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










Publication year range
1.
Cell Mol Biol Lett ; 29(1): 67, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724891

ABSTRACT

BACKGROUND: It is generally accepted that endothelial cells (ECs), primarily rely on glycolysis for ATP production, despite having functional mitochondria. However, it is also known that ECs are heterogeneous, and their phenotypic features depend on the vascular bed. Emerging evidence suggests that liver sinusoidal ECs (LSECs), located in the metabolically rich environment of the liver, show high metabolic plasticity. However, the substrate preference for energy metabolism in LSECs remains unclear. METHODS: Investigations were conducted in primary murine LSECs in vitro using the Seahorse XF technique for functional bioenergetic assays, untargeted mass spectrometry-based proteomics to analyse the LSEC proteome involved in energy metabolism pathways, liquid chromatography-tandem mass spectrometry-based analysis of acyl-carnitine species and Raman spectroscopy imaging to track intracellular palmitic acid. RESULTS: This study comprehensively characterized the energy metabolism of LSECs, which were found to depend on oxidative phosphorylation, efficiently fuelled by glucose-derived pyruvate, short- and medium-chain fatty acids and glutamine. Furthermore, despite its high availability, palmitic acid was not directly oxidized in LSEC mitochondria, as evidenced by the acylcarnitine profile and etomoxir's lack of effect on oxygen consumption. However, together with L-carnitine, palmitic acid supported mitochondrial respiration, which is compatible with the chain-shortening role of peroxisomal ß-oxidation of long-chain fatty acids before further degradation and energy generation in mitochondria. CONCLUSIONS: LSECs show a unique bioenergetic profile of highly metabolically plastic ECs adapted to the liver environment. The functional reliance of LSECs on oxidative phosphorylation, which is not a typical feature of ECs, remains to be determined.


Subject(s)
Endothelial Cells , Energy Metabolism , Fatty Acids , Liver , Oxidative Phosphorylation , Animals , Liver/metabolism , Liver/cytology , Endothelial Cells/metabolism , Mice , Fatty Acids/metabolism , Mitochondria/metabolism , Carnitine/metabolism , Carnitine/analogs & derivatives , Palmitic Acid/metabolism , Mice, Inbred C57BL , Male , Mitochondria, Liver/metabolism , Cells, Cultured , Oxidation-Reduction
2.
Int J Mol Sci ; 24(2)2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36675252

ABSTRACT

The aim of this study was to evaluate the anti-atherosclerotic effect of pomegranate seed oil as a source of conjugated linolenic acid (CLnA) (cis-9,trans-11,cis-13; punicic acid) compared to linolenic acid (LnA) and conjugated linoleic acid (CLA) (cis-9,trans-11) in apoE/LDLR-/- mice. In the LONG experiment, 10-week old mice were fed for the 18 weeks. In the SHORT experiment, 18-week old mice were fed for the 10 weeks. Diets were supplied with seed oils equivalent to an amount of 0.5% of studied fatty acids. In the SHORT experiment, plasma TCh and LDL+VLDL cholesterol levels were significantly decreased in animals fed CLnA and CLA compared to the Control. The expression of PPARα in liver was four-fold increased in CLnA group in the SHORT experiment, and as a consequence the expression of its target gene ACO was three-fold increased, whereas the liver's expression of SREBP-1 and FAS were decreased in CLnA mice only in the LONG experiment. Punicic acid and CLA isomers were determined in the adipose tissue and liver in animals receiving pomegranate seed oil. In both experiments, there were no effects on the area of atherosclerotic plaque in aortic roots. However, in the SHORT experiment, the area of atherosclerosis in the entire aorta in the CLA group compared to CLnA and LnA was significantly decreased. In conclusion, CLnA improved the lipid profile and affected the lipid metabolism gene expression, but did not have the impact on the development of atherosclerotic plaque in apoE/LDLR-/- mice.


Subject(s)
Atherosclerosis , Linoleic Acids, Conjugated , Plaque, Atherosclerotic , Pomegranate , Mice , Animals , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/metabolism , Pomegranate/metabolism , Lipid Metabolism , Linolenic Acids/pharmacology , Linolenic Acids/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Plant Oils/pharmacology , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Linoleic Acids, Conjugated/pharmacology , Linoleic Acids, Conjugated/metabolism
3.
FEBS J ; 288(22): 6563-6580, 2021 11.
Article in English | MEDLINE | ID: mdl-34058074

ABSTRACT

Monocyte chemoattractant protein-induced protein 1 (MCPIP1, alias Regnase 1) is a negative regulator of inflammation, acting through cleavage of transcripts coding for proinflammatory cytokines and by inhibition of NFκB activity. Moreover, it was demonstrated that MCPIP1 regulates lipid metabolism both in adipose tissue and in hepatocytes. In this study, we investigated the effects of tissue-specific Mcpip1 deletion on the regulation of hepatic metabolism and development of nonalcoholic fatty liver disease (NAFLD). We used control Mcpip1fl/fl mice and animals with deletion of Mcpip1 in myeloid leukocytes (Mcpip1fl/fl LysMCre ) and in hepatocytes (Mcpip1fl/fl AlbCre ), which were fed chow or a high-fat diet (HFD) for 12 weeks. Mcpip1fl/fl LysMCre mice fed a chow diet were characterized by a significantly reduced hepatic expression of genes regulating lipid and glucose metabolism, which subsequently resulted in low plasma glucose level and dyslipidemia. These animals also displayed systemic inflammation, demonstrated by increased concentrations of cytokines in the plasma and high Tnfa, Il6, IL1b mRNA levels in the liver and brown adipose tissue (BAT). Proinflammatory leukocyte infiltration into BAT, together with low expression of Ucp1 and Ppargc1a, resulted in hypothermia of 22-week-old Mcpip1fl/fl LysMCre mice. On the other hand, there were no significant changes in phenotype in Mcpip1fl/fl AlbCre mice. Although we detected a reduced hepatic expression of genes regulating glucose metabolism and ß-oxidation in these mice, they remained asymptomatic. Upon feeding with a HFD, Mcpip1fl/fl LysMCre mice did not develop obesity, glucose intolerance, nor hepatic steatosis, but were characterized by low plasma glucose level and dyslipidemia, along with proinflammatory phenotype. Mcpip1fl/fl AlbCre animals, following a HFD, became hypercholesterolemic, but accumulated lipids in the liver at the same level as Mcpip1fl/fl mice, and no changes in the level of soluble factors tested in the plasma were detected. We have demonstrated that Mcpip1 protein plays an important role in the liver homeostasis. Depletion of Mcpip1 in myeloid leukocytes, followed by systemic inflammation, has a more pronounced effect on controlling liver metabolism and homeostasis than the depletion of Mcpip1 in hepatocytes.


Subject(s)
Fatty Liver/metabolism , Liver/metabolism , Myeloid Cells/metabolism , Obesity/metabolism , Ribonucleases/metabolism , Animals , Mice , Mice, Knockout , Mice, Transgenic , Ribonucleases/blood , Ribonucleases/deficiency
4.
J Am Heart Assoc ; 9(21): e016929, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33073641

ABSTRACT

Background Long-term feeding with a high-fat diet (HFD) induces endothelial dysfunction in mice, but early HFD-induced effects on endothelium have not been well characterized. Methods and Results Using an magnetic resonance imaging-based methodology that allows characterization of endothelial function in vivo, we demonstrated that short-term (2 weeks) feeding with a HFD to C57BL/6 mice or to E3L.CETP mice resulted in the impairment of acetylcholine-induced response in the abdominal aorta (AA), whereas, in the thoracic aorta (TA), the acetylcholine-induced response was largely preserved. Similarly, HFD resulted in arterial stiffness in the AA, but not in the TA. The difference in HFD-induced response was ascribed to distinct characteristics of perivascular adipose tissue in the TA and AA, related to brown- and white-like adipose tissue, respectively, as assessed by histology, immunohistochemistry, and Raman spectroscopy. In contrast, short-term HFD-induced endothelial dysfunction could not be linked to systemic insulin resistance, changes in plasma concentration of nitrite, or concentration of biomarkers of glycocalyx disruption (syndecan-1 and endocan), endothelial inflammation (soluble form of vascular cell adhesion molecule 1, soluble form of intercellular adhesion molecule 1 and soluble form of E-selectin), endothelial permeability (soluble form of fms-like tyrosine kinase 1 and angiopoietin 2), and hemostasis (tissue plasminogen activator and plasminogen activator inhibitor 1). Conclusions Short-term feeding with a HFD induces endothelial dysfunction in the AA but not in the TA, which could be ascribed to a differential response of perivascular adipose tissue to a HFD in the AA versus TA. Importantly, early endothelial dysfunction in the AA is not linked to elevation of classical systemic biomarkers of endothelial dysfunction.


Subject(s)
Adipose Tissue/pathology , Aorta, Abdominal/diagnostic imaging , Aorta, Thoracic/diagnostic imaging , Diet, High-Fat , Endothelium, Vascular/physiopathology , Adipose Tissue/metabolism , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/physiopathology , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL
6.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165763, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32169502

ABSTRACT

Excess circulating fatty acids contribute to endothelial dysfunction that subsequently aggravates the metabolic conditions such as fatty liver diseases. However, the exact mechanism of this event is not fully understood, and the investigation on the effect of a direct exposure to fatty acids together with their subsequent fate is of interest. In this work we employed a chemically specific and label-free techniques such as Raman and CARS microscopies, to investigate the process of lipid droplets (LDs) formation in endothelial cells and hepatocytes after exposure to oleic and palmitic acid. We aimed to observe the changes in the composition of LDs associated with metabolism and degradation of lipids. We were able to characterize the diversity in the formation of LDs in endothelium as compared to hepatocytes, as well as the differences in the formation of LDs and degradation manner with respect to the used fatty acid. Thus, for the first time the spectral characteristics of LDs formed in endothelial cells after incubation with oleic and palmitic acid is presented, including the time-dependent changes in their chemical composition.


Subject(s)
Hepatocytes/metabolism , Lipid Droplets/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Fatty Acids/metabolism , Fatty Acids/pharmacology , Hepatocytes/drug effects , Humans , Lipid Droplets/drug effects , Liver/drug effects , Liver/pathology , Oleic Acid/pharmacology , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Spectrum Analysis, Raman
7.
Traffic ; 20(12): 932-942, 2019 12.
Article in English | MEDLINE | ID: mdl-31569283

ABSTRACT

Fenestrae are open transmembrane pores that are a structural hallmark of healthy liver sinusoidal endothelial cells (LSECs). Their key role is the transport of solutes and macromolecular complexes between the sinusoidal lumen and the space of Disse. To date, the biochemical nature of the cytoskeleton elements that surround the fenestrae and sieve plates in LSECs remain largely elusive. Herein, we took advantage of the latest developments in atomic force imaging and super-resolution fluorescence nanoscopy to define the organization of the supramolecular complex(es) that surround the fenestrae. Our data revealed that spectrin, together with actin, lines the inner cell membrane and provided direct structural support to the membrane-bound pores. We conclusively demonstrated that diamide and iodoacetic acid (IAA) affect fenestrae number by destabilizing the LSEC actin-spectrin scaffold. Furthermore, IAA induces rapid and repeatable switching between the open vs closed state of the fenestrae, indicating that the spectrin-actin complex could play an important role in controlling the pore number. Our results suggest that spectrin functions as a key regulator in the structural preservation of the fenestrae, and as such, it might serve as a molecular target for altering transendothelial permeability.


Subject(s)
Actins/metabolism , Cell Membrane/ultrastructure , Endothelial Cells/ultrastructure , Liver/ultrastructure , Spectrin/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cell Membrane/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Liver/blood supply , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Atomic Force , Single Molecule Imaging
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1458-1471, 2019 10.
Article in English | MEDLINE | ID: mdl-31185306

ABSTRACT

Monocyte chemoattractant protein-1-induced protein-1 (MCPIP1) acts as an endonuclease that degrades selected mRNAs, viral RNAs and pre-miRNAs. MCPIP1 inhibits adipogenesis by degradation of C/EBPß mRNA and adipogenesis-related miRNA, however its role in the regulation of hepatic lipid homeostasis is unknown. In this study, we investigated the role of MCPIP1 in the regulation of lipid metabolism in hepatocytes. C57BL/6 mice were fed a high-fat diet (HFD) for 2-20 weeks and next primary hepatocytes and adipose tissue were isolated. For in vitro experiments we used murine primary hepatocytes, control HepG2 cells and HepG2 with overexpressed or silenced MCPIP1. We found that Mcpip1 levels were lower in primary hepatocytes isolated from HFD-fed mice than in control cells starting at 4 weeks of a HFD. Level of Mcpip1 was also depleted in visceral fat isolated from obese and glucose-intolerant mice characterized by fatty liver disease. We showed that MCPIP1 overexpression in HepG2 cells treated with oleate induces the level and activity of peroxisome proliferator-activated receptor γ (PPARγ). This phenotype was reverted upon silencing of MCPIP1 in HepG2 cells and in primary hepatocytes lacking Mcpip1 protein. MCPIP1 activated the PPARγ transcription factor via the thioredoxin-interacting protein (TXNIP)/peroxisome proliferator-activated receptor γ coactivator 1- α (PGC-1α) pathway. MCPIP1 contributes to lipid metabolism in hepatocytes by regulating the TXNIP/PGC-1α/PPARγ pathway.


Subject(s)
Hepatocytes/metabolism , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism , Animals , Carrier Proteins/metabolism , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Signal Transduction
9.
Rocz Panstw Zakl Hig ; 70(1): 27-33, 2019.
Article in English | MEDLINE | ID: mdl-30837743

ABSTRACT

Background: Nutritional recommendations emphasize the need to limit consumption of saturated fatty acids and to increase the intake of polyunsaturated fatty acids in the prevention of non-communicable chronic diseases, particularly cardiovascular diseases. Among the fatty acids with health-related effects on the body, conjugated fatty acids are mentioned (i.e. CLA). Objective: The current study was designed to determine the effects of conjugated linoleic acid (CLA) on serum lipid profile, glucose, liver enzymes activity (AST and ALT), malonic dialdehyde (MDA) as well as lipid hydroperoxide (LPO) concentrations in rats fed diet differing in type of dietary fat. Material and methods: Male Wistar rats were divided into six groups and fed the following diets: control AIN-93G diet contained soybean oil (O) and diets with modification of fat source: butter (B) and margarine (M). The experimental diets were supplemented with 1% of conjugated linoleic acid (O+CLA, B+CLA, M+CLA). After 21 days the blood was collected and lipid profile, glucose, liver enzymes, MDA as well as LPO were analyzed. Results: The dietary treatments had no significant effect on the body weight and liver weight of the animals. The concentrations of total cholesterol (TC) and LDL+VLDL cholesterol were unchanged. Both experimental factors (fat source and CLA) had a significant influence on the TAG and HDL levels. Margarine (M) significantly increased the TAG concentration, whereas CLA had a significant impact on the TAG reduction (M+CLA). Glucose level was significantly decreased in all groups fed diets supplemented with CLA. Serum ALT significantly increased in all CLA groups. Fat source had statistically significant influence on the MDA concentration. The LPO level was significantly elevated in all CLA groups. There was statistically significant interaction of experimental factors (fat source and CLA supplementation) on LPO level. Conclusions: Margarine had an adverse effect on the rat's lipid profile. However, in the group fed with margarine, the addition of CLA decreased the concentration of TAG. Regardless of the type of the dietary fat, CLA supplementation increased the level of LPO in the blood serum of animals.


Subject(s)
Adipose Tissue/drug effects , Fatty Acids/metabolism , Linoleic Acids, Conjugated/pharmacology , Lipids/blood , Oxidative Stress/drug effects , Animals , Male , Rats , Rats, Wistar
10.
Front Physiol ; 10: 6, 2019.
Article in English | MEDLINE | ID: mdl-30809151

ABSTRACT

Healthy liver sinusoidal endothelial cells (LSECs) maintain liver homeostasis, while LSEC dysfunction was suggested to coincide with defenestration. Here, we have revisited the relationship between LSEC pro-inflammatory response, defenestration, and impairment of LSEC bioenergetics in non-alcoholic fatty liver disease (NAFLD) in mice. We characterized inflammatory response, morphology as well as bioenergetics of LSECs in early and late phases of high fat diet (HFD)-induced NAFLD. LSEC phenotype was evaluated at early (2-8 week) and late (15-20 week) stages of NAFLD progression induced by HFD in male C57Bl/6 mice. NAFLD progression was monitored by insulin resistance, liver steatosis and obesity. LSEC phenotype was determined in isolated, primary LSECs by immunocytochemistry, mRNA gene expression (qRT-PCR), secreted prostanoids (LC/MS/MS) and bioenergetics (Seahorse FX Analyzer). LSEC morphology was examined using SEM and AFM techniques. Early phase of NAFLD, characterized by significant liver steatosis and prominent insulin resistance, was related with LSEC pro-inflammatory phenotype as evidenced by elevated ICAM-1, E-selectin and PECAM-1 expression. Transiently impaired mitochondrial phosphorylation in LSECs was compensated by increased glycolysis. Late stage of NAFLD was featured by prominent activation of pro-inflammatory LSEC phenotype (ICAM-1, E-selectin, PECAM-1 expression, increased COX-2, IL-6, and NOX-2 mRNA expression), activation of pro-inflammatory prostaglandins release (PGE2 and PGF2α) and preserved LSEC bioenergetics. Neither in the early nor in the late phase of NAFLD, were LSEC fenestrae compromised. In the early and late phases of NAFLD, despite metabolic and pro-inflammatory burden linked to HFD, LSEC fenestrae and bioenergetics are functionally preserved. These results suggest prominent adaptive capacity of LSECs that might mitigate NAFLD progression.

11.
Hepatology ; 69(2): 876-888, 2019 02.
Article in English | MEDLINE | ID: mdl-30137644

ABSTRACT

The fenestrae of liver sinusoidal endothelial cells (LSECs) allow passive transport of solutes, macromolecules, and particulate material between the sinusoidal lumen and the liver parenchymal cells. Until recently, fenestrae and fenestrae-associated structures were mainly investigated using electron microscopy on chemically fixed LSECs. Hence, the knowledge about their dynamic properties has remained to date largely elusive. Recent progress in atomic force microscopy (AFM) has allowed the study of live cells in three dimensions (X, Y, and Z) over a prolonged time (t) and this at unprecedented speeds and resolving power. Hence, we employed the latest advances in AFM imaging on living LSECs. As a result, we were able to monitor the position, size, and number of fenestrae and sieve plates using four-dimensional AFM (X, Y, Z, and t) on intact LSECs in vitro. During these time-lapse experiments, dynamic data were collected on the origin and morphofunctional properties of the filtration apparatus of LSECs. We present structural evidence on single laying and grouped fenestrae, thereby elucidating their dynamic nature from formation to disappearance. We also collected data on the life span of fenestrae. More especially, the formation and closing of entire sieve plates were observed, and how the continuous rearrangement of sieve plates affects the structure of fenestrae within them was recorded. We observed also the dawn and rise of fenestrae-forming centers and defenestration centers in LSECs under different experimental conditions. Conclusion: Utilizing a multimodal biomedical high-resolution imaging technique we collected fine structural information on the life span, formation, and disappearance of LSEC fenestrae; by doing so, we also gathered evidence on three different pathways implemented in the loss of fenestrae that result in defenestrated LSECs.


Subject(s)
Endothelial Cells/physiology , Liver/cytology , Animals , Cytochalasin B , Depsipeptides , Mice , Microscopy, Atomic Force
12.
J Biophotonics ; 12(4): e201800290, 2019 04.
Article in English | MEDLINE | ID: mdl-30578586

ABSTRACT

Liver sinusoidal endothelial cells (LSECs), a type of endothelial cells with unique morphology and function, play an important role in the liver hemostasis, and LSECs dysfunction is involved in the development of nonalcoholic fatty liver disease (NAFLD). Here, we employed Raman imaging and chemometric data analysis in order to characterize the presence of lipid droplets (LDs) and their lipid content in primary murine LSECs, in comparison with hepatocytes, isolated from mice on high-fat diet. On NAFLD development, LDs content in LSECs changed toward more unsaturated lipids, and this response was associated with an increased expression of stearylo-CoA desaturase-1. To the best of our knowledge, this is a first report characterizing LDs in LSECs, where their chemical composition is analyzed along the progression of NAFLD at the level of single LD using Raman imaging.


Subject(s)
Endothelial Cells/cytology , Hepatocytes/cytology , Lipid Droplets/metabolism , Liver/cytology , Molecular Imaging , Spectrum Analysis, Raman , Animals , Disease Progression , Liver/diagnostic imaging , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology
13.
Analyst ; 143(19): 4723-4731, 2018 Sep 24.
Article in English | MEDLINE | ID: mdl-30191231

ABSTRACT

Raman spectroscopy via fiber optic probes omits some of the major limitations related to ex vivo preparation of tissue samples (e.g. fixation, freezing, cutting) and enables rapid registration of spectra, therefore, this technique has the potential to become a useful diagnostic tool. In this work, we evaluated the applicability of Raman spectroscopy via fiber optic probe for rapid assessment of lipid content in the liver in the context of its potential application as a tool to verify the degree of liver steatosis. Non-alcoholic fatty liver disease (NAFLD) is a common liver disorder that is characterized by excessive lipid accumulation within hepatic tissue and is associated with insulin resistance and metabolic syndrome. Raman spectroscopy via fiber optic probe was applied to investigate the biochemical status of the liver in mild (mice fed a high-fat diet) and severe (db/db mice) models of NAFLD. A considerable increase in lipid content without substantial alterations in composition was observed in mild liver steatosis. In contrast, more severe liver steatosis caused not only a significant lipid content increase, but also hepatic cholesterol accumulation accompanied by significant loss of hepatic vitamin A content. Chemometric analysis based on average Raman spectra recorded via fiber optic probe provided discrimination of mild and severe liver steatosis and control livers with high sensitivity and specificity. In conclusion, our work demonstrates that a relatively simple Raman setup equipped with a commercial fiber optic probe combined with basic chemometric analysis enables rapid quantification of liver steatosis.


Subject(s)
Fiber Optic Technology , Non-alcoholic Fatty Liver Disease/diagnosis , Spectrum Analysis, Raman , Animals , Cluster Analysis , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Pilot Projects , Vitamin A/analysis
14.
Pharmacol Rep ; 70(3): 463-469, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29631249

ABSTRACT

BACKGROUND: The impairment of liver sinusoidal endothelial cells (LSECs) function and diminished nitric oxide (NO) production has been regarded as an important pathogenic factor in liver steatosis. Restoring NO-dependent function was shown to counteract liver steatosis, obesity, and insulin resistance. However, it is not known whether restored liver perfusion and improvement in hepatic blood flow contributes to the anti-steatotic effects of NO. Taking advantage of in vivo MRI, we have examined the effects of short-term treatment with the hepatoselective NO donor V-PYRRO/NO on hepatic microcirculation in advanced liver steatosis. METHODS: Male C57BL/6 mice fed for six months a high fat diet (HFD; 60 kcal% of fat) were treated for 3 weeks with V-PYRRO/NO (twice a day 5mg/kg b.w. ip). An MRI assessment of liver perfusion using the FAIR-EPI method and a portal vein blood flow using the FLASH method were performed. Blood biochemistry, glucose tolerance tests, a histological evaluation of the liver, and liver NO concentrations were also examined. RESULTS: Short-term treatment with V-PYRRO/NO releasing NO selectively in the liver improved liver perfusion and portal vein blood flow. This effect was associated with a slight improvement in glucose tolerance but there was no effect on liver steatosis, body weight, white adipose tissue mass, plasma lipid profile, or aminotransferase activity. CONCLUSION: Short-term treatment with V-PYRRO/NO-derived NO improves perfusion in hepatic microcirculation and this effect may also contribute to the anti-steatotic effects of hepatoselective NO donors linked previously to the modulation of glucose and lipid metabolism in the liver.


Subject(s)
Fatty Liver/drug therapy , Liver/drug effects , Microcirculation/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Pyrrolidines/pharmacology , Regional Blood Flow/drug effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Body Weight/drug effects , Fatty Liver/metabolism , Glucose Tolerance Test/methods , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Transaminases/metabolism
15.
Nanoscale ; 9(47): 18867-18880, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29177344

ABSTRACT

The design of nanodelivery systems has been recently considered as a solution to the major challenge in pharmaceutical research - poor bioavailability of lipophilic drugs. Nanocapsules with liquid oil cores and shells based on amphiphilic polysaccharides were developed here as robust carriers of hydrophobic active compounds. A series of modified charged hyaluronates were synthesized and used as stabilizing shells ensuring also the biocompatibility of the nanocapsules that is crucial for applications related to the delivery of lipophilic drugs in vivo. Importantly, the oil nanodroplets were found to be stably suspended in water for at least 15 months without addition of low molar mass surfactants. Moreover, their size and stability may be tuned by varying the relative content of hydrophobic and hydrophilic groups in the hyaluronate derivatives as was confirmed by dynamic light scattering and nanoparticle tracking analysis as well as electron microscopy. In vivo studies demonstrated that hyaluronate-based nanocapsules accumulated preferentially in the liver as well as in the lungs. Moreover, their accumulation was dramatically potentiated in endotoxemic mice. In vitro studies showed that the nanocapsules were taken up by liver sinusoidal endothelial cells and by mouse lung vascular endothelial cells. Importantly, the capsules were found to be nontoxic in an acute oral toxicity experiment even at a dose of 2000 mg per kg b.w. Biocompatible hyaluronate-based nanocapsules with liquid cores described herein represent a promising and tunable nanodelivery system for lipophilic active compounds via both oral and intravenous administration.

16.
Analyst ; 142(20): 3948-3958, 2017 Oct 09.
Article in English | MEDLINE | ID: mdl-28944783

ABSTRACT

Non-Alcoholic Fatty Liver Disease (NAFLD) is the most prevalent liver disorder worldwide, involving pathogenic mechanisms of liver sinusoidal endothelial cells (LSECs), hepatocytes and other liver cells. Here, we used a novel approach of label-free Raman confocal imaging to study primary LSECs and hepatocytes freshly isolated from the livers of mice with NAFLD induced by a high fat diet (HFD), in comparison to healthy controls. Our aim was to characterize changes in the biochemical composition in LSECs and hepatocytes that occur in a single cell at the subcellular level. LSECs from NAFLD livers displayed a significant increase in the intensity of marker bands of nuclear DNA that was not associated with changes in LSEC nucleus size. A number of changes in the cytoplasm of hepatocytes were identified. However, the most prominent change in hepatocytes was a substantial increase in the degree of unsaturation of LBs' (lipid bodies) lipids in NAFLD, suggesting an increase in the de novo lipogenesis of unsaturated lipids. The confocal Raman imaging of single live cells isolated from the liver provided a unique tool to better understand disease-induced cell-specific changes in the biochemical phenotype of primary liver cells.


Subject(s)
Endothelial Cells/pathology , Hepatocytes/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Diet, High-Fat , Liver/cytology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Single-Cell Analysis , Spectrum Analysis, Raman
17.
Micron ; 101: 48-53, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28623691

ABSTRACT

Liver sinusoidal endothelial cells present unique morphology characterized by the presence of transmembrane pores called fenestrations. The size and number of fenestrations in live cells change dynamically in response to variety of chemical and physical factors. Although scanning electron microscopy is a well-established method for investigation of fixed liver sinusoidal endothelial cells morphology, atomic force microscopy is the interesting alternative providing detailed 3D topographical information. Moreover, simple sample preparation, only by wet-fixation, minimizing sample preparation artifacts enable high-resolution atomic force microscopy-based measurements. In this work, we apply imaging methods based on atomic force microscopy, to describe characteristic features of glutaraldehyde-fixed primary murine liver sinusoidal endothelial cells, namely: mean fenestration diameter, porosity, and fenestrations frequency. We also investigate the effect of different tip apex radius on evaluation of single fenestration diameter. By quantitative description of fenestrations, we demonstrate that atomic force microscopy became a well competing tool for nondestructive quantitative investigation of the liver sinusoidal endothelial cell morphology.


Subject(s)
Capillaries/cytology , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Liver/blood supply , Microscopy, Atomic Force , Animals , Biometry , Imaging, Three-Dimensional , Mice
18.
Front Pharmacol ; 8: 183, 2017.
Article in English | MEDLINE | ID: mdl-28443021

ABSTRACT

Although it is known that 1-methylnicotinamide (MNA) displays vasoprotective activity in mice, as yet the effect of MNA on endothelial function has not been demonstrated in vivo. Here, using magnetic resonance imaging (MRI) we profile the effects of MNA on endothelial phenotype in mice with atherosclerosis (ApoE/LDLR-/-) in vivo, in comparison to angiotensin (Ang) -converting enzyme (ACE) inhibitor (perindopril), with known vasoprotective activity. On a biochemical level, we analyzed whether MNA- or perindopril-induced improvement in endothelial function results in changes in ACE/Ang II-ACE2/Ang-(1-7) balance, and L-arginine/asymmetric dimethylarginine (ADMA) ratio. Endothelial function and permeability were evaluated in the brachiocephalic artery (BCA) in 4-month-old ApoE/LDLR-/- mice that were non-treated or treated for 1 month or 2 months with either MNA (100 mg/kg/day) or perindopril (10 mg/kg/day). The 3D IntraGate®FLASH sequence was used for evaluation of BCA volume changes following acetylcholine (Ach) administration, and for relaxation time (T1) mapping around BCA to assess endothelial permeability using an intravascular contrast agent. Activity of ACE/Ang II and ACE2/Ang-(1-7) pathways as well as metabolites of L-arginine/ADMA pathway were measured using liquid chromatography/mass spectrometry-based methods. In non-treated 6-month-old ApoE/LDLR-/- mice, Ach induced a vasoconstriction in BCA that amounted to -7.2%. 2-month treatment with either MNA or perindopril resulted in the reversal of impaired Ach-induced response to vasodilatation (4.5 and 5.5%, respectively) and a decrease in endothelial permeability (by about 60% for MNA-, as well as perindopril-treated mice). Improvement of endothelial function by MNA and perindopril was in both cases associated with the activation of ACE2/Ang-(1-7) and the inhibition of ACE/Ang II axes as evidenced by an approximately twofold increase in Ang-(1-9) and Ang-(1-7) and a proportional decrease in Ang II and its active metabolites. Finally, MNA and perindopril treatment resulted in an increase in L-arginine/ADMA ratio by 107% (MNA) and 140% (perindopril), as compared to non-treated mice. Functional and biochemical endothelial profiling in ApoE/LDLR-/- mice in vivo revealed that 2-month treatment with MNA (100 mg/kg/day) displayed a similar profile of vasoprotective effect as 2-month treatment with perindopril (10 mg/kg/day): i.e., the improvement in endothelial function that was associated with the beneficial changes in ACE/Ang II-ACE2/Ang (1-7) balance and in L-arginine/ADMA ratio in plasma.

19.
Pharmacol Rep ; 69(3): 560-565, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28364696

ABSTRACT

PURPOSE: To analyze the effect of liver steatosis and obesity on pharmacokinetic profile of two structurally-related liver-selective NO-donors - V-PYRRO/NO and V-PROLI/NO. METHODS: C57BL/6 mice were fed control or high-fat diet for 15 weeks to induced liver steatosis and obesity (HFD mice). Pharmacokinetics and renal elimination studies were conducted in vivo following iv dosing of V-PYRRO/NO and V-PROLI/NO (0.03mmol/kg). Hepatic clearance was evaluated ex vivo in the isolated perfused mice liver and in vitro with the use of liver microsomes. RESULTS: V-PYRRO/NO and V-PROLI/NO, despite similar structure, displayed different pharmacokinetic properties. V-PYRRO/NO was uptaken and metabolized by the liver, while V-PROLI/NO was eliminated unchanged with urine. In HFD mice, despite increased CYP450 metabolism of V-PYRRO/NO the elimination rate was slower most likely due to the impairment of hepatic microcirculation caused by liver fat accumulation. In turn, in HFD mice renal clearence of V-PROLI/NO was accelerated and volume of distribution was increased most likely due to additional intracellular water in HFD mice. CONCLUSIONS: The pharmacokinetics of V-PROLI/NO, the novel proline-based analog of V-PYRRO/NO with additional single carboxylic acid moiety, attached to the molecule of V-PYRRO/NO to improve the water solubility, was differently affected by liver steatosis and obesity as compared with the parent compound V-PYRRO/NO.


Subject(s)
Fatty Liver/physiopathology , Nitric Oxide Donors/pharmacokinetics , Obesity/complications , Pyrrolidines/pharmacokinetics , Triazenes/pharmacokinetics , Animals , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Liver/metabolism , Liver/physiopathology , Male , Mice , Mice, Inbred C57BL , Microcirculation , Microsomes, Liver/metabolism , Nitric Oxide Donors/chemistry , Pyrrolidines/chemistry , Solubility , Tissue Distribution , Triazenes/chemistry
20.
Pharmacol Rep ; 69(1): 112-118, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27915184

ABSTRACT

BACKGROUND: Cholesterol-dependent and independent mechanisms were proposed to explain anti-atherosclerotic action of statins in humans. However, their effects in murine models of atherosclerosis have not been consistently demonstrated. Here, we studied the effects of pravastatin on atherosclerosis in ApoE/LDLR-/- mice fed a control and atherogenic diet. METHODS: ApoE/LDLR-/- mice were fed a control (CHOW) or an atherogenic (Low Carbohydrate High Protein, LCHP) diet. Two doses of pravastatin (40mg/kg and 100mg/kg) were used. The anti-atherosclerotic effects of pravastatin in en face aorta, cross-sections of aortic roots and brachiocephalic artery (BCA) were analysed. The lipid profile was determined. Fourier Transform Infrared Spectroscopy followed by Fuzzy C-Means (FCM) clustering was used for the quantitative assessment of plaque composition. RESULTS: Treatment with pravastatin (100mg/kg) decreased total and LDL cholesterol only in the LCHP group, but displayed a pronounced anti-atherosclerotic effect in BCA and abdominal aorta. The anti-atherosclerotic effect of pravastatin (100mg/kg) in BCA was associated with significant alterations of the chemical plaque composition, including a fall in cholesterol and cholesterol esters contents independently on total cholesterol and LDL concentration in plasma. CONCLUSIONS: Pravastatin at high (100mg/kg), but not low dose displayed a pronounced anti-atherosclerotic effect in ApoE/LDLR-/- mice fed a CHOW or LCHP diet that was remarkable in BCA, visible in en face aorta, whereas it was not observed in aortic roots, suggesting that previous inconsistencies might have been due to the various sites of atherosclerotic plaque analysis.


Subject(s)
Aorta/drug effects , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Brachiocephalic Trunk/drug effects , Pravastatin/therapeutic use , Receptors, LDL/deficiency , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Aorta/metabolism , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Female , Mice , Mice, Knockout , Pravastatin/pharmacology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...