Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732080

ABSTRACT

Endothelial progenitor cells (EPCs) play a critical role in cardiovascular regeneration. Enhancement of their native properties would be highly beneficial to ensuring the proper functioning of the cardiovascular system. As androgens have a positive effect on the cardiovascular system, we hypothesized that dihydrotestosterone (DHT) could also influence EPC-mediated repair processes. To evaluate this hypothesis, we investigated the effects of DHT on cultured human EPCs' proliferation, viability, morphology, migration, angiogenesis, gene and protein expression, and ability to integrate into cardiac tissue. The results showed that DHT at different concentrations had no cytotoxic effect on EPCs, significantly enhanced the cell proliferation and viability and induces fast, androgen-receptor-dependent formation of capillary-like structures. DHT treatment of EPCs regulated gene expression of androgen receptors and the genes and proteins involved in cell migration and angiogenesis. Importantly, DHT stimulation promoted EPC migration and the cells' ability to adhere and integrate into murine cardiac slices, suggesting it has a role in promoting tissue regeneration. Mass spectrometry analysis further highlighted the impact of DHT on EPCs' functioning. In conclusion, DHT increases the proliferation, migration, and androgen-receptor-dependent angiogenesis of EPCs; enhances the cells' secretion of key factors involved in angiogenesis; and significantly potentiates cellular integration into heart tissue. The data offer support for potential therapeutic applications of DHT in cardiovascular regeneration and repair processes.


Subject(s)
Cell Movement , Cell Proliferation , Dihydrotestosterone , Endothelial Progenitor Cells , Neovascularization, Physiologic , Receptors, Androgen , Dihydrotestosterone/pharmacology , Humans , Cell Movement/drug effects , Receptors, Androgen/metabolism , Neovascularization, Physiologic/drug effects , Cell Proliferation/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/cytology , Animals , Cells, Cultured , Mice , Cell Survival/drug effects , Androgens/pharmacology , Androgens/metabolism , Male
2.
Int J Mol Sci ; 23(19)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36232476

ABSTRACT

Increased levels of low-density lipoproteins are the main risk factor in the initiation and progression of atherosclerosis. Although statin treatment can effectively lower these levels, there is still a residual risk of cardiovascular events. We hypothesize that a specific panel of stress-sensing molecules (alarmins) could indicate the persistence of silent atherosclerosis residual risk. New Zealand White rabbits were divided into: control group (C), a group that received a high-fat diet for twelve weeks (Au), and a treated hyperlipidemic group with a lipid diet for eight weeks followed by a standard diet and hypolipidemic treatment (atorvastatin and PCSK9 siRNA-inhibitor) for four weeks (Asi). Mass spectrometry experiments of left ventricle lysates were complemented by immunologic and genomic studies to corroborate the data. The hyperlipidemic diet determined a general alarmin up-regulation tendency over the C group. A significant spectral abundance increase was measured for specific heat shock proteins, S100 family members, HMGB1, and Annexin A1. The hypolipidemic treatment demonstrated a reversed regulation trend with non-significant spectral alteration over the C group for some of the identified alarmins. Our study highlights the discriminating potential of alarmins in hyperlipidemia or following hypolipidemic treatment. Data are available via ProteomeXchange with identifier PXD035692.


Subject(s)
Annexin A1 , Atherosclerosis , HMGB1 Protein , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Alarmins , Animals , Atherosclerosis/metabolism , Atorvastatin , HMGB1 Protein/metabolism , Heat-Shock Proteins/metabolism , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/therapeutic use , Lipoproteins, LDL/metabolism , Proprotein Convertase 9/metabolism , RNA, Small Interfering , Rabbits
3.
Int J Mol Sci ; 23(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35563680

ABSTRACT

Prognosis after myocardial infarction (MI) varies greatly depending on the extent of damaged area and the management of biological processes during recovery. Reportedly, the inhibition of the pro-inflammatory S100A9 reduces myocardial damage after MI. We hypothesize that a S100A9 blockade induces changes of major signaling pathways implicated in post-MI healing. Mass spectrometry-based proteomics and gene analyses of infarcted mice left ventricle were performed. The S100A9 blocker (ABR-23890) was given for 3 days after coronary ligation. At 3 and 7 days post-MI, ventricle samples were analyzed versus control and Sham-operated mice. Blockade of S100A9 modulated the expressed proteins involved in five biological processes: leukocyte cell-cell adhesion, regulation of the muscle cell apoptotic process, regulation of the intrinsic apoptotic signaling pathway, sarcomere organization and cardiac muscle hypertrophy. The blocker induced regulation of 36 proteins interacting with or targeted by the cellular tumor antigen p53, prevented myocardial compensatory hypertrophy, and reduced cardiac markers of post-ischemic stress. The blockade effect was prominent at day 7 post-MI when the quantitative features of the ventricle proteome were closer to controls. Blockade of S100A9 restores key biological processes altered post-MI. These processes could be valuable new pharmacological targets for the treatment of ischemic heart. Mass spectrometry data are available via ProteomeXchange with identifier PXD033683.


Subject(s)
Myocardial Infarction , Proteome , Alarmins/metabolism , Animals , Calgranulin B/genetics , Calgranulin B/metabolism , Heart Ventricles/metabolism , Hypertrophy/metabolism , Mice , Myocardial Infarction/metabolism , Myocardium/metabolism , Proteome/metabolism , Signal Transduction , Ventricular Remodeling
4.
Molecules ; 27(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36080270

ABSTRACT

Exosomes are small extracellular vesicles with a variable protein cargo in consonance with cell origin and pathophysiological conditions. Gestational diabetes mellitus (GDM) is characterized by different levels of chronic low-grade inflammation and vascular dysfunction; however, there are few data characterizing the serum exosomal protein cargo of GDM patients and associated signaling pathways. Eighteen pregnant women were enrolled in the study: 8 controls (CG) and 10 patients with GDM. Blood samples were collected from patients, for exosomes' concentration. Protein abundance alterations were demonstrated by relative mass spectrometric analysis and their association with clinical parameters in GDM patients was performed using Pearson's correlation analysis. The proteomics analysis revealed 78 significantly altered proteins when comparing GDM to CG, related to complement and coagulation cascades, platelet activation, prothrombotic factors and cholesterol metabolism. Down-regulation of Complement C3 (C3), Complement C5 (C5), C4-B (C4B), C4b-binding protein beta chain (C4BPB) and C4b-binding protein alpha chain (C4BPA), and up-regulation of C7, C9 and F12 were found in GDM. Our data indicated significant correlations between factors involved in the pathogenesis of GDM and clinical parameters that may improve the understanding of GDM pathophysiology. Data are available via ProteomeXchange with identifier PXD035673.


Subject(s)
Diabetes, Gestational , Exosomes , Blood Proteins/metabolism , Complement C4b-Binding Protein/metabolism , Complement System Proteins/metabolism , Exosomes/metabolism , Female , Humans , Lipid Metabolism , Pregnancy , Proteomics/methods
5.
J Cell Mol Med ; 24(20): 12131-12140, 2020 10.
Article in English | MEDLINE | ID: mdl-32935914

ABSTRACT

Nephropathy is a major chronic complication of diabetes. A crucial role in renal pathophysiology is played by hydrogen sulphide (H2 S) that is produced excessively by the kidney; however, the data regarding H2 S bioavailability are inconsistent. We hypothesize that early type 1 diabetes (T1D) increases H2 S production by a mechanism involving hyperglycaemia-induced alterations in sulphur metabolism. Plasma and kidney tissue collected from T1D double transgenic mice were subjected to mass spectrometry-based proteomic analysis, and the results were validated by immunological and gene expression assays.T1D mice exhibited a high concentration of H2 S in the plasma and kidney tissue and histological, showed signs of subtle kidney fibrosis, characteristic for early renal disease. The shotgun proteomic analyses disclosed that the level of enzymes implicated in sulphate activation modulators, H2 S-oxidation and H2 S-production were significantly affected (ie 6 up-regulated and 4 down-regulated). Gene expression results corroborated well with the proteomic data. Dysregulation of H2 S enzymes underly the changes occurring in H2 S production, which in turn could play a key role in the initiation of renal disease. The new findings lead to a novel target in the therapy of diabetic nephropathy. Mass spectrometry data are available via ProteomeXchange with identifier PXD018053.


Subject(s)
Diabetic Nephropathies/enzymology , Kidney/metabolism , Sulfur/metabolism , Animals , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/pathology , Diabetic Nephropathies/pathology , Disease Models, Animal , Gene Expression Regulation , Hydrogen Sulfide/metabolism , Metabolic Networks and Pathways , Mice, Inbred BALB C , Mice, Transgenic , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
6.
Expert Rev Proteomics ; 17(4): 257-273, 2020 04.
Article in English | MEDLINE | ID: mdl-32427033

ABSTRACT

INTRODUCTION: The importance of biomarkers for pharmaceutical drug development and clinical diagnostics is more significant than ever in the current shift toward personalized medicine. Biomarkers have taken a central position either as companion markers to support drug development and patient selection, or as indicators aiming to detect the earliest perturbations indicative of disease, minimizing therapeutic intervention or even enabling disease reversal. Protein biomarkers are of particular interest given their central role in biochemical pathways. Hence, capabilities to analyze multiple protein biomarkers in one assay are highly interesting for biomedical research. AREAS COVERED: We here review multiple methods that are suitable for robust, high throughput, standardized, and affordable analysis of protein biomarkers in a multiplex format. We describe innovative developments in immunoassays, the vanguard of methods in clinical laboratories, and mass spectrometry, increasingly implemented for protein biomarker analysis. Moreover, emerging techniques are discussed with potentially improved protein capture, separation, and detection that will further boost multiplex analyses. EXPERT COMMENTARY: The development of clinically applied multiplex protein biomarker assays is essential as multi-protein signatures provide more comprehensive information about biological systems than single biomarkers, leading to improved insights in mechanisms of disease, diagnostics, and the effect of personalized medicine.


Subject(s)
Biomarkers/chemistry , Proteomics/methods , Animals , Biomarkers/analysis , Humans , Immunoassay/methods , Mass Spectrometry/methods
7.
J Mol Cell Cardiol ; 133: 115-124, 2019 08.
Article in English | MEDLINE | ID: mdl-31201797

ABSTRACT

Scarcity of gender specific donor hearts highlights the importance of mesenchymal stem cells (MSCs) as a therapeutic tool for heart repair. However, inefficient incorporation, retention, and activity of MSCs in cardiac tissue remain an obstacle. Since surges in follicular estradiol (E2; µmolar-range) trigger tissue remodeling (e.g. ovulation) and E2 exerts beneficial actions on the cardiovascular system, we hypothesized that E2 may promote/improve MSC-mediated cardiac repair processes. Using Wharton's jelly (WJ)-derived MSCs we assessed the effects of E2 on MSC proliferation, directed migration, and engraftment in murine heart slices (using xCELLigence real-time cell-impedance system, DNA quantification, and microscopy) and on MSC-induced angiogenesis in vivo (matrigel plug assay). Protein expression was assessed by Western blotting, ELISA/Luminex, and proteomic analysis; whereas mRNA expression was assessed by qRT-PCR. MSCs expressed estrogen receptors (ERs) -alpha and -beta. E2 promoted MSC proliferation and up-regulated mRNA and protein expression of ER-alpha, ER-beta, extracellular matrix metalloproteinase inducer (EMMPRIN), and matrix metalloproteinase (MMP) -9, yet down-regulated MMP-2 expression. Moreover, E2 up-regulated expression of vascular endothelial growth factor (VEGF)-A, VEGFR-2, vascular cell adhesion protein-1 (VCAM-1), and angiogenin (ANG) and stimulated nitric oxide (NO) production via ER. Proteomic analysis of MSCs showed that E2 up-regulated 47 proteins, down-regulated 7 proteins, and increased the expression of key biochemical components/pathways involved in tissue repair. In MSCs co-cultured with murine heart-slices, E2 significantly induced MSC migration in an ER-alpha-dependent fashion and significantly increased the secretion of MMP-2, MMP-9, ANG, and VEGF. In an in vivo matrigel assay, E2-treated MSCs increased angiogenesis and hemoglobin content. In conclusion, E2-treatment increases the incorporation of MSCs in heart slices and promotes MSC-induced angiogenesis. These beneficial effects are mediated via increases in molecules/pathways involved in tissue remodeling and angiogenesis. We speculate that E2 may enhance MSC ability to repair/regenerate cardiac tissue.


Subject(s)
Cell Differentiation/drug effects , Estradiol/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Myocardium/cytology , Myocardium/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Female , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/genetics , Proteomics/methods
8.
J Proteome Res ; 15(9): 3377-87, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27480176

ABSTRACT

The study aimed to evaluate the proteomic changes in benign follicular adenoma versus malignant follicular variant of papillary thyroid carcinoma. Tumor and nontumor adjacent samples were analyzed by liquid nanochromatography mass spectrometry, and protein abundance was evaluated by label-free quantification. Western blotting and quantitative real-time polymerase chain reaction were used to validate and complement the mass spectrometry data. The results demonstrated deregulated expression of four endoplasmic reticulum chaperones (78 kDa glucose-regulated protein, endoplasmin, calnexin, protein disulfide-isomerase A4), glutathione peroxidase 3 and thyroglobulin, all of them involved in thyroid hormone synthesis pathway. The altered tissue abundance of endoplasmic reticulum chaperones in thyroid cancer was correlated with serum expression levels. The identified proteins significantly discriminate between adenoma and carcinoma in both thyroid tissue and corresponding sera. Data are available via ProteomeXchange with identifier PXD004322.


Subject(s)
Carcinogenesis/chemistry , Endoplasmic Reticulum/chemistry , Molecular Chaperones/analysis , Proteomics/methods , Thyroid Neoplasms/chemistry , Adenoma/chemistry , Adenoma/diagnosis , Biosynthetic Pathways , Carcinoma/chemistry , Carcinoma/diagnosis , Chromatography, Liquid , Humans , Mass Spectrometry , Molecular Chaperones/blood , Real-Time Polymerase Chain Reaction , Thyroid Hormones/biosynthesis , Thyroid Neoplasms/diagnosis
9.
Arch Biochem Biophys ; 583: 55-64, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26254814

ABSTRACT

Diabetes mellitus generates metabolic changes associated with inflammatory events that may eventually affect all body tissues. Both high-mobility group box 1 (HMGB1) and ß-catenin are active players in inflammation. The study aimed to determine whether HMGB1 modulates the ß-catenin activity in supporting inflammation, using an experimental type 1 diabetes mouse model. The protein and gene expression of HMGB1 were significantly increased (2-fold) in the diabetic lung compared to control and were positively correlated with the HMGB1 levels detected in serum. Co-immunoprecipitation of HMGB1 with RAGE co-exists with activation of PI3K/AKT1 and NF-kB signaling pathways. At the same time ß-catenin was increased in nuclear fraction (3.5 fold) while it was down-regulated in diabetic plasma membrane (2-fold). There was no difference of ß-catenin gene expression between the control and diabetic mice. ß-Catenin phosphorylation at Ser552 was higher in diabetic nuclear fraction, suggesting that AKT1 activation promotes ß-catenin nuclear translocation. In addition, c-Jun directly binds ß-catenin indicating the transcriptional activity of ß-catenin in diabetes, sustained by significantly COX2 increase by 6-fold in the cytosolic extract of diabetic lung compared to control. Taken together, the data support the new concept that HMGB1 maintains the inflammation through RAGE/AKT1/ß-catenin pathway in the diabetic lung.


Subject(s)
Diabetes Mellitus, Experimental/complications , HMGB1 Protein/physiology , Pneumonia/physiopathology , Animals , Cell Nucleus/metabolism , Mass Spectrometry , Mice , Mice, Transgenic , Phosphorylation , Pneumonia/complications , Pneumonia/metabolism , Proteome , Proto-Oncogene Proteins c-akt/metabolism , Receptor for Advanced Glycation End Products/metabolism , beta Catenin/metabolism
10.
Proteome Sci ; 13: 30, 2015.
Article in English | MEDLINE | ID: mdl-26628893

ABSTRACT

BACKGROUND: Membrane microdomains represent dynamic membrane nano-assemblies enriched in signaling molecules suggesting their active involvement in not only physiological but also pathological molecular processes. The hyperlipidemic stress is a major risk factor of atherosclerosis, but its exact mechanisms of action at the membrane microdomains level remain elusive. The aim of the present study was to determine whether membrane-cytoskeleton proteome in the pulmonary tissue could be modulated by the hyperlipidemic stress, a major risk factor of atherosclerosis. RESULTS: High resolution mass spectrometry based proteomics analysis was performed for detergent resistant membrane microdomains isolated from lung homogenates of control, ApoE deficient and statin treated ApoE deficient mice. The findings of the study allowed the identification with high confidence of 1925 proteins, 291 of which were found significantly altered by the modified genetic background, by the statin treatment or both conditions. Principal component analysis revealed a proximal partitioning of the biological replicates, but also a distinct spatial scattering of the sample groups, highlighting different quantitative profiles. The statistical significant over-representation of Regulation of actin cytoskeleton, Focal adhesion and Adherens junction Kyoto Encyclopedia of Genes and Genomes signaling pathways was demonstrated through bioinformatics analysis. The three inter-relation maps comprised 29 of regulated proteins, proving membrane-cytoskeleton coupling targeting and alteration by hyperlipidemia and/or statin treatment. CONCLUSIONS: The findings of the study allowed the identification with high confidence of the main proteins modulated by the hyperlipidemic stress involved in the actin-dependent pathways. Our study provides the basis for future work probing how the protein activities at the membrane-cytoskeleton interface are dependent upon genetic induced hyperlipidemia.

11.
Roum Arch Microbiol Immunol ; 73(3-4): 74-83, 2014.
Article in English | MEDLINE | ID: mdl-26201122

ABSTRACT

Infective endocarditis (IE) is an infection of the heart endothelium and valves and is frequently a consequence of a sanguine flow turbulence and injury of endocardium. Recent studies revealed an increase of Staphylococcus aureus strains involved in IE, but no evident correlations between the genetic background of this bacterium and IE involvement of certain strains have been found yet. In this study we analyzed the virulence profile, including adhesins, exotoxins, superantigens and biofilm determinants, along with agr type detection, for S. aureus strains isolated from IE, versus non-IE originating strains. We performed also bacterial typing (SCCmec typing, spa-typing and MLST typing), in order to compare our strains with international databases repositories. Although the study was carried out on a reduced number of isolates, our observations confirm the previous works, showing that no major differences were observed between the genetic backgrounds of the two groups of strains analyzed. Notably, the added value of this study was optimization of two new multiplex PCR protocols, and the enrichment of international databases with three new spa-types, three new MLST alleles and four new MLST sequence types.


Subject(s)
Endocarditis/microbiology , Staphylococcus aureus/isolation & purification , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Multilocus Sequence Typing , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence Factors/analysis
12.
Cell Tissue Res ; 354(3): 771-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23996201

ABSTRACT

A high-lipid diet is one of the main risk factors in atherosclerosis and can induce changes in the composition of plasma membrane microdomains. In response, important functions such as vesicle trafficking, protein docking, signaling and receptor recognition are significantly altered. In particular, interactions of heat-shock proteins (Hsps), acting as danger signals, with components of the membrane microdomains can influence signaling pathways and the inflammatory response of cells. Our study focuses on the composition of detergent-resistant membrane (DRM) isolated from ApoE-/- mice fed a standard or high-fat diet with and without fluvastatin treatment versus appropriate controls. Biochemical studies, immunoblotting and liquid chromatography mass spectrometric analysis were performed to investigate whether the structural components (such as caveolin and cavin) of the detergent-resistant microdomains were correlated with the expression and secretion of stress-inducible Hsps (Hsp70 and Hsp90) and AKT phosphorylation in experimental atherosclerosis. ApoE-/- mice challenged with a high-fat diet developed extensive atherosclerotic plaques in lesion-prone areas. DRM harvested from hyperlipidemic animals showed a modified biochemical composition with cholesterol, glycerolipids, caveolin-1 and phospho-AKT being up-regulated, whereas cavin-1 and dynamin were down-regulated. The data also demonstrated the co-fractionation of Hsps with caveolin-1 in isolated DRM, expression being positively correlated with their secretion into blood serum. Statin therapy significantly attenuated the processes induced by the development of atherosclerosis in ApoE-/- mice under a high-fat diet. Thus, high-lipid stress induces profound changes in DRM biochemistry and modifies the cellular response, supporting the systemic inflammatory onset of atherosclerosis.


Subject(s)
Caveolin 1/metabolism , Detergents/pharmacology , Dietary Fats/pharmacology , Heat-Shock Proteins/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Biologicals ; 41(3): 148-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23582120

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen causing various inflammatory diseases from skin and tissue local infections, to serious life threatening infections including endocarditis. Experimental models for endocarditis demonstrated that virulence factors of S. aureus, that are very important in infection of heart vegetations, are surface proteins which promote bacterial adherence. Until now, efforts to develop effective vaccines against S. aureus were unsuccessful, partly due to the fact that different vaccine formulations have targeted mainly B-cell immunity. Reverse vaccinology is applied here, in order to identify potential vaccine epitope candidates. The basic epitopes prediction strategy relied on detection of a common antigenic 9-mer epitope meant to be able to stimulate both the B-cell and T-cell mediated immunity. Ten surface exposed proteins were chosen for antigenicity testing. Using a web-based system, five T-cell epitopes corresponding to fibronectin binding protein A (FDFTLSNNV and YVDGYIETI), collagen adhesin (FSINYKTKI), serine-rich adhesin for platelets (LTFDSTNNT) and elastin binding protein (FAMDKSHPE) were selected as potential vaccine candidates. Epitopes sequences were found to be conserved among the different S. aureus genomes screened from NCBI GenBank. In vitro and in vivo immunological tests will be performed in order to validate the suitability of the epitopes for vaccine development.


Subject(s)
Bacterial Vaccines/immunology , Endocarditis, Bacterial/immunology , Epitopes, T-Lymphocyte/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Adhesins, Bacterial/immunology , Amino Acid Sequence , B-Lymphocytes/immunology , Bacterial Vaccines/administration & dosage , Endocarditis, Bacterial/microbiology , Endocarditis, Bacterial/prevention & control , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Host-Pathogen Interactions/immunology , Humans , Molecular Sequence Data , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/physiology , T-Lymphocytes/immunology
14.
J Clin Transl Hepatol ; 11(2): 284-294, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-36643050

ABSTRACT

Background and Aims: Nonalcoholic fatty liver disease (NAFLD) includes a range of progressive disorders generated by excess lipid accumulation in the liver leading to hepatic steatosis and eventually fibrosis. We aimed to identify by high performance mass spectrometry-based proteomics the main signaling pathways and liver proteome changes induced by hypercholesterolemia in a rabbit atherosclerotic model that induced high accumulation of lipids in the liver. Methods: The effect of combined lipid-lowering drugs (statins and anti-PCSK9 monoclonal antibody) were used after the interruption of the hypercholesterolemic diet to identify also the potential mediators, such as alarmins, responsible for the irreversible NAFLD build up under the hyperlipidemic sustained stress. Results: Proteomic analysis revealed a number of proteins whose abundance was altered. They were components of metabolic pathways including fatty-acid degradation, glycolysis/gluconeogenesis, and nonalcoholic fatty liver disease. Mitochondrial dysfunction indicated alteration at the mitochondrial respiratory chain level and down-regulation of NADH: ubiquinone oxidoreductase. The expression of a majority of cytochromes (P4502E1, b5, and c) were up-regulated by lipid-lowering treatment. Long-term hyperlipidemic stress, even with a low-fat diet and lipid-lowering treatment, was accompanied by alarmin release (annexins, galectins, HSPs, HMGB1, S100 proteins, calreticulin, and fibronectin) that generated local inflammation and induced liver steatosis and aggressive fibrosis (by high abundance of galectin 3, fibronectin, and calreticulin). Conclusions: The novel findings of this study were related to the residual effects of hyperlipidemic stress with consistent, combined lipid-lowering treatment with statin and inhibitor of PCSK9.

16.
Sci Rep ; 12(1): 2814, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35181730

ABSTRACT

Non-apoptotic regulated cell death (ferroptosis and necroptosis) leads to the release of damage-associated molecular patterns (DAMPs), which initiate and perpetuate a non-infectious inflammatory response. We hypothesize that DAMPs and non-apoptotic regulated cell death are critical players of atherosclerotic plaque progression with inadequate response to lipid-lowering treatment. We aimed to uncover the silent mechanisms that govern the existing residual risk of cardiovascular-related mortality in experimental atherosclerosis. Proteomic and genomic approaches were applied on the ascending aorta of hyperlipidemic rabbits and controls with and without lipid-lowering treatment. The hyperlipidemic animals, which presented numerous heterogeneous atherosclerotic lesions, exhibited high concentrations of serum lipids and increased lipid peroxidation oxidative stress markers. The analyses revealed the significant upregulation of DAMPs and proteins implicated in ferroptosis and necroptosis by hyperlipidemia. Some of them did not respond to lipid-lowering treatment. Dysregulation of five proteins involved in non-apoptotic regulated cell death proteins (VDAC1, VDAC3, FTL, TF and PCBP1) and nine associated DAMPs (HSP90AA1, HSP90AB1, ANXA1, LGALS3, HSP90B1, S100A11, FN, CALR, H3-3A) was not corrected by the treatment. These proteins could play a key role in the atherosclerotic silent evolution and may possess an unexplored therapeutic potential. Mass spectrometry data are available via ProteomeXchange with identifier PXD026379.


Subject(s)
Alarmins/genetics , Atherosclerosis/genetics , Lipids/blood , Plaque, Atherosclerotic/genetics , Alarmins/blood , Animals , Aorta/metabolism , Aorta/pathology , Apoptosis/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Progression , Gene Expression Regulation/genetics , Humans , Lipid Peroxidation/genetics , Lipids/genetics , Mass Spectrometry , Oxidative Stress/genetics , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/pathology , Proteome/metabolism , Rabbits
17.
Exp Ther Med ; 24(6): 710, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36382102

ABSTRACT

Gestational diabetes mellitus (GDM) is a metabolic complication of pregnancy. The pathogenesis of GDM is considered to involve ß-cell dysfunction and insulin resistance (IR). GDM is associated with a significant risk of macrosomia in addition to a high probability of metabolic complications for the offspring. The precise mechanism underlying GDM remains unclear. The aim of the present study was to analyse the factors associated with insulin resistance and ß-cell dysfunction involved in the pathophysiology of GDM complicated with macrosomia compared with GDM without macrosomia. In addition, another aim of the present study was to assess the relationship between GDM complicated with macrosomia and anthropometric, clinical and paraclinical parameters. The following group of patients were recruited as part of a case-control study: Patients with GDM without macrosomia, patients with GDM complicated with macrosomia and healthy gestational controls. Blood samples were collected at the third trimester of pregnancy and tested for adiponectin, leptin, insulin, proinsulin and C-peptide. Homeostatic model assessment-IR (HOMA-IR), steady state ß-cell function (HOMA%B), insulin sensitivity (HOMA%S) and body mass index (BMI) were also calculated. All patients diagnosed with GDM showed an impairment in HOMA%B and a decrease in C-peptide maternal serum concentration. Additionally, diabetic status leading to the birth of offspring with macrosomia did not induce changes in the maternal serum levels of insulin, proinsulin, adiponectin or leptin, which was also the case in patients with GDM but not macrosomia. HOMA%B presented a stronger positive correlation with pre-pregnancy BMI and maternal weight gain, and a stronger negative correlation with adiponectin. Furthermore, HOMA%S in this group exhibited strong positive correlations with maternal serum levels of high-density lipoprotein cholesterol (HDL) and aspartate aminotransferase, and a strong negative correlation with pre-pregnancy BMI. In the same patients, HOMA-IR was also found to have a high negative correlation with HDL levels, and highly positive correlations with gestational age and triglyceride levels. In conclusion, the present study suggests that the different correlations among the factors involved in the pathogenesis of GDM may explain the evolution of GDM pregnancy to macrosomia.

18.
Cell Tissue Res ; 346(3): 361-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22113336

ABSTRACT

Our aim was to evaluate the effect of hyperlipidemia on the activation of endogenous alarmin, the high mobility group box 1 (HMGB1) protein, related to systemic inflammation associated with the progression of experimental atherosclerosis and to establish whether statin treatment regulates the HMGB1 signaling pathway. Hyperlipidemia was induced in vivo in golden Syrian hamsters and in monocyte cell culture (U937) by feeding the animals with a high-fat Western diet and by exposing the cells to hyperlipidemic serum. Blood samples, heart, lung and cells were harvested for biochemical, morphological, Western blot, quantitative polymerase chain reaction and enzyme-linked immunosorbent assay analyses. The data revealed that, in the atherosclerotic animal model, the protein HMGB1 and its gene expression were increased and that fluvastatin treatment significantly reduced the release of HMGB1 into the extracellular space. The cell culture experiments demonstrated the relocation of HMGB1 protein from the nucleus to cytoplasm under hyperlipidemic stress. The high level of detected HMGB1 correlated positively with the up-regulation of the advanced glycation end product receptors (RAGE) in the lung tissue from hyperlipidemic animals. During hyperlipidemic stress, the AKT signaling pathway could be activated by HMGB1-RAGE interaction. These results support the existence of a direct correlation between experimentally induced hyperlipidemia and the extracellular release of HMGB1 protein; this might be controlled by statin treatment. Moreover, the data suggest new potentials for statin therapy, with improved effects on patients with systemic inflammation induced by hyperlipidemia.


Subject(s)
HMGB1 Protein/metabolism , Hyperlipidemias/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cells, Cultured , Cricetinae , Gene Expression , HMGB1 Protein/genetics , HMGB1 Protein/physiology , Hyperlipidemias/genetics , Male , Mesocricetus , Signal Transduction
19.
Cells ; 10(9)2021 08 24.
Article in English | MEDLINE | ID: mdl-34571830

ABSTRACT

Therapeutic use of mesenchymal stem cells (MSCs) for tissue repair has great potential. MSCs from multiple sources, including those derived from human umbilical matrix, namely Wharton's jelly, may serve as a resource for obtaining MSCs. However, low in vivo engraftment efficacy of MSCs remains a challenging limitation. To improve clinical outcomes using MSCs, an in-depth understanding of the mechanisms and factors involved in successful engraftment is required. We recently demonstrated that 17ß-estradiol (E2) improves MSCs in vitro proliferation, directed migration and engraftment in murine heart slices. Here, using a proteomics approach, we investigated the angiogenic potential of MSCs in vivo and the modulatory actions of E2 on mechanisms involved in tissue repair. Specifically, using a Matrigel® plug assay, we evaluated the effects of E2 on MSCs-induced angiogenesis in ovariectomized (OVX) mice. Moreover, using proteomics we investigated the potential pro-repair processes, pathways, and co-mechanisms possibly modified by the treatment of MSCs with E2. Using RT-qPCR, we evaluated mRNA expression of pro-angiogenic molecules, including endoglin, Tie-2, ANG, and VEGF. Hemoglobin levels, a marker for blood vessel formation, were increased in plugs treated with E2 + MSCs, suggesting increased capillary formation. This conclusion was confirmed by the histological analysis of capillary numbers in the Matrigel® plugs treated with E2 + MSC. The LC-MS screening of proteins obtained from the excised Matrigel® plugs revealed 71 proteins that were significantly altered following E2 exposure, 57 up-regulated proteins and 14 down-regulated proteins. A major result was the association of over 100 microRNA molecules (miRNAs) involved in cellular communication, vesicle transport, and metabolic and energy processes, and the high percentage of approximately 25% of genes involved in unknown biological processes. Together, these data provide evidence for increased angiogenesis by MSCs treated with the sex hormone E2. In conclusion, E2 treatment may increase the engraftment and repair potential of MSCs into tissue, and may promote MSC-induced angiogenesis after tissue injury.


Subject(s)
Estrogens/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Estradiol/metabolism , Female , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Proteomics/methods , Wharton Jelly/metabolism
20.
Microvasc Res ; 79(2): 154-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20004673

ABSTRACT

Diabetes and the associated hyperglycemia affect pulmonary physiology and biochemistry inducing endothelial impairment, as the first step in lung vascular dysfunction. Caveolin-1, a characteristic protein of endothelial caveolae, acts as a scaffolding protein involved in signal transduction, cholesterol homeostasis, and vesicular trafficking. To document the effect of hyperglycemia on lung endothelial cells, we designed experiments on streptozotocin-induced diabetes and on double transgenic diabetic mice and investigated (1) the early morphological changes occurring in endothelial cells, (2) the ACE activity and cholesterol content of caveolae-rich membrane microdomains, and (3) the protein and gene expression of caveolin-1. We provide evidence that in diabetic lung, the endothelial cell displays an increased number of caveolae and enlarged surface area and a well-developed synthetic machinery, changes that correlate with an overall augmented ACE activity and cholesterol content and overexpression (gene and protein) of caveolin-1. Targeting the endothelial cell surface molecules modulated by hyperglycemia, such as caveolin-1 and ACE could be an additional therapeutic strategy in diabetes.


Subject(s)
Caveolin 1/metabolism , Diabetes Mellitus, Experimental/pathology , Endothelium, Vascular/metabolism , Animals , Caveolin 1/genetics , Cell Fractionation , Cell Surface Extensions/ultrastructure , Cholesterol/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Endothelium, Vascular/ultrastructure , Gene Expression , Immunoblotting , Lung/blood supply , Male , Mice , Mice, Knockout , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL