Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Vasc Biol ; 4(1): 40-49, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36279189

ABSTRACT

Marfan syndrome (MFS) is a connective tissue disorder causing aortic aneurysm formation. Currently, only prophylactic aortic surgery and blood pressure-lowering drugs are available to reduce the risk of aortic rupture. Upon whole genome sequencing of a Marfan family, we identified a complement gene C1R variant (p.Ser152Leu), which is associated with severe aortic patients. Therefore, we assessed the role of complement activation in MFS aortic tissue. Expression of various complement genes and proteins was detected in human and murine MFS aneurysm tissue, which prompted us to study complement inhibition in MFS mice. Treatment of the Fbn1C1041G/+ MFS mice with human plasma-derived C1-esterase inhibitor Cetor® resulted in reduced complement deposition, decreased macrophage influx in the aorta, and lower circulating TNFα levels. However, in line with previous anti-inflammatory treatments, complement inhibition did not change the aortic dilatation rate in this MFS mouse model. Thus, while complement factors/component 3 activation were detected in human/murine MFS aorta, Cetor® had no effect on aortic dilatation in MFS mice, indicating that complement inhibition is not a suitable treatment strategy in MFS.

2.
Cardiovasc Pathol ; 38: 1-6, 2019.
Article in English | MEDLINE | ID: mdl-30359839

ABSTRACT

Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the fibrillin-1 gene (FBN1), resulting in aortic aneurysm formation and dissections. Interestingly, variable aortopathy is observed even within MFS families with the same mutation. Thus, additional risk factors determine disease severity. Here, we describe a case of a 2-month-old Fbn1C1039G/+ MFS mouse with extreme aortic dilatation and increased vascular inflammation, when compared to MFS siblings, which coincided with unilateral renal cystic disease. In addition, this mouse presented with increased serum levels of creatinine, angiotensin-converting enzyme, corticosterone, macrophage chemoattractant protein-1, and interleukin-6, which may have contributed to the vascular pathology. Possibly, cystic kidney disease is associated with aneurysm progression in MFS patients. Therefore, we propose that close monitoring of the presence of renal cysts in MFS patients, during regular vascular imaging of the whole aorta trajectory, may provide insight in the frequency of cystic kidney disease and its potential as a novel indicator of aneurysm progression in MFS patients.


Subject(s)
Aorta/pathology , Aortic Aneurysm/etiology , Fibrillin-1/genetics , Kidney Diseases, Cystic/etiology , Marfan Syndrome/genetics , Animals , Aorta/metabolism , Aortic Aneurysm/blood , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Aortitis/blood , Aortitis/etiology , Aortitis/genetics , Aortitis/pathology , Biomarkers/blood , Dilatation, Pathologic , Disease Models, Animal , Fibrillin-1/metabolism , Genetic Predisposition to Disease , Kidney Diseases, Cystic/blood , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Male , Marfan Syndrome/blood , Marfan Syndrome/complications , Marfan Syndrome/diagnosis , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
3.
Microbiology (Reading) ; 163(12): 1864-1879, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29139344

ABSTRACT

Disulfide bonds confer stability and activity to proteins. Bioinformatic approaches allow predictions of which organisms make protein disulfide bonds and in which subcellular compartments disulfide bond formation takes place. Such an analysis, along with biochemical and protein structural data, suggests that many of the extremophile Crenarachaea make protein disulfide bonds in both the cytoplasm and the cell envelope. We have sought to determine the oxidative folding pathways in the sequenced genomes of the Crenarchaea, by seeking homologues of the enzymes known to be involved in disulfide bond formation in bacteria. Some Crenarchaea have two homologues of the cytoplasmic membrane protein VKOR, a protein required in many bacteria for the oxidation of bacterial DsbAs. We show that the two VKORs of Aeropyrum pernix assume opposite orientations in the cytoplasmic membrane, when expressed in E. coli. One has its active cysteines oriented toward the E. coli periplasm (ApVKORo) and the other toward the cytoplasm (ApVKORi). Furthermore, the ApVKORo promotes disulfide bond formation in the E. coli cell envelope, while the ApVKORi promotes disulfide bond formation in the E. coli cytoplasm via a co-expressed archaeal protein ApPDO. Amongst the VKORs from different archaeal species, the pairs of VKORs in each species are much more closely related to each other than to the VKORs of the other species. The results suggest two independent occurrences of the evolution of the two topologically inverted VKORs in archaea. Our results suggest a mechanistic basis for the formation of disulfide bonds in the cytoplasm of Crenarchaea.


Subject(s)
Aeropyrum/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Disulfides/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Aeropyrum/chemistry , Aeropyrum/genetics , Archaeal Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Cysteine/chemistry , Cysteine/metabolism , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , Disulfides/metabolism , Membrane Proteins/genetics , Periplasm/genetics , Periplasm/metabolism , Protein Folding
4.
J Pathol ; 243(3): 294-306, 2017 11.
Article in English | MEDLINE | ID: mdl-28727149

ABSTRACT

Marfan syndrome (MFS) is a connective tissue disorder in which aortic rupture is the major cause of death. MFS patients with an aortic diameter below the advised limit for prophylactic surgery (<5 cm) may unexpectedly experience an aortic dissection or rupture, despite yearly monitoring. Hence, there is a clear need for improved prognostic markers to predict such aortic events. We hypothesize that elastin fragments play a causal role in aortic calcification in MFS, and that microcalcification serves as a marker for aortic disease severity. To address this hypothesis, we analysed MFS patient and mouse aortas. MFS patient aortic tissue showed enhanced microcalcification in areas with extensive elastic lamina fragmentation in the media. A causal relationship between medial injury and microcalcification was revealed by studies in vascular smooth muscle cells (SMCs); elastin peptides were shown to increase the activity of the calcification marker alkaline phosphatase (ALP) and reduce the expression of the calcification inhibitor matrix GLA protein in human SMCs. In murine Fbn1C1039G/+ MFS aortic SMCs, Alpl mRNA and activity were upregulated as compared with wild-type SMCs. The elastin peptide-induced ALP activity was prevented by incubation with lactose or a neuraminidase inhibitor, which inhibit the elastin receptor complex, and a mitogen-activated protein kinase kinase-1/2 inhibitor, indicating downstream involvement of extracellular signal-regulated kinase-1/2 (ERK1/2) phosphorylation. Histological analyses in MFS mice revealed macrocalcification in the aortic root, whereas the ascending aorta contained microcalcification, as identified with the near-infrared fluorescent bisphosphonate probe OsteoSense-800. Significantly, microcalcification correlated strongly with aortic diameter, distensibility, elastin breaks, and phosphorylated ERK1/2. In conclusion, microcalcification co-localizes with aortic elastin degradation in MFS aortas of humans and mice, where elastin-derived peptides induce a calcification process in SMCs via the elastin receptor complex and ERK1/2 activation. We propose microcalcification as a novel imaging marker to monitor local elastin degradation and thus predict aortic events in MFS patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Elastin/metabolism , Marfan Syndrome/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/metabolism , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Calcinosis/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Marfan Syndrome/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology
5.
Arterioscler Thromb Vasc Biol ; 36(8): 1618-26, 2016 08.
Article in English | MEDLINE | ID: mdl-27283746

ABSTRACT

OBJECTIVE: Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the fibrillin-1 gene. Patients with MFS are at risk of aortic aneurysm formation and dissection. Usually, blood pressure-lowering drugs are used to reduce aortic events; however, this is not sufficient for most patients. In the aorta of smooth muscle cell-specific sirtuin-1-deficient mice, spontaneous aneurysm formation and senescence are observed. Resveratrol is known to enhance sirtuin-1 activity and to reduce senescence, which prompted us to investigate the effectiveness of resveratrol in inhibition of aortic dilatation in the Fbn1(C1039G/+) MFS mouse model. APPROACH AND RESULTS: Aortic senescence strongly correlates with aortic root dilatation rate in MFS mice. However, although resveratrol inhibits aortic dilatation, it only shows a trend toward reduced aortic senescence. Resveratrol enhances nuclear localization of sirtuin-1 in the vessel wall and, in contrast to losartan, does not affect leukocyte infiltration nor activation of SMAD2 and extracellular signal-regulated kinases 1/2 (ERK1/2). Interestingly, specific sirtuin-1 activation (SRT1720) or inhibition (sirtinol) in MFS mice does not affect aortic root dilatation rate, although senescence is changed. Resveratrol reduces aortic elastin breaks and decreases micro-RNA-29b expression coinciding with enhanced antiapoptotic Bcl-2 expression and decreased number of terminal apoptotic cells. In cultured smooth muscle cells, the resveratrol effect on micro-RNA-29b downregulation is endothelial cell and nuclear factor κB-dependent. CONCLUSIONS: Resveratrol inhibits aortic root dilatation in MFS mice by promoting elastin integrity and smooth muscle cell survival, involving downregulation of the aneurysm-related micro-RNA-29b in the aorta. On the basis of these data, resveratrol holds promise as a novel intervention strategy for patients with MFS.


Subject(s)
Aorta/drug effects , Aortic Aneurysm/prevention & control , Fibrillin-1/genetics , Marfan Syndrome/drug therapy , Stilbenes/pharmacology , Active Transport, Cell Nucleus , Animals , Aorta/metabolism , Aorta/pathology , Aortic Aneurysm/etiology , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Apoptosis/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Dilatation, Pathologic , Disease Models, Animal , Elastin/metabolism , Female , Genetic Predisposition to Disease , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , Resveratrol , Sirtuin 1/metabolism
6.
PLoS One ; 9(9): e107221, 2014.
Article in English | MEDLINE | ID: mdl-25238161

ABSTRACT

AIMS: Patients with Marfan syndrome have an increased risk of life-threatening aortic complications, mostly preceded by aortic dilatation. In the FBN1(C1039G/+) Marfan mouse model, losartan decreases aortic root dilatation. We recently confirmed this beneficial effect of losartan in adult patients with Marfan syndrome. The straightforward translation of this mouse model to man is reassuring to test novel treatment strategies. A number of studies have shown signs of inflammation in aortic tissue of Marfan patients. This study examined the efficacy of anti-inflammatory therapies in attenuating aortic root dilation in Marfan syndrome and compared effects to the main preventative agent, losartan. METHODS AND RESULTS: To inhibit inflammation in FBN1(C1039G/+) Marfan mice, we treated the mice with losartan (angiotensin II receptor type 1 inhibitor), methylprednisolone (corticosteroid) or abatacept (T-cell-specific inhibitor). Treatment was initiated in adult Marfan mice with already existing aortic root dilatation, and applied for eight weeks. Methylprednisolone- or abatacept-treated mice did not reveal a reduction in aortic root dilatation. In this short time frame, losartan was the only treatment that significantly reduced aorta inflammation, transforming growth factor-beta (TGF-ß) signaling and aortic root dilatation rate in these adult Marfan mice. Moreover, the methylprednisolone-treated mice had significantly more aortic alcian blue staining as a marker for aortic damage. CONCLUSION: Anti-inflammatory agents do not reduce the aortic dilatation rate in Marfan mice, but possibly increase aortic damage. Currently, the most promising therapeutic drug in Marfan syndrome is losartan, by blocking the angiotensin II receptor type 1 and thereby inhibiting pSmad2 signaling.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Aorta/drug effects , Dilatation, Pathologic/drug therapy , Losartan/therapeutic use , Marfan Syndrome/drug therapy , Animals , Anti-Inflammatory Agents/adverse effects , Aorta/metabolism , Aorta/pathology , Marfan Syndrome/complications , Marfan Syndrome/pathology , Mice , Mice, Inbred C57BL , Smad2 Protein/metabolism
7.
Arterioscler Thromb Vasc Biol ; 33(10): 2380-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950142

ABSTRACT

OBJECTIVE: In aortic aneurysms the arterial vessel wall is dilated because of destruction of its integrity, which may lead to lethal vessel rupture. Chronic infiltration of inflammatory cells into the vessel wall is fundamental to aneurysm pathology. We aim to limit aneurysm growth by inhibition of inflammation and reducing endothelial cell (EC) activation with immunosuppressive drug azathioprine (Aza). APPROACH AND RESULTS: Aza and its metabolite 6-mercaptopurine have anti-inflammatory effects on leukocytes. We here demonstrate that treatment of ECs with 6-mercaptopurine inhibits cell activation as illustrated by reduced expression of interleukin-12, CCL5, CCL2, and vascular cell adhesion molecule-1 and inhibition of monocyte-EC adhesion. The underlying mechanism of 6-mercaptopurine involves suppression of GTPase Rac1 activation, resulting in reduced phosphorylation of c-Jun-terminal-N-kinase and c-Jun. Subsequently, the effect of Aza was investigated in aneurysm formation in the angiotensin II aneurysm mouse model in apolipoprotein E-deficient mice. We demonstrated that Aza decreases de novo aortic aneurysm formation from an average aneurysm severity score of 2.1 (control group) to 0.6 (Aza group), and that Aza effectively delays aorta pathology in a progression experiment, resulting in a reduced severity score from 2.8 to 1.7 in Aza-treated mice. In line with the in vitro observations, Aza-treated mice showed less c-Jun-terminal-N-kinase activation in ECs and reduced leukocyte influx in the aortic wall. CONCLUSIONS: The immunosuppressive drug Aza has an anti-inflammatory effect and in ECs inhibits Rac1 and c-Jun-terminal-N-kinase activation, which may explain the protective effect of Aza in aneurysm development and, most importantly for clinical implications, aneurysm severity.


Subject(s)
Aortic Aneurysm/prevention & control , Azathioprine/pharmacology , Endothelial Cells/drug effects , Immunosuppressive Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , rac1 GTP-Binding Protein/antagonists & inhibitors , Angiotensin II , Animals , Anti-Inflammatory Agents/pharmacology , Aortic Aneurysm/chemically induced , Aortic Aneurysm/enzymology , Aortic Aneurysm/genetics , Aortic Aneurysm/immunology , Aortic Aneurysm/pathology , Aortic Rupture/enzymology , Aortic Rupture/immunology , Aortic Rupture/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cell Line, Tumor , Coculture Techniques , Disease Models, Animal , Disease Progression , Endothelial Cells/enzymology , Endothelial Cells/immunology , Enzyme Activation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mercaptopurine/metabolism , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/enzymology , Monocytes/immunology , Neuropeptides/metabolism , Phosphorylation , Signal Transduction/drug effects , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...