Network


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

Hotspot


Dive into the research topics where Kitty B. J. M. Cleutjens is active.

Publication


Featured researches published by Kitty B. J. M. Cleutjens.


The FASEB Journal | 2007

Cathepsin cysteine proteases in cardiovascular disease

Suzanne P. M. Lutgens; Kitty B. J. M. Cleutjens; Mat J.A.P. Daemen; Sylvia Heeneman

Extracellular matrix (ECM) remodeling is one of the underlying mechanisms in cardiovascular diseases. Cathepsin cysteine proteases have a central role in ECM remodeling and have been implicated in the development and progression of cardiovascular diseases. Cathepsins also show differential expression in various stages of atherosclerosis, and in vivo knockout studies revealed that deficiency of cathepsin K or S reduces atherosclerosis. Furthermore, cathepsins are involved in lipid metabolism. Cathepsins have the capability to degrade low‐density lipoprotein and reduce cholesterol efflux from macrophages, aggravating foam cell formation. Although expression studies also demonstrated differential expression of cathepsins in cardiovascular diseases like aneurysm formation, neoin‐tima formation, and neovascularization, in vivo studies to define the exact role of cathepsins in these processes are lacking. Evaluation of the feasibility of cathepsins as a diagnostic tool revealed that serum levels of cathepsins L and S seem to be promising as biomarkers in the diagnosis of atherosclerosis, whereas cathepsin B shows potential as an imaging tool. Furthermore, ca‐thepsin K and S inhibitors showed effectiveness in (pre) clinical evaluation for the treatment of osteoporosis and osteoarthritis, suggesting that cathepsin inhibitors may also have therapeutic effects for the treatment of atherosclerosis.—Lutgens, S. P. M., Cleutjens, K. B. J. M., Daemen, M. J. A. P., Heeneman, S. Cathepsin cysteine proteases in cardiovascular disease. FASEB J. 21, 3029–3041 (2007)


Circulation | 2005

Disruption of the Cathepsin K Gene Reduces Atherosclerosis Progression and Induces Plaque Fibrosis but Accelerates Macrophage Foam Cell Formation

Esther Lutgens; Suzanne P. M. Lutgens; Birgit C.G. Faber; Sylvia Heeneman; M.M.J. Gijbels; M.P.J. de Winther; P. Frederik; I. van der Made; A. Daugherty; Anneke M Sijbers; A. Fisher; C.J. Long; Paul Saftig; D. Black; Mat J.A.P. Daemen; Kitty B. J. M. Cleutjens

Background— Cathepsin K (catK), a lysosomal cysteine protease, was identified in a gene-profiling experiment that compared human early plaques, advanced stable plaques, and advanced atherosclerotic plaques containing a thrombus, where it was highly upregulated in advanced stable plaques. Methods and Results— To assess the function of catK in atherosclerosis, catK−/−/apolipoprotein (apo) E−/− mice were generated. At 26 weeks of age, plaque area in the catK−/−/apoE−/− mice was reduced (41.8%) owing to a decrease in the number of advanced lesions as well as a decrease in individual advanced plaque area. This suggests an important role for catK in atherosclerosis progression. Advanced plaques of catK−/−/apoE−/− mice showed an increase in collagen content. Medial elastin fibers were less prone to rupture than those of apoE−/− mice. Although the relative macrophage content did not differ, individual macrophage size increased. In vitro studies of bone marrow derived–macrophages confirmed this observation. Scavenger receptor–mediated uptake (particularly by CD36) of modified LDL increased in the absence of catK, resulting in an increased macrophage size because of increased cellular storage of cholesterol esters, thereby enlarging the lysosomes. Conclusions— A deficiency of catK reduces plaque progression and induces plaque fibrosis but aggravates macrophage foam cell formation in atherosclerosis.


Journal of Bone and Mineral Research | 2006

Osteoclastic Bone Degradation and the Role of Different Cysteine Proteinases and Matrix Metalloproteinases: Differences Between Calvaria and Long Bone

Vincent Everts; Wolf Korper; Kees A. Hoeben; Ineke D. C. Jansen; Dieter Brömme; Kitty B. J. M. Cleutjens; Sylvia Heeneman; Christoph Peters; Thomas Reinheckel; Paul Saftig; Wouter Beertsen

Osteoclastic bone degradation involves the activity of cathepsin K. We found that in addition to this enzyme other, yet unknown, cysteine proteinases participate in digestion. The results support the notion that osteoclasts from different bone sites use different enzymes to degrade the collagenous bone matrix.


The Journal of Pathology | 2003

The dynamic extracellular matrix: intervention strategies during heart failure and atherosclerosis

Sylvia Heeneman; Jack P.M. Cleutjens; Birgit C.G. Faber; Esther E. Creemers; Robert-Jan van Suylen; Esther Lutgens; Kitty B. J. M. Cleutjens; Mat J.A.P. Daemen

The extracellular matrix is no longer seen as the static embedding in which cells reside; it has been shown to be involved in cell proliferation, migration and cell–cell interactions. Turnover of the different extracellular matrix components is an active process with multiple levels of regulation. Collagen, a major extracellular matrix constituent of the myocardium and the arterial vascular wall, is synthesized by (myo)fibroblasts in the myocardium and smooth muscle cells in the medial arterial vascular wall. Its degradation is controlled by proteinases, which include matrix metalloproteinases. This review will focus on the impact of fibrosis and especially collagen turnover on the progression of heart failure and atherosclerosis, two of the main cardiovascular pathologies. We will discuss data from human studies and animal models, with an emphasis on the effects of interventions on collagen synthesis and degradation. We conclude that there is a dynamic (dis)balance in the rate of collagen synthesis and degradation during heart failure and atherosclerosis, which makes the outcome of interventions not always predictable. Alternative approaches for intervening in collagen metabolism will be discussed as possible therapeutic intervention strategies. Copyright


Circulation | 2005

Gene Profiling in Atherosclerosis Reveals a Key Role for Small Inducible Cytokines Validation Using a Novel Monocyte Chemoattractant Protein Monoclonal Antibody

Esther Lutgens; Birgit C.G. Faber; Kitty Schapira; Chris T. Evelo; Rachel I. M. van Haaften; Sylvia Heeneman; Kitty B. J. M. Cleutjens; Ann Pascale Bijnens; Linda Beckers; J. Gordon Porter; Charles R. Mackay; Paul D. Rennert; Veronique Bailly; Matthew Jarpe; Brian M. Dolinski; Victor Koteliansky; Tony de Fougerolles; Mat J.A.P. Daemen

Background—Pathological aspects of atherosclerosis are well described, but gene profiles during atherosclerotic plaque progression are largely unidentified. Methods and Results—Microarray analysis was performed on mRNA of aortic arches of ApoE−/− mice fed normal chow (NC group) or Western-type diet (WD group) for 3, 4.5, and 6 months. Of 10 176 reporters, 387 were differentially (>2×) expressed in at least 1 group compared with a common reference (ApoE−/−, 3- month NC group). The number of differentially expressed genes increased during plaque progression. Time-related expression clustering and functional grouping of differentially expressed genes suggested important functions for genes involved in inflammation (especially the small inducible cytokines monocyte chemoattractant protein [MCP]-1, MCP-5, macrophage inflammatory protein [MIP]-1&agr;, MIP-1&bgr;, MIP-2, and fractalkine) and matrix degradation (cathepsin-S, matrix metalloproteinase-2/12). Validation experiments focused on the gene cluster of small inducible cytokines. Real-time polymerase chain reaction revealed a plaque progression–dependent increase in mRNA levels of MCP-1, MCP-5, MIP-1&agr;, and MIP-1&bgr;. ELISA for MCP-1 and MCP-5 showed similar results. Immunohistochemistry for MCP-1, MCP-5, and MIP-1&agr; located their expression to plaque macrophages. An inhibiting antibody for MCP-1 and MCP-5 (11K2) was designed and administered to ApoE−/− mice for 12 weeks starting at the age of 5 or 17 weeks. 11K2 treatment reduced plaque area and macrophage and CD45+ cell content and increased collagen content, thereby inducing a stable plaque phenotype. Conclusions—Gene profiling of atherosclerotic plaque progression in ApoE−/− mice revealed upregulation of the gene cluster of small inducible cytokines. Further expression and in vivo validation studies showed that this gene cluster mediates plaque progression and stability.


Journal of Magnetic Resonance Imaging | 2004

In vivo detection of hemorrhage in human atherosclerotic plaques with magnetic resonance imaging

Vincent C. Cappendijk; Kitty B. J. M. Cleutjens; Sylvia Heeneman; Geert Willem H. Schurink; Rob J. Th. J. Welten; Alfons G. H. Kessels; Robert J. van Suylen; Mat J.A.P. Daemen; Jos M. A. van Engelshoven; M. Eline Kooi

To investigate the performance of high‐resolution T1‐weighted (T1w) turbo field echo (TFE) magnetic resonance imaging (MRI) for the identification of the high‐risk component intraplaque hemorrhage, which is described in the literature as a troublesome component to detect.


Journal of Magnetic Resonance Imaging | 2008

Comparison of lipid-rich necrotic core size in symptomatic and asymptomatic carotid atherosclerotic plaque: Initial results.

Vincent C. Cappendijk; Alfons G. H. Kessels; Sylvia Heeneman; Kitty B. J. M. Cleutjens; Geert Willem H. Schurink; Rob J. Th. J. Welten; Werner H. Mess; Robert-Jan van Suylen; Tim Leiner; Mat J.A.P. Daemen; Jos M. A. van Engelshoven; M. Eline Kooi

To investigate the potential difference in the size of the lipid‐rich necrotic core (LRNC) in carotid plaques of symptomatic patients versus asymptomatic patients. Pathological studies established that a large LRNC is an important feature of vulnerable atherosclerotic plaque. Previously, we have demonstrated a high correlation between semiquantitative analysis of the LRNC size in T1‐weighted (w) turbo field echo (TFE) MR images and histology.


Annals of Medicine | 2003

Inflammation and restenosis: implications for therapy

Marjo M. P. C. Donners; Mat J.A.P. Daemen; Kitty B. J. M. Cleutjens; Sylvia Heeneman

Restenosis is the process of luminal narrowing in an atherosclerotic artery after an intra‐arterial intervention such as balloon angioplasty and stenting. It is believed that this process is mainly characterized by migration and proliferation of smooth muscle cells and extracellular matrix accumulation. However, there is now increasing evidence for a role of inflammation in the development of restenosis. The underlying molecular mechanisms of restenosis are, in fact, most probably regulated by inflammatory mediators, such as cytokines. Understanding the molecular mechanisms in restenosis is crucial for the development of a suitable therapy for this disease. Recently, the use of immunosuppressives in drug‐eluting stents has provided very promising results in the treatment of restenosis. In this review, we will describe the molecular mechanisms involved in restenosis with a focus on the role of inflammation and the use of immunosuppressive therapy.


PLOS ONE | 2012

Chemokines CCL3/MIP1α, CCL5/RANTES and CCL18/PARC are Independent Risk Predictors of Short-Term Mortality in Patients with Acute Coronary Syndromes

Saskia C.A. de Jager; Brenda W. C. Bongaerts; Michael Weber; A.O. Kraaijeveld; Mat Rousch; Stefanie Dimmeler; Marja P. van Dieijen-Visser; Kitty B. J. M. Cleutjens; Patty J. Nelemans; Theo J.C. van Berkel; Erik A.L. Biessen

Cytokines play an important role in ischemic injury and repair. However, little is known about their prognostic value in cardiovascular disease. The aim of this study was to investigate the prognostic importance of chemokines CCL3/MIP-1α, CCL5/RANTES and CCL18/PARC for the risk of future cardiovascular events in patients with acute coronary syndromes (ACS). Baseline levels of CCL3/MIP-1α, CCL5/RANTES and CCL18/PARC were determined in ACS patients from the Bad Nauheim ACS II registry (n = 609). During the following 200 days, patients were monitored for the occurrence of fatal and non-fatal cardiovascular events. Patients with CCL3/MIP1α, CCL5/RANTES and CCL18/PARC concentrations in the highest tertile were associated with an increased risk of a fatal event during follow-up (HR: 2.19, 95%CI: 1.04–4.61 for CCL3/MIP1α, HR: 3.45, 95%CI: 1.54–7.72 for CCL5/RANTES and HR: 3.14, 95%CI: 1.33–7.46 for CCL18/PARC). This risk was highest for patients with all three biomarkers concentrations in the upper tertile (HR: 2.52, 95%CI: 1.11–5.65). Together with known risk predictors of cardiovascular events, CCL3/MIP-1α, CCL5/RANTES and CCL18/PARC combined improved the c-statistics from 0.74 to 0.81 (p = 0.007). In conclusion, CCL3/MIP-1α, CCL5/RANTES and CCL18/PARC are independently associated with the risk of short-term mortality in ACS patients. Combining all three biomarkers further increased their prognostic value.


Journal of Magnetic Resonance Imaging | 2008

Comparison of single‐sequence T1w TFE MRI with multisequence MRI for the quantification of lipid‐rich necrotic core in atherosclerotic plaque

Vc Cappendijk; Sylvia Heeneman; Alfons G. H. Kessels; Kitty B. J. M. Cleutjens; Geert Willem H. Schurink; Rob J. Th. J. Welten; Werner H. Mess; Robert-Jan van Suylen; Tim Leiner; Mat J.A.P. Daemen; Jos M. A. van Engelshoven; M. Eline Kooi

To prospectively determine the accuracy of semiquantitative analysis of the amount of lipid‐rich necrotic core (LRNC) in atherosclerotic plaque using multi‐ as well as single‐sequence T1‐weighted (w) turbo field echo (TFE) MRI. Histology served as a reference standard.

Collaboration


Dive into the Kitty B. J. M. Cleutjens's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge