Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
JVS Vasc Sci ; 4: 100111, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37519334

RESUMO

Objective: Atherosclerosis is a leading cause of mortality in the rapidly growing population with diabetes mellitus. Vascular interventions in patients with diabetes can lead to complications attributed to defective vascular remodeling and impaired healing response in the vessel wall. In this study, we aim to elucidate the molecular differences in the vascular healing response over time using a rat model of arterial injury applied to healthy and diabetic conditions. Methods: Wistar (healthy) and Goto-Kakizaki (GK, diabetic) rats (n = 40 per strain) were subjected to left common carotid artery (CCA) balloon injury and euthanized at different timepoints: 0 and 20 hours, 5 days, and 2, 4, and 6 weeks. Noninvasive morphological and physiological assessment of the CCA was performed with ultrasound biomicroscopy (Vevo 2100) and corroborated with histology. Total RNA was isolated from the injured CCA at each timepoint, and microarray profiling was performed (n = 3 rats per timepoint; RaGene-1_0-st-v1 platform). Bioinformatic analyses were conducted using R software, DAVID bioinformatic tool, online STRING database, and Cytoscape software. Results: Significant increase in the neointimal thickness (P < .01; two-way analysis of variance) as well as exaggerated negative remodeling was observed after 2 weeks of injury in GK rats compared with heathy rats, which was confirmed by histological analyses. Bioinformatic analyses showed defective expression patterns for smooth muscle cells and immune cell markers, along with reduced expression of key extracellular matrix-related genes and increased expression of pro-thrombotic genes, indicating potential faults on cell regulation level. Transcription factor-protein-protein interaction analysis provided mechanistic evidence with an array of transcription factors dysregulated in diabetic rats. Conclusions: In this study, we have demonstrated that diabetic rats exhibit impaired arterial remodeling characterized by a delayed healing response. We show that increased contractile smooth muscle cell marker expression coincided with decreased matrix metalloproteinase expression, indicating a potential mechanism for a lack of extracellular matrix reorganization in the impaired vascular healing in GK rats. These results further corroborate the higher prevalence of restenosis in patients with diabetes and provide vital molecular insights into the mechanisms contributing to the impaired arterial healing response in diabetes. Moreover, the presented study provides the research community with the valuable longitudinal gene expression data bank for further exploration of diabetic vasculopathy.

2.
Biomolecules ; 13(6)2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37371462

RESUMO

BACKGROUND: Intraplaque hemorrhage (IPH) is a hallmark of atherosclerotic plaque instability. Biliverdin reductase B (BLVRB) is enriched in plasma and plaques from patients with symptomatic carotid atherosclerosis and functionally associated with IPH. OBJECTIVE: We explored the biomarker potential of plasma BLVRB through (1) its correlation with IPH in carotid plaques assessed by magnetic resonance imaging (MRI), and with recurrent ischemic stroke, and (2) its use for monitoring pharmacotherapy targeting IPH in a preclinical setting. METHODS: Plasma BLVRB levels were measured in patients with symptomatic carotid atherosclerosis from the PARISK study (n = 177, 5 year follow-up) with and without IPH as indicated by MRI. Plasma BLVRB levels were also measured in a mouse vein graft model of IPH at baseline and following antiangiogenic therapy targeting vascular endothelial growth factor receptor 2 (VEGFR-2). RESULTS: Plasma BLVRB levels were significantly higher in patients with IPH (737.32 ± 693.21 vs. 520.94 ± 499.43 mean fluorescent intensity (MFI), p = 0.033), but had no association with baseline clinical and biological parameters. Plasma BLVRB levels were also significantly higher in patients who developed recurrent ischemic stroke (1099.34 ± 928.49 vs. 582.07 ± 545.34 MFI, HR = 1.600, CI [1.092-2.344]; p = 0.016). Plasma BLVRB levels were significantly reduced following prevention of IPH by anti-VEGFR-2 therapy in mouse vein grafts (1189 ± 258.73 vs. 1752 ± 366.84 MFI; p = 0.004). CONCLUSIONS: Plasma BLVRB was associated with IPH and increased risk of recurrent ischemic stroke in patients with symptomatic low- to moderate-grade carotid stenosis, indicating the capacity to monitor the efficacy of IPH-preventive pharmacotherapy in an animal model. Together, these results suggest the utility of plasma BLVRB as a biomarker for atherosclerotic plaque instability.


Assuntos
Doenças das Artérias Carótidas , AVC Isquêmico , Placa Aterosclerótica , Animais , Humanos , Camundongos , Biomarcadores/sangue , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/complicações , Hemorragia/sangue , Hemorragia/diagnóstico por imagem , Hemorragia/etiologia , AVC Isquêmico/sangue , AVC Isquêmico/etiologia , Placa Aterosclerótica/sangue , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
3.
Vascul Pharmacol ; 150: 107167, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36958707

RESUMO

BACKGROUND: Calcification, a key feature of advanced human atherosclerosis, is positively associated with vascular disease burden and adverse events. We showed that macrocalcification can be a stabilizing factor for carotid plaque molecular biology, due to inverse association with immune processes. Mast cells (MCs) are important contributors to plaque instability, but their relationship with macrocalcification is unexplored. With a hypothesis that MC activation negatively associates with carotid plaque macrocalcification, we aimed to investigate the link between MCs and carotid plaque vulnerability, and study MC role in plaque calcification via smooth muscle cells (SMCs). METHODS: Pre-operative computed tomography angiographies of patients (n = 40) undergoing surgery for carotid stenosis were used to characterize plaque morphology. Plaque microarrays (n = 40 and n = 126) were used for bioinformatic deconvolution of immune cell populations. Tissue microarrays (n = 103) were used to histologically validate the contribution of activated and resting MCs in plaques. RESULTS: Activated MCs and their typical markers were negatively correlated with macrocalcification. The ratio of activated vs. resting MCs was increased in low-calcified plaques from symptomatic patients. There was no modulating effect of medication on MC ratios. In vitro experiments showed that SMC calcification attenuated MC activation, while both active and resting MCs stimulated SMC calcification and induced dedifferentiation towards a pro-inflammatory-, osteochondrocyte-like phenotype, without modulating their migro-proliferative function. CONCLUSIONS: Integrative analyses from human plaques showed that MC activation is inversely associated with macrocalcification and positively with parameters of plaque vulnerability. Mechanistically, MCs induce SMC osteogenic reprograming, while matrix calcification in turn attenuates MC activation, offering new therapeutic avenues for exploration.


Assuntos
Aterosclerose , Estenose das Carótidas , Placa Aterosclerótica , Calcificação Vascular , Humanos , Placa Aterosclerótica/patologia , Mastócitos/patologia , Estenose das Carótidas/complicações , Aterosclerose/patologia , Miócitos de Músculo Liso/patologia , Calcificação Vascular/diagnóstico por imagem , Calcificação Vascular/genética
4.
Comput Biol Med ; 152: 106364, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36525832

RESUMO

OBJECTIVE: Guidance for preventing myocardial infarction and ischemic stroke by tailoring treatment for individual patients with atherosclerosis is an unmet need. Such development may be possible with computational modeling. Given the multifactorial biology of atherosclerosis, modeling must be based on complete biological networks that capture protein-protein interactions estimated to drive disease progression. Here, we aimed to develop a clinically relevant scale model of atherosclerosis, calibrate it with individual patient data, and use it to simulate optimized pharmacotherapy for individual patients. APPROACH AND RESULTS: The study used a uniquely constituted plaque proteomic dataset to create a comprehensive systems biology disease model for simulating individualized responses to pharmacotherapy. Plaque tissue was collected from 18 patients with 6735 proteins at two locations per patient. 113 pathways were identified and included in the systems biology model of endothelial cells, vascular smooth muscle cells, macrophages, lymphocytes, and the integrated intima, altogether spanning 4411 proteins, demonstrating a range of 39-96% plaque instability. After calibrating the systems biology models for individual patients, we simulated intensive lipid-lowering, anti-inflammatory, and anti-diabetic drugs. We also simulated a combination therapy. Drug response was evaluated as the degree of change in plaque stability, where an improvement was defined as a reduction of plaque instability. In patients with initially unstable lesions, simulated responses varied from high (20%, on combination therapy) to marginal improvement, whereas patients with initially stable plaques showed generally less improvement. CONCLUSION: In this pilot study, proteomics-based system biology modeling was shown to simulate drug response based on atherosclerotic plaque instability with a power of 90%, providing a potential strategy for improved personalized management of patients with cardiovascular disease.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Placa Aterosclerótica , Humanos , Doenças Cardiovasculares/tratamento farmacológico , Proteômica , Medicina de Precisão , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Calibragem , Projetos Piloto , Aterosclerose/tratamento farmacológico , Simulação por Computador
5.
Cells ; 11(20)2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36291144

RESUMO

Intimal calcification and vascular stiffening are predominant features of end-stage atherosclerosis. However, their role in atherosclerotic plaque instability and how the extent and spatial distribution of calcification influence plaque biology remain unclear. We recently showed that extensive macro calcification can be a stabilizing feature of late-stage human lesions, associated with a reacquisition of more differentiated properties of plaque smooth muscle cells (SMCs) and extracellular matrix (ECM) remodeling. Here, we hypothesized that biomechanical forces related to macro-calcification within plaques influence SMC phenotype and contribute to plaque stabilization. We generated a finite element modeling (FEM) pipeline to assess plaque tissue stretch based on image analysis of preoperative computed tomography angiography (CTA) of carotid atherosclerotic plaques to visualize calcification and soft tissues (lipids and extracellular matrix) within the lesions. Biomechanical stretch was significantly reduced in tissues in close proximity to macro calcification, while increased levels were observed within distant soft tissues. Applying this data to an in vitro stretch model on primary vascular SMCs revealed upregulation of typical markers for differentiated SMCs and contractility under low stretch conditions but also impeded SMC alignment. In contrast, high stretch conditions in combination with calcifying conditions induced SMC apoptosis. Our findings suggest that the load bearing capacities of macro calcifications influence SMC differentiation and survival and contribute to atherosclerotic plaque stabilization.


Assuntos
Calcinose , Doenças das Artérias Carótidas , Placa Aterosclerótica , Humanos , Placa Aterosclerótica/patologia , Miócitos de Músculo Liso/patologia , Doenças das Artérias Carótidas/diagnóstico por imagem , Calcinose/patologia , Fenótipo , Lipídeos
6.
Arterioscler Thromb Vasc Biol ; 42(5): 659-676, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35321563

RESUMO

BACKGROUND: Understanding the processes behind carotid plaque instability is necessary to develop methods for identification of patients and lesions with stroke risk. Here, we investigated molecular signatures in human plaques stratified by echogenicity as assessed by duplex ultrasound. METHODS: Lesion echogenicity was correlated to microarray gene expression profiles from carotid endarterectomies (n=96). The findings were extended into studies of human and mouse atherosclerotic lesions in situ, followed by functional investigations in vitro in human carotid smooth muscle cells (SMCs). RESULTS: Pathway analyses highlighted muscle differentiation, iron homeostasis, calcification, matrix organization, cell survival balance, and BCLAF1 (BCL2 [B-cell lymphoma 2]-associated transcription factor 1) as the most significant signatures. BCLAF1 was downregulated in echolucent plaques, positively correlated to proliferation and negatively to apoptosis. By immunohistochemistry, BCLAF1 was found in normal medial SMCs. It was repressed early during atherogenesis but reappeared in CD68+ cells in advanced plaques and interacted with BCL2 by proximity ligation assay. In cultured SMCs, BCLAF1 was induced by differentiation factors and mitogens and suppressed by macrophage-conditioned medium. BCLAF1 silencing led to downregulation of BCL2 and SMC markers, reduced proliferation, and increased apoptosis. Transdifferentiation of SMCs by oxLDL (oxidized low-denisty lipoprotein) was accompanied by upregulation of BCLAF1, CD36, and CD68, while oxLDL exposure with BCLAF1 silencing preserved MYH (myosin heavy chain) 11 expression and prevented transdifferentiation. BCLAF1 was associated with expression of cell differentiation, contractility, viability, and inflammatory genes, as well as the scavenger receptors CD36 and CD68. BCLAF1 expression in CD68+/BCL2+ cells of SMC origin was verified in plaques from MYH11 lineage-tracing atherosclerotic mice. Moreover, BCLAF1 downregulation associated with vulnerability parameters and cardiovascular risk in patients with carotid atherosclerosis. CONCLUSIONS: Plaque echogenicity correlated with enrichment of distinct molecular pathways and identified BCLAF1, previously not described in atherosclerosis, as the most significant gene. Functionally, BCLAF1 seems necessary for survival and transdifferentiation of SMCs into a macrophage-like phenotype. The role of BCLAF1 in plaque vulnerability should be further evaluated.


Assuntos
Aterosclerose , Placa Aterosclerótica , Proteínas Repressoras/metabolismo , Animais , Aterosclerose/diagnóstico por imagem , Aterosclerose/genética , Aterosclerose/metabolismo , Transdiferenciação Celular , Humanos , Lipídeos , Camundongos , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Repressoras/genética , Transcriptoma , Proteínas Supressoras de Tumor/genética , Ultrassonografia
7.
Clin Transl Med ; 12(2): e682, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35184400

RESUMO

RATIONALE: Vascular calcification is a prominent feature of late-stage diabetes, renal and cardiovascular disease (CVD), and has been linked to adverse events. Recent studies in patients reported that plasma levels of osteomodulin (OMD), a proteoglycan involved in bone mineralisation, associate with diabetes and CVD. We hypothesised that OMD could be implicated in these diseases via vascular calcification as a common underlying factor and aimed to investigate its role in this context. METHODS AND RESULTS: In patients with chronic kidney disease, plasma OMD levels correlated with markers of inflammation and bone turnover, with the protein present in calcified arterial media. Plasma OMD also associated with cardiac calcification and the protein was detected in calcified valve leaflets by immunohistochemistry. In patients with carotid atherosclerosis, circulating OMD was increased in association with plaque calcification as assessed by computed tomography. Transcriptomic and proteomic data showed that OMD was upregulated in atherosclerotic compared to control arteries, particularly in calcified plaques, where OMD expression correlated positively with markers of smooth muscle cells (SMCs), osteoblasts and glycoproteins. Immunostaining confirmed that OMD was abundantly present in calcified plaques, localised to extracellular matrix and regions rich in α-SMA+ cells. In vivo, OMD was enriched in SMCs around calcified nodules in aortic media of nephrectomised rats and in plaques from ApoE-/- mice on warfarin. In vitro experiments revealed that OMD mRNA was upregulated in SMCs stimulated with IFNγ, BMP2, TGFß1, phosphate and ß-glycerophosphate, and by administration of recombinant human OMD protein (rhOMD). Mechanistically, addition of rhOMD repressed the calcification process of SMCs treated with phosphate by maintaining their contractile phenotype along with enriched matrix organisation, thereby attenuating SMC osteoblastic transformation. Mechanistically, the role of OMD is exerted likely through its link with SMAD3 and TGFB1 signalling, and interplay with BMP2 in vascular tissues. CONCLUSION: We report a consistent association of both circulating and tissue OMD levels with cardiovascular calcification, highlighting the potential of OMD as a clinical biomarker. OMD was localised in medial and intimal α-SMA+ regions of calcified cardiovascular tissues, induced by pro-inflammatory and pro-osteogenic stimuli, while the presence of OMD in extracellular environment attenuated SMC calcification.


