Network


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

Hotspot


Dive into the research topics where Hetty C. de Boer is active.

Publication


Featured researches published by Hetty C. de Boer.


Journal of Cellular and Molecular Medicine | 2009

Antagomir-mediated silencing of endothelial cell specific microRNA-126 impairs ischemia-induced angiogenesis.

Coen van Solingen; Leonard Seghers; Roel Bijkerk; Jacques M.G.J. Duijs; Marko K. Roeten; Annemarie M. van Oeveren-Rietdijk; Hans J. Baelde; Matthieu Monge; Joost B. Vos; Hetty C. de Boer; Paul H.A. Quax; Ton J. Rabelink; Anton Jan van Zonneveld

MicroRNAs are negative regulators of gene expression that play a key role in cell‐type specific differentiation and modulation of cell function and have been proposed to be involved in neovascularization. Previously, using an extensive cloning and sequencing approach, we identified miR‐126 to be specifically and highly expressed in human endothelial cells (EC). Here, we demonstrate EC‐specific expression of miR‐126 in capillaries and the larger vessels in vivo. We therefore explored the potential role of miR‐126 in arteriogenesis and angiogenesis. Using miR‐reporter constructs, we show that miR‐126 is functionally active in EC in vitro and that it could be specifically repressed using antagomirs specifically targeting miR‐126. To study the consequences of miR‐126 silencing on vascular regeneration, mice were injected with a single dose of antagomir‐126 or a control ‘scramblemir’ and exposed to ischemia of the left hindlimb by ligation of the femoral artery. Although miR‐126 was effectively silenced in mice treated with a single, high dose (HD) of antagomir‐126, laser Doppler perfusion imaging did not show effects on blood flow recovery. In contrast, quantification of the capillary density in the gastrocnemius muscle revealed that mice treated with a HD of antagomir‐126 had a markedly reduced angiogenic response. Aortic explant cultures of the mice confirmed the role of miR‐126 in angiogenesis. Our data demonstrate a facilitary function for miR‐126 in ischemia‐induced angiogenesis and show the efficacy and specificity of antagomir‐induced silencing of EC‐specific microRNAs in vivo.


Journal of Clinical Investigation | 2007

Shear stress–induced changes in atherosclerotic plaque composition are modulated by chemokines

Caroline Cheng; Dennie Tempel; Rien van Haperen; Hetty C. de Boer; Dolf Segers; Martin Huisman; Anton Jan van Zonneveld; Pieter J. M. Leenen; Anton F.W. van der Steen; Patrick W. Serruys; Rini de Crom; Rob Krams

We previously found that low shear stress (LSS) induces atherosclerotic plaques in mice with increased lipid and matrix metalloproteinase content and decreased vascular smooth muscle and collagen content. Here, we evaluated the role of chemokines in this process, using an extravascular device inducing regions of LSS, high shear stress, and oscillatory shear stress (OSS) in the carotid artery. One week of shear stress alterations induced expression of IFN-gamma-inducible protein-10 (IP-10) exclusively in the LSS region, whereas monocyte chemoattractant protein-1 (MCP-1) and the mouse homolog of growth-regulated oncogene alpha (GRO-alpha) were equally upregulated in both LSS and OSS regions. After 3 weeks, GRO-alpha and IP-10 were specifically upregulated in LSS regions. After 9 weeks, lesions with thinner fibrous caps and larger necrotic cores were found in the LSS region compared with the OSS region. Equal levels of MCP-1 expression were observed in both regions, while expression of fractalkine was found in the LSS region only. Blockage of fractalkine inhibited plaque growth and resulted in striking differences in plaque composition in the LSS region. We conclude that LSS or OSS triggers expression of chemokines involved in atherogenesis. Fractalkine upregulation is critically important for the composition of LSS-induced atherosclerotic lesions.


Aging Cell | 2006

Haematopoietic stem cells and endothelial progenitor cells in healthy men: effect of aging and training

Dick H. J. Thijssen; Joost B. Vos; Caroline Verseyden; Anton Jan van Zonneveld; Paul Smits; Fred C.G.J. Sweep; Maria T. E. Hopman; Hetty C. de Boer

The number of hematopoietic stem cells (HSC) and endothelial progenitor cells (EPC) is thought to be a marker for neovascularization and vascular repair. Because physical inactivity and aging are risk factors for cardiovascular diseases, these factors may influence the numbers of HSCs and EPCs. Therefore, we examined baseline and exercise‐induced levels of HSCs and EPCs in sedentary and trained young and older men. To study the role of aging in eight sedentary young (19–28 years) and eight sedentary older men (67–76 years), baseline and acute exercise‐induced numbers of HSCs (CD34+‐cells) and EPCs (CD34+/VEGFR‐2+‐cells) were quantified by fluorescence‐activated cell sorter (FACS) analysis. To examine the effect of chronic training, eight age‐matched trained young men (18–28 years) were compared with sedentary young men, whereas older men performed an 8‐week endurance training. Older men showed significantly lower baseline and exercise‐induced levels of HSCs/EPCs than the young men (P < 0.05). In young and older men, acute exercise significantly increased HSCs (P < 0.01), but not EPCs. The absolute increase in numbers of HSCs was attenuated in older men (P = 0.03). Apart from the lower baseline numbers of EPCs after chronic training in older men, training status did not alter baseline or exercise‐induced levels of HSCs/EPCs in young and older men. We concluded that advancing age results in lower circulating numbers of HSCs and EPCs and attenuates the acute exercise‐induced increase in HSCs. Interestingly, in young as well as in older men chronic endurance training does not affect baseline and exercise‐induced numbers of HSCs and EPCs.


Journal of Immunology | 2008

Properdin Binds to Late Apoptotic and Necrotic Cells Independently of C3b and Regulates Alternative Pathway Complement Activation

Wei Xu; Stefan P. Berger; Leendert A. Trouw; Hetty C. de Boer; Nicole Schlagwein; Chantal A. Mutsaers; Mohamed R. Daha; Cees van Kooten

Cells that undergo apoptosis or necrosis are promptly removed by phagocytes. Soluble opsonins such as complement can opsonize dying cells, thereby promoting their removal by phagocytes and modulating the immune response. The pivotal role of the complement system in the handling of dying cells has been demonstrated for the classical pathway (via C1q) and lectin pathway (via mannose-binding lectin and ficolin). Herein we report that the only known naturally occurring positive regulator of complement, properdin, binds predominantly to late apoptotic and necrotic cells, but not to early apoptotic cells. This binding occurs independently of C3b, which is additional to the standard model wherein properdin binds to preexisting clusters of C3b on targets and stabilizes the convertase C3bBb. By binding to late apoptotic or necrotic cells, properdin serves as a focal point for local amplification of alternative pathway complement activation. Furthermore, properdin exhibits a strong interaction with DNA that is exposed on the late stage of dying cells. Our data indicate that direct recognition of dying cells by properdin is essential to drive alternative pathway complement activation.


Nature Reviews Nephrology | 2010

Endothelial activation and circulating markers of endothelial activation in kidney disease

Ton J. Rabelink; Hetty C. de Boer; Anton Jan van Zonneveld

The recognition of a central role for the endothelium in the development of kidney disease or the development of vascular lesions in patients with established renal dysfunction has led to the emergence of methods to test different aspects of endothelium function, including in endothelium injury and repair. Endothelial-cell activation is associated with the shedding of components of the glycocalyx, adhesion molecules and endothelial microparticles into the circulation. This process may eventually result in the detachment of endothelial cells and recruitment of circulating myeloid and progenitor cells that are involved in vascular remodeling and repair. Circulating markers of endothelium activation may therefore represent novel markers of vessel wall injury. This Review describes the biology of these circulating markers of vessel wall injury, the methodologies used to measure them, and their possible relevance to patients with kidney disease.


Molecular Medicine | 2009

Differentiation of bone marrow-derived endothelial progenitor cells is shifted into a proinflammatory phenotype by hyperglycemia.

Cindy J.M. Loomans; Rien van Haperen; Jacques M.G.J. Duijs; C. Verseyden; Rini de Crom; Pieter J. M. Leenen; Hemmo A. Drexhage; Hetty C. de Boer; Eelco J.P. de Koning; Ton J. Rabelink; Frank J. T. Staal; Anton Jan van Zonneveld

Bone marrow (BM)-derived endothelial progenitor cells (EPC) contribute to vascular maintenance by participating in angiogenesis, re-endothelialization, and remodeling. Myeloid progenitor cells in the BM are functionally and quantitatively an important precursor pool for cells that contribute to these processes. However, these precursor pools in the BM also give rise to important effector cells of the innate immune system, such as macrophages and dendritic cells.We hypothesized that the disturbed repair responses that are being observed in diabetes mellitus are also related to an effect on functional and differentiation characteristics at the level of this bone marrow precursor pool. Indeed, we observed that bone marrow differentiation cultures for EPC, macrophages (Mph), or dendritic cells (DC) from hyperglycemic BM yielded 40% fewer EPC and 50% more Mph compared with control BM. These changes were directly related to the hemoglobin A1C levels of the donor mice. BM-derived DC numbers were not affected by hyperglycemia. The composition of the BM was not altered; in particular, the numbers of CD31+/Ly6C+ cells, which serve as common progenitors for EPC, Mph, and DC, were unaffected. In addition, BM-derived EPC from hyperglycemic mice were less angiogenic and more proinflammatory in regards to endocytosis, T-cell activation, and interleukin 12 production. HMG-CoA (3-hydroxy-3-methylglutaryl coenzyme A) reductase inhibition by statin supplementation of the culture medium counteracted these hyperglycemia-induced changes. Our study results show that hyperglycemia alters the differentiation fate of BM precursor cells, reducing the potential to generate vascular regenerative cells and favoring the development of proinflammatory cells.


European Heart Journal | 2013

Aspirin treatment hampers the use of plasma microRNA-126 as a biomarker for the progression of vascular disease

Hetty C. de Boer; Coen van Solingen; Jurriën Prins; Jacques M.G.J. Duijs; Menno V. Huisman; Ton J. Rabelink; Anton Jan van Zonneveld

AIMS MicroRNA-126 (miR-126) facilitates angiogenesis and regulates endothelial cell function. Recent data suggest that miR-126 can serve as a biomarker for vascular disease. Although endothelial cells are enriched for miR-126, platelets also contain miR-126. In this paper, we investigated the contribution of platelets to the pool of miR-126 in plasma of patients with type 2 diabetes (DM2) and how this is affected by aspirin. METHODS AND RESULTS In vitro platelet activation resulted in the transfer of miR-126 from the platelet to the plasma compartment, which was prevented by aspirin. In vivo platelet activation, monitored in patients with DM2 by measuring soluble P-selectin, correlated directly with circulating levels of miR-126. The administration of aspirin resulted both in platelet inhibition and concomitantly reduced circulating levels of platelet-derived microRNAs including miR-126. CONCLUSION Platelets are a major source of circulating miR-126. Consequently, in patho-physiological conditions associated with platelet activation, such as diabetes type 2, the administration of aspirin may lead to reduced levels of circulating miR-126. Thus, the use of platelet inhibitors should be taken into account when using plasma levels of miR-126 as a biomarker.


Cardiovascular Research | 2011

MicroRNA-126 modulates endothelial SDF-1 expression and mobilization of Sca-1+/Lin− progenitor cells in ischaemia

Coen van Solingen; Hetty C. de Boer; Roel Bijkerk; Matthieu Monge; Annemarie M. van Oeveren-Rietdijk; Leonard Seghers; Margreet R. de Vries; Eric P. van der Veer; Paul H.A. Quax; Ton J. Rabelink; Anton Jan van Zonneveld

AIMS MicroRNA-126 (miR-126), which is enriched in endothelial cells, plays a role in angiogenesis. Based on the seed sequence, miR-126 can also be predicted to regulate vasculogenesis by modulating the endothelial expression of stromal cell-derived factor-1 (SDF-1). METHODS AND RESULTS Using miR-reporter constructs, we first validated that miR-126 inhibits SDF-1 expression in endothelial cells in vitro. Next, we investigated the potential relevance of this observation with respect to the mobilization of progenitor cells. For this, we studied the migration of human CD34+ progenitor cells towards chemotactic factors present in endothelial cell-conditioned medium. Antagomir-induced silencing of miR-126 elevated SDF-1 expression by human umbilical vein endothelial cells and enhanced migration of the CD34+ cells. In a murine model of hind limb ischaemia, a striking increase in the number of circulating Sca-1(+)/Lin(-) progenitor cells in antagomir-126-treated mice was observed when compared with scramblemir-treated controls. Immunohistochemical staining of capillaries in the post-ischaemic gastrocnemius muscle of miR-126-silenced mice revealed elevated SDF-1 expressing CD31-positive capillaries, whereas a mobilizing effect of miR-126 inhibition was not detected in healthy control animals. CONCLUSION miR-126 can regulate the expression of SDF-1 in endothelial cells. In the context of an ischaemic event, systemic silencing of miR-126 leads to the mobilization of Sca-1(+)/Lin(-) progenitor cells into the peripheral circulation, potentially in response to elevated SDF-1 expression by endothelial cells present in the ischaemic tissue.


Journal of The American Society of Nephrology | 2014

Hematopoietic MicroRNA-126 Protects against Renal Ischemia/Reperfusion Injury by Promoting Vascular Integrity

Roel Bijkerk; Coen van Solingen; Hetty C. de Boer; Pieter van der Pol; Meriem Khairoun; Ruben G. de Bruin; Annemarie M. van Oeveren-Rietdijk; Ellen Lievers; Nicole Schlagwein; Daniëlle J. van Gijlswijk; Marko K. Roeten; Zeinab Neshati; Antoine A.F. de Vries; Mark Rodijk; Karin Pike-Overzet; Yascha W. van den Berg; Eric P. van der Veer; Henri H. Versteeg; Marlies E.J. Reinders; Frank J. T. Staal; Cees van Kooten; Ton J. Rabelink; Anton Jan van Zonneveld

Ischemia/reperfusion injury (IRI) is a central phenomenon in kidney transplantation and AKI. Integrity of the renal peritubular capillary network is an important limiting factor in the recovery from IRI. MicroRNA-126 (miR-126) facilitates vascular regeneration by functioning as an angiomiR and by modulating mobilization of hematopoietic stem/progenitor cells. We hypothesized that overexpression of miR-126 in the hematopoietic compartment could protect the kidney against IRI via preservation of microvascular integrity. Here, we demonstrate that hematopoietic overexpression of miR-126 increases neovascularization of subcutaneously implanted Matrigel plugs in mice. After renal IRI, mice overexpressing miR-126 displayed a marked decrease in urea levels, weight loss, fibrotic markers, and injury markers (such as kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin). This protective effect was associated with a higher density of the peritubular capillary network in the corticomedullary junction and increased numbers of bone marrow-derived endothelial cells. Hematopoietic overexpression of miR-126 increased the number of circulating Lin(-)/Sca-1(+)/cKit(+) hematopoietic stem and progenitor cells. Additionally, miR-126 overexpression attenuated expression of the chemokine receptor CXCR4 on Lin(-)/Sca-1(+)/cKit(+) cells in the bone marrow and increased renal expression of its ligand stromal cell-derived factor 1, thus favoring mobilization of Lin(-)/Sca-1(+)/cKit(+) cells toward the kidney. Taken together, these results suggest overexpression of miR-126 in the hematopoietic compartment is associated with stromal cell-derived factor 1/CXCR4-dependent vasculogenic progenitor cell mobilization and promotes vascular integrity and supports recovery of the kidney after IRI.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Annexin A5 Therapy Attenuates Vascular Inflammation and Remodeling and Improves Endothelial Function in Mice

Mark M Ewing; Margreet R. de Vries; Mariette Nordzell; Knut Pettersson; Hetty C. de Boer; Anton Jan van Zonneveld; Johan Frostegård; J. Wouter Jukema; Paul H.A. Quax

Objective—Annexin A5 (AnxA5) has antithrombotic, antiapoptotic, and antiinflammatory properties; we investigated its effectiveness against vascular inflammation, remodeling, and dysfunction in accelerated atherosclerosis. Methods and Results—AnxA5 (1 mg/kg per day or vehicle) was investigated in vascular injury models in hypercholesterolemic apolipoprotein E (ApoE)3*Leiden mice. AnxA5 treatment reduced adhesion and infiltration of leukocytes by 71% to 69% (P=0.015, P=0.031) and macrophages by 51% to 87% (P=0.014, P=0.018), as well as monocyte chemotactic protein-1 and tumor necrosis factor-&agr; expression in a femoral artery inflammation model (perivascular cuff for 3 days), indicating reduced vascular inflammation. In a vein graft model, 28 days of AnxA5 treatment reduced vein graft thickening (48%; P=0.006) and leukocyte infiltration (46%; P=0.003). In these mice, reduced plasma concentrations of IFN-&ggr; (−72%; P=0.040), granulocyte colony–stimulating factor (−41%; P=0.010), and macrophage inflammatory protein-1&bgr; (MIP-1&bgr;) (−66%; P=0.020) were measured, indicating reduced systemic inflammation. An in vitro endothelial cell model shows the importance of AnxA5s anticoagulant properties in reducing vascular inflammation. Endothelium-mediated dilatation in hypercholesterolemic ApoE(−/−) mice was improved by 3 days of AnxA5 treatment, shown by improved systolic and diastolic blood pressure reductions in response to metacholine, which could be abolished by l-Nitro-Arginine-Methyl Ester (l-NAME), indicating nitric oxide involvement. Conclusion—AnxA5 reduced local vascular and systemic inflammation and vascular remodeling and improved vascular function, indicating that it has a therapeutic potential against atherosclerotic cardiovascular diseases.

Collaboration


Dive into the Hetty C. de Boer's collaboration.

Top Co-Authors

Avatar

Anton Jan van Zonneveld

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ton J. Rabelink

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul H.A. Quax

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Margreet R. de Vries

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cees van Kooten

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eric P. van der Veer

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erna Peters

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Coen van Solingen

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge