Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Med Rep ; 18(1): 41-48, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749489

RESUMEN

Osteoprotegerin (OPG), additionally termed tumor necrosis factor receptor superfamily member 11B, is produced by vascular smooth muscle cells (VSMCs) and endothelial cells in the vasculature, and its release may be modulated by pro­inflammatory cytokines, including interleukin­1ß and tumor necrosis factor­α. The present study investigated the effects of treatment with low­dose human recombinant OPG on abdominal aortic aneurysm (AAA) development in mice. Mice were treated with 1 µg human recombinant OPG four times (or vehicle) for 2 weeks prior to inducing AAA. A total of two different models for inducing AAA were used to investigate the hypothesis as to whether OPG is involved in key events of AAA development, using osmotic mini­pumps with angiotensin II in apolipoprotein­E (ApoE­/­) mice for 28 days or using periaortic application of CaCl2 on the aorta in C57Bl/6J mice for 14 days. OPG was continuously administered during the experimental period. Histological staining using Masson's trichrome, Verhoeff's van­Gieson and picro­sirius red, in addition to reverse transcription­quantitative polymerase chain reaction analysis of various markers, were used to analyze phenotypic alterations. Treatment with OPG had no inhibitory effect on AAA development in the angiotensin II model in ApoE­/­ mice, which developed suprarenal aneurysms, although it increased vessel wall thickness of the aorta and total collagen in C57Bl/6J mice using the CaCl2 model that induced infrarenal dilation of the aorta. Treatment with OPG did not inhibit aneurysm development and key events, including inflammation, extracellular matrix or VSMC remodeling, in aortas from OPG­treated mice with periaortic treatment with CaCl2. The results indicated that mice treated with low levels of human recombinant OPG may have a more stable aneurysmal phenotype due to compensatory production of collagen and increased vessel wall thickness of the aorta, potentially protecting the aneurysm from rupture. Further studies investigating rupture models of AAA in addition to using higher levels of OPG are require to verify this speculation. Furthermore, treatment with low levels of OPG in patients with AAA may represent a novel therapeutic strategy for the treatment of AAA as well as attenuate the adverse effects associated with the administration of normal and high dosages of OPG.


Asunto(s)
Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Osteoprotegerina/farmacología , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Cloruro de Calcio/toxicidad , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas Recombinantes/farmacología
2.
Int J Mol Med ; 42(1): 471-478, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29658561

RESUMEN

Atherosclerosis is the main underlying causes of cardiovascular disease. There is a well­established association between high blood cholesterol levels and the extent of atherosclerosis. Furthermore, atherosclerosis has been proposed to augment abdominal aortic aneurysm (AAA) formation. As patients with AAA often have parallel atherosclerotic disease and are therefore often on cholesterol­lowering therapy, it is not possible to fully address the independent effects of plasma cholesterol lowering (PCL) treatment on AAA. The present study investigated the effect of angiotensin II (AngII)­infusion in modestly hypercholesterolemic Ldlr­/­Apob100/100Mttpflox/floxMx1­Cre mice with or without PCL treatment on a morphological and molecular level, in terms of atherosclerosis and AAA development. AngII infusion in the study mice resulted in an increased atherosclerotic lesion area and increased infiltration of inflammatory leukocytes, which was not observed in mice with PCL induced prior to AngII infusion. This suggested that AngII infusion in this mouse model induced atherosclerosis development, and that plasma cholesterol levels represent a controlling factor. Furthermore, AngII infusion in Ldlr­/­Apob100/100Mttpflox/floxMx1­Cre mice caused a modest aneurysmal phenotype, and no differences in AAA development were observed between the different study groups. However, the fact that modest hypercholesterolemic mice did not develop AAA in a classical aneurysmal model indicated that plasma cholesterol levels are important for disease development.