Assuntos
Proteínas da Matriz Extracelular/farmacologia , Músculo Liso/efeitos dos fármacos , Osteogênese/genética , Proteoglicanas/farmacologia , Calcificação Vascular/etiologia , Análise de Variância , Estudos de Coortes , Estudos Transversais , Proteínas da Matriz Extracelular/metabolismo , Humanos , Modelos Lineares , Músculo Liso/fisiologia , Países Baixos , Osteogênese/fisiologia , Estudos Prospectivos , Proteoglicanas/metabolismo , Estatísticas não Paramétricas , Suécia , Calcificação Vascular/genética
8.
Eur J Vasc Endovasc Surg ; 62(5): 716-726, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34511314

RESUMO

OBJECTIVE: Ischaemic strokes can be caused by unstable carotid atherosclerosis, but methods for identification of high risk lesions are lacking. Carotid plaque morphology imaging using software for visualisation of plaque components in computed tomography angiography (CTA) may improve assessment of plaque phenotype and stroke risk, but it is unknown if such analyses also reflect the biological processes related to lesion stability. Here, we investigated how carotid plaque morphology by image analysis of CTA is associated with biological processes assessed by transcriptomic analyses of corresponding carotid endarterectomies (CEAs). METHODS: Carotid plaque morphology was assessed in patients undergoing CEA for symptomatic or asymptomatic carotid stenosis consecutively enrolled between 2006 and 2015. Computer based analyses of pre-operative CTA was performed to define calcification, lipid rich necrotic core (LRNC), intraplaque haemorrhage (IPH), matrix (MATX), and plaque burden. Plaque morphology was correlated with molecular profiles obtained from microarrays of corresponding CEAs and models were built to assess the ability of plaque morphology to predict symptomatology. RESULTS: Carotid plaques (n = 93) from symptomatic patients (n = 61) had significantly higher plaque burden and LRNC compared with plaques from asymptomatic patients (n = 32). Lesions selected from the transcriptomic cohort (n = 40) with high LRNC, IPH, MATX, or plaque burden were characterised by molecular signatures coupled with inflammation and extracellular matrix degradation, typically linked with instability. In contrast, highly calcified plaques had a molecular signature signifying stability with enrichment of profibrotic pathways and repressed inflammation. In a cross validated prediction model for symptoms, plaque morphology by CTA alone was superior to the degree of stenosis. CONCLUSION: The study demonstrates that CTA image analysis for evaluation of carotid plaque morphology, also reflects prevalent biological processes relevant for assessment of plaque phenotype. The results support the use of CTA image analysis of plaque morphology for risk stratification and management of patients with carotid stenosis.


Assuntos
Estenose das Carótidas/diagnóstico por imagem , Estenose das Carótidas/metabolismo , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/metabolismo , Idoso , Estenose das Carótidas/etiologia , Estudos de Coortes , Angiografia por Tomografia Computadorizada , Endarterectomia das Carótidas , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Placa Aterosclerótica/etiologia , Sensibilidade e Especificidade
9.
Cells ; 10(6)2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34063989

RESUMO

Calcification is a prominent feature of late-stage atherosclerosis, but the mechanisms driving this process are unclear. Using a biobank of carotid endarterectomies, we recently showed that Proteoglycan 4 (PRG4) is a key molecular signature of calcified plaques, expressed in smooth muscle cell (SMC) rich regions. Here, we aimed to unravel the PRG4 role in vascular remodeling and intimal calcification. PRG4 expression in human carotid endarterectomies correlated with calcification assessed by preoperative computed tomographies. PRG4 localized to SMCs in early intimal thickening, while in advanced lesions it was found in the extracellular matrix, surrounding macro-calcifications. In experimental models, Prg4 was upregulated in SMCs from partially ligated ApoE-/- mice and rat carotid intimal hyperplasia, correlating with osteogenic markers and TGFb1. Furthermore, PRG4 was enriched in cells positive for chondrogenic marker SOX9 and around plaque calcifications in ApoE-/- mice on warfarin. In vitro, PRG4 was induced in SMCs by IFNg, TGFb1 and calcifying medium, while SMC markers were repressed under calcifying conditions. Silencing experiments showed that PRG4 expression was driven by transcription factors SMAD3 and SOX9. Functionally, the addition of recombinant human PRG4 increased ectopic SMC calcification, while arresting cell migration and proliferation. Mechanistically, it suppressed endogenous PRG4, SMAD3 and SOX9, and restored SMC markers' expression. PRG4 modulates SMC function and osteogenic phenotype during intimal remodeling and macro-calcification in response to TGFb1 signaling, SMAD3 and SOX9 activation. The effects of PRG4 on SMC phenotype and calcification suggest its role in atherosclerotic plaque stability, warranting further investigations.


Assuntos
Calcinose , Miócitos de Músculo Liso , Proteoglicanas/metabolismo , Remodelação Vascular , Animais , Diferenciação Celular , Estudos de Coortes , Humanos , Masculino , Camundongos , Camundongos Knockout para ApoE , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos , Fatores de Transcrição SOX9/metabolismo , Proteína Smad3/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 40(11): 2700-2713, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32907367

RESUMO

OBJECTIVE: There is no medical treatment to prevent abdominal aortic aneurysm (AAA) growth and rupture, both of which are linked to smoking. Our objective was to map the tunica-specific pathophysiology of AAA with consideration of the intraluminal thrombus, age, and sex, and to subsequently identify which mechanisms were linked to smoking and diameter growth rate. Approach and Results: Microarray analyses were performed on 246 samples from 76 AAA patients and 13 controls. In media and adventitia, there were 5889 and 2701 differentially expressed genes, respectively. Gene sets related to adaptive and innate immunity were upregulated in both tunicas. Media-specific gene sets included increased matrix disassembly and angiogenesis, as well as decreased muscle cell development, contraction, and differentiation. Genes implicated in previous genome-wide association studies were dysregulated in media. The intraluminal thrombus had a pro-proteolytic and proinflammatory effect on the underlying media. Active smoking resulted in increased inflammation, oxidative stress, and angiogenesis in all tissues and enriched lipid metabolism in adventitia. Processes enriched with active smoking in control aortas overlapped to a high extent with those differentially expressed between AAAs and controls. The AAA diameter growth rate (n=24) correlated with T- and B-cell expression in media, as well as lipid-related processes in the adventitia. CONCLUSIONS: This tunica-specific analysis of gene expression in a large study enabled the detection of features not previously described in AAA disease. Smoking was associated with increased expression of aneurysm-related processes, of which adaptive immunity and lipid metabolism correlated with growth rate.


Assuntos
Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/genética , Fumar/efeitos adversos , Trombose/genética , Transcriptoma , Túnica Média/metabolismo , Remodelação Vascular/genética , Imunidade Adaptativa/genética , Adulto , Idoso , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Estudos de Casos e Controles , Dilatação Patológica , Progressão da Doença , Feminino , Redes Reguladoras de Genes , Interação Gene-Ambiente , Humanos , Metabolismo dos Lipídeos/genética , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Fumar/genética , Fumar/metabolismo , Fumar/patologia , Trombose/metabolismo , Trombose/patologia , Túnica Média/patologia
11.
PLoS One ; 15(1): e0227889, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31971988

RESUMO

BACKGROUND: Abdominal aortic aneurysm (AAA) is a potentially life-threatening disease, and until today there is no other treatment available than surgical intervention. Dipeptidyl peptidase-4 (DPP4)-inhibitors, used clinically to treat type 2 diabetes, have in murine models been shown to attenuate aneurysm formation and decrease aortic wall matrix degradation, inflammation and apoptosis. Our aim was to investigate if DPP4 is present, active and differentially expressed in human AAA. METHODS AND RESULTS: DPP4 gene expression was elevated in both media and adventitia of AAA tissue compared with control tissue, as measured by microarrays and qPCR, with consistent findings in external data. The plasma activity of DPP4 was however lower in male patients with AAA compared with age- and gender-matched controls, independently of comorbidity or medication. Immunohistochemical double staining revealed co-localization of DPP4 with cells positive for CD68, CD4 and -8, CD20, and SMA. Gene set enrichment analysis demonstrated that expression of DPP4 in AAA tissue correlated with expression of biological processes related to B- and T-cells, extracellular matrix turnover, peptidase activity, oxidative stress and angiogenesis whereas it correlated negatively with muscle-/actin-related processes. CONCLUSION: DPP4 is upregulated in both media and adventitia of human AAA and correlates with aneurysm pathophysiological processes. These results support previous murine mechanistic studies and implicate DPP4 as a target in AAA disease.


Assuntos
Aorta/metabolismo , Aneurisma da Aorta Abdominal/genética , Vasos Sanguíneos/metabolismo , Dipeptidil Peptidase 4/genética , Actinas/genética , Adulto , Idoso , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Aorta/patologia , Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/patologia , Apoptose/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Vasos Sanguíneos/patologia , Antígenos CD4/genética , Antígenos CD8/genética , Dipeptidil Peptidase 4/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/genética , Feminino , Regulação da Expressão Gênica/genética , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Linfócitos T/metabolismo
12.
Circ Res ; 126(5): 571-585, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31893970

RESUMO

RATIONALE: PCSKs (Proprotein convertase subtilisins/kexins) are a protease family with unknown functions in vasculature. Previously, we demonstrated PCSK6 upregulation in human atherosclerotic plaques associated with smooth muscle cells (SMCs), inflammation, extracellular matrix remodeling, and mitogens. OBJECTIVE: Here, we applied a systems biology approach to gain deeper insights into the PCSK6 role in normal and diseased vessel wall. METHODS AND RESULTS: Genetic analyses revealed association of intronic PCSK6 variant rs1531817 with maximum internal carotid intima-media thickness progression in high-cardiovascular risk subjects. This variant was linked with PCSK6 mRNA expression in healthy aortas and plaques but also with overall plaque SMA+ cell content and pericyte fraction. Increased PCSK6 expression was found in several independent human cohorts comparing atherosclerotic lesions versus healthy arteries, using transcriptomic and proteomic datasets. By immunohistochemistry, PCSK6 was localized to fibrous cap SMA+ cells and neovessels in plaques. In human, rat, and mouse intimal hyperplasia, PCSK6 was expressed by proliferating SMA+ cells and upregulated after 5 days in rat carotid balloon injury model, with positive correlation to PDGFB (platelet-derived growth factor subunit B) and MMP (matrix metalloprotease) 2/MMP14. Here, PCSK6 was shown to colocalize and cointeract with MMP2/MMP14 by in situ proximity ligation assay. Microarrays of carotid arteries from Pcsk6-/- versus control mice revealed suppression of contractile SMC markers, extracellular matrix remodeling enzymes, and cytokines/receptors. Pcsk6-/- mice showed reduced intimal hyperplasia response upon carotid ligation in vivo, accompanied by decreased MMP14 activation and impaired SMC outgrowth from aortic rings ex vivo. PCSK6 silencing in human SMCs in vitro leads to downregulation of contractile markers and increase in MMP2 expression. Conversely, PCSK6 overexpression increased PDGFBB (platelet-derived growth factor BB)-induced cell proliferation and particularly migration. CONCLUSIONS: PCSK6 is a novel protease that induces SMC migration in response to PDGFB, mechanistically via modulation of contractile markers and MMP14 activation. This study establishes PCSK6 as a key regulator of SMC function in vascular remodeling. Visual Overview: An online visual overview is available for this article.


Assuntos
Miócitos de Músculo Liso/metabolismo , Pró-Proteína Convertases/genética , Serina Endopeptidases/genética , Remodelação Vascular , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Masculino , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/fisiologia , Polimorfismo de Nucleotídeo Único , Pró-Proteína Convertases/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Ratos , Ratos Sprague-Dawley , Serina Endopeptidases/metabolismo , Transcriptoma
13.
JVS Vasc Sci ; 1: 13-27, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34617037

RESUMO

OBJECTIVE: Endovascular interventions cause arterial injury and induce a healing response to restore vessel wall homeostasis. Complications of defective or excessive healing are common and result in increased morbidity and repeated interventions. Experimental models of intimal hyperplasia are vital for understanding the vascular healing mechanisms and resolving the clinical problems of restenosis, vein graft stenosis, and dialysis access failure. Our aim was to systematically investigate the transcriptional, histologic, and systemic reaction to vascular injury during a prolonged time. METHODS: Balloon injury of the left common carotid artery was performed in male rats. Animals (n = 69) were euthanized before or after injury, either directly or after 2 hours, 20 hours, 2 days, 5 days, 2 weeks, 6 weeks, and 12 weeks. Both injured and contralateral arteries were subjected to microarray profiling, followed by bioinformatic exploration, histologic characterization of the biopsy specimens, and plasma lipid analyses. RESULTS: Immune activation and coagulation were key mechanisms in the early response, followed by cytokine release, tissue remodeling, and smooth muscle cell modulation several days after injury, with reacquisition of contractile features in later phases. Novel pathways related to clonal expansion, inflammatory transformation, and chondro-osteogenic differentiation were identified and immunolocalized to neointimal smooth muscle cells. Analysis of uninjured arteries revealed a systemic component of the reaction after local injury, underlined by altered endothelial signaling, changes in overall tissue bioenergy metabolism, and plasma high-density lipoprotein levels. CONCLUSIONS: We demonstrate that vascular injury induces dynamic transcriptional landscape and metabolic changes identifiable as early, intermediate, and late response phases, reaching homeostasis after several weeks. This study provides a temporal "roadmap" of vascular healing as a publicly available resource for the research community.

14.
Thromb Haemost ; 119(12): 2014-2024, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31634957

RESUMO

Inflammatory processes contribute to intimal hyperplasia (IH) and long-term failure of vein grafts used in bypass surgery. Leukocyte recruitment on endothelial cells of vessels during inflammation is regulated by P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1), which also mediates the interaction between platelets and endothelial cells in vein grafts transferred to arteries. However, how this pathway causes IH in vein grafts is unclear. In this study, we used a murine model of vein grafting to investigate P-selectin-mediated platelet adhesion, followed by IH. On the luminal surface of the vein graft, leukocyte recruitment occurred mainly in areas with adhered platelets rather than on endothelial cells without adherent platelets 1 hour after vein grafting. Blockage of either P-selectin or PSGL-1 reduced platelet adhesion and leukocyte recruitment on the luminal surface of vein grafts. Inhibition of the P-selectin pathway in vein grafts significantly reduced platelet-mediated leukocyte recruitment and IH of vein grafts 28 days after surgery. The study demonstrates that functional blockage of the P-selectin/PSGL-1 pathway in the early inflammatory phase after vein grafting reduced leukocyte invasion in the vein graft wall and later IH development. The findings imply an attractive early time window for prevention of vein graft failure by manipulating platelet adhesion.


Assuntos
Hiperplasia/prevenção & controle , Glicoproteínas de Membrana/sangue , Selectina-P/sangue , Adesividade Plaquetária , Túnica Íntima/patologia , Veias/transplante , Animais , Plaquetas/patologia , Modelos Animais de Doenças , Células Endoteliais , Feminino , Inflamação , Leucócitos/metabolismo , Ligantes , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P/antagonistas & inibidores
15.
Atherosclerosis ; 288: 175-185, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31109707

RESUMO

BACKGROUND AND AIMS: Unstable carotid atherosclerosis causes stroke, but methods to identify patients and lesions at risk are lacking. We recently found enrichment of genes associated with calcification in carotid plaques from asymptomatic patients. Here, we hypothesized that calcification represents a stabilising feature of plaques and investigated how macro-calcification, as estimated by computed tomography (CT), correlates with gene expression profiles in lesions. METHODS: Plaque calcification was measured in pre-operative CT angiographies. Plaques were sorted into high- and low-calcified, profiled with microarrays, followed by bioinformatic analyses. Immunohistochemistry and qPCR were performed to evaluate the findings in plaques and arteries with medial calcification from chronic kidney disease patients. RESULTS: Smooth muscle cell (SMC) markers were upregulated in high-calcified plaques and calcified plaques from symptomatic patients, whereas macrophage markers were downregulated. The most enriched processes in high-calcified plaques were related to SMCs and extracellular matrix (ECM) organization, while inflammation, lipid transport and chemokine signaling were repressed. These findings were confirmed in arteries with high medial calcification. Proteoglycan 4 (PRG4) was identified as the most upregulated gene in association with plaque calcification and found in the ECM, SMA+ and CD68+/TRAP + cells. CONCLUSIONS: Macro-calcification in carotid lesions correlated with a transcriptional profile typical for stable plaques, with altered SMC phenotype and ECM composition and repressed inflammation. PRG4, previously not described in atherosclerosis, was enriched in the calcified ECM and localized to activated macrophages and smooth muscle-like cells. This study strengthens the notion that assessment of calcification may aid evaluation of plaque phenotype and stroke risk.


Assuntos
Estenose das Carótidas/diagnóstico por imagem , Estenose das Carótidas/genética , Angiografia por Tomografia Computadorizada , Perfilação da Expressão Gênica , Músculo Liso Vascular/diagnóstico por imagem , Placa Aterosclerótica , Transcriptoma , Calcificação Vascular/diagnóstico por imagem , Calcificação Vascular/genética , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/patologia , Estenose das Carótidas/complicações , Estenose das Carótidas/patologia , Humanos , Músculo Liso Vascular/patologia , Valor Preditivo dos Testes , Proteoglicanas/genética , Medição de Risco , Fatores de Risco , Ruptura Espontânea , Acidente Vascular Cerebral/etiologia , Suécia , Calcificação Vascular/complicações , Calcificação Vascular/patologia
16.
Atherosclerosis ; 283: 127-136, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30665614

RESUMO

BACKGROUND AND AIMS: Calcification is a hallmark of advanced atherosclerosis and an active process akin to bone remodeling. Heparanase (HPSE) is an endo-ß-glucuronidase, which cleaves glycosaminoglycan chains of heparan sulfate proteoglycans. The role of HPSE is controversial in osteogenesis and bone remodeling while it is unexplored in vascular calcification. Previously, we reported upregulation of HPSE in human carotid endarterectomies from symptomatic patients and showed correlation of HPSE expression with markers of inflammation and increased thrombogenicity. The present aim is to investigate HPSE expression in relation to genes associated with osteogenesis and osteolysis and the effect of elevated HPSE expression on calcification and osteolysis in vitro. METHODS: Transcriptomic and immunohistochemical analyses were performed using the Biobank of Karolinska Endarterectomies (BiKE). In vitro calcification and osteolysis were analysed in human carotid smooth muscle cells overexpressing HPSE and bone marrow-derived osteoclasts from HPSE-transgenic mice respectively. RESULTS: HPSE expression correlated primarily with genes coupled to osteoclast differentiation and function in human carotid atheromas. HPSE was expressed in osteoclast-like cells in atherosclerotic lesions, and HPSE-transgenic bone marrow-derived osteoclasts displayed a higher osteolytic activity compared to wild-type cells. Contrarily, human carotid SMCs with an elevated HPSE expression demonstrated markedly increased mineralization upon osteogenic differentiation. CONCLUSIONS: We suggest that HPSE may have dual functions in vascular calcification, depending on the stage of the disease and presence of inflammatory cells. While HPSE plausibly enhances mineralization and osteogenic differentiation of vascular smooth muscle cells, it is associated with inflammation-induced osteoclast differentiation and activity in advanced atherosclerotic plaques.


Assuntos
Doenças das Artérias Carótidas/genética , Regulação da Expressão Gênica , Glucuronidase/genética , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/genética , Calcificação Vascular/genética , Western Blotting , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Diferenciação Celular , Células Cultivadas , Citometria de Fluxo , Glucuronidase/biossíntese , Glucuronidase/metabolismo , Humanos , Imuno-Histoquímica , Miócitos de Músculo Liso/patologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , RNA/genética , Calcificação Vascular/metabolismo , Calcificação Vascular/patologia
17.
JACC Basic Transl Sci ; 3(4): 464-480, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30175270

RESUMO

Clinical tools to identify individuals with unstable atherosclerotic lesions are required to improve prevention of myocardial infarction and ischemic stroke. Here, a systems-based analysis of atherosclerotic plaques and plasma from patients undergoing carotid endarterectomy for stroke prevention was used to identify molecular signatures with a causal relationship to disease. Local plasma collected in the lesion proximity following clamping prior to arteriotomy was profiled together with matched peripheral plasma. This translational workflow identified biliverdin reductase B as a novel marker of intraplaque hemorrhage and unstable carotid atherosclerosis, which should be investigated as a potential predictive biomarker for cardiovascular events in larger cohorts.

18.
Maturitas ; 96: 39-44, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28041593

RESUMO

OBJECTIVE: Male sex is a significant risk factor for abdominal aortic aneurysm (AAA). Female sex hormones have been reported to prevent aneurysm formation in animal models. The study aims to describe the expression profile of sex hormone receptors in the aneurysm wall of men and women with AAA and compare with unaffected controls. METHODS: Aneurysm wall biopsies were obtained during elective open repair of AAA in men and women (n=16+16). Aortic vessel wall from controls were obtained at organ donation (n=6). Western blot-, mRNA expression- and immunohistochemical analyses were performed to assess the expression profile of the sex hormone receptors - androgen receptor (AR), progesterone receptor (PR), estrogen receptor α (ERα) and ß (ERß). RESULTS: The mRNA- and protein expression levels of AR were higher in AAA compared with control aorta (7.26 vs. 5.14, P=0.001). mRNA- and protein expression levels of ERß were lower in AAA compared with control aorta (9.15 vs. 12.29, P<0.001). mRNA expression levels of PR were higher in AAA compared with control aorta (8.73 vs. 6.21, P=0.003), but could not be confirmed on protein level. The expression profile of sex hormone receptors in men and women with AAA was similar. CONCLUSION: Expression of sex hormone receptors differs in the aneurysmal aorta compared with unaffected aorta in men and women. A higher expression of AR and a lower expression of ERß suggest that sex hormone activity could be associated with aneurysm development.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Receptor alfa de Estrogênio/análise , Receptor beta de Estrogênio/análise , Receptores Androgênicos/análise , Receptores de Progesterona/análise , Idoso , Aorta/química , Estudos de Casos e Controles , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Receptores Androgênicos/genética , Receptores de Progesterona/genética , Fatores de Risco , Fatores Sexuais
19.
J Cardiovasc Pharmacol ; 69(2): 101-109, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27875385

RESUMO

Diabetic patients suffer an increased risk of restenosis and late stent thrombosis after angioplasty, complications which are related to a defective reendothelialization. Dipeptidyl peptidase-4 inhibitors have been suggested to exert a direct effect on endothelial and smooth muscle cells (SMCs). Therefore, the objective was to study if the dipeptidyl peptidase-4 inhibitor linagliptin could influence vascular repair and accelerate reendothelialization after arterial injury in healthy and diabetic animals. Diabetic Goto-Kakizaki and healthy Wistar rats were subjected to arterial injury and treated with linagliptin or vehicle. Vessel wall healing was monitored noninvasively using ultrasound, and on sacrifice, with Evans blue staining and immunohistochemistry. The effect of linagliptin on SMCs was also studied in vitro. We found that linagliptin reduced the proliferation and dedifferentiation of SMCs in vitro, and modulated the inflammatory response in the SMCs after arterial injury in vivo. However, these effects of linagliptin did not affect the neointima formation or the reendothelialization under normal and diabetic conditions. Although linagliptin did not influence vessel wall healing, it seems to possess a desirable antiproliferative influence on SMCs in vitro and an antiinflammatory effect in vivo. These pharmacological properties might carry a potential significance for favorable outcome after vascular interventions in diabetic patients.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Artéria Carótida Externa/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Linagliptina/uso terapêutico , Cicatrização/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Lesões das Artérias Carótidas/sangue , Artéria Carótida Externa/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Diabetes Mellitus Tipo 2/sangue , Relação Dose-Resposta a Droga , Hipoglicemiantes/farmacologia , Linagliptina/farmacologia , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Resultado do Tratamento , Cicatrização/fisiologia
20.
Arterioscler Thromb Vasc Biol ; 36(9): 1947-61, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27470516

RESUMO

OBJECTIVE: Key augmented processes in atherosclerosis have been identified, whereas less is known about downregulated pathways. Here, we applied a systems biology approach to examine suppressed molecular signatures, with the hypothesis that they may provide insight into mechanisms contributing to plaque stability. APPROACH AND RESULTS: Muscle contraction, muscle development, and actin cytoskeleton were the most downregulated pathways (false discovery rate=6.99e-21, 1.66e-6, 2.54e-10, respectively) in microarrays from human carotid plaques (n=177) versus healthy arteries (n=15). In addition to typical smooth muscle cell (SMC) markers, these pathways also encompassed cytoskeleton-related genes previously not associated with atherosclerosis. SYNPO2, SYNM, LMOD1, PDLIM7, and PLN expression positively correlated to typical SMC markers in plaques (Pearson r>0.6, P<0.0001) and in rat intimal hyperplasia (r>0.8, P<0.0001). By immunohistochemistry, the proteins were expressed in SMCs in normal vessels, but largely absent in human plaques and intimal hyperplasia. Subcellularly, most proteins localized to the cytoskeleton in cultured SMCs and were regulated by active enhancer histone modification H3K27ac by chromatin immunoprecipitation-sequencing. Functionally, the genes were downregulated by PDGFB (platelet-derived growth factor beta) and IFNg (interferron gamma), exposure to shear flow stress, and oxLDL (oxidized low-density lipoprotein) loading. Genetic variants in PDLIM7, PLN, and SYNPO2 loci associated with progression of carotid intima-media thickness in high-risk subjects without symptoms of cardiovascular disease (n=3378). By eQTL (expression quantitative trait locus), rs11746443 also associated with PDLIM7 expression in plaques. Mechanistically, silencing of PDLIM7 in vitro led to downregulation of SMC markers and disruption of the actin cytoskeleton, decreased cell spreading, and increased proliferation. CONCLUSIONS: We identified a panel of genes that reflect the altered phenotype of SMCs in vascular disease and could be early sensitive markers of SMC dedifferentiation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autoantígenos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Doenças das Artérias Carótidas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas dos Microfilamentos/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Autoantígenos/genética , Proteínas de Ligação ao Cálcio/genética , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Artérias Carótidas/fisiopatologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Doenças das Artérias Carótidas/fisiopatologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Estudos de Casos e Controles , Desdiferenciação Celular , Células Cultivadas , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Regulação para Baixo , Estudos de Associação Genética , Humanos , Proteínas de Filamentos Intermediários/genética , Proteínas com Domínio LIM/genética , Masculino , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/patologia , Neointima , Fenótipo , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo , Transfecção , Vasoconstrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...