SwePub
Tyck till om SwePub Sök här!
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Suur Bianca E) "

Sökning: WFRF:(Suur Bianca E)

  • Resultat 1-5 av 5
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Folkersen, Lasse, et al. (författare)
  • Genomic and drug target evaluation of 90 cardiovascular proteins in 30,931 individuals.
  • 2020
  • Ingår i: Nature metabolism. - : Springer Science and Business Media LLC. - 2522-5812. ; 2:10, s. 1135-1148
  • Tidskriftsartikel (refereegranskat)abstract
    • Circulating proteins are vital in human health and disease and are frequently used as biomarkers for clinical decision-making or as targets for pharmacological intervention. Here, we map and replicate protein quantitative trait loci (pQTL) for 90 cardiovascular proteins in over 30,000 individuals, resulting in 451 pQTLs for 85 proteins. For each protein, we further perform pathway mapping to obtain trans-pQTL gene and regulatory designations. We substantiate these regulatory findings with orthogonal evidence for trans-pQTLs using mouse knockdown experiments (ABCA1 and TRIB1) and clinical trial results (chemokine receptors CCR2 and CCR5), with consistent regulation. Finally, we evaluate known drug targets, and suggest new target candidates or repositioning opportunities using Mendelian randomization. This identifies 11 proteins with causal evidence of involvement in human disease that have not previously been targeted, including EGF, IL-16, PAPPA, SPON1, F3, ADM, CASP-8, CHI3L1, CXCL16, GDF15 and MMP-12. Taken together, these findings demonstrate the utility of large-scale mapping of the genetics of the proteome and provide a resource for future precision studies of circulating proteins in human health.
  •  
2.
  • Rykaczewska, Urszula, et al. (författare)
  • PCSK6 Is a Key Protease in the Control of Smooth Muscle Cell Function in Vascular Remodeling
  • 2020
  • Ingår i: Circulation Research. - : LIPPINCOTT WILLIAMS & WILKINS. - 0009-7330 .- 1524-4571. ; 126:5, s. 571-585
  • Tidskriftsartikel (refereegranskat)abstract
    • 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.
  •  
3.
  • Röhl, Samuel, et al. (författare)
  • Transcriptomic profiling of experimental arterial injury reveals new mechanisms and temporal dynamics in vascular healing response
  • 2020
  • Ingår i: JVS-Vascular Science. - : Elsevier BV. - 2666-3503. ; 315, s. E14-E14
  • Tidskriftsartikel (refereegranskat)abstract
    • 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.
  •  
4.
  • Suur, Bianca E (författare)
  • The role of proprotein convertases in vascular disease
  • 2022
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • To date, the role of the proprotein convertase subtilisin/kexin (PCSK) family in vascular disease is largely unknown, with the exception of PCSK9 and its key function in lipid metabolism. The PCSK family constitutes of 9 different proteases, of which the first 7 show remarkable structural and functional overlap. While the first 7 members have not been thoroughly investigated in vascular disease, they have been extensively studied in cancer. Primarily, they have been shown to proteolytically cleave various substrates, including cytokines, growth factors, hormones and other proteases, which are all typically involved in vascular disease as well. The only PCSK that has received broad attention in the cardiovascular field before the initiation of this Ph.D. project was PCSK9, due to its involvement in lipid metabolism through regulation of the surface expression of the low-density lipoprotein receptor on hepatocytes. Previously, we demonstrated upregulation of PCSK6 in atherosclerotic plaques, particularly those from symptomatic patients, localization to smooth muscle cells (SMCs) in the fibrous cap and positive correlations with inflammation, extracellular matrix (ECM) remodeling and cytokines. In this Ph.D. thesis we aimed to further expand upon this finding and to investigate the expression and localization of the entire PCSK family in vascular disease, as well as their mechanisms and translational potential. In Study I we hypothesized that PCSK6 could be involved in flow-mediated vascular remodeling. Using various in vivo and ex vivo models, we evaluated the role of PCSK6 in the physiology of this process. Pcsk6-/- and control mice were subjected to carotid ligation of the left common carotid artery, resulting in increased blood flow in the contralateral right common carotid artery. Using ultrasound and ex vivo wire myography, we detected increased flow in the right common carotid artery, concomitant with a progressive increase in vascular circumference in the knockout mice, but not in control mice. Furthermore, these changes were accompanied by a reduction in active tension in knockout mice, while control vessels exhibited an increase in active tension over time. On a cellular level, these changes could be attributed to decreased expression of the typical contractile SMC markers, SMA, MYH11 and LMOD1. In Study II we aimed to elucidate the mechanistic role of PCSK6 in SMCs in the context of vascular injury, a process leading to the disruption of ECM and release of the matrix-bound growth factors TGF-b1 and PDGF-BB, also implicated in the early stages of atherosclerosis. Using various human biobanks, we investigated the genetic locus surrounding the PCSK6 gene and identified one SNP in particular, rs1531817 to be associated with cIMT progression, a surrogate marker for atherosclerosis. Next, our previously published results were further extended using microarray analysis of end-stage carotid plaques as well as several publicly available datasets, all showing increased PCSK6 expression in diseased tissues. PCSK6 protein was located particularly to SMCs in the fibrous cap, remnants of the media and mature neovessels. In the rat carotid artery balloon injury model, Pcsk6 expression was low in the initial timepoints, but increasing over-time to reach the peak at day 5 after injury. Based on microarray analysis, we uncovered positive correlations on day 5 after injury between Pcsk6 and matrix metalloproteases Mmp2 and Mmp14. Neointima formation in response to carotid artery ligation was found to be decreased in Pcsk6-/- mice compared to control, concomitant with decreased MMP14 activity. Taken together, our results implicate PCSK6 as a key protease in modulating SMC control in vascular injury. Mechanistically, this likely happens through activation of the PCSK6-MMP14-MMP2 axis, allowing for dedifferentiation of SMCs, degradation of the surrounding ECM and subsequent migration out of the media to contribute to the neointima formation. Expression and localization of PCSK6 was also associated positively with typical markers of T lymphocytes and macrophages in atherosclerotic plaques. Since PCSK6 is highly expressed in healthy human spleens, this together led to a hypothesis for Study III that PCSK6 may be involved in modulating immune responses, particularly in vascular diseases. At baseline, Pcks6-/- plasma showed enrichment of pro-inflammatory cytokines CCL2, CCL3 and in particular IL-17A and IL-17F. Pcks6-/- spleens had an increased number of germinal centers and contained significantly more CD8+ T cells. Microarray analysis of spleens confirmed that expression of T cell markers Cd4, Cd3e and Cd3g was upregulated in Pcks6-/- mice vs. controls. In vitro, splenocytes isolated from Pcks6-/- mice secreted higher levels of IFN-g, IL-2, IL-17 and IL-10 upon stimulation with a-CD3 and a-CD28 antibodies, and were more proliferative. Moreover, peritoneal macrophages from Pcks6-/- mice secreted more TNF-a, CCL2, IL-6 and IL-10 upon LPS stimulation. Bone marrow-derived macrophages from Pcks6- /- mice were also prone to lipid uptake. Finally, in vivo transplantation of Pcks6-/- bone marrow to Ldlr-/- mice led to increased atherosclerotic plaque burden compared to controls. However, these plaques were more stable, attributed to increased collagen deposition, SMC presence and IL-17 content. In Study IV we aimed to evaluate the vascular expression and cellular localization of all the members of the PCSK family and assess their therapeutic targeting potential in this context. To first establish a genetic link between the PCSKs and CVD phenotypes, we interrogated publicly available data for any SNPs surrounding the PCSK genetic loci. With the exception of PCSK4, the genetic locus surrounding every PCSK contained SNPs associated with CVD. To further assess the expression and localization of PCSKs within tissue biopsies of pathologies underlying cardiovascular disease, we interrogated three large-scale, independent vascular disease biobanks. We found that PCSK1, PCSK2, PCSK4 and PCSK9 were expressed only in moderate levels in disease biopsies and did not observe differences in expression between healthy and diseased tissues. In carotid plaques vs. control arteries FURIN, PCSK5, MBTPS1 were downregulated, while PCSK6 and 7 were upregulated. In abdominal aneurysms, FURIN, PCSK5, PCSK7, MBTPS1 were downregulated, while PCSK6 was enriched in diseased media. In thoracic aneurysms, only FURIN was significantly upregulated. Cell type correlation analyses and immunohistochemistry showed that PCSK transcripts and protein levels parallel each other, except for PCSK9 where the transcript was not detected, while protein was abundant in vascular biopsies. Integrating our results lead to the development of a novel ‘molecular’ 5D framework for early evaluation of target druggability potential. Our results using this translational pipeline revealed primarily PCSK6, followed by PCSK5, PCSK7 and FURIN, as proprotein convertases with the highest novel therapeutic potential. Overall, in this thesis we have identified PCSK6 as one of the key players in vascular disease and assessed in detail its function and mechanisms in vascular injury, atherosclerosis and aneurysms. Moreover, we evaluated the expression and localization of the entire PCSK family in various vascular disease pathologies. From a translational perspective, a novel druggability scoring system or ‘molecular’ 5D framework was constructed, aimed at early molecular evaluation of potential novel drug targets in the context of cardiovascular disease.
  •  
5.
  • Suur, Bianca E., et al. (författare)
  • Therapeutic potential of the Proprotein Convertase Subtilisin/Kexin family in vascular disease
  • 2022
  • Ingår i: Frontiers in Pharmacology. - : Frontiers Media SA. - 1663-9812. ; 13
  • Tidskriftsartikel (refereegranskat)abstract
    • Proprotein convertase subtilisin/kexins (PCSKs) constitute a family of nine related proteases: PCSK1-7, MBTPS1, and PCSK9. Apart from PCSK9, little is known about PCSKs in cardiovascular disease. Here, we aimed to investigate the expression landscape and druggability potential of the entire PCSK family for CVD. We applied an integrative approach, combining genetic, transcriptomic and proteomic data from three vascular biobanks comprising carotid atherosclerosis, thoracic and abdominal aneurysms, with patient clinical parameters and immunohistochemistry of vascular biopsies. Apart from PCSK4, all PCSK family members lie in genetic regions containing variants associated with human cardiovascular traits. Transcriptomic analyses revealed that FURIN, PCSK5, MBTPS1 were downregulated, while PCSK6/7 were upregulated in plaques vs. control arteries. In abdominal aneurysms, FURIN, PCSK5, PCSK7, MBTPS1 were downregulated, while PCSK6 was enriched in diseased media. In thoracic aneurysms, only FURIN was significantly upregulated. Network analyses of the upstream and downstream pathways related to PCSKs were performed on the omics data from vascular biopsies, revealing mechanistic relationships between this protein family and disease. Cell type correlation analyses and immunohistochemistry showed that PCSK transcripts and protein levels parallel each other, except for PCSK9 where transcript was not detected, while protein was abundant in vascular biopsies. Correlations to clinical parameters revealed a positive association between FURIN plaque levels and serum LDL, while PCSK6 was negatively associated with Hb. PCSK5/6/7 were all positively associated with adverse cardiovascular events. Our results show that PCSK6 is abundant in plaques and abdominal aneurysms, while FURIN upregulation is characteristic for thoracic aneurysms. PCSK9 protein, but not the transcript, was present in vascular lesions, suggesting its accumulation from circulation. Integrating our results lead to the development of a novel 'molecular' 5D framework. Here, we conducted the first integrative study of the proprotein convertase family in this context. Our results using this translational pipeline, revealed primarily PCSK6, followed by PCSK5, PCSK7 and FURIN, as proprotein convertases with the highest novel therapeutic potential.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-5 av 5

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy