Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
BMC Cardiovasc Disord ; 24(1): 211, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627621

ABSTRACT

BACKGROUND: C-reactive protein (CRP) is an acute inflammatory protein detected in obese patients with metabolic syndrome. Moreover, increased CRP levels have been linked with atherosclerotic disease, congestive heart failure, and ischemic heart disease, suggesting that it is not only a biomarker but also plays an active role in the pathophysiology of cardiovascular diseases. Since endothelial dysfunction plays an essential role in various cardiovascular pathologies and is characterized by increased expression of cell adhesion molecules and inflammatory markers, we aimed to detect specific markers of endothelial dysfunction, inflammation, and oxidative stress in spontaneously hypertensive rats (SHR) expressing human CRP. This model is genetically predisposed to the development of the metabolic syndrome. METHODS: Transgenic SHR male rats (SHR-CRP) and non-transgenic SHR (SHR) at the age of 8 months were used. Metabolic profile (including serum and tissue triglyceride (TAG), serum insulin concentrations, insulin-stimulated incorporation of glucose, and serum non-esterified fatty acids (NEFA) levels) was measured. In addition, human serum CRP, MCP-1 (monocyte chemoattractant protein-1), and adiponectin were evaluated by means of ELISA, histological analysis was used to study morphological changes in the aorta, and western blot analysis of aortic tissue was performed to detect expression of endothelial, inflammatory, and oxidative stress markers. RESULTS: The presence of human CRP was associated with significantly decreased insulin-stimulated glycogenesis in skeletal muscle, increased muscle and hepatic accumulation of TAG and decreased plasmatic cGMP concentrations, reduced adiponectin levels, and increased monocyte chemoattractant protein-1 (MCP-1) levels in the blood, suggesting pro-inflammatory and presence of multiple features of metabolic syndrome in SHR-CRP animals. Histological analysis of aortic sections did not reveal any visible morphological changes in animals from both SHR and SHR-CRP rats. Western blot analysis of the expression of proteins related to the proper function of endothelium demonstrated significant differences in the expression of p-eNOS/eNOS in the aorta, although endoglin (ENG) protein expression remained unaffected. In addition, the presence of human CRP in SHR in this study did not affect the expression of inflammatory markers, namely p-NFkB, P-selectin, and COX2 in the aorta. On the other hand, biomarkers related to oxidative stress, such as HO-1 and SOD3, were significantly changed, indicating the induction of oxidative stress. CONCLUSIONS: Our findings demonstrate that CRP alone cannot fully induce the expression of endothelial dysfunction biomarkers, suggesting other risk factors of cardiovascular disorders are necessary to be involved to induce endothelial dysfunction with CRP.


Subject(s)
Hypertension , Insulins , Metabolic Syndrome , Animals , Humans , Male , Rats , Adiponectin , Aorta , Biomarkers/metabolism , C-Reactive Protein/metabolism , Chemokine CCL2 , Inflammation , Insulins/metabolism , Metabolic Syndrome/diagnosis , Metabolic Syndrome/genetics , Oxidative Stress , Rats, Inbred SHR
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 166990, 2024 03.
Article in English | MEDLINE | ID: mdl-38110128

ABSTRACT

Liver sinusoidal endothelial cells (LSECs) play a crucial role in regulating the hepatic function. Endoglin (ENG), a transmembrane glycoprotein, was shown to be related to the development of endothelial dysfunction. In this study, we hypothesized the relationship between changes in ENG expression and markers of liver sinusoidal endothelial dysfunction (LSED) during liver impairment. Male C57BL/6J mice aged 9-12 weeks were fed with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet (intrahepatic cholestasis) or choline-deficient l-amino acid defined high-fat diet (CDAA-HFD) (non-alcoholic steatohepatitis (NASH)). Significant increases in liver enzymes, fibrosis, and inflammation biomarkers were observed in both cholestasis and NASH. Decreased p-eNOS/eNOS and VE-cadherin protein expression and a significant increase in VCAM-1 and ICAM-1 expression were detected, indicating LSED in both mouse models of liver damage. A significant reduction of ENG in the DDC-fed mice, while a significant increase of ENG in the CDAA-HFD group was observed. Both DDC and CDAA-HFD-fed mice showed a significant increase in MMP-14 protein expression, which is related to significantly increased levels of soluble endoglin (sENG) in the plasma. In conclusion, we demonstrated that intrahepatic cholestasis and NASH result in an altered ENG expression, predominantly in LSECs, suggesting a critical role of ENG expression for the proper function of liver sinusoids. Both pathologies resulted in elevated sENG levels, cleaved by MMP-14 expressed predominantly from LSECs, indicating sENG as a liver injury biomarker.


Subject(s)
Acetamides , Cholestasis, Intrahepatic , Non-alcoholic Fatty Liver Disease , Animals , Male , Mice , Diet, High-Fat/adverse effects , Endoglin/metabolism , Endothelial Cells/metabolism , Matrix Metalloproteinase 14 , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology
3.
J Cardiovasc Dev Dis ; 8(12)2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34940528

ABSTRACT

Endoglin is a 180 kDa transmembrane glycoprotein that was demonstrated to be present in two different endoglin forms, namely membrane endoglin (Eng) and soluble endoglin (sEng). Increased sEng levels in the circulation have been detected in atherosclerosis, arterial hypertension, and type II diabetes mellitus. Moreover, sEng was shown to aggravate endothelial dysfunction when combined with a high-fat diet, suggesting it might be a risk factor for the development of endothelial dysfunction in combination with other risk factors. Therefore, this study hypothesized that high sEng levels exposure for 12 months combined with aging (an essential risk factor of atherosclerosis development) would aggravate vascular function in mouse aorta. Male transgenic mice with high levels of human sEng in plasma (Sol-Eng+) and their age-matched male transgenic littermates that do not develop high soluble endoglin (Control) on a chow diet were used. The aging process was initiated to contribute to endothelial dysfunction/atherosclerosis development, and it lasted 12 months. Wire myograph analysis showed impairment contractility in the Sol-Eng+ group when compared to the control group after KCl and PGF2α administration. Endothelium-dependent responsiveness to Ach was not significantly different between these groups. Western blot analysis revealed significantly decreased protein expression of Eng, p-eNOS, and ID1 expression in the Sol-Eng+ group compared to the control group suggesting reduced Eng signaling. In conclusion, we demonstrated for the first time that long-term exposure to high levels of sEng during aging results in alteration of vasoconstriction properties of the aorta, reduced eNOS phosphorylation, decreased Eng expression, and altered Eng signaling. These findings suggest that sEng can be considered a risk factor for the development of vascular dysfunction during aging and a potential therapeutical target for pharmacological intervention.

4.
Curr Microbiol ; 78(5): 2104-2111, 2021 May.
Article in English | MEDLINE | ID: mdl-33765192

ABSTRACT

The yeasts of the genus Candida are among the most clinically significant fungal pathogenic agents. One of the unique features of the Candida species' pathogenicity is their ability to form biofilms. Generally, infections caused by biofilm-forming microorganisms tend to have chronic course and are difficult to treat. This fact highlights the need to search for drugs with anti-biofilm activities. At present, there are variety of protocols for performing antifungal anti-biofilm activity testing in which fundamental differences, especially in the choice of cultivation media for biofilm formation, can be noted. In our study, we focused on the effect of four different culture media on biofilm biomass formation in ten Candida spp. strains. With emphasis placed on clinical significance, strains of the C. albicans species were predominantly included in this study. Based on our results, we can conclude that the availability of other components in the culture media, such as amino acids or proteins, and not just the commonly mentioned glucose availability, helps promote the transition of Candida yeasts into a sessile form and leads to in vitro robust biofilm formation. We revealed that biofilm formation in C. albicans strains was enhanced, especially in media supplemented with fetal bovine serum (FBS). The nutritionally balanced cultivation medium with 10 g/L glucose and 10% (v/v) FBS evidently showed the most significant benefit for in vitro biofilm production in C. albicans strains.


Subject(s)
Candida albicans , Candida , Antifungal Agents , Biofilms , Biomass , Culture Media
5.
J Cell Mol Med ; 24(4): 2402-2415, 2020 02.
Article in English | MEDLINE | ID: mdl-31957261

ABSTRACT

Arthrospira platensis, a blue-green alga, is a popular nutraceutical substance having potent antioxidant properties with potential anti-carcinogenic activities. The aim of our study was to assess the possible anti-angiogenic effects of A platensis in an experimental model of pancreatic cancer. The effects of an A platensis extract were investigated on human pancreatic cancer cells (PA-TU-8902) and immortalized endothelial-like cells (Ea.hy926). PA-TU-8902 pancreatic tumours xenografted to athymic mice were also examined. In vitro migration and invasiveness assays were performed on the tested cells. Multiple angiogenic factors and signalling pathways were analysed in the epithelial, endothelial and cancer cells, and tumour tissue. The A platensis extract exerted inhibitory effects on both migration and invasion of pancreatic cancer as well as endothelial-like cells. Tumours of mice treated with A platensis exhibited much lesser degrees of vascularization as measured by CD31 immunostaining (P = .004). Surprisingly, the VEGF-A mRNA and protein expressions were up-regulated in pancreatic cancer cells. A platensis inhibited ERK activation upstream of Raf and suppressed the expression of ERK-regulated proteins. Treatment of pancreatic cancer with A platensis was associated with suppressive effects on migration and invasiveness with various anti-angiogenic features, which might account for the anticancer effects of this blue-green alga.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/drug therapy , Spirulina/chemistry , Animals , Antioxidants/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/drug effects , Humans , Mice , Mice, Nude , Signal Transduction/drug effects , Up-Regulation/drug effects , Pancreatic Neoplasms
6.
Microb Pathog ; 136: 103666, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31412284

ABSTRACT

One of the pathways for the delivery of virulence effector molecules into the extracellular environment of Candida albicans relies on the release of membrane-bound carriers which are called extracellular vesicles (EVs). Only a few studies aimed at investigating Candida albicans extracellular vesicles protein cargo and its potential contribution to the pathogenesis of C. albicans infections have been conducted to date. In this study, we mainly focused on a search for proteins with a demonstrated linkage to pathogenesis in EVs isolated from two C. albicans strains, the model strain ATCC 90028 and the clinical isolate from a woman suffering from vulvovaginal candidiasis. For the purpose of mimicking one of many hostile conditions during a host-pathogen interaction, C. albicans strains in a nutrient-limited medium were cultivated. We have hypothesized that this unfavourable, stressful condition could contribute to the induction of virulence effector molecules being released at a more extensive rate. In conclusion, 34 proteins with an undisputed linkage to C. albicans pathogenesis were detected in the extracellular vesicle cargoes of both strains. In case of the clinical isolate strain, no unique virulence-associated proteins were detected. In the C. albicans ATCC 90028 model strain, three unique proteins were detected, namely: agglutinin-like protein 3 (Als3), secreted aspartic protease 8 (Sap8) and cell surface superoxide dismutase [Cu-Zn] 6 (Sod6).


Subject(s)
Candida albicans/metabolism , Extracellular Vesicles/metabolism , Fungal Proteins/metabolism , Virulence Factors/metabolism , Candida albicans/growth & development , Culture Media/chemistry , Humans , Protein Transport
7.
Atherosclerosis ; 271: 15-25, 2018 04.
Article in English | MEDLINE | ID: mdl-29459262

ABSTRACT

BACKGROUND AND AIMS: Increased plasma levels of soluble endoglin (sEng) were detected in patients with endothelial dysfunction-related disorders and hypercholesterolemia. In this study, we hypothesized that high levels of sEng accompanied by mild hypercholesterolemia could aggravate endothelial and vessel wall dysfunction and affect endoglin/eNOS signaling in mouse aorta. METHODS: Three-month-old female transgenic mice on CBAxC57BL/6J background, with high levels of sEng (Sol-Eng+high HFD), and their littermates with low levels of sEng (Sol-Eng+low HFD), were fed a high fat diet for six months. Plasma samples were used for biochemical, ELISA and Luminex analyses of total cholesterol, sEng and inflammatory markers. Functional parameters of aorta were assessed with wire myograph 620M. Western blot analyses of membrane endoglin/eNOS signaling and endothelial dysfunction/inflammation markers in aorta were performed. RESULTS: Functional analysis of aorta showed impaired KCl induced vasoconstriction, endothelial-dependent relaxation after the administration of acetylcholine as well as endothelial-independent relaxation induced by sodium nitroprusside in the Sol-Eng+high HFD group compared to the Sol-Eng+low HFD group. Ach-induced vasodilation after administration of l-NAME was significantly higher in the Sol-Eng+high HFD group compared to the Sol-Eng+low HFD group. The expression of endoglin, p-eNOS/eNOS, pSmad2/3/Smad2/3 signaling pathway was significantly lower in the Sol-Eng+high HFD group compared to the Sol-Eng+low HFD group. CONCLUSIONS: The results indicate that long-term hypercholesterolemia combined with high levels of sEng leads to the aggravation of endothelial and vessel wall dysfunction in aorta, with possible alterations of the membrane endoglin/eNOS signaling, suggesting that high levels of soluble endoglin might be considered as a risk factor of cardiovascular diseases.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Endoglin/metabolism , Endothelium, Vascular/metabolism , Hypercholesterolemia/complications , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Diet, High-Fat , Disease Models, Animal , Endoglin/genetics , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Humans , Hypercholesterolemia/etiology , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Up-Regulation , Vasoconstriction , Vasodilation
8.
Heart Vessels ; 32(8): 1013-1024, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28382387

ABSTRACT

A soluble form of endoglin (sEng) released into the circulation was suggested to be a direct inducer of endothelial dysfunction, inflammation and contributed to the development of hypertension by interfering with TGF-ß signaling in cardiovascular pathologies. In the present study, we assessed the hypothesis that high sEng level-induced hypertension via a possible sEng interference with TGF-ß signaling pathways may result in inflammatory, structural or fibrotic changes in hearts of Sol-Eng+ mice (mice with high levels of soluble endoglin) fed either chow or high-fat diet. Female Sol-Eng+ mice and their age matched littermates with low plasma levels of sEng were fed either chow or high-fat diet (HFD). Heart samples were subsequently analyzed by histology, qRT-PCR and Western blot analysis. In this study, no differences in myocardial morphology/hypertrophy and possible fibrotic changes between Sol-Eng+ mice and control mice were detected on both chow and HFD. The presence of sEng did not significantly affect the expression of selected members of TGF-ß signaling (membrane endoglin, TGFßRII, ALK-5, ALK-1, Id-1, PAI-1 and activated Smad proteins-pSmad 1,5 and pSmad 2,3), inflammation, heart remodeling (PDGFb, Col1A1) and endothelial dysfunction (VCAM-1, ICAM-1) in the hearts of Sol-Eng+ mice compared to control mice on both chow and high-fat diet. High levels of soluble endoglin did not affect microscopic structure (profibrotic and degenerative cardiomyocyte changes), and specific parts of TGF-ß signaling, endothelial function and inflammation in the heart of Sol-Eng+ mice fed both chow diet or HFD. However, we cannot rule out a possibility that a long-term chronic exposure (9 months and more) to soluble endoglin alone or combined with other cardiovascular risk factors may contribute to alterations of heart function and structure in Sol-Eng+ mice, which is the topic in our lab in ongoing experiments.


Subject(s)
Cardiomyopathy, Hypertrophic/blood , Endoglin/genetics , Gene Expression Regulation , Hypertension/blood , Myocardium/metabolism , RNA/genetics , Animals , Blotting, Western , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/pathology , Disease Models, Animal , Endoglin/biosynthesis , Female , Gene Expression Profiling , Heart , Hypertension/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Oxidative Stress , Real-Time Polymerase Chain Reaction , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
9.
Life Sci ; 175: 52-60, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28336397

ABSTRACT

AIMS: Endoglin is a transmembrane glycoprotein, that plays an important role in regulating endothelium. Proteolytic cleavage of membrane endoglin releases soluble endoglin (sEng), whose increased plasma levels have been detected in diseases related to the cardiovascular system. It was proposed that sEng might damage vascular endothelium, but detailed information about the potential mechanisms involved is not available. Thus, we hypothesized that sEng contributes to endothelial dysfunction, leading to a pro-inflammatory phenotype by a possible modulation of the TGF-ß and/or inflammatory pathways. MAIN METHODS: Human umbilical vein endothelial cells (HUVECs) and Human embryonic kidney cell line (HEK293T) were treated with different sEng concentration and time in order to reveal possible effect on biomarkers of inflammation and TGF-ß signaling. IL6 and NFκB reporter luciferase assays, quantitative real-time PCR analysis, Western blot analysis and immunofluorescence flow cytometry were used. KEY FINDINGS: sEng treatment results in activation of NF-κB/IL-6 expression, increased expression of membrane endoglin and reduced expression of Id-1. On the other hand, no significant effects on other markers of endothelial dysfunction and inflammation, including eNOS, peNOSS1177, VCAM-1, COX-1, COX-2 and ICAM-1 were detected. SIGNIFICANCE: As a conclusion, sEng treatment resulted in an activation of NF-κB, IL-6, suggesting activation of pro-inflammatory phenotype in endothelial cells. The precise mechanism of this activation and its consequence remains to be elucidated. A combined treatment of sEng with other cardiovascular risk factors will be necessary in order to reveal whether sEng is not only a biomarker of cardiovascular diseases, but also a protagonist of endothelial dysfunction.


Subject(s)
Endoglin/biosynthesis , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Interleukin-6/biosynthesis , NF-kappa B/biosynthesis , Signal Transduction , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inhibitor of Differentiation Protein 1/biosynthesis , Solubility
10.
J Vasc Res ; 53(3-4): 149-162, 2016.
Article in English | MEDLINE | ID: mdl-27721318

ABSTRACT

AIMS: A soluble form of endoglin (sEng) was proposed to participate in the induction of endothelial dysfunction in small blood vessels. Here, we tested the hypothesis that high levels of sEng combined with a high-fat diet induce endothelial dysfunction in an atherosclerosis-prone aorta. METHODS AND RESULTS: Six-month-old female and male transgenic mice overexpressing human sEng (Sol-Eng+) with low (Sol-Eng+low) or high (Sol-Eng+high) levels of plasma sEng were fed a high-fat rodent diet containing 1.25% cholesterol and 40% fat for 3 months. The plasma cholesterol and mouse sEng levels did not differ in the Sol-Eng+high and Sol-Eng+low mice. The expression of proinflammatory (P-selectin, ICAM-1, pNFκB and COX-2) and oxidative-stress-related markers (HO-1, NOX-1 and NOX-2) in the aortas of Sol-Eng+high female mice was significantly higher than in Sol-Eng+low female mice. Endothelium-dependent vasodilatation induced by acetylcholine was preserved better in the Sol-Eng+ high female mice than in the Sol-Eng+low female mice. CONCLUSION: These results suggest that high concentrations of sEng in plasma in combination with a high-fat diet induce the simultaneous activation of proinflammatory, pro-oxidative and vasoprotective mechanisms in mice aorta and the balance of these biological processes determines whether the final endothelial phenotype is adaptive or maladaptive.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Diet, High-Fat , Endoglin/metabolism , Inflammation/metabolism , Nitric Oxide/metabolism , Oxidative Stress , Vasodilation , Adaptation, Physiological , Animals , Aorta/drug effects , Aorta/physiopathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/physiopathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Biomarkers/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endoglin/blood , Endoglin/genetics , Female , Genetic Predisposition to Disease , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/physiopathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Phenotype , Up-Regulation , Vasodilation/drug effects , Vasodilator Agents/pharmacology
11.
PLoS One ; 11(7): e0158817, 2016.
Article in English | MEDLINE | ID: mdl-27391242

ABSTRACT

Non Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH) are the hepatic manifestations of the metabolic syndrome; worrisome is the booming increase in pediatric age. To recreate the full spectrum of juvenile liver pathology and investigate the gender impact, male and female C57Bl/6 mice were fed with high fat diet plus fructose in the drinking water (HFHC) immediately after weaning (equal to 3-years old human), and disease progression followed for 16 weeks, until adults (equal to 30-years old human). 100% of subjects of both genders on HFHC diet developed steatosis in 4weeks, and some degree of fibrosis in 8weeks, with the 86% of males and 15% of females presenting a stage 2 fibrosis at 16weeks. Despite a similar final liver damage both groups, a sex difference in the pathology progression was observed. Alterations in glucose homeostasis, dyslipidemia, hepatomegaly and obese phenotype were evident from the very beginning in males with an increased hepatic inflammatory activity. Conversely, such alterations were present in females only at the end of the HFHC diet (with the exception of insulin resistance and the hepatic inflammatory state). Interestingly, only females showed an altered hepatic redox state. This juvenile model appears a good platform to unravel the underlying gender dependent mechanisms in the progression from NAFLD to NASH, and to characterize novel therapeutic approaches.


Subject(s)
Dietary Fats/adverse effects , Disease Models, Animal , Fructose/adverse effects , Non-alcoholic Fatty Liver Disease/chemically induced , Sex Characteristics , Adult , Animals , Dietary Fats/pharmacology , Dyslipidemias/chemically induced , Dyslipidemias/metabolism , Dyslipidemias/pathology , Dyslipidemias/physiopathology , Female , Fructose/pharmacology , Humans , Male , Mice , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/blood , Obesity/chemically induced , Obesity/pathology , Obesity/physiopathology
12.
Atherosclerosis ; 243(2): 383-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26520890

ABSTRACT

A soluble form of endoglin (sEng) is known to be an extracellular domain of the full-length membrane endoglin, which is elevated during various pathological conditions related to vascular endothelium. In the current review, we tried to summarize a possible role of soluble endoglin in cardiovascular pathologies, focusing on its relation to endothelial dysfunction and cholesterol levels. We discussed sEng as a proposed biomarker of cardiovascular disease progression, cardiovascular disease treatment and endothelial dysfunction. We also addressed a potential interaction of sEng with TGF-ß/eNOS or BMP-9 signaling. We suggest soluble endoglin levels to be monitored, because they reflect the progression/treatment efficacy of cardiovascular diseases related to endothelial dysfunction and hypercholesterolemia. A possible role of soluble endoglin as an inducer of endothelial dysfunction however remains to be elucidated.


Subject(s)
Antigens, CD/blood , Cardiovascular Diseases/blood , Endothelium, Vascular/metabolism , Hypercholesterolemia/blood , Receptors, Cell Surface/blood , Animals , Biomarkers/blood , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Endoglin , Endothelium, Vascular/physiopathology , Growth Differentiation Factor 2 , Growth Differentiation Factors/metabolism , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/physiopathology , Hypercholesterolemia/therapy , Nitric Oxide Synthase Type III/metabolism , Prognosis , Signal Transduction , Transforming Growth Factor beta/metabolism
13.
PLoS One ; 10(3): e0119665, 2015.
Article in English | MEDLINE | ID: mdl-25768936

ABSTRACT

Increased levels of a soluble form of endoglin (sEng) circulating in plasma have been detected in various pathological conditions related to cardiovascular system. High concentration of sEng was also proposed to contribute to the development of endothelial dysfunction, but there is no direct evidence to support this hypothesis. Therefore, in the present work we analyzed whether high sEng levels induce endothelial dysfunction in aorta by using transgenic mice with high expression of human sEng. Transgenic mice with high expression of human sEng on CBAxC57Bl/6J background (Sol-Eng+) and age-matched transgenic littermates that do not develop high levels of human soluble endoglin (control animals in this study) on chow diet were used. As expected, male and female Sol-Eng+ transgenic mice showed higher levels of plasma concentrations of human sEng as well as increased blood arterial pressure, as compared to control animals. Functional analysis either in vivo or ex vivo in isolated aorta demonstrated that the endothelium-dependent vascular function was similar in Sol-Eng+ and control mice. In addition, Western blot analysis showed no differences between Sol-Eng+ and control mice in the protein expression levels of endoglin, endothelial NO-synthase (eNOS) and pro-inflammatory ICAM-1 and VCAM-1 from aorta. Our results demonstrate that high levels of soluble endoglin alone do not induce endothelial dysfunction in Sol-Eng+ mice. However, these data do not rule out the possibility that soluble endoglin might contribute to alteration of endothelial function in combination with other risk factors related to cardiovascular disorders.


Subject(s)
Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Intracellular Signaling Peptides and Proteins/blood , Animals , Aorta , Arterial Pressure/physiology , Cardiovascular Diseases/blood , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Endoglin , Female , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
14.
Histol Histopathol ; 30(2): 233-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25208891

ABSTRACT

Endoglin (TGF-ß receptor III), has been demonstrated to affect vascular endothelium and atherosclerosis. Moreover, it was also demonstrated that endoglin is involved in inflammation and plays a role in leukocyte adhesion and transmigration in vitro and in vivo but not in atherosclerosis related vessels. In this study, we wanted to evaluate endoglin expression in two different parts of the aorta (heart aortic sinus and ascending aorta) and assess its potential simultaneous expression with cell adhesion molecules in non-atherosclerotic and atherosclerotic aortas of apoE-deficient mice. Ten-week-old female apolipoprotein E-deficient mice on a C57BL/6J background (n=24) were randomly subdivided into three groups and were fed either chow diet (for another two months) or Western type diet (for another two or four months). Immunohistochemical staining of endoglin, VCAM-1 and P-selectin in aortic sinus and ascending aorta was performed. Endoglin expression was detected only in endothelial cells and varied during atherogenic process in aorta but not in aortic sinus. Moreover, its expression seemed to be weaker in aorta when compared to aortic sinus and the positivity was detected only in endothelium covering atherosclerotic lesions but not in non-atherosclerotic endothelium regardless of the plaque size. Endoglin was not expressed with P selectin and VCAM-1 in aortic endothelium in any studied group. This study shows that endothelial expression of endoglin is related to the atherogenic process predominantly in aorta outside the heart. Moreover, endoglin is not localized with cell adhesion molecules involved in atherosclerosis, suggesting it might not participate in leukocyte accumulation in aorta of apoE-deficient mice during atherogenesis.


Subject(s)
Aorta/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cell Adhesion Molecules/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , Animals , Diet, Atherogenic , Endoglin , Female , Lipids/blood , Macrophages/metabolism , Mice , Mice, Inbred C57BL , P-Selectin/biosynthesis , Plaque, Atherosclerotic/pathology , Vascular Cell Adhesion Molecule-1/biosynthesis
15.
Heart Vessels ; 30(2): 241-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24633496

ABSTRACT

C57BL/6J (B6) mice were demonstrated to be the most susceptible and C3H/HeJ (C3H) mice the most resistant to development of atherosclerosis. We hypothesized, whether pro-atherogenic (P-selectin, VCAM-1, and ICAM-1) and anti-atherogenic (endoglin and eNOS) proteins are expressed differently in aorta before the onset of atherosclerosis in these two mouse strains. B6 mice (n = 16) and C3H mice (n = 16) sustained on either chow or cholesterol (1 %) diet for 8 weeks. Biochemical analysis of lipoprotein profile and Western blot analysis of P-selectin, VCAM-1, ICAM-1, eNOS, endoglin, peNOS and TGF-ßRII in aorta were performed. Western blot analysis revealed a lower expression of P-selectin by 7 %, VCAM-1 by 51 %, ICAM-1 by 6 %, and a higher expression of eNOS (by 18 %) in C3H mice in comparison with B6 mice after cholesterol diet. Further analysis revealed that cholesterol diet significantly increased the expression of endoglin (by 97 %), TGF-ßRII (by 50 %), eNOS (by 21 %) and peNOS (by 122 %) in C3H mice, but not in B6 mice. We propose that lower expression of P-selectin, VCAM-1 and ICAM-1 and higher expression of eNOS in vivo in aorta of C3H mice might represent another potential mechanism for C3H mice being less susceptible to atherosclerosis when compared to B6 mice. In addition, endoglin seems to be involved in an upregulation of eNOS only in C3H mice. Thus, we propose that aorta of C3H mice is less prone to the expression of pro-inflammatory and endothelial dysfunction markers when compared to B6 mice, regardless of lipoprotein profile and before any signs of atherosclerotic process.


Subject(s)
Aorta/enzymology , Atherosclerosis/enzymology , Cell Adhesion Molecules/metabolism , Inflammation Mediators/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Female , Genetic Predisposition to Disease , Mice, Inbred C3H , Mice, Inbred C57BL , Phenotype , Protective Factors , Risk Factors , Species Specificity
16.
Biomed Res Int ; 2014: 291531, 2014.
Article in English | MEDLINE | ID: mdl-25162006

ABSTRACT

Rats transgenic for Huntington's disease (tgHD51 CAG rats), surviving up to two years, represent an animal model of HD similar to the late-onset form of human disease. This enables us to follow histopathological changes in course of neurodegenerative process (NDP) within the striatum and compare them with postmortem samples of human HD brains. A basic difference between HD pathology in human and tgHD51 rats is in the rate of NDP progression that originates primarily from slow neuronal degeneration consequently resulting in lesser extent of concomitant reactive gliosis in the brain of tgHD51 rats. Although larger amount of striatal neurons displays only gradual decrease in their size, their number is significantly reduced in the oldest tgHD51 rats. Our quantitative analysis proved that the end of the first year represents the turn in the development of morphological changes related to the progression of NDP in tgHD51 rats. Our data also support the view that all types of CNS glial cells play an important, irreplaceable role in NDP. To the best of our knowledge, our findings are the first to document that tgHD51 CAG rats can be used as a valid animal model for detailed histopathological studies related to HD in human.


Subject(s)
Corpus Striatum/pathology , Huntington Disease/pathology , Nerve Degeneration/pathology , Trinucleotide Repeat Expansion/genetics , Animals , Disease Models, Animal , Humans , Huntington Disease/genetics , Nerve Degeneration/genetics , Neurons/pathology , Rats , Rats, Transgenic
17.
Food Funct ; 4(11): 1586-94, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056745

ABSTRACT

Spirulina platensis, a water blue-green alga, has been associated with potent biological effects, which might have important relevance in atheroprotection. We investigated whether S. platensis or phycocyanobilin (PCB), its tetrapyrrolic chromophore, can activate atheroprotective heme oxygenase-1 (Hmox1), a key enzyme in the heme catabolic pathway responsible for generation of a potent antioxidant bilirubin, in endothelial cells and in a mouse model of atherosclerosis. In vitro experiments were performed on EA.hy926 endothelial cells exposed to extracts of S. platensis or PCB. In vivo studies were performed on ApoE-deficient mice fed a cholesterol diet and S. platensis. The effect of these treatments on Hmox1, as well as other markers of oxidative stress and endothelial dysfunction, was then investigated. Both S. platensis and PCB markedly upregulated Hmox1 in vitro, and a substantial overexpression of Hmox1 was found in aortic atherosclerotic lesions of ApoE-deficient mice fed S. platensis. In addition, S. platensis treatment led to a significant increase in Hmox1 promoter activity in the spleens of Hmox-luc transgenic mice. Furthermore, both S. platensis and PCB were able to modulate important markers of oxidative stress and endothelial dysfunction, such as eNOS, p22 NADPH oxidase subunit, and/or VCAM-1. Both S. platensis and PCB activate atheroprotective HMOX1 in endothelial cells and S. platensis increased the expression of Hmox1 in aortic atherosclerotic lesions in ApoE-deficient mice, and also in Hmox-luc transgenic mice beyond the lipid lowering effect. Therefore, activation of HMOX1 and the heme catabolic pathway may represent an important mechanism of this food supplement for the reduction of atherosclerotic disease.


Subject(s)
Atherosclerosis/prevention & control , Heme Oxygenase-1/genetics , Phycobilins/administration & dosage , Phycocyanin/administration & dosage , Spirulina/chemistry , Animals , Aorta/drug effects , Aorta/enzymology , Atherosclerosis/drug therapy , Atherosclerosis/enzymology , Female , Heme Oxygenase-1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Up-Regulation/drug effects , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...