Asunto(s)
Aterosclerosis/sangre , Aterosclerosis/complicaciones , Colesterol/sangre , Hipercolesterolemia/sangre , Hipercolesterolemia/complicaciones , Angiotensina II , Animales , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/complicaciones , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Apolipoproteína B-100/metabolismo , Aterosclerosis/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hipercolesterolemia/genética , Integrasas/metabolismo , Masculino , Receptores de LDL/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(8): 1907-1912, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29432192

RESUMEN

Cysteinyl-leukotrienes (cys-LTs) are 5-lipoxygenase-derived lipid mediators involved in the pathogenesis and progression of inflammatory disorders, in particular asthma. We have previously found evidence linking these mediators to increased levels of proteolytic enzymes in tissue specimens of human abdominal aortic aneurysm (AAA). Here we show that antagonism of the CysLT1 receptor by montelukast, an established antiasthma drug, protects against a strong aorta dilatation (>50% increase = aneurysm) in a mouse model of CaCl2-induced AAA at a dose comparable to human medical practice. Analysis of tissue extracts revealed that montelukast reduces the levels of matrix metalloproteinase-9 (MMP-9) and macrophage inflammatory protein-1α (MIP-1α) in the aortic wall. Furthermore, aneurysm progression was specifically mediated through CysLT1 signaling since a selective CysLT2 antagonist was without effect. A significantly reduced vessel dilatation is also observed when treatment with montelukast is started days after aneurysm induction, suggesting that the drug not only prevents but also stops and possibly reverts an already ongoing degenerative process. Moreover, montelukast reduced the incidence of aortic rupture and attenuated the AAA development in two additional independent models, i.e., angiotensin II- and porcine pancreatic elastase-induced AAA, respectively. Our results indicate that cys-LTs are involved in the pathogenesis of AAA and that antagonism of the CysLT1 receptor is a promising strategy for preventive and therapeutic treatment of this clinically silent and highly lethal disease.


Asunto(s)
Acetatos/farmacología , Aneurisma de la Aorta Abdominal/prevención & control , Modelos Animales de Enfermedad , Antagonistas de Leucotrieno/farmacología , Quinolinas/farmacología , Receptores de Leucotrienos/metabolismo , Angiotensina II/administración & dosificación , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Quimiocina CCL3/metabolismo , Ciclopropanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados para ApoE , Receptores de Leucotrienos/genética , Sulfuros
4.
PLoS One ; 12(6): e0178729, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28570682

RESUMEN

INTRODUCTION: Extracellular matrix degradation is a hallmark of abdominal aortic aneurysm (AAA). Among proteases that are capable of degrading extracellular matrix are a disintegrin and metalloproteases with thrombospondin motifs (ADAMTS). Pathogenesis of these proteases in AAA has not been investigated until date. METHODS AND RESULTS: Human aneurysmal and control aortas were collected and analyzed with RT-PCR measuring the ADAMTS-1, 4,5,6,8,9,10,13,17 and ADAMTSL-1. Expression of a majority of the investigated ADAMTS members on mRNA level was decreased in aneurysm compared to control aorta. ADAMTS-1 was one of the members that was reduced most. Protein analysis using immunohistochemistry and western blot for localization and expression of ADAMTS-1 revealed that ADAMTS-1 was present predominantly in areas of SMCs and macrophages in aneurysmal aorta and higher expressed in AAA compared to control aortas. The role of ADAMTS-1 in AAA disease was further examined using ADAMTS-1 transgenic/apoE-/- mice with the experimental angiotensin II induced aneurysmal model. Transgenic mice overexpressing ADAMTS-1 showed to be similar to ADAMTS-1 wild type mice pertaining collagen, elastin content and aortic diameter. CONCLUSION: Several of the ADAMTS members, and especially ADAMTS-1, are down regulated at mRNA level in AAA, due to unknown mechanisms, at the same time ADAMTS-1 protein is induced. The cleavage of its substrates, don't seem to be crucial for the pathogenesis of AAA but rather more important in the development of thoracic aortic aneurysm and atherosclerosis as shown in previous studies.


Asunto(s)
Proteína ADAMTS1/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Animales , Humanos , Masculino , Ratones
5.
J Vasc Surg ; 65(4): 1171-1179.e1, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26960947

RESUMEN

BACKGROUND: Abdominal aortic aneurysm (AAA) is a deadly irreversible weakening and distension of the abdominal aortic wall. The pathogenesis of AAA remains poorly understood. Investigation into the physical and molecular characteristics of perivascular adipose tissue (PVAT) adjacent to AAA has not been done before and is the purpose of this study. METHODS AND RESULTS: Human aortae, periaortic PVAT, and fat surrounding peripheral arteries were collected from patients undergoing elective surgical repair of AAA. Control aortas were obtained from recently deceased healthy organ donors with no known arterial disease. Aorta and PVAT was found in AAA to larger extent compared with control aortas. Immunohistochemistry revealed neutrophils, macrophages, mast cells, and T-cells surrounding necrotic adipocytes. Gene expression analysis showed that neutrophils, mast cells, and T-cells were found to be increased in PVAT compared with AAA as well as cathepsin K and S. The concentration of ceramides in PVAT was determined using mass spectrometry and correlated with content of T-cells in the PVAT. CONCLUSIONS: Our results suggest a role for abnormal necrotic, inflamed, proteolytic adipose tissue to the adjacent aneurysmal aortic wall in ongoing vascular damage.


Asunto(s)
Adipocitos/enzimología , Tejido Adiposo/enzimología , Aneurisma de la Aorta Abdominal/enzimología , Ceramidas/análisis , Mastocitos/enzimología , Neutrófilos/enzimología , Péptido Hidrolasas/análisis , Linfocitos T/enzimología , Adipocitos/inmunología , Adipocitos/patología , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Antígenos CD/análisis , Antígenos CD/genética , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/genética , Citocinas/análisis , Citocinas/genética , Proteínas Ligadas a GPI/análisis , Proteínas Ligadas a GPI/genética , Regulación Enzimológica de la Expresión Génica , Humanos , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/patología , Mastocitos/inmunología , Mastocitos/patología , Necrosis , Neutrófilos/inmunología , Neutrófilos/patología , Péptido Hidrolasas/genética , Linfocitos T/inmunología , Linfocitos T/patología
6.
Atherosclerosis ; 256: 82-88, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27993388

RESUMEN

BACKGROUND AND AIMS: MicroRNA (miR) are important regulators of gene expression and biological processes and have recently been suggested as possible biomarkers for abdominal aortic aneurysm (AAA) disease. The aim of the present study was to assess the role of miR as biomarkers for initiation and progression of AAA disease, through evaluation of a wide range of miRs in a large population-based cohort, with AAA patients with linked clinical data regarding risk factors, AAA size and growth, as well as controls. METHODS: The expression of the 172 most commonly expressed miRs in plasma was analyzed by real-time PCR in samples from 169 screening-detected AAA patients and 48 age-matched controls. RESULTS: For 103 miRs, there was a significant difference in expression between AAA and controls. Of these, 20 miRs were differently expressed between fast and slow growing aneurysms. These miRs target genes known to be involved in AAA disease as well as novel genes and pathways. By combining the top altered miRs together with clinical variables, strong predictive values, determining growth of AAA, were obtained (area under curve = 0.86, p < 0.001). CONCLUSIONS: This large cohort study identified several novel miRs with altered expression in AAA patients when compared to controls. Assessment of miR expression may offer an opportunity to predict disease progression and aneurysm growth.


Asunto(s)
Aneurisma de la Aorta Abdominal/sangre , MicroARNs/sangre , Anciano , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/epidemiología , Aneurisma de la Aorta Abdominal/genética , Área Bajo la Curva , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Marcadores Genéticos , Humanos , Masculino , MicroARNs/genética , Valor Predictivo de las Pruebas , Prevalencia , Curva ROC , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suecia/epidemiología , Factores de Tiempo
7.
Sci Rep ; 6: 31414, 2016 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-27659201

RESUMEN

Abdominal aortic aneurysm (AAA) is a degenerative disease characterized by aortic dilation and rupture leading to sudden death. Currently, no non-surgical treatments are available and novel therapeutic targets are needed to prevent AAA. We investigated whether increasing plasma levels of adiponectin (APN), a pleiotropic adipokine, provides therapeutic benefit to prevent AngII-induced advanced AAA in a well-established preclinical model. In the AngII-infused hyperlipidemic low-density lipoprotein receptor-deficient mouse (LDLR-/-) model, we induced plasma APN levels using a recombinant adenovirus expressing mouse APN (AdAPN) and as control, adenovirus expressing green florescent protein (AdGFP). APN expression produced sustained and significant elevation of total and high-molecular weight APN levels and enhanced APN localization in the artery wall. AngII infusion for 8 weeks induced advanced AAA development in AdGFP mice. Remarkably, APN inhibited the AAA development in AdAPN mice by suppressing aortic inflammatory cell infiltration, medial degeneration and elastin fragmentation. APN inhibited the angiotensin type-1 receptor (AT1R), inflammatory cytokine and mast cell protease expression, and induced lysyl oxidase (LOX) in the aortic wall, improved systemic cytokine profile and attenuated adipose inflammation. These studies strongly support APN therapeutic actions through multiple mechanisms inhibiting AngII-induced AAA and increasing plasma APN levels as a strategy to prevent advanced AAA.

8.
J Vasc Res ; 53(1-2): 17-26, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27344146

RESUMEN

INTRODUCTION: Abdominal aortic aneurysm (AAA) is a complex and deadly vascular disorder. The pathogenesis of AAA includes destruction and phenotypic alterations of the vascular smooth muscle cells (VSMCs) and aortic tissues. PPARγ coactivator-1 alpha (PGC1α) regulates VSMC migration and matrix formation and is a major inducer of mitochondrial biogenesis and function, including oxidative metabolism. METHODS: Protein and gene expression of PGC1α and markers for mitochondria biogenesis and cell type-specificity were analysed in AAA aortas from humans and mice and compared against control aortas. RESULTS: Gene expression of PPARGC1A was decreased in human AAA and angiotensin (Ang) II-induced AAA in mice when compared to control vessels. However, high expression of PGC1α was detected in regions of neovascularisation in the adventitia layer. In contrast, the intima/media layer of AAA vessel exhibited defective mitochondrial biogenesis as indicated by low expression of PPARGC1A, VDAC, ATP synthase and citrate synthase. CONCLUSION: Our results suggest that mitochondrial biogenesis is impaired in AAA in synthetic SMCs in the media, with the exception of newly formed supporting vessels in the adventitia where the mitochondrial markers seem to be intact. To our knowledge, this is the first study investigating PGC1α and mitochondria biogenesis in AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Mitocondrias/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Angiotensina II , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Citrato (si)-Sintasa/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Ratones Noqueados , Mitocondrias/patología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Neovascularización Patológica , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Canales Aniónicos Dependientes del Voltaje/metabolismo
9.
Atherosclerosis ; 249: 101-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27085160

RESUMEN

BACKGROUND: Abdominal aortic aneurysm (AAA) is characterized by vascular remodeling with increased infiltration of inflammatory cells and apoptosis/modulation of vascular smooth muscle cells (SMCs). Imatinib is a selective inhibitor of several tyrosine kinases, including PDGF receptors, Abl, and c-kit. The objective of this study was to characterize the potential protective role of imatinib on AAA development and the molecular mechanisms involved. METHODS: Male ApoE(-/-) mice were infused with angiotensin (Ang) II (1000 ng/kg/min) for 4 weeks to induce AAA or saline as controls. Daily treatment with 10 mg/kg imatinib, or tap water as control, was provided via gavage for 4 weeks. RESULTS: Treatment with imatinib was found to decrease the aortic diameter and vessel wall thickness, mediated by multiple effects. Imatinib treatment in AngII infused mice resulted in a reduced cellular infiltration of CD3ε positive T lymphocytes by 86% and reduced gene expression of mast cell chymase by 50% compared with AngII infused mice lacking imatinib. Gene expression analysis of SMC marker SM22α demonstrated an increase by 48% together with a more intact medial layer after treatment with imatinib as evaluated with SM22α immunostaining. CONCLUSION: Present findings highlight the importance of tyrosine kinase pathways in the development of AAA. Our results show, that imatinib treatment inhibits essential mast cell, T lymphocyte and SMC mediated processes in experimental AAA. Thus, our results support the idea that tyrosine kinase inhibitors may be useful in the treatment of pathological vascular inflammation and remodeling in conditions like AAA.


Asunto(s)
Angiotensina II/efectos adversos , Aorta/patología , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Mesilato de Imatinib/uso terapéutico , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Apolipoproteínas E/genética , Apoptosis , Complejo CD3/metabolismo , Quimasas/metabolismo , Humanos , Inflamación , Masculino , Mastocitos/enzimología , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
10.
Pharmacol Rep ; 68(1): 1-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26721343

RESUMEN

BACKGROUND: Imatinib mesylate (Glivec®, formerly STI-571) is a selective tyrosine kinase inhibitor used for the treatment of chronic myeloid leukemia and gastrointestinal stromal tumors. However, there are reports suggesting that imatinib could be atheroprotective by lowering plasma low-density lipoprotein (LDL). AIM: To investigate the potential inhibitory effect of imatinib on cholesterol uptake in human macrophages as well as its effect on matrix metalloproteinase (MMP) activity. METHODS AND RESULTS: Uptake of fluorescence-labeled LDL was analyzed using flow cytometry. Macrophages treated with imatinib showed a 23.5%, 27%, and 15% decrease in uptake of native LDL (p<0.05), acetylated LDL (p<0.01), and copper-modified oxidized LDL (p<0.01), respectively. Gel-based zymography showed that secretion and activity of MMP-2 and MMP-9 were inhibited by imatinib. Using GeneChip Whole Transcript Expression array analysis, no obvious gene candidates involved in the mechanisms of cholesterol metabolism or MMP regulation were found to be affected by imatinib. Instead, we found that imatinib up-regulated microRNA 155 (miR155) by 43.8% and down-regulated ADAM metallopeptidase domain 28 (ADAM28) by 41.4%. Both genes could potentially play an atheroprotective role and would be interesting targets in future studies. CONCLUSION: Our results indicate that imatinib causes post-translational inhibition with respect to cholesterol uptake and regulation of MMP-2 and MMP-9. More research is needed to further evaluate the role of imatinib in the regulation of other genes and processes.


Asunto(s)
Colesterol/metabolismo , Mesilato de Imatinib/farmacología , Macrófagos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Macrófagos/efectos de los fármacos
11.
Atherosclerosis ; 241(2): 561-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26100679

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is characterized by inflammation, loss of smooth muscle cells (SMCs), and degradation of the extracellular matrix in the vessel wall. Innate immune receptors such as Toll-like receptors (TLRs) were recently shown to regulate immunological processes leading to the formation and progression of atherosclerotic plaques as well as to other cardiovascular pathologies. Our aim was to investigate whether blockage of TLR signaling, under the control of TIR domain-containing adaptor protein including IFN-ß (TRIF), could inhibit the inflammatory response and AAA development in mice. RESULTS: In human AAA, an increased TLR3 and TLR4 expression in association with macrophages and T lymphocytes was demonstrated with immunohistochemical analysis. Angiotensin (Ang) II-induced aneurysm formation was significantly reduced by 30% in ApoE(-/-)Trif(-/-) mice compared to ApoE(-/-) mice. Morphologically, AngII-infused ApoE(-/-)Trif(-/-) mice had a more intact cellular and extracellular matrix while ApoE(-/-) mice infused with AngII displayed an increased medial thickness associated with aortic dissection, thrombus formation, and a more disorganized vessel wall. Gene expression analysis of the abdominal aorta revealed a profound decrease of the inflammatory genes CD68 (P < 0.05), CD11b (P < 0.05), and TNF-α (P < 0.05) and the protease gene MMP-12 (P < 0.01) in ApoE(-/-)Trif(-/-) mice compared to ApoE(-/-) mice infused with AngII. CONCLUSION: Our results suggest that signaling through TRIF is important for the inflammatory response of AngII-induced AAA and that blockage of the TRIF pathway reduces vascular inflammation and protects against AAA formation.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Regulación de la Expresión Génica , Proteínas Adaptadoras del Transporte Vesicular/genética , Angiotensina II/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Aorta/metabolismo , Aorta/patología , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Complejo CD3/metabolismo , Elastina/metabolismo , Humanos , Inflamación , Ratones , Ratones Noqueados , Transducción de Señal , Receptores Toll-Like/metabolismo
12.
BMC Med Genet ; 16: 37, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-26068642

RESUMEN

BACKGROUND: It is important to identify cardiovascular diseases in patients at high risk. To include genetics into routine cardiological patients has therefore been discussed recently. We wanted to evaluate the association between high-molecular weight adiponectin and cardiovascular risk, and secondly in the same population evaluate if specific genotype differences regarding risk could be observed, and thirdly if gender differences could be seen. METHOD: Four hundred seventy-six elderly participants recruited from a rural community were included. All participants underwent a clinical examination, echocardiography, and blood sampling and the single nucleotide polymorphism (SNP) (rs266729) of adiponectin was analysed. Follow-up time was 6.7 years. RESULTS: Those with high serum concentration of adiponectin had a more 2 fold increased cardiovascular risk, and it might be that females exhibits even higher risk where a more than 5 fold increased risk could be seen. The result could be demonstrated even in a multivariate model adjusting for well-known clinical risk factors. However, as the sample size was small the gender differences should be interpreted with caution. In the genotype evaluation the C/C carriers of the female group had a more than 9-fold increased risk of cardiovascular mortality, however the confidence interval was wide. Such genotype difference could not be found in the male group. CONCLUSION: High level of adiponectin was associated with increased cardiovascular risk. Also a gender difference in the genotype evaluation could be seen where the C/C carriers obtained higher risk in the female group but not in the male group. Thus, in order to identify patients at risk early, genetic analyses may add to the armamentarium used in the clinical routine. However, information should be regarded as hypothesis generating as the sample size was small and should stimulate further research in individualized cardiovascular prevention and treatment.


Asunto(s)
Adiponectina/genética , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/mortalidad , Polimorfismo de Nucleótido Simple , Caracteres Sexuales , Adiponectina/química , Adiponectina/metabolismo , Anciano , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/fisiopatología , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Masculino , Peso Molecular , Población Rural
13.
Int J Colorectal Dis ; 30(7): 883-90, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26008729

RESUMEN

PURPOSE: Cluster of differentiation 93 (CD93) is involved in apoptosis and inflammation and has a suggested role in angiogenesis, and all of which are involved in the development and dissemination of cancer. We evaluated the expression of CD93 and the association with two single nucleotide polymorphisms (SNPs), rs2749812 and rs2749817, as possible biomarkers in colorectal cancer (CRC). METHODS: Tissue levels and plasma levels of CD93 were measured using an enzyme-linked immunosorbent assay (ELISA). Expression of CD93 was determined by immunohistochemistry, western blot and gene expression analysis. Genotype frequencies were established for the SNPs by real-time polymerase chain reaction (PCR), and the association with tumour stage and survival was analysed. RESULTS: Total CD93 levels were 82% higher (P < 0.001) in tumours compared to matched normal tissues. Mean levels of soluble CD93 in plasma were 30% lower (P < 0.001) in the patients compared to the controls. The T/T genotype of SNP rs2749817 was more common in stage IV patients, with consequently higher risk of CRC death (T/T vs. C/C and C/T; hazard ratio (HR) = 1.73, 95% confidence interval (CI) = 1.11-2.67, P = 0.014), and was associated with a higher risk of CRC recurrence after radical operation (T/T vs. C/C and C/T; HR = 2.07, CI = 1.22-3.51, P = 0.007). CONCLUSIONS: We showed that the T/T genotype of SNP rs2749817 is associated with disseminated cancer at diagnosis and an increased recurrence rate after radical operation. Patients with this genotype may benefit from early identification.


Asunto(s)
Neoplasias Colorrectales/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glicoproteínas de Membrana/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Complemento/genética , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Estudios de Casos y Controles , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Frecuencia de los Genes/genética , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Glicoproteínas de Membrana/sangre , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Complemento/sangre , Solubilidad
14.
Circ Res ; 116(4): 642-52, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25556206

RESUMEN

RATIONALE: In human genetic studies a single nucleotide polymorphism within the salt-inducible kinase 1 (SIK1) gene was associated with hypertension. Lower SIK1 activity in vascular smooth muscle cells (VSMCs) leads to decreased sodium-potassium ATPase activity, which associates with increased vascular tone. Also, SIK1 participates in a negative feedback mechanism on the transforming growth factor-ß1 signaling and downregulation of SIK1 induces the expression of extracellular matrix remodeling genes. OBJECTIVE: To evaluate whether reduced expression/activity of SIK1 alone or in combination with elevated salt intake could modify the structure and function of the vasculature, leading to higher blood pressure. METHODS AND RESULTS: SIK1 knockout (sik1(-/-)) and wild-type (sik1(+/+)) mice were challenged to a normal- or chronic high-salt intake (1% NaCl). Under normal-salt conditions, the sik1(-/-) mice showed increased collagen deposition in the aorta but similar blood pressure compared with the sik1(+/+) mice. During high-salt intake, the sik1(+/+) mice exhibited an increase in SIK1 expression in the VSMCs layer of the aorta, whereas the sik1(-/-) mice exhibited upregulated transforming growth factor-ß1 signaling and increased expression of endothelin-1 and genes involved in VSMC contraction, higher systolic blood pressure, and signs of cardiac hypertrophy. In vitro knockdown of SIK1 induced upregulation of collagen in aortic adventitial fibroblasts and enhanced the expression of contractile markers and of endothelin-1 in VSMCs. CONCLUSIONS: Vascular SIK1 activation might represent a novel mechanism involved in the prevention of high blood pressure development triggered by high-salt intake through the modulation of the contractile phenotype of VSMCs via transforming growth factor-ß1-signaling inhibition.


Asunto(s)
Aorta/enzimología , Presión Arterial , Hipertensión/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Remodelación Vascular , Adventicia/enzimología , Adventicia/patología , Animales , Aorta/patología , Aorta/fisiopatología , Células Cultivadas , Colágeno/metabolismo , Endotelina-1/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Genotipo , Humanos , Hipertensión/etiología , Hipertensión/genética , Hipertensión/patología , Hipertensión/fisiopatología , Ratones Noqueados , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Natriuresis , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal , Cloruro de Sodio Dietético , Sistema Nervioso Simpático/fisiopatología , Transfección , Factor de Crecimiento Transformador beta1/metabolismo , Vasoconstricción
15.
Int J Mol Med ; 30(2): 288-94, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22580763

RESUMEN

Remodeling of extracellular matrix (ECM) plays an important role in both atherosclerosis and aneurysm disease. Serine protease inhibitor A3 (serpinA3) is an inhibitor of several proteases such as elastase, cathepsin G and chymase derived from mast cells and neutrophils. In this study, we investigated the putative role of serpinA3 in atherosclerosis and aneurysm formation. SerpinA3 was expressed in endothelial cells and medial smooth muscle cells in human atherosclerotic lesions and a 14-fold increased expression of serpinA3n mRNA was found in lesions from Apoe-/- mice compared to lesion-free vessels. In contrast, decreased mRNA expression (-80%) of serpinA3 was found in biopsies of human abdominal aortic aneurysm (AAA) compared to non-dilated aortas. Overexpression of serpinA3n in transgenic mice did not influence the development of atherosclerosis or CaCl2-induced aneurysm formation. In situ zymography analysis showed that the transgenic mice had lower cathepsin G and elastase activity, and more elastin in the aortas compared to wild-type mice, which could indicate a more stable aortic phenotype. Differential vascular expression of serpinA3 is clearly associated with human atherosclerosis and AAA but serpinA3 had no major effect on experimentally induced atherosclerosis or AAA development in mouse. However, serpinA3 may be involved in a phenotypic stabilization of the aorta.


Asunto(s)
Aneurisma/metabolismo , Aterosclerosis/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , alfa 1-Antiquimotripsina/metabolismo , Aneurisma/genética , Animales , Aterosclerosis/genética , Cloruro de Calcio/farmacología , Catepsina G/metabolismo , Línea Celular , Citocinas/farmacología , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mediadores de Inflamación/farmacología , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Elastasa Pancreática/metabolismo , ARN Mensajero/metabolismo , alfa 1-Antiquimotripsina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...