Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
BMJ Open ; 13(7): e072040, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37451717

ABSTRACT

INTRODUCTION: Prevention of cardiovascular disease (CVD) is of key importance in reducing morbidity, disability and mortality worldwide. Observational studies suggest that digital health interventions can be an effective strategy to reduce cardiovascular (CV) risk. However, evidence from large randomised clinical trials is lacking. METHODS AND ANALYSIS: The CV-PREVITAL study is a multicentre, prospective, randomised, controlled, open-label interventional trial designed to compare the effectiveness of an educational and motivational mobile health (mHealth) intervention versus usual care in reducing CV risk. The intervention aims at improving diet, physical activity, sleep quality, psycho-behavioural aspects, as well as promoting smoking cessation and adherence to pharmacological treatment for CV risk factors. The trial aims to enrol approximately 80 000 subjects without overt CVDs referring to general practitioners' offices, community pharmacies or clinics of Scientific Institute for Research, Hospitalization and Health Care (Italian acronym IRCCS) affiliated with the Italian Cardiology Network. All participants are evaluated at baseline and after 12 months to assess the effectiveness of the intervention on short-term endpoints, namely improvement in CV risk score and reduction of major CV risk factors. Beyond the funded life of the study, a long-term (7 years) follow-up is also planned to assess the effectiveness of the intervention on the incidence of major adverse CV events. A series of ancillary studies designed to evaluate the effect of the mHealth intervention on additional risk biomarkers are also performed. ETHICS AND DISSEMINATION: This study received ethics approval from the ethics committee of the coordinating centre (Monzino Cardiology Center; R1256/20-CCM 1319) and from all other relevant IRBs and ethics committees. Findings are disseminated through scientific meetings and peer-reviewed journals and via social media. Partners are informed about the study's course and findings through regular meetings. TRIAL REGISTRATION NUMBER: NCT05339841.


Subject(s)
Cardiovascular Diseases , Humans , Prospective Studies , Cardiovascular Diseases/prevention & control , Diet , Exercise
2.
Antioxidants (Basel) ; 12(3)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36978789

ABSTRACT

The process of adipogenesis involves the differentiation of preadipocytes into mature adipocytes. Excessive adipogenesis promotes obesity, a condition that increasingly threatens global health and contributes to the rapid rise of obesity-related diseases. We have recently shown that prenylcysteine oxidase 1 (PCYOX1) is a regulator of atherosclerosis-disease mechanisms, which acts through mechanisms not exclusively related to its pro-oxidant activity. To address the role of PCYOX1 in the adipogenic process, we extended our previous observations confirming that Pcyox1-/-/Apoe-/- mice fed a high-fat diet for 8 or 12 weeks showed significantly lower body weight, when compared to Pcyox1+/+/Apoe-/- mice, due to an evident reduction in visceral adipose content. We herein assessed the role of PCYOX1 in adipogenesis. Here, we found that PCYOX1 is expressed in adipose tissue, and, independently from its pro-oxidant enzymatic activity, is critical for adipogenesis. Pcyox1 gene silencing completely prevented the differentiation of 3T3-L1 preadipocytes, by acting as an upstream regulator of several key players, such as FABP4, PPARγ, C/EBPα. Proteomic analysis, performed by quantitative label-free mass spectrometry, further strengthened the role of PCYOX1 in adipogenesis by expanding the list of its downstream targets. Finally, the absence of Pcyox1 reduces the inflammatory markers in adipose tissue. These findings render PCYOX1 a novel adipogenic factor with possible pathophysiological or therapeutic potential.

3.
Int J Mol Sci ; 21(12)2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32630608

ABSTRACT

Platelets are a heterogeneous small anucleate blood cell population with a central role both in physiological haemostasis and in pathological states, spanning from thrombosis to inflammation, and cancer. Recent advances in proteomic studies provided additional important information concerning the platelet biology and the response of platelets to several pathophysiological pathways. Platelets circulate systemically and can be easily isolated from human samples, making proteomic application very interesting for characterizing the complexity of platelet functions in health and disease as well as for identifying and quantifying potential platelet proteins as biomarkers and novel antiplatelet therapeutic targets. To date, the highly dynamic protein content of platelets has been studied in resting and activated platelets, and several subproteomes have been characterized including platelet-derived microparticles, platelet granules, platelet releasates, platelet membrane proteins, and specific platelet post-translational modifications. In this review, a critical overview is provided on principal platelet proteomic studies focused on platelet biology from signaling to granules content, platelet proteome changes in several diseases, and the impact of drugs on platelet functions. Moreover, recent advances in quantitative platelet proteomics are discussed, emphasizing the importance of targeted quantification methods for more precise, robust and accurate quantification of selected proteins, which might be used as biomarkers for disease diagnosis, prognosis and therapy, and their strong clinical impact in the near future.


Subject(s)
Blood Platelets/metabolism , Blood Platelets/physiology , Biomarkers/metabolism , Humans , Platelet Activation , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Protein Processing, Post-Translational , Proteome/metabolism , Proteomics/methods , Signal Transduction
4.
Arterioscler Thromb Vasc Biol ; 39(7): 1330-1342, 2019 07.
Article in English | MEDLINE | ID: mdl-31092014

ABSTRACT

Clinical and preclinical studies over the past 3 decades have uncovered a multitude of signaling pathways involved in the initiation and progression of atherosclerosis. From these studies, signaling by proteins of the Wnt family has recently emerged as an important player in the development of atherosclerosis. Wnt signaling is characterized by a large number of ligands, receptors, and coreceptors and can be regulated at many different levels. Among Wnt modulators, the evolutionary conserved Dkk (Dickkopf) proteins, and especially Dkk-1, the founding member of the family, are the best characterized. The role of Dkks in the pathophysiology of the arterial wall is only partially understood, but their involvement in atherosclerosis is becoming increasingly evident. This review introduces recent key findings on Dkk proteins and their functions in atherosclerosis and discusses the potential importance of modulating Dkk signaling as part of a novel, improved strategy for preventing and treating atherosclerosis-related diseases. Visual Overview- An online visual overview is available for this article.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Atherosclerosis/etiology , Intercellular Signaling Peptides and Proteins/physiology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Apolipoproteins E/physiology , Atherosclerosis/drug therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Vascular Calcification/etiology , Wnt Signaling Pathway/physiology , beta Catenin/physiology
5.
Sci Rep ; 8(1): 16671, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420710

ABSTRACT

This study shows that DKK-1, a member of the Dickkopf family and a regulator of the Wnt pathways, represents a novel target of statins which, through the inhibition of HMG-CoA reductase and of non-steroidal isoprenoid intermediates, exert extra-beneficial effect in preventing atherosclerosis beyond their effect on the lipid profile. We found that atorvastatin downregulates DKK-1 protein (-88.3 ± 4.1%) and mRNA expression (-90 ± 4.2%) through the inhibition of Cdc42, Rho and Rac geranylgeranylated proteins. Further, a combined approach based on the integration of label-free quantitative mass spectrometry based-proteomics and gene silencing allowed us to demonstrate that DKK-1 itself mediates, at least in part, statin effects on human endothelial cells. Indeed, DKK-1 is responsible for the regulation of the 21% of the statin-modulated proteins, which include, among others, clusterin/apoJ, plasminogen activator inhibitor type 1 (PAI-1), myristoylated alanine-rich C-kinase substrate (MARCKS), and pentraxin 3 (PTX3). The Gene Ontology enrichment annotation revealed that DKK-1 is also a potential mediator of the extracellular matrix organization, platelet activation and response to wounding processes induced by statin. Finally, we found that plasma level of DKK-1 from cholesterol-fed rabbits treated with atorvastatin (2.5 mg/kg/day for 8 weeks) was lower (-42 ± 23%) than that of control animals. Thus, DKK-1 is not only a target of statin but it directly regulates the expression of molecules involved in a plethora of biological functions, thus expanding its role, which has been so far restricted mainly to cancer.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Atorvastatin/pharmacology , C-Reactive Protein/metabolism , Cell Line , Cholesterol/pharmacology , Gene Ontology , Human Umbilical Vein Endothelial Cells , Humans , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Platelet Activation/drug effects , Prenylation/drug effects , Proteomics , Rabbits , Serum Amyloid P-Component/metabolism
6.
J Proteomics ; 178: 57-72, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29622522

ABSTRACT

Cardiovascular diseases (CVDs) represent the most important cause of mortality in women and in men. Contrary to the long-standing notion that the effects of the major risk factors on CVD outcomes are the same in both sexes, recent evidence recognizes new, potentially independent, sex/gender-related risk factors for CVDs, and sex/gender-differences in the clinical presentation of CVDs have been demonstrated. Furthermore, some therapeutic options may not be equally effective and safe in men and women. In this context, proteomics offers an extremely useful and versatile analytical platform for biomedical researches that expand from the screening of early diagnostic and prognostic biomarkers to the investigation of the molecular mechanisms underlying CDVs. In this review, we summarized the current applications of proteomics in the cardiovascular field, with emphasis on sex and gender-related differences in CVDs. SIGNIFICANCE: Increasing evidence supports the profound effect of sex and gender on cardiovascular physio-pathology and the response to drugs. A clear understanding of the mechanisms underlying sexual dimorphisms in CVDs would not only improve our knowledge of the etiology of these diseases, but could also inform health policy makers and guideline committees in tailoring specific interventions for the prevention, treatment and management of CVDs in both men and women.

7.
J Proteomics ; 173: 62-76, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29180046

ABSTRACT

Cardiovascular diseases (CVDs) represent the most important cause of mortality in women and in men. Contrary to the long-standing notion that the effects of the major risk factors on CVD outcomes are the same in both sexes, recent evidence recognizes new, potentially independent, sex/gender-related risk factors for CVDs, and sex/gender-differences in the clinical presentation of CVDs have been demonstrated. Furthermore, some therapeutic options may not be equally effective and safe in men and women. In this context, proteomics offers an extremely useful and versatile analytical platform for biomedical researches that expand from the screening of early diagnostic and prognostic biomarkers to the investigation of the molecular mechanisms underlying CDVs. In this review, we summarized the current applications of proteomics in the cardiovascular field, with emphasis on sex and gender-related differences in CVDs. SIGNIFICANCE: Increasing evidence supports the profound effect of sex and gender on cardiovascular physio-pathology and the response to drugs. A clear understanding of the mechanisms underlying sexual dimorphisms in CVDs would not only improve our knowledge of the etiology of these diseases, but could also inform health policy makers and guideline committees in tailoring specific interventions for the prevention, treatment and management of CVDs in both men and women.


Subject(s)
Cardiovascular Diseases , Proteomics/methods , Sex Factors , Female , Humans , Male , Precision Medicine/methods , Risk Factors
8.
Drug Discov Today ; 22(6): 848-869, 2017 06.
Article in English | MEDLINE | ID: mdl-28284830

ABSTRACT

Proteomic-based techniques provide a powerful tool for identifying the full spectrum of protein targets of a drug, elucidating its mechanism(s) of action, and identifying biomarkers of its efficacy and safety. Herein, we outline the technological advancements in the field, and illustrate the contribution of proteomics to the definition of the pharmacological profile of statins, which represent the cornerstone of the prevention and treatment of cardiovascular diseases (CVDs). Statins act by inhibiting 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase, thus reducing cholesterol biosynthesis and consequently enhancing the clearance of low-density lipoproteins from the blood; however, HMG-CoA reductase inhibition can result in a multitude of additional effects beyond lipid lowering, known as 'pleiotropic effects'. The case of statins highlights the unique contribution of proteomics to the target profiling of a drug molecule.


Subject(s)
Drug Discovery , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Proteomics , Animals , Biomarkers , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Molecular Targeted Therapy , Treatment Outcome
9.
Atherosclerosis ; 253: 214-224, 2016 10.
Article in English | MEDLINE | ID: mdl-27477186

ABSTRACT

BACKGROUND AND AIMS: Proprotein convertase subtilisin kexin type 9 (PCSK9) induces degradation of the low-density lipoprotein-receptor (LDLR). Smooth muscle cells (SMCs) in human atherosclerotic plaques and cultured SMCs express PCSK9. The present study aimed at defining the role of PCSK9 on vascular response to injury. METHODS: Carotid neointimal lesions were induced by positioning a non-occlusive collar in PCSK9 knock-out (PCSK9-/-) and wild type littermate (PCSK9+/+) mice. RESULTS: In PCSK9-/- mice, we observed a significantly less intimal thickening (p < 0.05), a lower intimal media ratio (p < 0.02), and a tendency to higher lumen area, compared to PCSK9+/+ mice. When compared with PCSK9-/-, lesions of PCSK9+/+ mice had a higher content of SMCs (p < 0.05) and collagen (p < 0.05), while no difference was observed in the accumulation of macrophages. PCSK9 was detectable in both left and right carotids artery in regions occupied by medial and neointimal SMCs. SMCs freshly isolated from PCSK9-/-, when compared to PCSK9+/+ cells, showed higher levels of α-smooth muscle actin (α-SMA; 2.24 ± 0.36 fold; p < 0.01) and myosin heavy chain II (MHC-II; 8.65 ± 1.55 fold; p < 0.01), and lower levels of caldesmon mRNA(-54 ± 14%; p < 0.01). PCSK9-/- cells also showed a slower proliferation rate, and an impaired migratory capacity and G1/S progression of the cell cycle. The reconstitution of PCSK9 expression, by retroviral infection of PCSK9-/- SMCs, led to a downregulation of α-SMA (-56 ± 2%; p < 0.01), MHC-II (-45% ± 25.5 fold: p = 0.06) and calponin (-25% ± 0.8 fold: p < 0.05) and induction of caldesmon mRNA (1.46 ± 0.3 fold; p < 0.05). Proliferation rate of SMCs PCSK9-/- was significantly lower compared to PCSK9 reconstituted cells. CONCLUSIONS: Taken together, the present results suggest that PCSK9, by sustaining SMC synthetic phenotype, proliferation, and migration, may play a pro-atherogenic role in the arterial wall.


Subject(s)
Carotid Arteries/pathology , Carotid Artery Injuries/pathology , Neointima/pathology , Proprotein Convertase 9/genetics , Proprotein Convertase 9/physiology , Animals , Aorta/metabolism , Becaplermin , Cell Differentiation , Cell Movement , Cell Proliferation , Chemotaxis , Cholesterol, LDL/metabolism , Cytoskeleton/metabolism , Female , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Phenotype , Proto-Oncogene Proteins c-sis/metabolism , Tunica Intima/pathology
10.
Electrophoresis ; 37(20): 2633-2643, 2016 10.
Article in English | MEDLINE | ID: mdl-27450324

ABSTRACT

The mitral valve is a highly complex structure which regulates blood flow from the left atrium to the left ventricle (LV) avoiding a significant forward gradient during diastole or regurgitation during systole. The integrity of the mitral valve is also essential for the maintenance of normal LV size, geometry, and function. Significant advances in the comprehension of the biological, functional, and mechanical behavior of the mitral valve have recently been made. However, current knowledge of protein components in the normal human mitral valve is still limited and complicated by the low cellularity of this tissue and the presence of high abundant proteins from the extracellular matrix. We employed here an integrated proteomic approach to analyse the protein composition of the normal human mitral valve and reported confident identification of 422 proteins, some of which have not been previously described in this tissue. In particular, we described the ability of pre-MS separation technique based on liquid-phase IEF and SDS-PAGE to identify the largest number of proteins. We also demonstrated that some of these proteins, e.g. αB-Crystallin, septin-11, four-and-a-half LIM domains protein 1, and dermatopontin, are synthesised by interstitial cells isolated from human mitral valves. These initial results provide a valuable basis for future studies aimed at analysing in depth the mitral valve protein composition and at investigating potential pathogenetic molecular mechanisms. Data are available via ProteomeXchange with identifier PXD004397.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Mitral Valve/chemistry , Proteome/analysis , Proteomics/methods , Adult , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Proteome/chemistry
11.
Vascul Pharmacol ; 67-69: 38-47, 2015.
Article in English | MEDLINE | ID: mdl-25849951

ABSTRACT

BACKGROUND: The long pentraxin PTX3 is an acute-phase multi-functional protein that might play both positive and detrimental effects under different pathophysiological conditions. We previously showed that statins down-regulate the release of PTX3 in human endothelial cells (ECs). The present study investigated the mechanism mediating this effect, its occurrence in other cells involved in atherogenesis, and whether it takes place in experimental atherosclerosis. METHODS AND RESULTS: We found that atorvastatin (1-5 µmol/L) decreased the production and release of PTX3 in human ECs through a post-transcriptional effect. Co-incubation with mevalonate or geranylgeranyl pyrophosphate prevented this effect. Direct blockade of geranylgeranyl transferase I by GGTI-286, treatment with the Rac inhibitor NSC23766 or silencing of the geranylgeranylated GTPase Rac2 by siRNA closely mimicked the action of atorvastatin. In contrast, inactivation of other geranylgeranylated proteins such as RhoA, RhoB, and RhoC or Rac1 did not affect PTX3 release. In addition, we found that atorvastatin also decreased PTX3 secretion in aortic SMCs through a mechanism likely dependent on protein geranylgeranylation, while no effect was observed in monocytes. Finally, we found that atherosclerotic lesions from cholesterol-fed rabbits treated with atorvastatin (2.5 mg/kg/day for 8 weeks) showed less immunoreactive PTX3 than lesions from control animals. CONCLUSIONS: Results suggest that statins may interfere with PTX3 expression in vascular cells via inhibition of protein geranylgeranylation. Since PTX3 is increasingly regarded as an important mediator of the inflammatory response underlying atherosclerosis and its complications, these results highlight the need for further studies of the role of PTX3 and its potential pharmacological modulation in cardiovascular disease.


Subject(s)
Atorvastatin/pharmacology , C-Reactive Protein/biosynthesis , Endothelial Cells/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Protein Prenylation/physiology , Serum Amyloid P-Component/biosynthesis , Animals , C-Reactive Protein/antagonists & inhibitors , Cells, Cultured , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Protein Prenylation/drug effects , Rabbits , Serum Amyloid P-Component/antagonists & inhibitors
12.
Thromb Haemost ; 114(1): 186-97, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25881051

ABSTRACT

The primary transcript of fibronectin undergoes alternative splicing in the cassette-type EDA and EDB exons and in the IIICs segment to generate different protein isoforms. Human carotid atherosclerotic plaques with a more stable phenotype are enriched with EDA containing fibronectin (FN-EDA). The aim of this study was to investigate the role of EDA containing fibronectin during atherogenesis. Mice constitutively expressing or lacking the EDA domain of fibronectin (EDA+/+ or EDA-/-)were crossed with ApoE-/- or LDL-R-/- mice and fed with a western type diet for 12 weeks. Lack of FN-EDA resulted in reduced atherosclerosis and in a plaque phenotype characterised by decreased calponin positive VSMC's (-15 %) and increased macrophages (+20 %). This was paralleled by increased MMP2, MMP9, and reduced TIMP2, collagen 1A1, 1A2 and 3A1 gene expression compared to that of wild-type and EDA+/+ mice. In vitro, VSMCs and macrophages isolated from EDA-/- miceshowed increased MMPs expression and activity compared to wild-type or EDA+/+ mice. Albumin-Cre recombinase/EDA+/+/ApoE-/- mice, which produceEDA containing FN only in peripheral tissues, presented an extension, a composition and a gene expression pattern in the atherosclerotic lesions similar to that of controls. The inclusion of EDA in FN results in larger atherosclerotic plaques compared to mice lacking EDA but with a more favourable phenotype in two animals models of atherosclerosis. This effect depends on the EDA-containing fibronectin produced by cells in the vasculature but not in the liver. These observations set the stage for investigating the properties of circulating EDA containing FN in improving plaque stability.


Subject(s)
Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Fibronectins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Collagen/metabolism , Diet, High-Fat , Disease Models, Animal , Fibronectins/deficiency , Fibronectins/genetics , Genotype , Macrophages/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Knockout , Microfilament Proteins/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Receptors, LDL/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Calponins
13.
Cardiovasc Drugs Ther ; 27(3): 211-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23400638

ABSTRACT

PURPOSE: Polymorphonuclear neutrophils, the first leukocytes to infiltrate the inflamed tissue, can make important contributions to vascular inflammatory processes driving the development of atherosclerosis. We herein investigated the effects of atorvastatin and NCX 6560 (a nitric oxide (NO)-donating atorvastatin derivative that has completed a successful phase 1b study) on neutrophilic inflammation in carotid arteries of normocholesterolemic rabbits subjected to perivascular collar placement. METHODS: Atorvastatin or NCX 6560 were administered orally (5 mg/kg/day or equimolar dose) to New Zealand White rabbits for 6 days, followed by collar implantation 1 h after the last dose. Twenty-four hours later carotids were harvested for neutrophil quantification by immunostaining. RESULTS: Treatment with NCX 6560 was associated with a lower neutrophil infiltration (-39.5 %), while atorvastatin did not affect neutrophil content. The result was independent of effects on plasma cholesterol or differences in atorvastatin bioavailability, which suggests an important role of NO-related mechanisms in mediating this effect. Consistent with these in vivo findings, in vitro studies showed that NCX 6560, as compared to atorvastatin, had greater inhibitory activity on processes involved in neutrophil recruitment, such as migration in response to IL-8 and IL-8 release by endothelial cells and by neutrophils themselves. Pretreatment with NCX 6560, but not with atorvastatin, reduced the ability of neutrophil supernatants to promote monocyte chemotaxis, a well-known pro-inflammatory activity of neutrophils. CONCLUSION: Experimental data suggest a potential role of NO-releasing statins in the control of the vascular inflammatory process mediated by polymorphonuclear neutrophils.


Subject(s)
Atherosclerosis/prevention & control , Cholesterol/blood , Heptanoic Acids/therapeutic use , Neutrophil Infiltration/drug effects , Nitric Oxide Donors/therapeutic use , Pyrroles/therapeutic use , Acute Disease , Animals , Atherosclerosis/blood , Atherosclerosis/immunology , Atherosclerosis/pathology , Atorvastatin , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Survival/drug effects , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Heptanoic Acids/administration & dosage , Heptanoic Acids/pharmacokinetics , Heptanoic Acids/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-8/immunology , Male , Molecular Structure , Monocytes/cytology , Monocytes/drug effects , Neutrophil Infiltration/immunology , Neutrophils/cytology , Neutrophils/drug effects , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide Donors/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Rabbits
14.
Drugs ; 71(11): 1441-67, 2011 Jul 30.
Article in English | MEDLINE | ID: mdl-21812507

ABSTRACT

The dipeptidyl peptidase (DPP)-4 inhibitors, which enhance glucose-dependent insulin secretion from pancreatic ß cells by preventing DPP-4-mediated degradation of endogenously released incretin hormones, represent a new therapeutic approach to the management of type 2 diabetes mellitus. The 'first-in-class' DPP-4 inhibitor, sitagliptin, was approved in 2006; it was followed by vildagliptin (available in the EU and many other countries since 2007, although approval in the US is still pending), saxagliptin (in 2009), alogliptin (in 2010, presently only in Japan) and linagliptin, which was approved in the US in May 2011 and is undergoing regulatory review in Japan and the EU. As the number of DPP-4 inhibitors on the market increases, potential differences among the different members of the class become important when deciding which agent is best suited for an individual patient. The aim of this review is to provide a comprehensive and updated comparison of the pharmacodynamic and pharmacokinetic properties of DPP-4 inhibitors, and to pinpoint pharmacological differences of potential interest for their use in therapy. Despite their common mechanism of action, these agents show significant structural heterogeneity that could translate into different pharmacological properties. At the pharmacokinetic level, DPP-4 inhibitors have important differences, including half-life, systemic exposure, bioavailability, protein binding, metabolism, presence of active metabolites and excretion routes. These differences could be relevant, especially in patients with renal or hepatic impairment, and when considering combination therapy. At the pharmacodynamic level, the data available so far indicate a similar glucose-lowering efficacy of DPP-4 inhibitors, either as monotherapy or in combination with other hypoglycaemic drugs, a similar weight-neutral effect, and a comparable safety and tolerability profile. Data on nonglycaemic parameters are scant at present and do not allow a comparison among DPP-4 inhibitors. Several phase III trials of DPP-4 inhibitors are currently ongoing; these trials, along with post-marketing surveillance data, will hopefully increase our knowledge about the long-term efficacy and safety of DPP-4 inhibitor therapy, the effect on pancreatic cell function and peripheral glucose metabolism, and the effect on cardiovascular outcomes in patients with type 2 diabetes.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Molecular Targeted Therapy , Age Factors , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Drug Approval , Female , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Male , Treatment Outcome
15.
Atherosclerosis ; 210(1): 1-13, 2010 May.
Article in English | MEDLINE | ID: mdl-19931081

ABSTRACT

Contrary to the long-standing and widely accepted belief that polymorphonuclear neutrophils (PMN) are of marginal relevance in atherosclerosis, evidence revealing a previously unappreciated role of PMN in the process of atherosclerosis is being accumulating. Systemic inflammation involving activated PMN is clearly associated with unstable conditions of coronary artery disease and an increased number of circulating neutrophils is a well-known risk indicator of future cardiovascular outcomes. Furthermore, PMN are activated in a number of clinical conditions associated with high risk of developing atherosclerosis and are detectable into culprit lesions of patients with coronary artery disease. At present, pharmacological interventions aimed at blocking neutrophil emigration from the blood into the arterial wall and/or inhibiting neutrophil-mediated inflammatory functions are not an option for treating atherosclerosis. Nevertheless, several lines of evidence suggest that part of the atheroprotective effects of statins as well as HDL and HDL apolipoproteins may be related to their ability to modulate neutrophilic inflammation in the arterial wall. These hypotheses are not definitely established and warrant for further study. This Review describes the evidence suggesting that PMN may have a causative role in atherogenesis and atheroprogression and discusses the potential importance of modulating neutrophilic inflammation as part of a novel, improved strategy for preventing and treating atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Neutrophils/physiology , Animals , Atherosclerosis/drug therapy , Coronary Disease/etiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation , Neutrophils/drug effects
16.
J Pharmacol Exp Ther ; 328(2): 419-25, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19023042

ABSTRACT

Monocytes/macrophages recruited into the arterial wall during atherogenesis are crucial in the initiation and progression of atherosclerosis and play a fundamental role in the destabilization process that is the main causal event of acute coronary syndromes. In the present study, we investigated the effect of the mammalian target of rapamycin inhibitor everolimus on macrophage accumulation within carotid lesions elicited by perivascular collar placement in cholesterol-fed rabbits. Everolimus (1.5 mg/kg given 1 day before collaring followed by 1 mg/kg/day for 14 days, administered by oral gavage) markedly decreased lesion macrophage content as compared with vehicle control (-65%; p < 0.01). This effect was associated with a reduction in intimal thickening and occurred in the absence of changes in plasma cholesterol concentrations. To gain insights on the potential mechanism(s) underlying this effect, we investigated the influence of everolimus on chemoattractant-induced migration of human monocytes in vitro. Pretreatment with therapeutic concentrations of everolimus (10 nM) significantly lowered monocyte chemotaxis in response to various chemotactic factors (i.e., monocyte chemoattractant protein-1/CCL2, fractalkine/CX3CL1, interleukin-8/CXCL8, complement fragment 5a, or N-formyl-Met-Leu-Phe) without inducing monocyte cell death. These results suggest that everolimus may favorably influence the atherosclerotic process by affecting the recruitment of monocytes into early lesions.


Subject(s)
Carotid Artery Diseases/pathology , Cell Movement/drug effects , Cholesterol/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Sirolimus/analogs & derivatives , Animals , Carotid Artery Diseases/metabolism , Cell Movement/physiology , Chemotactic Factors/pharmacology , Cholesterol/metabolism , Everolimus , Humans , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Macrophages/pathology , Macrophages/physiology , Monocytes/physiology , Rabbits , Sirolimus/pharmacokinetics , Sirolimus/pharmacology
17.
Pharmacol Res ; 56(2): 160-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17582780

ABSTRACT

Secretion of matrix metalloproteinases (MMPs) by macrophages and smooth muscle cells (SMC) may impair atherosclerotic cap integrity leading to atherosclerosis complications. Selective estrogen receptor modulators (SERMs) have favourable impact on plasma lipid levels, but their role in the prevention of atherosclerosis still remains unclear. We investigated the effects of raloxifene, a second generation SERM, on MMP expression and activity in cultured macrophages and SMC, and in rabbit carotid lesions. Human monocyte-derived macrophages were isolated from blood of healthy donors. SMC were isolated from the intima-media layers of collared rabbit carotid arteries. Cells were incubated for 24h with increasing concentrations of raloxifene. Ovariectomized rabbits fed a 1% cholesterol-rich diet were subjected to pericarotid collar placement and treated with or without 10mgkg(-1)d(-1) raloxifene for 2 weeks. In macrophages, raloxifene treatment (0.1-10microM) significantly reduced MMP-9 gelatinolytic potential in a concentration-dependent manner, without affecting MMP-9 activation. This effect was estrogen receptor (ER)-dependent and due to the inhibition of MMP-9 promoter-driven transcription following an interaction with NF-kB pathway. Similarly, in cultured SMC, raloxifene inhibited up to 40% MMP-2 gelatinolytic activity. In vivo, raloxifene decreased the expression of MMP-2, MMP-3, and MMP-9 by intimal cells and the total gelatinolytic activity of collared carotids. These effects were accompanied by reduction of lesion size and inhibition of macrophage accumulation. Overall, results indicate that raloxifene may reduce MMPs expression and activity in macrophages and smooth muscle cells and favourably affect lesion formation.


Subject(s)
Macrophages, Peritoneal/drug effects , Macrophages/drug effects , Matrix Metalloproteinase Inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protease Inhibitors/pharmacology , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Atherosclerosis/enzymology , Atherosclerosis/etiology , Atherosclerosis/prevention & control , CHO Cells , Carotid Arteries/drug effects , Carotid Arteries/enzymology , Cells, Cultured , Cholesterol, Dietary , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Macrophages/enzymology , Macrophages, Peritoneal/enzymology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NF-kappa B/metabolism , Ovariectomy , Promoter Regions, Genetic/drug effects , Protease Inhibitors/therapeutic use , Rabbits , Raloxifene Hydrochloride/therapeutic use , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Transcription, Genetic/drug effects , Transfection
18.
Atherosclerosis ; 195(1): e112-24, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17482195

ABSTRACT

We investigated the influence of apolipoprotein E deficiency and Western-type diet feeding on the development and composition of neointimal lesions induced by periadventitial carotid placement of a non-occlusive collar in mice. ApoE-/- and wild-type mice were fed a Western-type diet or chow diet for 4 weeks before collar surgery. Diets were continued after collar placement for 6 or 12 weeks. Compared to sham-operated arteries, collared carotids showed significant neointima formation, lumen loss, and outward remodeling in both apoE-/- and wild-type mice. These changes were not affected by either the genotype or the diet. Conversely, significant differences in neointima composition were detected between the two genotypes, with apoE-/- mice showing greater lipid deposition and lower SMC accumulation compared to wild-type mice, independent of the dietetic regimen. Altogether, the results of the present study indicate that although lesion composition may be influenced by genotype, neointima formation and arterial remodeling in the murine perivascular carotid collar model occur independent of the exposure to atherogenic diet or the presence of a sensitized genotype such as apoE-/-. The murine perivascular carotid collar model would thus be suitable for investigating neointima formation, arterial remodeling, and their potential pharmacological modulation in the setting of different genetic and dietary conditions.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Carotid Arteries/pathology , Tunica Intima/pathology , Animal Feed , Animals , Arteries/pathology , Diet , Genotype , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Neutrophils/metabolism
19.
J Pharmacol Exp Ther ; 319(3): 1444-51, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16943257

ABSTRACT

Previous studies reported the ability of raloxifene to acutely relax arterial and venous vessels, but the underlying mechanisms are controversial. Anti-inflammatory effects of the drug have been reported in nonvascular tissues. Therefore, the aim of this study was to investigate the nature of short- and long-term effects of raloxifene on selected aspects of vascular function in rat aorta. Isometric tension changes in response to raloxifene were recorded in aortic rings from ovariectomized female rats that underwent estrogen replacement, whereas long-term experiments were performed in isolated aortic smooth muscle cells (SMCs). Raloxifene (0.1 pM-0.1 microM) induced acute vasorelaxation through endothelium- and nitric oxide (NO)-dependent, prostanoid-independent mechanisms. The relaxant response to raloxifene was significantly weaker than that to 17beta-estradiol and was sensitive to neither the nonselective estrogen receptor antagonist ICI 182,780 [7,17-[9[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17-diol] nor a selective estrogen receptor (ER) alpha antagonist. This rapid vasorelaxant effect was retained in aortic rings from rats treated with 0.1 mg/kg, but not 1 mg/kg, lipopolysaccharide, 4 h before sacrifice. In cultured aortic SMCs, raloxifene treatment (1 nM-1 microM) for 24 h reduced inducible NO synthase activation in response to cytokines. This effect was prevented by the selective ERalpha antagonist and was associated with up-regulation of ERalpha protein levels, which dropped markedly upon cytokine stimulation. These findings illustrate the relevance of classic ER-dependent pathways to the vascular anti-inflammatory effects rather than to the nongenomic vasorelaxation induced by raloxifene and may assist in the design of novel ER isoform-selective estrogen-receptor modulators targeted to the vascular system.


Subject(s)
Anti-Inflammatory Agents , Muscle, Smooth, Vascular/drug effects , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Aorta, Thoracic/drug effects , Blotting, Western , Cell Survival/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Estrogen Receptor beta/metabolism , Female , Fulvestrant , In Vitro Techniques , Isometric Contraction/drug effects , Kinetics , Lipopolysaccharides/pharmacology , Muscle Relaxation/drug effects , Nitrites/metabolism , Ovariectomy , Rats
20.
Semin Vasc Med ; 4(4): 347-56, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15861311

ABSTRACT

Clinical trials have firmly established that 3-hydroxy-3-methylglutaryl-coenzyme-A reductase inhibitors (statins) can induce regression of vascular atherosclerosis as well as reduction of cardiovascular-related morbidity and death in patients with and without coronary artery disease. These beneficial effects of statins are usually assumed to result from their ability to reduce cholesterol synthesis. However, because mevalonic acid is the precursor not only of cholesterol but also of many nonsteroidal isoprenoid compounds, inhibition of 3-hydroxy-3-methylglutaryl-coenzyme-A reductase may result in pleiotropic effects. Indeed, statins can interfere with major events involved in the formation and the evolution of atherosclerotic lesions, such as arterial myocyte migration and proliferation and cholesterol accumulation, independent of their hypolipidemic properties. The aim of this article is to focus on clinical and experimental data that show that statins possess effects beyond cholesterol lowering, particularly on arterial smooth muscle cell proliferation. The contribution of these direct vascular effects to the reduction of cardiovascular events observed in clinical trials with statins represents one of the major challenges for future studies to understand the antiatherosclerotic benefits of these agents.


Subject(s)
Cell Proliferation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Muscle, Smooth, Vascular/pathology , Animals , Arteries/drug effects , Arteries/pathology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Arteriosclerosis/prevention & control , Cholesterol/biosynthesis , Humans , Muscle, Smooth, Vascular/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...