and des-G, its unacylated isoform, on glucose and fatty acid uptake and to identify des-G-specific binding sites in cardiomyocytes. In the murine HL-1 adult cardiomyocyte line, ghrelin and des-G had opposing metabolic effects: des-G increased medium-chain fatty acid uptake (BODIPY fluorescence intensity), whereas neither ghrelin alone nor in combination with des-G did so. Ghrelin inhibited the increase in glucose uptake normally induced by insulin (rate of 2-[(3)H]deoxy-d-glucose incorporation), but des-G did not; des-G was also able to partially reverse the inhibitory effect of ghrelin. In HL-1 cells and primary cultures of neonatal rat cardiomyocytes, des-G but not ghrelin increased insulin-induced translocation of glucose transporter-4 from nuclear to cytoplasmic compartments (immunohistochemistry and quantitative confocal analysis). AKT was phosphorylated by insulin but not affected by ghrelin or des-G, whereas neither AMP-activated protein kinase nor phosphatase and tensin homolog deleted from chromosome 10 was phosphorylated by any treatments. HL-1 and primary-cultured mouse and rat cardiomyocytes each possessed two independent specific binding sites for des-G not recognized by ghrelin (radioreceptor assays). Neither ghrelin nor des-G affected viability (dimethylthiazol diphenyltetrazolium bromide assays), whereas both isoforms were equally protective against apoptosis. Therefore, in cardiomyocytes, des-G binds to specific receptors and has effects on glucose and medium-chain fatty acid uptake that are distinct from those of ghrelin. Real-time PCR indicated that expression levels of ghrelin O-acyltransferase RNA were comparable between HL-1 cells, human myocardial tissue, and human and murine stomach tissue, indicating the possibility of des-G conversion to ghrelin within our model.


Subject(s)
Ghrelin/metabolism , Ghrelin/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Binding Sites , Biological Transport/drug effects , Cell Line , Gastric Mucosa/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Immunoblotting , Immunohistochemistry , Insulin/pharmacology , Lauric Acids/metabolism , Mice , Microscopy, Confocal , Myocytes, Cardiac/cytology , PTEN Phosphohydrolase/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...