Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Erna Peters is active.

Publication


Featured researches published by Erna Peters.


Circulation Research | 2014

Inhibition of 14q32 MicroRNAs miR-329, miR-487b, miR-494, and miR-495 Increases Neovascularization and Blood Flow Recovery After Ischemia

Sabine M.J. Welten; A.J.N.M. Bastiaansen; Rob C. M. de Jong; Margreet R. de Vries; Erna Peters; Martin C. Boonstra; Søren Paludan Sheikh; Nicola La Monica; Ekambar R. Kandimalla; Paul H.A. Quax; A. Yaël Nossent

Rationale: Effective neovascularization is crucial for recovery after cardiovascular events. Objective: Because microRNAs regulate expression of up to several hundred target genes, we set out to identify microRNAs that target genes in all pathways of the multifactorial neovascularization process. Using www.targetscan.org, we performed a reverse target prediction analysis on a set of 197 genes involved in neovascularization. We found enrichment of binding sites for 27 microRNAs in a single microRNA gene cluster. Microarray analyses showed upregulation of 14q32 microRNAs during neovascularization in mice after single femoral artery ligation. Methods and Results: Gene silencing oligonucleotides (GSOs) were used to inhibit 4 14q32 microRNAs, miR-329, miR-487b, miR-494, and miR-495, 1 day before double femoral artery ligation. Blood flow recovery was followed by laser Doppler perfusion imaging. All 4 GSOs clearly improved blood flow recovery after ischemia. Mice treated with GSO-495 or GSO-329 showed increased perfusion already after 3 days (30% perfusion versus 15% in control), and those treated with GSO-329 showed a full recovery of perfusion after 7 days (versus 60% in control). Increased collateral artery diameters (arteriogenesis) were observed in adductor muscles of GSO-treated mice, as well as increased capillary densities (angiogenesis) in the ischemic soleus muscle. In vitro, treatment with GSOs led to increased sprout formation and increased arterial endothelial cell proliferation, as well as to increased arterial myofibroblast proliferation. Conclusions: The 14q32 microRNA gene cluster is highly involved in neovascularization. Inhibition of 14q32 microRNAs miR-329, miR-487b, miR-494, and miR-495 provides a promising tool for future therapeutic neovascularization.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2005

CD34+ Cells Home, Proliferate, and Participate in Capillary Formation, and in Combination With CD34− Cells Enhance Tube Formation in a 3-Dimensional Matrix

Maarten B. Rookmaaker; Marianne C. Verhaar; Cindy J.M. Loomans; Robert Verloop; Erna Peters; Peter E. Westerweel; Toyoaki Murohara; Frank J. T. Staal; Anton Jan van Zonneveld; Pieter Koolwijk; Ton J. Rabelink; Victor W.M. van Hinsbergh

Objective—Emerging evidence suggests that human blood contains bone marrow (BM)-derived endothelial progenitor cells that contribute to postnatal neovascularization. Clinical trials demonstrated that administration of BM-cells can enhance neovascularization. Most studies, however, used crude cell populations. Identifying the role of different cell populations is important for developing improved cellular therapies. Methods and Results—Effects of the hematopoietic stem cell–containing CD34+ cell population on migration, proliferation, differentiation, stimulation of, and participation in capillary-like tubule formation were assessed in an in vitro 3-dimensional matrix model using human microvascular endothelial cells. During movement over the endothelial monolayer, CD34+ cells remained stuck at sites of capillary tube formation and time- and dose-dependently formed cell clusters. Immunohistochemistry confirmed homing and proliferation of CD34+ cells in and around capillary sprouts. CD34+ cells were transduced with the LNGFR marker gene to allow tracing. LNGFR gene–transduced CD34+ cells integrated in the tubular structures and stained positive for CD31 and UEA-1. CD34+ cells alone stimulated neovascularization by 17%. Coculture with CD34− cells led to 68% enhancement of neovascularization, whereas CD34− cells displayed a variable response by themselves. Cell–cell contact between CD34+ and CD34− cells facilitated endothelial differentiation of CD34+ cells. Conclusions—Our data suggest that administration of CD34+-enriched cell populations may significantly improve neovascularization and point at an important supportive role for (endogenous or exogenous) CD34− cells.


Circulation Research | 2013

Quaking, an RNA-Binding Protein, Is a Critical Regulator of Vascular Smooth Muscle Cell Phenotype

Eric P. van der Veer; Ruben G. de Bruin; Adriaan O. Kraaijeveld; Margreet R. de Vries; Ilze Bot; Tonio Pera; Filip M. Segers; Stella Trompet; Janine M. van Gils; Marko K. Roeten; Cora Beckers; Peter J. van Santbrink; Anique Janssen; Coen van Solingen; Jim Swildens; Hetty C. de Boer; Erna Peters; Roel Bijkerk; Mat Rousch; Merijn Doop; Johan Kuiper; Martin J. Schalij; Allard C. van der Wal; Stéphane Richard; Theo J.C. van Berkel; J. Geoffrey Pickering; Pieter S. Hiemstra; Marie-José Goumans; Ton J. Rabelink; Antoine A.F. de Vries

Rationale: RNA-binding proteins are critical post-transcriptional regulators of RNA and can influence pre-mRNA splicing, RNA localization, and stability. The RNA-binding protein Quaking (QKI) is essential for embryonic blood vessel development. However, the role of QKI in the adult vasculature, and in particular in vascular smooth muscle cells (VSMCs), is currently unknown. Objective: We sought to determine the role of QKI in regulating adult VSMC function and plasticity. Methods and Results: We identified that QKI is highly expressed by neointimal VSMCs of human coronary restenotic lesions, but not in healthy vessels. In a mouse model of vascular injury, we observed reduced neointima hyperplasia in Quaking viable mice, which have decreased QKI expression. Concordantly, abrogation of QKI attenuated fibroproliferative properties of VSMCs, while potently inducing contractile apparatus protein expression, rendering noncontractile VSMCs with the capacity to contract. We identified that QKI localizes to the spliceosome, where it interacts with the myocardin pre-mRNA and regulates the splicing of alternative exon 2a. This post-transcriptional event impacts the Myocd_v3/Myocd_v1 mRNA balance and can be modulated by mutating the quaking response element in exon 2a of myocardin. Furthermore, we identified that arterial damage triggers myocardin alternative splicing and is tightly coupled with changes in the expression levels of distinct QKI isoforms. Conclusions: We propose that QKI is a central regulator of VSMC phenotypic plasticity and that intervention in QKI activity can ameliorate pathogenic, fibroproliferative responses to vascular injury.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Lysine Acetyltransferase PCAF Is a Key Regulator of Arteriogenesis

A.J.N.M. Bastiaansen; Mark M Ewing; Hetty C. de Boer; Tineke C. T. M. van der Pouw Kraan; Margreet R. de Vries; Erna Peters; Sabine M.J. Welten; Ramon Arens; Scott M. Moore; James E. Faber; J. Wouter Jukema; Jaap F. Hamming; A. Yaël Nossent; Paul H.A. Quax

Objective—Therapeutic arteriogenesis, that is, expansive remodeling of preexisting collaterals, using single-action factor therapies has not been as successful as anticipated. Modulation of factors that act as a master switch for relevant gene programs may prove more effective. Transcriptional coactivator p300-CBP–associated factor (PCAF) has histone acetylating activity and promotes transcription of multiple inflammatory genes. Because arteriogenesis is an inflammation-driven process, we hypothesized that PCAF acts as multifactorial regulator of arteriogenesis. Approach and Results—After induction of hindlimb ischemia, blood flow recovery was impaired in both PCAF−/− mice and healthy wild-type mice treated with the pharmacological PCAF inhibitor Garcinol, demonstrating an important role for PCAF in arteriogenesis. PCAF deficiency reduced the in vitro inflammatory response in leukocytes and vascular cells involved in arteriogenesis. In vivo gene expression profiling revealed that PCAF deficiency results in differential expression of 3505 genes during arteriogenesis and, more specifically, in impaired induction of multiple proinflammatory genes. Additionally, recruitment from the bone marrow of inflammatory cells, in particular proinflammatory Ly6Chi monocytes, was severely impaired in PCAF−/− mice. Conclusions—These findings indicate that PCAF acts as master switch in the inflammatory processes required for effective arteriogenesis.


PLOS ONE | 2013

TLR accessory molecule RP105 (CD180) is involved in post-interventional vascular remodeling and soluble RP105 modulates neointima formation

Jacco C. Karper; Mark M Ewing; Margreet R. de Vries; Saskia C.A. de Jager; Erna Peters; Hetty C. de Boer; Anton-Jan van Zonneveld; Johan Kuiper; Eric G. Huizinga; T. Harma C. Brondijk; J. Wouter Jukema; Paul H.A. Quax

Background RP105 (CD180) is TLR4 homologue lacking the intracellular TLR4 signaling domain and acts a TLR accessory molecule and physiological inhibitor of TLR4-signaling. The role of RP105 in vascular remodeling, in particular post-interventional remodeling is unknown. Methods and Results TLR4 and RP105 are expressed on vascular smooth muscle cells (VSMC) as well as in the media of murine femoral artery segments as detected by qPCR and immunohistochemistry. Furthermore, the response to the TLR4 ligand LPS was stronger in VSMC from RP105−/− mice resulting in a higher proliferation rate. In RP105−/− mice femoral artery cuff placement resulted in an increase in neointima formation as compared to WT mice (4982±974 µm2 vs.1947±278 µm2,pu200a=u200a0.0014). Local LPS application augmented neointima formation in both groups, but in RP105−/− mice this effect was more pronounced (10316±1243 µm2 vs.4208±555 µm2,pu200a=u200a0.0002), suggesting a functional role for RP105. For additional functional studies, the extracellular domain of murine RP105 was expressed with or without its adaptor protein MD1 and purified. SEC-MALSanalysis showed a functional 2∶2 homodimer formation of the RP105-MD1 complex. This protein complex was able to block the TLR4 response in whole blood ex-vivo. In vivo gene transfer of plasmid vectors encoding the extracellular part of RP105 and its adaptor protein MD1 were performed to initiate a stable endogenous soluble protein production. Expression of soluble RP105-MD1 resulted in a significant reduction in neointima formation in hypercholesterolemic mice (2500±573 vs.6581±1894 µm2,p<0.05), whereas expression of the single factors RP105 or MD1 had no effect. Conclusion RP105 is a potent inhibitor of post-interventional neointima formation.


Anti-Cancer Drugs | 2008

Alkylphospholipids inhibit capillary-like endothelial tube formation in vitro: Antiangiogenic properties of a new class of antitumor agents

S.F. Zerp; Stefan R. Vink; Gerald A. Ruiter; Pieter Koolwijk; Erna Peters; Arnold H. van der Luit; Daphne de Jong; Marianne Budde; Harry Bartelink; Wim J. van Blitterswijk; Marcel Verheij

Synthetic alkylphospholipids (APLs), such as edelfosine, miltefosine and perifosine, constitute a new class of antineoplastic compounds with various clinical applications. Here we have evaluated the antiangiogenic properties of APLs. The sensitivity of three types of vascular endothelial cells (ECs) (bovine aortic ECs, human umbilical vein ECs and human microvascular ECs) to APL-induced apoptosis was dependent on the proliferative status of these cells and correlated with the cellular drug incorporation. Although confluent, nondividing ECs failed to undergo apoptosis, proliferating ECs showed a 3–4-fold higher uptake and significant levels of apoptosis after APL treatment. These findings raised the question of whether APLs interfere with new blood vessel formation. To test the antiangiogenic properties in vitro, we studied the effect of APLs using two different experimental models. The first one tested the ability of human microvascular ECs to invade a three-dimensional human fibrin matrix and form capillary-like tubular networks. In the second model, bovine aortic ECs were grown in a collagen gel sandwich to allow tube formation. We found that all three APLs interfered with endothelial tube formation in a dose-dependent manner, with a more than 50% reduction at 25u2009μmol/l. Interference with the angiogenic process represents a novel mode of action of APLs and might significantly contribute to the antitumor effect of these compounds.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Blocking Toll-Like Receptors 7 and 9 Reduces Postinterventional Remodeling via Reduced Macrophage Activation, Foam Cell Formation, and Migration

Jacco C. Karper; Mark M Ewing; Kim L.L. Habets; Margreet R. de Vries; Erna Peters; Annemarie M. van Oeveren-Rietdijk; Hetty C. de Boer; Jaap F. Hamming; Johan Kuiper; Ekambar R. Kandimalla; Nicola La Monica; J. Wouter Jukema; Paul H.A. Quax

Objective—The role of toll-like receptors (TLRs) in vascular remodeling is well established. However, the involvement of the endosomal TLRs is unknown. Here, we study the effect of combined blocking of TLR7 and TLR9 on postinterventional remodeling and accelerated atherosclerosis. Methods and Results—In hypercholesterolemic apolipoprotein E*3-Leiden mice, femoral artery cuff placement led to strong increase of TLR7 and TLR9 presence demonstrated by immunohistochemistry. Blocking TLR7/9 with a dual antagonist in vivo reduced neointimal thickening and foam cell accumulation 14 days after surgery by 65.6% (P=0.0079). Intima/media ratio was reduced by 64.5% and luminal stenosis by 62.8%. The TLR7/9 antagonist reduced the arterial wall inflammation, with reduced macrophage infiltration, decreased cytoplasmic high-mobility group box 1 expression, and altered serum interleukin-10 levels. Stimulation of cultured macrophages with TLR7 and TLR9 ligands enhanced tumor necrosis factor-&agr; expression, which is decreased by TLR7/9 antagonist coadministration. Additionally, the antagonist abolished the TLR7/9-enhanced low-density lipoprotein uptake. The antagonist also reduced oxidized low-density lipoprotein–induced foam cell formation, most likely not via decreased influx but via increased efflux, because CD36 expression was unchanged whereas interleukin-10 levels were higher (36.1±22.3 pg/mL versus 128.9±6.6 pg/mL; P=0.008). Conclusion—Blocking TLR7 and TLR9 reduced postinterventional vascular remodeling and foam cell accumulation indicating TLR7 and TLR9 as novel therapeutic targets.


PLOS ONE | 2014

TLR4 Accessory Molecule RP105 (CD180) Regulates Monocyte-Driven Arteriogenesis in a Murine Hind Limb Ischemia Model

A.J.N.M. Bastiaansen; Jacco C. Karper; Anouk Wezel; Hetty C. de Boer; Sabine M.J. Welten; Rob C. M. de Jong; Erna Peters; Margreet R. de Vries; Annemarie M. van Oeveren-Rietdijk; Anton Jan van Zonneveld; Jaap F. Hamming; A. Yaël Nossent; Paul H.A. Quax

Aims We investigated the role of the TLR4-accessory molecule RP105 (CD180) in post-ischemic neovascularization, i.e. arteriogenesis and angiogenesis. TLR4-mediated activation of pro-inflammatory Ly6Chi monocytes is crucial for effective neovascularization. Immunohistochemical analyses revealed that RP105+ monocytes are present in the perivascular space of remodeling collateral arterioles. As RP105 inhibits TLR4 signaling, we hypothesized that RP105 deficiency would lead to an unrestrained TLR4-mediated inflammatory response and hence to enhanced blood flow recovery after ischemia. Methods and Results RP105−/− and wild type (WT) mice were subjected to hind limb ischemia and blood flow recovery was followed by Laser Doppler Perfusion Imaging. Surprisingly, we found that blood flow recovery was severely impaired in RP105−/− mice. Immunohistochemistry showed that arteriogenesis was reduced in these mice compared to the WT. However, both in vivo and ex vivo analyses showed that circulatory pro-arteriogenic Ly6Chi monocytes were more readily activated in RP105−/− mice. FACS analyses showed that Ly6Chi monocytes became activated and migrated to the affected muscle tissues in WT mice following induction of hind limb ischemia. Although Ly6Chi monocytes were readily activated in RP105−/− mice, migration into the ischemic tissues was hampered and instead, Ly6Chi monocytes accumulated in their storage compartments, bone marrow and spleen, in RP105−/− mice. Conclusions RP105 deficiency results in an unrestrained inflammatory response and monocyte over-activation, most likely due to the lack of TLR4 regulation. Inappropriate, premature systemic activation of pro-inflammatory Ly6Chi monocytes results in reduced infiltration of Ly6Chi monocytes in ischemic tissues and in impaired blood flow recovery.


Scientific Reports | 2016

Deficiency of the TLR4 analogue RP105 aggravates vein graft disease by inducing a pro-inflammatory response

Anouk Wezel; Margreet R. de Vries; Johanna M. Maassen; Peter Kip; Erna Peters; Jacco C. Karper; Johan Kuiper; Ilze Bot; Paul H.A. Quax

Venous grafts are often used to bypass occlusive atherosclerotic lesions; however, poor patency leads to vein graft disease. Deficiency of TLR4, an inflammatory regulator, reduces vein graft disease. Here, we investigate the effects of the accessory molecule and TLR4 analogue RadioProtective 105 (RP105) on vein graft disease. RP105 deficiency resulted in a 90% increase in vein graft lesion area compared to controls. In a hypercholesterolemic setting (LDLr−/−/RP105−/− versus LDLr−/− mice), which is of importance as vein graft disease is usually characterized by excessive atherosclerosis, total lesion area was not affected. However we did observe an increased number of unstable lesions and intraplaque hemorrhage upon RP105 deficiency. In both setups, lesional macrophage content, and lesional CCL2 was increased. In vitro, RP105−/− smooth muscle cells and mast cells secreted higher levels of CCL2. In conclusion, aggravated vein graft disease caused by RP105 deficiency results from an increased local inflammatory response.


Thrombosis and Haemostasis | 2003

Aminopeptidase inhibitor bestatin stimulates microvascular endothelial cell invasion in a fibrin matrix

Yvette van Hensbergen; Henk J. Broxterman; Erna Peters; Sareena Rana; Yvonne W. Elderkamp; Victor W.M. van Hinsbergh; Pieter Koolwijk

The aminopeptidase inhibitor bestatin has been shown to have anti-angiogenic effects in a number of model systems. These effects are thought to result from inhibition of CD13 activity. Because tumor angiogenesis can evolve in a fibrin-rich stroma matrix we have studied for the first time the effects of bestatin on microvascular endothelial capillary-like tube formation in a fibrin matrix. Bestatin enhanced the formation of capillary-like tubes dose-dependently. Its effects were apparent at 8 micro M; the increase was 3.7-fold at 125 micro M; while high concentrations (>250 micro M), that were shown to have anti-angiogenic effects in other systems, caused extensive matrix degradation. Specific CD13-blocking antibodies WM15 and MY-7, and the aminopeptidase inhibitors amastatin and actinonin also enhanced capillary-like tube formation (maximally 1.5-fold), but these effects did not reach statistical significance. The effect of bestatin was not due to a change in uPAR availability because the relative involvement of the u-PA/u-PAR activity was not altered by bestatin. In view of the present findings we hypothesize that aminopeptidases other than CD13 predominantly contribute to the observed pro-angiogenic effect of bestatin in a fibrin matrix. The identification of this novel effect of bestatin is important in the light of the proposed use of bestatin as anti-angiogenic and/or anti-tumor agent.

Collaboration


Dive into the Erna Peters's collaboration.

Top Co-Authors

Avatar

Paul H.A. Quax

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sabine M.J. Welten

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jacco C. Karper

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Margreet R. de Vries

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hetty C. de Boer

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jaap F. Hamming

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pieter Koolwijk

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

A. Yaël Nossent

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

A.J.N.M. Bastiaansen

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge