Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Basic Res Cardiol ; 119(3): 481-503, 2024 06.
Article in English | MEDLINE | ID: mdl-38517482

ABSTRACT

Hypercholesterolemia is a major risk factor for coronary artery diseases and cardiac ischemic events. Cholesterol per se could also have negative effects on the myocardium, independently from hypercholesterolemia. Previously, we reported that myocardial ischemia-reperfusion induces a deleterious build-up of mitochondrial cholesterol and oxysterols, which is potentiated by hypercholesterolemia and prevented by translocator protein (TSPO) ligands. Here, we studied the mechanism by which sterols accumulate in cardiac mitochondria and promote mitochondrial dysfunction. We performed myocardial ischemia-reperfusion in rats to evaluate mitochondrial function, TSPO, and steroidogenic acute regulatory protein (STAR) levels and the related mitochondrial concentrations of sterols. Rats were treated with the cholesterol synthesis inhibitor pravastatin or the TSPO ligand 4'-chlorodiazepam. We used Tspo deleted rats, which were phenotypically characterized. Inhibition of cholesterol synthesis reduced mitochondrial sterol accumulation and protected mitochondria during myocardial ischemia-reperfusion. We found that cardiac mitochondrial sterol accumulation is the consequence of enhanced influx of cholesterol and not of the inhibition of its mitochondrial metabolism during ischemia-reperfusion. Mitochondrial cholesterol accumulation at reperfusion was related to an increase in mitochondrial STAR but not to changes in TSPO levels. 4'-Chlorodiazepam inhibited this mechanism and prevented mitochondrial sterol accumulation and mitochondrial ischemia-reperfusion injury, underlying the close cooperation between STAR and TSPO. Conversely, Tspo deletion, which did not alter cardiac phenotype, abolished the effects of 4'-chlorodiazepam. This study reveals a novel mitochondrial interaction between TSPO and STAR to promote cholesterol and deleterious sterol mitochondrial accumulation during myocardial ischemia-reperfusion. This interaction regulates mitochondrial homeostasis and plays a key role during mitochondrial injury.


Subject(s)
Mitochondria, Heart , Myocardial Reperfusion Injury , Phosphoproteins , Animals , Rats , Benzodiazepinones , Cholesterol/metabolism , Disease Models, Animal , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Rats, Wistar , Receptors, GABA/metabolism , Receptors, GABA/genetics , Receptors, GABA-A
2.
Epilepsia ; 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39367795

ABSTRACT

OBJECTIVE: Epilepsy surgery is a treatment option for patients with seizures that do not respond to pharmacotherapy. The histopathological characterization of the resected tissue has an important prognostic value to define postoperative seizure outcome in these patients. However, the diagnostic classification process based on microscopic assessment remains challenging, particularly in the case of focal cortical dysplasia (FCD). Imaging mass spectrometry is a spatial omics technique that could improve tissue phenotyping and patient stratification by investigating hundreds of biomolecules within a single tissue sample, without the need for target-specific reagents. METHODS: An in situ proteomic technique called matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) is here investigated as a potential new tool to expand conventional diagnosis on standard paraffin brain tissue sections. Unsupervised and region of interest-based MALDI-MSI analyses of sections from 10 FCD type IIb (FCDIIb) cases were performed, and the results were validated by immunohistochemistry. RESULTS: MALDI-MSI identified distinct histopathological features and the boundaries of the dysplastic lesion. The capability to visualize the spatial distribution of well-known diagnostic markers enabling multiplex measurements on single tissue sections was demonstrated. Finally, a fingerprint list of potential discriminant peptides that distinguish FCD core from peri-FCD tissue was generated. SIGNIFICANCE: This is the first study that explores the potential application of MALDI-MSI in epilepsy postsurgery fixed tissue, by utilizing the well-characterized FCDIIb features as a model. Extending these preliminary analyses to a larger cohort of patients will generate spectral libraries of molecular signatures that discriminate tissue features and will contribute to patient phenotyping.

3.
Pharmacol Res ; 194: 106823, 2023 08.
Article in English | MEDLINE | ID: mdl-37336430

ABSTRACT

Evidence that Huntington's disease (HD) is characterized by impaired cholesterol biosynthesis in the brain has led to strategies to increase its level in the brain of the rapidly progressing R6/2 mouse model, with a positive therapeutic outcome. Here we tested the long-term efficacy of chronic administration of cholesterol to the brain of the slowly progressing zQ175DN knock-in HD mice in preventing ("early treatment") or reversing ("late treatment") HD symptoms. To do this we used the most advanced formulation of cholesterol loaded brain-permeable nanoparticles (NPs), termed hybrid-g7-NPs-chol, which were injected intraperitoneally. We show that one cycle of treatment with hybrid-g7-NPs-chol, administered in the presymptomatic ("early treatment") or symptomatic ("late treatment") stages is sufficient to normalize cognitive defects up to 5 months, as well as to improve other behavioral and neuropathological parameters. A multiple cycle treatment combining both early and late treatments ("2 cycle treatment") lasting 6 months generates therapeutic effects for more than 11 months, without severe adverse reactions. Sustained cholesterol delivery to the brain of zQ175DN mice also reduces mutant Huntingtin aggregates in both the striatum and cortex and completely normalizes synaptic communication in the striatal medium spiny neurons compared to saline-treated HD mice. Furthermore, through a meta-analysis of published and current data, we demonstrated the power of hybrid-g7-NPs-chol and other strategies able to increase brain cholesterol biosynthesis, to reverse cognitive decline and counteract the formation of mutant Huntingtin aggregates. These results demonstrate that cholesterol delivery via brain-permeable NPs is a therapeutic option to sustainably reverse HD-related behavioral decline and neuropathological signs over time, highlighting the therapeutic potential of cholesterol-based strategies in HD patients. DATA AVAILABILITY: This study does not include data deposited in public repositories. Data are available on request to the corresponding authors.


Subject(s)
Huntington Disease , Mice , Animals , Huntington Disease/drug therapy , Huntington Disease/pathology , Brain/pathology , Cholesterol , Corpus Striatum/pathology , Cognition , Disease Models, Animal , Mice, Transgenic
4.
Brain ; 144(10): 3175-3190, 2021 11 29.
Article in English | MEDLINE | ID: mdl-33974044

ABSTRACT

Brain cholesterol is produced mainly by astrocytes and is important for neuronal function. Its biosynthesis is severely reduced in mouse models of Huntington's disease. One possible mechanism is a diminished nuclear translocation of the transcription factor sterol regulatory element-binding protein 2 (SREBP2) and, consequently, reduced activation of SREBP2-controlled genes in the cholesterol biosynthesis pathway. Here we evaluated the efficacy of a gene therapy based on the unilateral intra-striatal injection of a recombinant adeno-associated virus 2/5 (AAV2/5) targeting astrocytes specifically and carrying the transcriptionally active N-terminal fragment of human SREBP2 (hSREBP2). Robust hSREBP2 expression in striatal glial cells in R6/2 Huntington's disease mice activated the transcription of cholesterol biosynthesis pathway genes, restored synaptic transmission, reversed dopamine receptor D2 (Drd2) transcript levels decline, cleared mutant huntingtin aggregates and attenuated behavioural deficits. We conclude that glial SREBP2 participates in Huntington's disease brain pathogenesis in vivo and that AAV-based delivery of SREBP2 to astrocytes counteracts key features of the disease.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Huntington Disease/therapy , Sterol Regulatory Element Binding Protein 2/administration & dosage , Animals , Astrocytes/pathology , Corpus Striatum/pathology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Phenotype , Sterol Regulatory Element Binding Protein 2/biosynthesis , Sterol Regulatory Element Binding Protein 2/genetics
5.
Int J Mol Sci ; 23(18)2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36142751

ABSTRACT

Exercise induces cardioprotection against myocardial infarction, despite obesity, by restoring pro-survival pathways and increasing resistance of mitochondrial permeability transition pore (mPTP) opening at reperfusion. Among the mechanisms involved in the inactivation of these pathways, oxysterols appear interesting. Thus, we investigated the influence of regular exercise on the reperfusion injury salvage kinase (RISK) pathway, oxysterols, and mitochondria, in the absence of ischemia-reperfusion. We also studied 7ß-hydroxycholesterol (7ßOH) concentration (mass spectrometry) in human lean and obese subjects. Wild-type (WT) and obese (ob/ob) mice were assigned to sedentary conditions or regular treadmill exercise. Exercise significantly increased Akt phosphorylation, whereas 7ßOH concentration was reduced. Moreover, exercise induced the translocation of PKCε from the cytosol to mitochondria. However, exercise did not affect the calcium concentration required to open mPTP in the mitochondria, neither in WT nor in ob/ob animals. Finally, human plasma 7ßOH concentration was consistent with observations made in mice. In conclusion, regular exercise enhanced the RISK pathway by increasing kinase phosphorylation and PKCε translocation and decreasing 7ßOH concentration. This activation needs the combination with stress conditions, i.e., ischemia-reperfusion, in order to inhibit mPTP opening at the onset of reperfusion. The human findings suggest 7ßOH as a candidate marker for evaluating cardiovascular risk factors in obesity.


Subject(s)
Myocardial Reperfusion Injury , Oxysterols , Animals , Humans , Mice , Calcium/metabolism , Mice, Obese , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Obesity/metabolism , Oxysterols/metabolism , Protein Kinase C-epsilon/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
6.
J Transl Med ; 17(1): 237, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337415

ABSTRACT

BACKGROUND: Obesity is a chronic and systemic inflammatory disorder and an important risk factor for the onset of several chronic syndromes. Adipose tissue (AT) plays a crucial role in the development of obesity, promoting the infiltration and accumulation of leukocytes in the tissue and sustaining adipocyte expansion. Anthocyanins exert a broad range of health benefits, but their effect in improving obesity-related inflammation in vivo has been poorly characterized. We examined the effects of a purple corn cob extract in the context of AT inflammation in a murine diet-induced obesity (DIO) model. METHODS: Male C57BL/6J mice were subjected to control diet (CTR + H2O), high fat diet (HF + H2O) or high fat diet plus purple corn extract (HF + RED) for 12 weeks. Blood glucose, AT, and liver gene expression, metabolism, biochemistry, and histology were analysed and flow cytometry was performed on AT leukocytes and Kupffer cells. RESULTS: RED extract intake resulted in lower MCP-1 mediated recruitment and proliferation of macrophages into crown-like structures in the AT. AT macrophages (ATM) of HF + RED group upregulated M2 markers (ArgI, Fizz1, TGFß), downregulating inflammatory mediators (TNF-α, IL-6, IL-1ß, COX-2) thanks to the suppression of NF-kB signalling. ATM also increased the expression of iron metabolism-related genes (FABP4, Hmox1, Ferroportin, CD163, TfR1, Ceruloplasmin, FtL1, FtH1) associated with a reduction in iron storage and increased turnover. ATM from HF + RED mice did not respond to LPS treatment ex vivo, confirming the long-lasting effects of the treatment on M2 polarization. Adipocytes of HF + RED group improved lipid metabolism and displayed a lower inflammation grade. Liver histology revealed a remarkable reduction of steatosis in the HF + RED group, and Kupffer cell profiling displayed a marked switch towards the M2 phenotype. CONCLUSIONS: RED extract attenuated AT inflammation in vivo, with a long-lasting reprogramming of ATM and adipocyte profiles towards the anti-inflammatory phenotype, therefore representing a valuable supplement in the context of obesity-associated disorders.


Subject(s)
Adipose Tissue/cytology , Cellular Reprogramming , Macrophages/drug effects , Plant Extracts/pharmacology , Zea mays/chemistry , Adipocytes/cytology , Adipocytes/drug effects , Alanine Transaminase/metabolism , Animals , Anthocyanins/chemistry , Blood Glucose/analysis , Body Weight , Diet, High-Fat , Gene Expression Regulation , Glucose Tolerance Test , Inflammation , Insulin Resistance , Lipopolysaccharides , Liver/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity , Phenotype
7.
Molecules ; 24(18)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514417

ABSTRACT

Neurodegenerative diseases are characterized by oxidative stress, mitochondrial damage, and death of neuronal cells. To counteract such damage and to favor neurogenesis, neurotrophic factors could be used as therapeutic agents. Octadecaneuropeptide (ODN), produced by astrocytes, is a potent neuroprotective agent. In N2a cells, we studied the ability of ODN to promote neuronal differentiation. This parameter was evaluated by phase contrast microscopy, staining with crystal violet, cresyl blue, and Sulforhodamine 101. The effect of ODN on cell viability and mitochondrial activity was determined with fluorescein diacetate and DiOC6(3), respectively. The impact of ODN on the topography of mitochondria and peroxisomes, two tightly connected organelles involved in nerve cell functions and lipid metabolism, was evaluated by transmission electron microscopy and fluorescence microscopy: detection of mitochondria with MitoTracker Red, and peroxisome with an antibody directed against the ABCD3 peroxisomal transporter. The profiles in fatty acids, cholesterol, and cholesterol precursors were determined by gas chromatography, in some cases coupled with mass spectrometry. Treatment of N2a cells with ODN (10-14 M, 48 h) induces neurite outgrowth. ODN-induced neuronal differentiation was associated with modification of topographical distribution of mitochondria and peroxisomes throughout the neurites and did not affect cell viability and mitochondrial activity. The inhibition of ODN-induced N2a differentiation with H89, U73122, chelerythrine and U0126 supports the activation of a PKA/PLC/PKC/MEK/ERK-dependent signaling pathway. Although there is no difference in fatty acid profile between control and ODN-treated cells, the level of cholesterol and some of its precursors (lanosterol, desmosterol, lathosterol) was increased in ODN-treated cells. The ability of ODN to induce neuronal differentiation without cytotoxicity reinforces the interest for this neuropeptide with neurotrophic properties to overcome nerve cell damage in major neurodegenerative diseases.


Subject(s)
Cell Differentiation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diazepam Binding Inhibitor/pharmacology , Lipids/chemistry , Mitochondria/metabolism , Neuropeptides/pharmacology , Peptide Fragments/pharmacology , Peroxisomes/metabolism , Protein Kinase C/metabolism , Type C Phospholipases/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , MAP Kinase Signaling System/drug effects , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Peroxisomes/drug effects , Peroxisomes/ultrastructure , Rhodamines/chemistry , Rhodamines/metabolism , Signal Transduction/drug effects
8.
Neurobiol Dis ; 98: 66-76, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27913290

ABSTRACT

Cholesterol precursors and cholesterol levels are reduced in brain regions of Huntington's disease (HD) mice. Here we quantified the rate of in vivo de novo cholesterol biosynthesis in the HD brain. Samples from different brain regions and blood of the heterozygous knock-in mouse model carrying 175 CAG repeats (Q175) at different phenotypic stages were processed independently by two research units to quantify cholesterol synthesis rate by 2H2O labeling and measure the concentrations of lathosterol, cholesterol and its brain-specific cholesterol catabolite 24-hydroxy-cholesterol (24OHC) by isotope dilution mass spectrometry. The daily synthesis rate of cholesterol and the corresponding concentration of lathosterol were significantly reduced in the striatum of heterozygous Q175 mice early in the disease course. We also report that the decrease in lathosterol was inversely correlated with CAG-size at symptomatic stage, as observed in striatal samples from an allelic series of HD mice. There was also a significant correlation between the fractional synthesis rates of total cholesterol and 24OHC in brain of wild-type (WT) and Q175 mice, supporting the evidence that plasma 24OHC may reflect cholesterol synthesis in the adult brain. This comprehensive analysis demonstrates consistent cholesterol biosynthesis defects in HD mouse models and suggests that plasma 24OHC may serve as a biomarker of brain cholesterol metabolism.


Subject(s)
Brain/metabolism , Cholesterol/biosynthesis , Huntington Disease/metabolism , Animals , Biomarkers/blood , Disease Models, Animal , Disease Progression , Female , Gene Knock-In Techniques , Male , Mice, Inbred C57BL , Mice, Transgenic , Sex Characteristics
9.
Biochim Biophys Acta ; 1851(8): 1095-105, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25596342

ABSTRACT

Huntington disease (HD), an autosomal dominant neurodegenerative disorder caused by an abnormal expansion of CAG trinucleotide repeat in the Huntingtin (HTT) gene, is characterized by extensive neurodegeneration of striatum and cortex and severe diffuse atrophy at MRI. The expression of genes involved in the cholesterol biosynthetic pathway and the amount of cholesterol, lanosterol, lathosterol and 24S-hydroxycholesterol were reduced in murine models of HD. In case of HD-patients, the decrease of plasma 24OHC follows disease progression proportionally to motor and neuropsychiatric dysfunction and MRI brain atrophy, together with lanosterol and lathosterol (markers of cholesterol synthesis), and 27-hydroxycholesterol. A significant reduction of total plasma cholesterol was observed only in advanced stages. It is likely that mutant HTT decreases the maturation of SREBP and the up-regulation LXR and LXR-targeted genes (SREBP, ABCG1 and ABCG4, HMGCoA reductase, ApoE) resulting into a lower synthesis and transport of cholesterol from astrocytes to neurons via ApoE. In primary oligodendrocytes, mutant HTT inhibited the regulatory effect of PGC1α on cholesterol metabolism and on the expression of MBP. HTT seems to play a regulatory role in lipid metabolism. The impairment of the cholesterol metabolism was found to be proportional to the CAG repeat length and to the load of mutant HTT. A dysregulation on PGC1α and mitochondria dysfunction may be involved in an overall reduction of acetyl-CoA and ATP synthesis, contributing to the cerebral and whole body cholesterol impairment. This article is part of a Special Issue entitled Brain Lipids.


Subject(s)
Cerebral Cortex/metabolism , Cholesterol/metabolism , Corpus Striatum/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Animals , Cerebral Cortex/pathology , Corpus Striatum/pathology , Gene Expression Regulation , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Hydroxycholesterols/metabolism , Lanosterol/metabolism , Liver X Receptors , Mice , Mutation , Nerve Tissue Proteins/metabolism , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Trinucleotide Repeats
10.
Biochem Biophys Res Commun ; 446(3): 697-701, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24525128

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an abnormal expansion of a CAG repeat in the huntingtin gene. Neurodegeneration of striatum and cortex with a severe atrophy at MRI are common findings in HD. The expression of genes involved in the cholesterol biosynthetic pathway such as HMG-CoA reductase and the levels of cholesterol, lanosterol, lathosterol and 24S-hydroxycholesterol are reduced in the brain, striatum and cortex in several HD mouse models. Mutant huntingtin affects the maturation and translocation of SREBP and cannot up-regulate LXR. There is a lower synthesis and transport of cholesterol from astrocytes to neurons via ApoE. In primary oligodendrocytes, mutant huntingtin inhibits the regulatory effect of PGC1α on cholesterol metabolism and the expression of Myelin Basic Protein. In humans the decrease of plasma 24S-hydroxycholesterol follows disease progression proportionally to motor and neuropsychiatric dysfunctions and MRI brain atrophy. Huntingtin seems to play a regulatory role in lipid metabolism. Dysregulation of PGC1α and mitochondrial dysfunction may reduce synthesis of Acetyl-CoA and ATP contributing to the cerebral and whole body impairment of cholesterol metabolism.


Subject(s)
Brain/metabolism , Cholesterol/metabolism , Huntington Disease/metabolism , Hydroxycholesterols/blood , Animals , Disease Models, Animal , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
11.
Mol Genet Metab ; 105(3): 463-71, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22221393

ABSTRACT

Pantothenate kinase-associated neurodegeneration (PKAN) is a rare, inborn error of metabolism characterized by iron accumulation in the basal ganglia and by the presence of dystonia, dysarthria, and retinal degeneration. Mutations in pantothenate kinase 2 (PANK2), the rate-limiting enzyme in mitochondrial coenzyme A biosynthesis, represent the most common genetic cause of this disorder. How mutations in this core metabolic enzyme give rise to such a broad clinical spectrum of pathology remains a mystery. To systematically explore its pathogenesis, we performed global metabolic profiling on plasma from a cohort of 14 genetically defined patients and 18 controls. Notably, lactate is elevated in PKAN patients, suggesting dysfunctional mitochondrial metabolism. As predicted, but never previously reported, pantothenate levels are higher in patients with premature stop mutations in PANK2. Global metabolic profiling and follow-up studies in patient-derived fibroblasts also reveal defects in bile acid conjugation and lipid metabolism, pathways that require coenzyme A. These findings raise a novel therapeutic hypothesis, namely, that dietary fats and bile acid supplements may hold potential as disease-modifying interventions. Our study illustrates the value of metabolic profiling as a tool for systematically exploring the biochemical basis of inherited metabolic diseases.


Subject(s)
Coenzyme A/deficiency , Mitochondria/enzymology , Neuroaxonal Dystrophies/metabolism , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adolescent , Adult , Bile Acids and Salts/metabolism , Child , Child, Preschool , Codon, Nonsense , Coenzyme A/biosynthesis , Coenzyme A/genetics , Cohort Studies , Female , Humans , Iron Metabolism Disorders , Lactic Acid/blood , Lipid Metabolism/genetics , Lipid Metabolism Disorders/genetics , Lipid Metabolism Disorders/metabolism , Male , Metabolome , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neuroaxonal Dystrophies/diagnosis , Neuroaxonal Dystrophies/enzymology , Pantothenate Kinase-Associated Neurodegeneration/enzymology , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenic Acid/blood , Sphingomyelins/blood , Young Adult
12.
Steroids ; 183: 109032, 2022 07.
Article in English | MEDLINE | ID: mdl-35381271

ABSTRACT

Peroxisomes play an important role in regulating cell metabolism and RedOx homeostasis. Peroxisomal dysfunctions favor oxidative stress and cell death. The ability of 7ß-hydroxycholesterol (7ß-OHC; 50 µM, 24 h), known to be increased in patients with age-related diseases such as sarcopenia, to trigger oxidative stress, mitochondrial and peroxisomal dysfunction was studied in murine C2C12 myoblasts. The capacity of milk thistle seed oil (MTSO, 100 µg/mL) as well as α-tocopherol (400 µM; reference cytoprotective agent) to counteract the toxic effects of 7ß-OHC, mainly at the peroxisomal level were evaluated. The impacts of 7ß-OHC, in the presence or absence of MTSO or α-tocopherol, were studied with complementary methods: measurement of cell density and viability, quantification of reactive oxygen species (ROS) production and transmembrane mitochondrial potential (ΔΨm), evaluation of peroxisomal mass as well as topographic, morphologic and functional peroxisomal changes. Our results indicate that 7ß-OHC induces a loss of cell viability and a decrease of cell adhesion associated with ROS overproduction, alterations of mitochondrial ultrastructure, a drop of ΔΨm, and several peroxisomal modifications. In the presence of 7ß-OHC, comparatively to untreated cells, important quantitative and qualitative peroxisomal modifications were also identified: a) a reduced number of peroxisomes with abnormal sizes and shapes, mainly localized in cytoplasmic vacuoles, were observed; b) the peroxisomal mass was decreased as indicated by lower protein and mRNA levels of the peroxisomal ABCD3 transporter; c) lower mRNA level of Pex5 involved in peroxisomal biogenesis as well as higher mRNA levels of Pex13 and Pex14, involved in peroxisomal biogenesis and/or pexophagy, was found; d) lower levels of ACOX1 and MFP2 enzymes, implicated in peroxisomal ß-oxidation, were detected; e) higher levels of very-long-chain fatty acids, which are substrates of peroxisomal ß-oxidation, were found. These different cytotoxic effects were strongly attenuated by MTSO, in the same range of order as with α-tocopherol. These findings underline the interest of MTSO and α-tocopherol in the prevention of peroxisomal damages (pexotherapy).


Subject(s)
Silybum marianum , alpha-Tocopherol , Animals , Antioxidants/pharmacology , Flavonoids , Humans , Hydroxycholesterols , Mice , Silybum marianum/metabolism , Myoblasts/metabolism , Plant Oils , RNA, Messenger , Reactive Oxygen Species/metabolism , alpha-Tocopherol/pharmacology
13.
Article in English | MEDLINE | ID: mdl-34886549

ABSTRACT

The present work proposes several modifications to optimize both emissions and consumption in a commercial marine diesel engine. A numerical model was carried out to characterize the emissions and consumption of the engine under several performance parameters. Particularly, five internal modifications were analyzed: water addition; exhaust gas recirculation; and modification of the intake valve closing, overlap timing, and cooling water temperature. It was found that the result on the emissions and consumption presents conflicting criteria, and thus, a multiple-criteria decision-making model was carried out to characterize the most appropriate parameters. In order to analyze a high number of possibilities in a reasonable time, an artificial neural network was developed.


Subject(s)
Gasoline , Vehicle Emissions , Environmental Pollution , Neural Networks, Computer , Temperature
14.
J Steroid Biochem Mol Biol ; 210: 105854, 2021 06.
Article in English | MEDLINE | ID: mdl-33631373

ABSTRACT

Hepatitis B virus (HBV) infection is a global health problem with different immunological phases and therapeutic approaches. The serological condition of inactive carrier (IC) was recently well defined as a clinical and virological stable status, in which specific treatment is usually deferred, while the active chronic hepatitis B (CHB) condition requires an immediate treatment strategy. Recently, a possible broad antiviral effect of oxysterols, in particular 25-hydroxycholesterol (25OHC) and 27-hydroxycholesterol (27OHC), was observed, as most likely linked to the positive modulation of innate immunity, but no clear evidence is available about their possible role in chronic HBV infection. Thus, we examined the relationship between the plasma levels of oxysterols and the disease condition of 40 HBV patients, without treatment at the start of the study. Of these, 33 were ICs and 7 were active CHB subjects. A marked reduction of 25OHC and 27OHC plasma levels was detectable in all active CHB recruited patients, while the plasma values observed in ICs all remained within the physiological range. No difference was observed between the two groups of patients with regard to the plasma levels of 24-hydroxycholesterol (24OHC). Further, the plasma level of 27OHC ≥ 140 µg/L was shown to be predictive of an inactive carrier status. This cohort study points to 27OHC as a good candidate biomarker to differentiate active and inactive CHB status. An increasing bulk of research reports is supporting the very likely contribution of this oxysterol to the immunological control of chronic hepatitis B.


Subject(s)
Carrier State/blood , Hepatitis B, Chronic/blood , Hepatitis B, Chronic/virology , Hydroxycholesterols/blood , Adult , Biomarkers/blood , Carrier State/virology , Elasticity Imaging Techniques , Female , Genotype , Hepatitis B Antigens/blood , Hepatitis B virus/genetics , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/physiopathology , Humans , Liver/physiopathology , Liver/virology , Male , Prospective Studies
15.
Redox Biol ; 39: 101837, 2021 02.
Article in English | MEDLINE | ID: mdl-33360775

ABSTRACT

Among Alzheimer's disease (AD) brain hallmarks, the presence of reactive astrocytes was demonstrated to correlate with neuronal loss and cognitive deficits. Evidence indeed supports the role of reactive astrocytes as mediators of changes in neurons, including synapses. However, the complexity and the outcomes of astrocyte reactivity are far from being completely elucidated. Another key role in AD pathogenesis is played by alterations in brain cholesterol metabolism. Oxysterols (cholesterol oxidation products) are crucial for brain cholesterol homeostasis, and we previously demonstrated that changes in the brain levels of various oxysterols correlate with AD progression. Moreover, oxysterols have been shown to contribute to various pathological mechanisms involved in AD pathogenesis. In order to deepen the role of oxysterols in AD, we investigated whether they could contribute to astrocyte reactivity, and consequently impact on neuronal health. Results showed that oxysterols present in mild or severe AD brains induce a clear morphological change in mouse primary astrocytes, accompanied by the upregulation of some reactive astrocyte markers, including lipocalin-2 (Lcn2). Moreover, astrocyte conditioned media analysis revealed a significant increase in the release of Lcn2, cytokines, and chemokines in response to oxysterols. A significant reduction of postsynaptic density protein 95 (PSD95) and a concurrent increase in cleaved caspase-3 protein levels have been demonstrated in neurons co-cultured with oxysterol-treated astrocytes, pointing out that mediators released by astrocytes have an impact on neurons. Among these mediators, Lcn2 has been demonstrated to play a major role on synapses, affecting neurite morphology and decreasing dendritic spine density. These data demonstrated that oxysterols present in the AD brain promote astrocyte reactivity, determining the release of several mediators that affect neuronal health and synapses. Lcn2 has been shown to exert a key role in mediating the synaptotoxic effect of oxysterol-treated astrocytes.


Subject(s)
Alzheimer Disease , Oxysterols , Animals , Astrocytes/metabolism , Brain/metabolism , Lipocalin-2/metabolism , Mice
16.
J Control Release ; 330: 587-598, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33412229

ABSTRACT

Supplementing brain cholesterol is emerging as a potential treatment for Huntington's disease (HD), a genetic neurodegenerative disorder characterized, among other abnormalities, by inefficient brain cholesterol biosynthesis. However, delivering cholesterol to the brain is challenging due to the blood-brain barrier (BBB), which prevents it from reaching the striatum, especially, with therapeutically relevant doses. Here we describe the distribution, kinetics, release, and safety of novel hybrid polymeric nanoparticles made of PLGA and cholesterol which were modified with an heptapeptide (g7) for BBB transit (hybrid-g7-NPs-chol). We show that these NPs rapidly reach the brain and target neural cells. Moreover, deuterium-labeled cholesterol from hybrid-g7-NPs-chol is released in a controlled manner within the brain and accumulates over time, while being rapidly removed from peripheral tissues and plasma. We confirm that systemic and repeated injections of the new hybrid-g7-NPs-chol enhanced endogenous cholesterol biosynthesis, prevented cognitive decline, and ameliorated motor defects in HD animals, without any inflammatory reaction. In summary, this study provides insights about the benefits and safety of cholesterol delivery through advanced brain-permeable nanoparticles for HD treatment.


Subject(s)
Huntington Disease , Nanoparticles , Animals , Brain , Cholesterol , Huntington Disease/drug therapy , Kinetics
17.
Cells ; 9(4)2020 04 04.
Article in English | MEDLINE | ID: mdl-32260362

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare cancer with poor prognosis. Mitotane, the standard treatment for ACC, impairs adrenocortical steroid biosynthesis and cholesterol metabolism. In the H295R cell line, a standard ACC in vitro model, mitotane was previously reported to enhance the production of some oxysterols. To verify the possible mechanistic involvement of oxysterols in the anti-ACC effect of mitotane, a gas chromatography mass spectrometry (GC-MS) profiling of oxysterols and the main cholesterol precursors was carried out in H295R cells. Among the oxysterols detected in mitotane-treated cells, 27OHC was markedly produced, as well as lanosterol and lathosterol cholesterol precursors. In this cell model, mitotane was confirmed to affect mitochondrial transmembrane potential and induce apoptosis. Such cytotoxic effects were perfectly matched by H295R cell treatment with a single identical micromolar amount of 27OHC. The mitotane-dependent strong increase in 27OHC was confirmed in vivo, in the plasma of ACC patients under treatment with the drug. Moreover, lanosterol, lathosterol, desmosterol and, to a minor extent, 24-hydroxycholesterol and 25-hydroxycholesterol plasma levels were significantly increased in those patients. The cytotoxic effect of mitotane on ACC cells may be partly related to the increased intracellular level of 27OHC induced by the drug itself.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Hydroxycholesterols/metabolism , Mitotane/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mitotane/pharmacology , Oxidation-Reduction , Oxysterols/metabolism
18.
Redox Biol ; 36: 101682, 2020 09.
Article in English | MEDLINE | ID: mdl-32810737

ABSTRACT

There is an urgent need to identify antivirals against the coronavirus SARS-CoV-2 in the current COVID-19 pandemic and to contain future similar emergencies early on. Specific side-chain cholesterol oxidation products of the oxysterols family have been shown to inhibit a large variety of both enveloped and non-enveloped human viral pathogens. Here we report on the in vitro inhibitory activity of the redox active oxysterol 27-hydroxycholesterol against SARS-CoV-2 and against one of the common cold agents HCoV-OC43 human coronavirus without significant cytotoxicity. Interestingly, physiological serum levels of 27-hydroxycholesterol in SARS-CoV-2 positive subjects were significantly decreased compared to the matched control group, reaching a marked 50% reduction in severe COVID-19 cases. Moreover, no correlation at all was observed between 24-hydroxycholesterol and 25-hydroxycholesterol serum levels and the severity of the disease. Opposite to that of 27-hydroxycholesterol was the behaviour of two recognized markers of redox imbalance, i.e. 7-ketocholesterol and 7ß-hydroxycholesterol, whose serum levels were significantly increased especially in severe COVID-19. The exogenous administration of 27-hydroxycholesterol may represent in the near future a valid antiviral strategy in the worsening of diseases caused by present and emerging coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/blood , Hydroxycholesterols/blood , Pneumonia, Viral/blood , Aged , Animals , Biomarkers/blood , COVID-19 , Chlorocebus aethiops , Coronavirus Infections/pathology , Female , Hep G2 Cells , Humans , Hydroxycholesterols/pharmacology , Male , Middle Aged , Pandemics , Pneumonia, Viral/pathology , SARS-CoV-2 , Vero Cells
19.
EMBO Mol Med ; 12(10): e12519, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32959531

ABSTRACT

A variety of pathophysiological mechanisms are implicated in Huntington's disease (HD). Among them, reduced cholesterol biosynthesis has been detected in the HD mouse brain from pre-symptomatic stages, leading to diminished cholesterol synthesis, particularly in the striatum. In addition, systemic injection of cholesterol-loaded brain-permeable nanoparticles ameliorates synaptic and cognitive function in a transgenic mouse model of HD. To identify an appropriate treatment regimen and gain mechanistic insights into the beneficial activity of exogenous cholesterol in the HD brain, we employed osmotic mini-pumps to infuse three escalating doses of cholesterol directly into the striatum of HD mice in a continuous and rate-controlled manner. All tested doses prevented cognitive decline, while amelioration of disease-related motor defects was dose-dependent. In parallel, we found morphological and functional recovery of synaptic transmission involving both excitatory and inhibitory synapses of striatal medium spiny neurons. The treatment also enhanced endogenous cholesterol biosynthesis and clearance of mutant Huntingtin aggregates. These results indicate that cholesterol infusion to the striatum can exert a dose-dependent, disease-modifying effect and may be therapeutically relevant in HD.


Subject(s)
Huntington Disease , Animals , Cholesterol , Corpus Striatum , Disease Models, Animal , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Mice , Mice, Transgenic , Synapses
20.
J Clin Med ; 9(3)2020 Mar 03.
Article in English | MEDLINE | ID: mdl-32138288

ABSTRACT

Niemann-Pick type C (NPC) disease is an autosomal recessive lysosomal storage disorder caused by mutations in NPC1 or NPC2 genes. In 2009, the molecular characterization of 44 NPC Italian patients has been published. Here, we present an update of the genetic findings in 105 Italian NPC patients belonging to 83 unrelated families (77 NPC1 and 6 NPC2). NPC1 and NPC2 genes were studied following an algorithm recently published. Eighty-four different NPC1 and five NPC2 alleles were identified. Only two NPC1 alleles remained non detected. Sixty-two percent of NPC1 alleles were due to missense variants. The most frequent NPC1 mutation was the p.F284Lfs*26 (5.8% of the alleles). All NPC2 mutations were found in the homozygous state, and all but one was severe. Among newly diagnosed patients, 18 novel NPC1 mutations were identified. The pathogenic nature of 7/9 missense alleles and 3/4 intronic variants was confirmed by filipin staining and NPC1 protein analysis or mRNA expression in patient's fibroblasts. Taken together, our previous published data and new results provide an overall picture of the molecular characteristics of NPC patients diagnosed so far in Italy.

SELECTION OF CITATIONS
SEARCH DETAIL