Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Int J Mol Sci ; 24(21)2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37958823

ABSTRACT

Age-related alterations in cardiac function, metabolic, inflammatory and antioxidant profiles are associated with an increased risk of cardiovascular mortality and morbidity. Here, we examined cardiac and metabolic phenotypes in relation to inflammatory status and antioxidant capacity in young, middle-aged and old mice. Real-time reverse transcription-polymerase chain reactions were performed on myocardium and immunoassays on plasma. Left ventricular (LV) structure and function were assessed by echocardiography using high-frequency ultrasound. Middle-aged mice exhibited an altered metabolic profile and antioxidant capacity compared to young mice, whereas myocardial expression of inflammatory factors (TNFα, IL1ß, IL6 and IL10) remained unchanged. In contrast, old mice exhibited increased expression of inflammatory cytokines and plasma levels of resistin compared to young and middle-aged mice (p < 0.05). The pro-inflammatory signature of aged hearts was associated with alterations in glutathione redox homeostasis and elevated contents of 4-hydroxynonenal (4-HNE), a marker of lipid peroxidation and oxidative stress. Furthermore, echocardiographic parameters of LV systolic and diastolic functions were significantly altered in old mice compared to young mice. Taken together, these findings suggest age-related shifts in cardiac phenotype encompass the spectrum of metabo-inflammatory abnormalities and altered redox homeostasis.


Subject(s)
Antioxidants , Cytokines , Mice , Animals , Antioxidants/metabolism , Cytokines/metabolism , Heart , Myocardium/metabolism , Oxidative Stress
2.
Int J Mol Sci ; 24(14)2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37511465

ABSTRACT

Environmental stress can disturb the integrative functioning of the cardiovascular system and trigger a number of adaptive and/or maladaptive cell responses. Concomitant with the expanding use of mobile communication systems, public exposure to electromagnetic fields (EMFs) raises the question of the impact of 900 MHz EMFs on cardiovascular health. Therefore, in this study, we experimentally investigated whether 915 MHz EMF exposure influenced cardiac metabolic, antioxidant, apoptotic, and fibro-inflammatory profiles in a mouse model. Healthy mice were sham-exposed or exposed to EMF for 14 days. Western blot analysis using whole cardiac tissue lysates demonstrated that there was no significant change in the expression of oxidative phosphorylation (OXPHOS) complexes between the control and EMF-exposed mice. In addition, the myocardial expression of fibro-inflammatory cytokines, antioxidant enzymes, and apoptosis-related markers remained unchanged in the EMF-challenged hearts. Finally, the structural integrity of the cardiac tissues was preserved among the groups. These findings suggest that the apoptotic, antioxidant, metabolic, and fibro-inflammatory profiles of the heart remained stable under conditions of EMF exposure in the analyzed mice.


Subject(s)
Electromagnetic Fields , Fibromyalgia , Mice , Animals , Electromagnetic Fields/adverse effects , Antioxidants/metabolism , Heart , Oxidative Stress , Myocardium/metabolism , Fibromyalgia/metabolism
4.
Arterioscler Thromb Vasc Biol ; 42(8): 987-1004, 2022 08.
Article in English | MEDLINE | ID: mdl-35708031

ABSTRACT

BACKGROUND: Secretory granules are key elements for platelet functions. Their biogenesis and integrity are regulated by fine-tuned mechanisms that need to be fully characterized. Here, we investigated the role of the phosphoinositide 5-kinase PIKfyve and its lipid products, PtdIns5P (phosphatidylinositol 5 monophosphate) and PtdIns(3,5)P2 (phosphatidylinositol (3,5) bisphosphate) in granule homeostasis in megakaryocytes and platelets. METHODS: For that, we invalidated PIKfyve by pharmacological inhibition or gene silencing in megakaryocytic cell models (human MEG-01 cell line, human imMKCLs, mouse primary megakaryocytes) and in human platelets. RESULTS: We unveiled that PIKfyve expression and its lipid product levels increased with megakaryocytic maturation. In megakaryocytes, PtdIns5P and PtdIns(3,5)P2 were found in alpha and dense granule membranes with higher levels in dense granules. Pharmacological inhibition or knock-down of PIKfyve in megakaryocytes decreased PtdIns5P and PtdIns(3,5)P2 synthesis and induced a vacuolar phenotype with a loss of alpha and dense granule identity. Permeant PtdIns5P and PtdIns(3,5)P2 and the cation channel TRPML (transient receptor potential mucolipin) 1 and TPC (two pore segment channel) 2 activation were able to accelerate alpha and dense granule integrity recovery following release of PIKfyve pharmacological inhibition. In platelets, PIKfyve inhibition specifically impaired the integrity of dense granules culminating in defects in their secretion, platelet aggregation, and thrombus formation. CONCLUSIONS: These data demonstrated that PIKfyve and its lipid products PtdIns5P and PtdIns(3,5)P2 control granule integrity both in megakaryocytes and platelets.


Subject(s)
Megakaryocytes , Phosphatidylinositol 3-Kinases , Phosphatidylinositols , Animals , Blood Platelets/metabolism , Cytoplasmic Granules/metabolism , Humans , Megakaryocytes/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism
5.
Front Pharmacol ; 13: 869179, 2022.
Article in English | MEDLINE | ID: mdl-35431947

ABSTRACT

The regulatory peptide galanin is broadly distributed in the central nervous systems and peripheral tissues where it modulates numerous physiological and pathological processes through binding to its three G-protein-coupled receptors, GalR1-3. However, the function and identity of the galaninergic system in the heart remain unclear. Therefore, we investigated the expression of the galanin receptors in cardiac cells and tissues and found that GalR2 is the dominant receptor subtype in adult mouse hearts, cardiomyocytes and H9C2 cardiomyoblasts. In vivo, genetic suppression of GalR2 promotes cardiac hypertrophy, fibrosis and mitochondrial oxidative stress in the heart. In vitro, GalR2 silencing by siRNA abolished the beneficial effects of galanin on cell hypertrophy and mitochondrial reactive oxygen species (ROS) production. These findings unravel new insights into the role of galaninergic system in the heart and suggest novel therapeutic strategies in heart disease.

6.
Elife ; 112022 01 18.
Article in English | MEDLINE | ID: mdl-35040777

ABSTRACT

Cell surface receptors control how cells respond to their environment. Many cell surface receptors recycle from endosomes to the plasma membrane via a recently discovered pathway, which includes sorting-nexin SNX17, Retriever, WASH, and CCC complexes. Here, using mammalian cells, we discover that PIKfyve and its upstream PI3-kinase VPS34 positively regulate this pathway. VPS34 produces phosphatidylinositol 3-phosphate (PI3P), which is the substrate for PIKfyve to generate PI3,5P2. We show that PIKfyve controls recycling of cargoes including integrins, receptors that control cell migration. Furthermore, endogenous PIKfyve colocalizes with SNX17, Retriever, WASH, and CCC complexes on endosomes. Importantly, PIKfyve inhibition results in displacement of Retriever and CCC from endosomes. In addition, we show that recruitment of SNX17 is an early step and requires VPS34. These discoveries suggest that VPS34 and PIKfyve coordinate an ordered pathway to regulate recycling from endosomes and suggest how PIKfyve functions in cell migration.


Subject(s)
Cell Membrane/metabolism , Endosomes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Animals , Cell Line , Cell Membrane/chemistry , Class III Phosphatidylinositol 3-Kinases/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice
7.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502314

ABSTRACT

Diabetes is a major risk factor for the development of cardiovascular disease with a higher incidence of myocardial infarction. This study explores the role of metformin, a first-line antihyperglycemic agent, in postinfarction fibrotic and inflammatory remodeling in mice. Three-month-old C57BI/6J mice were submitted to 30 min cardiac ischemia followed by reperfusion for 14 days. Intraperitoneal treatment with metformin (5 mg/kg) was initiated 15 min after the onset of reperfusion and maintained for 14 days. Real-time PCR was used to determine the levels of COL3A1, αSMA, CD68, TNF-α and IL-6. Increased collagen deposition and infiltration of macrophages in heart tissues are associated with upregulation of the inflammation-associated genes in mice after 14 days of reperfusion. Metformin treatment markedly reduced postinfarction fibrotic remodeling and CD68-positive cell population in mice. Moreover, metformin resulted in reduced expression of COL3A1, αSMA and CD68 after 14 days of reperfusion. Taken together, these results open new perspectives for the use of metformin as a drug that counteracts adverse myocardial fibroticand inflammatory remodeling after MI.


Subject(s)
Fibrosis/drug therapy , Hypoglycemic Agents/pharmacology , Inflammation/drug therapy , Metformin/pharmacology , Myocardial Infarction/complications , Myocardium/pathology , Animals , Fibrosis/etiology , Fibrosis/pathology , Inflammation/etiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Ventricular Remodeling
8.
Theranostics ; 11(13): 6491-6506, 2021.
Article in English | MEDLINE | ID: mdl-33995670

ABSTRACT

Rationale: TGFß signaling pathway controls tissue fibrotic remodeling, a hallmark in many diseases leading to organ injury and failure. In this study, we address the role of Apilimod, a pharmacological inhibitor of the lipid kinase PIKfyve, in the regulation of cardiac pathological fibrotic remodeling and TGFß signaling pathway. Methods: The effects of Apilimod treatment on myocardial fibrosis, hypertrophy and cardiac function were assessed in vivo in a mouse model of pressure overload-induced heart failure. Primary cardiac fibroblasts and HeLa cells treated with Apilimod as well as genetic mutation of PIKfyve in mouse embryonic fibroblasts were used as cell models. Results: When administered in vivo, Apilimod reduced myocardial interstitial fibrosis development and prevented left ventricular dysfunction. In vitro, Apilimod controlled TGFß-dependent activation of primary murine cardiac fibroblasts. Mechanistically, both Apilimod and genetic mutation of PIKfyve induced TGFß receptor blockade in intracellular vesicles, negatively modulating its downstream signaling pathway and ultimately dampening TGFß response. Conclusions: Altogether, our findings propose a novel function for PIKfyve in the control of myocardial fibrotic remodeling and the TGFß signaling pathway, therefore opening the way to new therapeutic perspectives to prevent adverse fibrotic remodeling using Apilimod treatment.


Subject(s)
Heart Failure/drug therapy , Hydrazones/therapeutic use , Morpholines/therapeutic use , Phosphatidylinositol 3-Kinases/physiology , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Fibroblasts/drug effects , Fibrosis , HEK293 Cells , HeLa Cells , Heart Failure/pathology , Humans , Hydrazones/pharmacology , Male , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Myocardium/pathology , Pyrimidines/pharmacology , Rats , Receptor, Transforming Growth Factor-beta Type II/drug effects , Single-Blind Method , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling/drug effects
9.
Redox Biol ; 40: 101866, 2021 04.
Article in English | MEDLINE | ID: mdl-33493902

ABSTRACT

Autophagy and apoptosis are powerful regulators of multiple facets of cellular metabolism and homeostasis. Here, we uncover that galanin, a pleiotropic peptide, regulates cardiac autophagy and deactivates apoptotic cell death through the Forkhead box protein O1 (FoxO1) pathway. In hypertrophied heart, galanin promotes autophagy and metabolic shift from fatty acid (FA) to glucose oxidation and preserves mitochondrial integrity. In cardiomyoblasts, galanin triggers autophagosome formation and alleviates hypertrophy, apoptotic cell death, and mitochondrial stress. Mechanistically, galanin dictates cell autophagic and anti-apoptotic phenotypes through FoxO1 pathway. Together, these findings uncover a previously unknown role for galanin in the regulation of cardiac autophagy and provide new insights into the molecular mechanisms supporting cell survival in the hypertrophic reprogramming of the heart.


Subject(s)
Galanin , Signal Transduction , Apoptosis , Autophagy , Cardiomegaly , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans
10.
J Mol Cell Cardiol ; 138: 165-174, 2020 01.
Article in English | MEDLINE | ID: mdl-31836542

ABSTRACT

AIMS: Apelin and vitamin E have been proposed as signaling molecules, but their synergistic role is unknown. The aim of this work was to develop vitamin E TPGS/Apelin system to test their cardioprotective and metabolic efficacy in vitro and in vivo. METHODS: FDA-approved surfactant D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS-1000) and Apelin complex were characterized by physico-chemical methods (CMC determination, dynamic light scattering and circular dichroism). In vitro studies were carried out on H9C2 cardiomyoblasts and isolated murine cardiomyocytes. In vivo studies were performed in isoproterenol- and high-fat diet-induced cardiac remodeling models in mice. RESULTS: We found that vitamin E TPGS/Apelin provide cardioprotective and metabolic efficacy in vitro and in vivo. In vitro studies revealed that vitamin E TPGS/Apelin reduces hypoxia-induced mitochondrial ROS production in cultured cardiomyocytes and H9C2 cardiomyoblasts. In addition, vitamin E TPGS/Apelin confers apoptotic response to hypoxic stress in cells. In a mouse model of isoproterenol-induced cardiac injury, TPGS is not able to affect cardiac remodeling, however combination of vitamin E TPGS and Apelin counteracts myocardial apoptosis, oxidative stress, hypertrophy and fibrosis. Furthermore, combination treatment attenuated obesity-induced cardiometabolic and fibrotic remodeling in mice. CONCLUSION: Together, our data demonstrated the therapeutic benefits of vitamin E TPGS/Apelin complex to combat cardiovascular and metabolic disorders.


Subject(s)
Apelin/pharmacology , Cardiotonic Agents/pharmacology , Vitamin E/pharmacology , Animals , Apoptosis/drug effects , Cardiomegaly/complications , Cardiomegaly/pathology , Cell Hypoxia/drug effects , Cell Line , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/pathology , Diet, High-Fat , Fibrosis , Isoproterenol , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Vascular Remodeling/drug effects
11.
Front Pharmacol ; 10: 154, 2019.
Article in English | MEDLINE | ID: mdl-30873028

ABSTRACT

Cardiovascular complications are the most prevalent cause of morbidity and mortality in diabetic patients. Metformin is currently the first-line blood glucose-lowering agent with potential relevance to cardiovascular diseases. However, the underpinning mechanisms of action remain elusive. Here, we report that metformin represses cardiac apoptosis at least in part through inhibition of Forkhead box O1 (FoxO1) pathway. In a mouse model of ischemia-reperfusion (I/R), treatment with metformin attenuated cardiac and hypertrophic remodeling after 14 days of post-reperfusion. Additionally, cardiac expression of brain-like natriuretic peptide (BNP) was significantly reduced in metformin-treated mice after 14 days of cardiac I/R. In cultured H9C2 cells, metformin counteracted hypertrophic and apoptotic responses to metabolic or hypoxic stress. FoxO1 silencing by siRNA abolished anti-apoptotic effect of metformin under hypoxic stress in H9C2 cells. Taken together, these results suggest that metformin protects the heart against hypertrophic and apoptotic remodeling after myocardial infarction.

12.
Int J Chron Obstruct Pulmon Dis ; 13: 2569-2575, 2018.
Article in English | MEDLINE | ID: mdl-30197513

ABSTRACT

Background: Chronic inflammatory process is the main link in COPD pathogenesis, which causes structural changes in the respiratory tract and lungs. In overweight patients, an adipose tissue could contribute to activation of the inflammatory process. Therefore, it is highly important to identify potential biomarkers of inflammation for patients with COPD and obesity. The aim of this study was to investigate the role of interleukin-26 (IL-26) and evaluate the relationship between the level of systemic inflammation, lung function, and body mass index (BMI) in patients with COPD. Patients and methods: Eighty-three patients with COPD in the stable condition (stage 2 according to the 2016 Global Initiative for Chronic Obstructive Lung Disease recommendations), aged 40-70 years, were included in the study. All patients were divided into 2 groups: obese (n=53) (BMI - 30.0-39.9 kg/m2) and non-obese (n=30) (BMI - 18.5-24.9 kg/m2). We conducted patients' examination, spirometry, induced the sputum, determined the level of C-reactive protein (CRP), leptin in serum and IL-26 in sputum. Results: Obese and non-obese COPD patients had a significant increase in IL-26 compared with healthy subjects by 2.3 and 2.6 (P=0.0003). We also observed a higher level of CRP by 1.38 times (P=0.0008), compared with the rate in non-obese COPD patients, and by 1.8 times (P=0.015) higher concentration level of leptin compared with healthy subjects. The sputum IL-26 level had positive correlation with BMI, CRP, and leptin, and a negative - with forced expiratory volume in 1 second (FEV1) and FEV1/forced vital capacity. Leptin level had positive correlation with BMI and CRP, and negative with FEV1, FEV1/forced vital capacity. Conclusion: Obese COPD patients had a higher level of persistent systemic inflammation than non-obese ones, which is confirmed by a significant increase of CRP and leptin in serum. The data confirm that IL-26 can be considered as a perspective marker to detect the inflammation level in lung tissue of COPD patients.


Subject(s)
Body Weight , Interleukins/analysis , Obesity/complications , Pneumonia/diagnosis , Pulmonary Disease, Chronic Obstructive/etiology , Sputum/chemistry , Adult , Aged , Biomarkers/analysis , Body Mass Index , C-Reactive Protein/analysis , Female , Forced Expiratory Volume , Germany , Humans , Inflammation/blood , Inflammation/complications , Inflammation/diagnosis , Leptin/blood , Male , Middle Aged , Obesity/metabolism , Pneumonia/blood , Pneumonia/complications , Pulmonary Disease, Chronic Obstructive/physiopathology
13.
Sci Rep ; 8(1): 7894, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29784904

ABSTRACT

Identification of novel circulating biomarkers predicting death and major cardio-metabolic events in obese patients with heart failure (HF) remains a research priority. In this study, we compared multi-marker profile of non-obese (NOB) and obese (OB) HF patients in relation to mortality outcome. The new multiplex proximity extension assay technology was used to analyze the levels of 92 proteins in plasma samples from HF patients according to body mass index (BMI) categories. At 2-year follow-up, all-cause mortality rates were significantly greater in NOB patients (BMI < 30 kg/m2) compared to the OB patients (BMI > 30 kg/m2) with HF (odds ratio 26; 95% CI: 1.14-624, p < 0,04). Quantitative proteomic analysis revealed thirteen distinct proteins expression profiles of OB and NOB HF patients. Among these proteins, RAGE, CXCL6, CXCL1, CD40, NEMO, VEGF-A, KLK6, PECAM1, PAR1, MMP1, BNP and NTproBNP were down-regulated, whereas leptin was up-regulated in OB HF patients. In addition, an inverse correlation between plasma BNP levels and leptin in OB HF patients was observed (r = -0.58 p = 0.02). This study identifies specific plasma protein signature in OB and NOB patients with HF in relation to mortality outcome.


Subject(s)
Biomarkers/blood , Heart Failure/diagnosis , Obesity/complications , Proteome/analysis , Proteome/metabolism , Aged , Heart Failure/blood , Heart Failure/etiology , Humans , Male , Middle Aged , Retrospective Studies
14.
Oncotarget ; 8(60): 101659-101671, 2017 Nov 24.
Article in English | MEDLINE | ID: mdl-29254194

ABSTRACT

BACKGROUND AND PURPOSE: Galanin is an endogenous peptide involved in diverse physiological functions in the central nervous system including central cardiovascular regulation. The present study was designed to evaluate the potential effects of the short N-terminal galanin fragment 2-15 (G) on cardiac ischemia/reperfusion (I/R) injury. EXPERIMENTAL APPROACH: Peptide G was synthesized by the automatic solid phase method and identified by 1H-NMR spectroscopy and mass spectrometry. Experiments were performed on cultured rat cardiomyoblast (H9C2) cells, isolated perfused working rat hearts and anaesthetized open-chest rats. KEY RESULTS: Cell viability increased significantly after treatment with 10 and 50 nM of G peptide. In hypoxia and reoxygenation conditions, exposure of H9C2 cells to G peptide decreased cell apoptosis and mitochondrial reactive oxygen species (ROS) production. Postischemic infusion of G peptide reduced cell membrane damage and improved functional recovery in isolated hearts during reperfusion. These effects were accompanied by enhanced restoration of myocardial metabolic state. Treatment with G peptide at the onset of reperfusion induced minor changes in hemodynamic variables but significantly reduced infarct size and plasma levels of necrosis markers. CONCLUSION AND IMPLICATIONS: These findings suggest that G peptide is effective in mitigating cardiac I/R injury, thereby providing a rationale for promising tool for the treatment of cardiovascular diseases.

15.
EMBO Mol Med ; 9(6): 770-785, 2017 06.
Article in English | MEDLINE | ID: mdl-28396567

ABSTRACT

PIKfyve is an evolutionarily conserved lipid kinase that regulates pleiotropic cellular functions. Here, we identify PIKfyve as a key regulator of cardiometabolic status and mitochondrial integrity in chronic diet-induced obesity. In vitro, we show that PIKfyve is critical for the control of mitochondrial fragmentation and hypertrophic and apoptotic responses to stress. We also provide evidence that inactivation of PIKfyve by the selective inhibitor STA suppresses excessive mitochondrial ROS production and apoptosis through a SIRT3-dependent pathway in cardiomyoblasts. In addition, we report that chronic STA treatment improves cardiometabolic profile in a mouse model of cardiomyopathy linked to obesity. We provide evidence that PIKfyve inhibition reverses obesity-induced cardiac mitochondrial damage and apoptosis by activating SIRT3. Furthermore, treatment of obese mice with STA improves left ventricular function and attenuates cardiac hypertrophy. In contrast, STA is not able to reduce isoproterenol-induced cardiac hypertrophy in SIRT3.KO mice. Altogether, these results unravel a novel role for PIKfyve in obesity-associated cardiomyopathy and provide a promising therapeutic strategy to combat cardiometabolic complications in obesity.


Subject(s)
Apoptosis , Hypertrophy/pathology , Myocardium/pathology , Phosphoinositide-3 Kinase Inhibitors , Sirtuin 3/metabolism , Animals , Cardiomyopathies/pathology , Cell Line , Disease Models, Animal , Mice, Knockout , Mice, Obese , Obesity/complications , Phosphatidylinositol 3-Kinases
16.
Oncotarget ; 8(13): 21241-21252, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28177906

ABSTRACT

BACKGROUND AND PURPOSE: Galanin is a multifunctional neuropeptide with pleiotropic roles. The present study was designed to evaluate the potential effects of galanin (2-11) (G1) on functional and metabolic abnormalities in response to myocardial ischemia-reperfusion (I/R) injury. EXPERIMENTAL APPROACH: Peptide G1 was synthesized by the 9-fluorenylmethoxycarbonyl (Fmoc)-based solid-phase method. The chemical structure was identified by 1H-NMR spectroscopy and mass spectrometry. Experiments were conducted using a rat model of I/R injury in vivo, isolated perfused rat hearts ex vivo and cultured rat cardiomyoblast H9C2 cells in vitro. Cardiac function, infarct size, myocardial energy metabolism, hemodynamic parameters, plasma levels of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH) were measured in order to evaluate the effects of G1 on myocardial I/R injury. KEY RESULTS: Treatment with G1 increased cell viability in a dose-dependent manner, inhibited cell apoptosis and excessive mitochondrial reactive oxygen species (ROS) production in response to oxidative stress in H9C2 cells. Pre- or postischemic infusion of G1 enhanced functional and metabolic recovery during reperfusion of the ischemic isolated rat heart. Administration of G1 at the onset of reperfusion significantly reduced infarct size and plasma levels of CK-MB and LDH in rats subjected to myocardial I/R injury. CONCLUSIONS AND IMPLICATIONS: These data provide the first evidence for cardioprotective activity of galanin G1 against myocardial I/R injury. Therefore, peptide G1 may represent a promising treatment strategy for ischemic heart disease.


Subject(s)
Galanin/pharmacology , Myocardial Reperfusion Injury/pathology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Disease Models, Animal , In Situ Nick-End Labeling , Isolated Heart Preparation , Male , Rats , Rats, Wistar
17.
Bio Protoc ; 7(5): e2169, 2017 Mar 05.
Article in English | MEDLINE | ID: mdl-34458479

ABSTRACT

Phosphoinositides are rare membrane lipids involved in the control of the major cellular functions and signaling pathways. They are able to recruit specific effector proteins to the cytosolic face of plasma membrane and organelles to coordinate a vast variety of signaling and trafficking processes, as well to maintain specific identity of the different subcellular compartments (Di Paolo and De Camilli, 2006; Lemmon, 2003). Therefore, analysis of these effectors' binding properties and specificity towards different phosphoinositides is crucial for the understanding of their cellular functions. This protocol describes a method to characterize the binding of proteins to different phosphoinositide-containing vesicles.

18.
Nat Commun ; 7: 12203, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27417143

ABSTRACT

The accumulation of lipid droplets (LD) is frequently observed in hepatitis C virus (HCV) infection and represents an important risk factor for the development of liver steatosis and cirrhosis. The mechanisms of LD biogenesis and growth remain open questions. Here, transcriptome analysis reveals a significant upregulation of septin 9 in HCV-induced cirrhosis compared with the normal liver. HCV infection increases septin 9 expression and induces its assembly into filaments. Septin 9 regulates LD growth and perinuclear accumulation in a manner dependent on dynamic microtubules. The effects of septin 9 on LDs are also dependent on binding to PtdIns5P, which, in turn, controls the formation of septin 9 filaments and its interaction with microtubules. This previously undescribed cooperation between PtdIns5P and septin 9 regulates oleate-induced accumulation of LDs. Overall, our data offer a novel route for LD growth through the involvement of a septin 9/PtdIns5P signalling pathway.


Subject(s)
Hepacivirus/pathogenicity , Lipid Droplets/metabolism , Microtubules/metabolism , Phosphatidylinositol Phosphates/metabolism , Septins/metabolism , Cell Line, Tumor , Gene Expression Regulation , Hepacivirus/physiology , Hepatitis C/metabolism , Host-Pathogen Interactions/physiology , Humans , Lipid Droplets/drug effects , Lipid Metabolism/physiology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Microtubules/virology , Oleic Acid/pharmacology , Septins/genetics , Virus Replication
19.
Biochem Biophys Res Commun ; 476(3): 167-73, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27155155

ABSTRACT

Phosphoinositides play a key role in the spatiotemporal control of central intracellular processes and several specific kinases and phosphatases regulating the level of these lipids are implicated in human diseases. Myotubularins are a family of 3-phosphatases acting specifically on phosphatidylinositol 3-monophosphate and phosphatidylinositol 3,5 bisphosphate. Members of this family are mutated in genetic diseases including myotubularin 1 (MTM1) and myotubularin-related protein 2 (MTMR2) which mutations are responsible of X-linked centronuclear myopathy and Charcot-Marie-Tooth neuropathy, respectively. Here we show that MTM1 is expressed in blood platelets and that hundred microliters of blood is sufficient to detect the protein by western blotting. Since the most severe cases of pathogenic mutations of MTM1 lead to loss of expression of the protein, we propose that a minimal amount of blood can allow a rapid diagnostic test of X-linked myotubular myopathy, which is currently based on histopathology of muscle biopsy and molecular genetic testing. In platelets, MTM1 is a highly active 3-phosphatase mainly associated to membranes and found on the dense granules and to a lesser extent on alpha-granules. However, deletion of MTM1 in mouse had no significant effect on platelet count and on platelet secretion and aggregation induced by thrombin or collagen stimulation. Potential compensation by other members of the myotubularin family is conceivable since MTMR2 was easily detectable by western blotting and the mRNA of several members of the family increased during in vitro differentiation of human megakaryocytes and MEG-01 cells. In conclusion, we show the presence of several myotubularins in platelets and propose that minimal amounts of blood can be used to develop a rapid diagnostic test for genetic pathologies linked to loss of expression of these phosphatases.


Subject(s)
Blood Platelets/pathology , Myopathies, Structural, Congenital/diagnosis , Protein Tyrosine Phosphatases, Non-Receptor/analysis , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Line , Gene Expression Regulation , Humans , Mice, Inbred C57BL , Mice, Knockout , Myopathies, Structural, Congenital/blood , Myopathies, Structural, Congenital/genetics , Platelet Aggregation , Protein Tyrosine Phosphatases, Non-Receptor/blood , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RNA, Messenger/genetics
20.
Br J Pharmacol ; 173(11): 1850-63, 2016 06.
Article in English | MEDLINE | ID: mdl-27005319

ABSTRACT

BACKGROUND AND PURPOSE: Apelin-13, an endogenous ligand for the apelin (APJ) receptor, behaves as a potent modulator of metabolic and cardiovascular disorders. Here, we examined the effects of apelin-13 on myocardial injury in a mouse model combining ischaemia/reperfusion (I/R) and obesity and explored their underlying mechanisms. EXPERIMENTAL APPROACH: Adult male C57BL/6J mice were fed a normal diet (ND) or high-fat diet (HFD) for 6 months and then subjected to cardiac I/R. The effects of apelin-13 post-treatment on myocardial injury were evaluated in HFD-fed mice after 24 h I/R. Changes in protein abundance, phosphorylation, subcellular localization and mRNA expression were determined in cardiomyoblast cell line H9C2, primary cardiomyocytes and cardiac tissue from ND- and HFD-fed mice. Apoptosis was evaluated by TUNEL staining and caspase-3 activity. Mitochondrial ultrastructure was analysed by electron microscopy. KEY RESULTS: In HFD-fed mice subjected to cardiac I/R, i.v. administration of apelin-13 significantly reduced infarct size, myocardial apoptosis and mitochondrial damage compared with vehicle-treated animals. In H9C2 cells and primary cardiomyocytes, apelin-13 induced FoxO1 phosphorylation and nuclear exclusion. FoxO1 silencing by siRNA abolished the protective effects of apelin-13 against hypoxia-induced apoptosis and mitochondrial ROS generation. Finally, apelin deficiency in mice fed a HFD resulted in reduced myocardial FoxO1 expression and impaired FoxO1 distribution. CONCLUSIONS AND IMPLICATIONS: These data reveal apelin as a novel regulator of FoxO1 in cardiac cells and provide evidence for the potential of apelin-13 in prevention of apoptosis and mitochondrial damage in conditions combining I/R injury and obesity.


Subject(s)
Apoptosis/drug effects , Diet, High-Fat/adverse effects , Forkhead Box Protein O1/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Myocardial Reperfusion Injury/metabolism , Obesity/drug therapy , Animals , Intercellular Signaling Peptides and Proteins/administration & dosage , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Obesity/metabolism , Obesity/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...