Network


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

Hotspot


Dive into the research topics where T. J. C. Van Berkel is active.

Publication


Featured researches published by T. J. C. Van Berkel.


Circulation | 2005

Blockade of Interleukin-12 Function by Protein Vaccination Attenuates Atherosclerosis

Arnaud D. Hauer; Catherine Uyttenhove; P. de Vos; Vincent Stroobant; J.-C. Renauld; T. J. C. Van Berkel; J Van Snick; Johan Kuiper

Background—Interleukin-12 (IL-12) has been identified as a key inducer of a type 1 T-helper cell cytokine pattern, which is thought to contribute to the development of atherosclerosis. We sought to study the role of IL-12 in atherosclerosis by inhibition of IL-12 using a newly developed vaccination technique that fully blocks the action of IL-12. Methods and Results—LDL receptor–deficient (LDLr−/−) mice were vaccinated against IL-12 by 5 intramuscular injections of IL-12–PADRE complex in combination with adjuvant oil-in-water emulsion (low dose)/MPL/QS21 every 2 weeks. Two weeks thereafter, atherogenesis was initiated in the carotid artery by perivascular placement of silicone elastomer collars. IL-12 vaccination resulted in the induction of anti–IL-12 antibodies that functionally blocked the action of IL-12 as determined in an IL-12 bioassay. Blockade of IL-12 by vaccination of LDLr−/− mice resulted in significantly reduced (68.5%; P<0.01) atherogenesis compared with control mice without a change in serum cholesterol levels. IL-12 vaccination also resulted in a significant decrease in intima/media ratios (66.7%; P<0.01) and in the degree of stenosis (57.8%; P<0.01). On IL-12 vaccination, smooth muscle cell and collagen content in the neointima increased 2.8-fold (P<0.01) and 4.2-fold (P<0.01), respectively. Conclusions—Functional blockade of endogenous IL-12 by vaccination resulted in a significant 68.5% reduction in atherogenesis in LDLr−/− mice. Vaccination against IL-12 also improved plaque stability, from which we conclude that the blockade of IL-12 by vaccination may be considered a promising new strategy in the treatment of atherosclerosis.


Circulation | 2007

CC chemokine ligand-5 (CCL5/RANTES) and CC chemokine ligand-18 (CCL18/PARC) are specific markers of refractory unstable angina pectoris and are transiently raised during severe ischemic symptoms

A.O. Kraaijeveld; S.C.A. de Jager; W.J. de Jager; Berent Prakken; I. Haspels; Hein Putter; T. J. C. Van Berkel; L. Nagelkerken; J.W. Jukema; E.A.L. Biessen

Background— Chemokines play an important role in atherogenesis and in ischemic injury and repair; however, prospective data on individual chemokines in unstable angina pectoris (UAP) are scarce. Therefore, we assessed chemokine patterns in a prospective cohort of patients with UAP. Methods and Results— Plasma samples of 54 patients with Braunwald class IIIB UAP were examined at baseline for 11 chemokines and 5 inflammatory mediators via multiplex analysis. Levels of CC chemokine ligand (CCL)-5 (also known as RANTES [regulated on activation, normally T-cell expressed, and secreted]; 32.7 versus 23.1 ng/mL, P=0.018) and CCL18 (also known as PARC [pulmonary and activation-regulated chemokine]; 104.4 versus 53.7 ng/mL, P=0.011) were significantly elevated in patients with refractory ischemic symptoms versus stabilized patients. Temporal monitoring by ELISA of CCL5, CCL18, and soluble CD40 ligand (sCD40) levels revealed a drop in CCL5 and sCD40L levels in all UAP patients from day 2 onward (CCL5 12.1 ng/mL, P<0.001; sCD40L 1.35 ng/mL, P<0.05), whereas elevated CCL18 levels were sustained for at least 2 days, then were decreased at 180 days after inclusion (34.5 ng/mL, P<0.001). Peripheral blood mononuclear cells showed increased protein expression of chemokine receptors CCR3 and CCR5 in CD3+ and CD14+ cells at baseline compared with 180 days after inclusion, whereas mRNA levels were downregulated, which was attributable in part to a postischemic release of human neutrophil peptide-3–positive neutrophils and in part to negative feedback. Finally, elevated CCL5 and CCL18 levels predicted future cardiovascular adverse events, whereas C-reactive protein and sCD40L levels did not. Conclusions— We are the first to report that CCL18 and CCL5 are transiently raised during episodes of UAP, and peak levels of both chemokines are indicative of refractory symptoms. Because levels of both chemokines, as well as of cognate receptor expression by circulating peripheral blood mononuclear cells, are increased during cardiac ischemia, this may point to an involvement of CCL5/CCL18 in the pathophysiology of UAP and/or post-UAP responses.


Atherosclerosis | 2010

Vaccination against Foxp3(+) regulatory T cells aggravates atherosclerosis.

T. van Es; G.H.M. van Puijvelde; Amanda C. Foks; Kim L.L. Habets; Ilze Bot; Eli Gilboa; T. J. C. Van Berkel; Johan Kuiper

OBJECTIVE Regulatory T cells are crucial for immune homeostasis and an impaired regulatory T cell function results in many pathological conditions. Regulatory T cells have already been described to be protective in atherosclerosis. However the exact contribution of Foxp3-expressing natural regulatory T cells in atherosclerosis has not been elucidated yet. METHODS AND RESULTS In this study we vaccinated LDL receptor deficient mice with dendritic cells which are transfected with Foxp3 encoding mRNA and studied the effect on initial atherosclerosis. Vaccination against Foxp3 resulted in a reduction of Foxp3(+) regulatory T cells in several organs and in an increase in initial atherosclerotic lesion formation. Furthermore we observed an increase in plaque cellularity and increased T cell proliferation in the Foxp3 vaccinated mice. CONCLUSION We further establish the protective role of Tregs in atherosclerosis. The results illustrate the important role for Foxp3-expressing regulatory T cells in atherosclerosis, thereby providing a potential opportunity for therapeutic intervention against this disease.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1984

In vivo catabolism of biologically modified LDL.

J.F. Nagelkerke; L.M. Havekes; V.W.M. van Hinsbergh; T. J. C. Van Berkel

Incubation of human low density lipoprotein (LDL) at 37 degrees C in the presence of human umbilical vein endothelial cells (EC) caused a time-dependent shift in the charge and density of LDL. The physical changes of the human LDL occurred parallel with an increase in its clearance from the serum and uptake in the liver when injected into rats. The serum decay of the EC-modified LDL (44 hours incubation) was 20 times faster than for control LDL. EC-modified LDL, cleared from the blood, was quantitatively recovered in the liver. Isolation of the different liver cell types (parenchymal, Kupffer, and endothelial cells) after in vivo injection of 125I-EC-modified LDL showed that approximately 30 times more radioactivity was associated with the endothelial cells than with the parenchymal cells (per milligram of cell protein). In vitro experiments indicated that EC-modified-LDL was processed by the rat liver endothelial cells via a high affinity, saturable pathway related to the pathway by which these cells processed acetyl-LDL. We concluded that, if EC-modified LDL is generated in vivo, the liver, and in particular the endothelial cells, forms the major protection system against the occurrence of atherogenic particles in the blood.


Clinica Chimica Acta | 1995

Enhanced susceptibility of low-density lipoproteins to oxidation in coronary bypass patients with progression of atherosclerosis

Y.B. de Rijke; Harriette F. Verwey; C.J.M. Vogelezang; E. A. van der Velde; H.M.G. Princen; A. van der Laarse; A. V. G. Bruschke; T. J. C. Van Berkel

Oxidation of low-density lipoprotein (LDL) may play a causal role in atherosclerosis. In this study we analyzed whether the severity of progression of coronary atherosclerosis is related to the susceptibility of LDL to oxidative modification. On the basis of repeated coronary angiography, 28 coronary bypass patients were divided into two groups: group A, 12 patients with, and group B, 16 patients without progression of coronary atherosclerosis. The lag time, reflecting the resistance of LDL to oxidative modification, was significantly smaller in group A as compared with group B (81 +/- 10 and 93 +/- 15 min, respectively). Besides differences in cholesterol and apolipoprotein B concentrations, the difference in susceptibility of LDL to oxidation significantly contributes to the differences between the progression and the non-progression group (P = 0.02). In the combined groups of patients, the lag phase of LDL for oxidation was positively correlated with LDL cholesterol ester to protein ratio (r = 0.53, P = 0.01). It is concluded that LDL samples obtained from coronary bypass patients differ with respect to their oxidizability depending on progression of atherosclerosis following coronary bypass surgery.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2000

Low density lipoprotein receptor of macrophages facilitates atherosclerotic lesion formation in C57Bl/6 mice.

Nicole Herijgers; M. Van Eck; P.H.E. Groot; P.M. Hoogerbrugge; T. J. C. Van Berkel

Macrophage-derived foam cells play an important role in the initiation and progression of atherosclerosis. To examine the role of the macrophage low density lipoprotein receptor (LDLr) in atherosclerotic lesion formation, bone marrow from LDLr knockout [LDLr(-/-)] mice was transplanted into irradiated wild-type C57Bl/6 [LDLr(+/+)] mice. After 3 months on an atherogenic diet, C57Bl/6 mice, reconstituted with LDLr(-/-) bone marrow, showed a mean lesion area of 34.7 x 10(3)+/-22.4 x 10(3) microm(2) compared with 100. 8 x 10(3)+/-33.0 x 10(3) microm(2) (P<0.001) in control C57Bl/6 mice that were transplanted with LDLr(+/+) bone marrow. There were no significant differences in total serum cholesterol, triglyceride levels, and lipoprotein profiles between the 2 groups. Histochemical analysis of macrophage LDLr expression in the atherosclerotic lesions indicated that C57Bl/6 mice, reconstituted with LDLr(+/+) bone marrow, showed extensive staining of the foam cells in the atherosclerotic lesions, whereas mice reconstituted with LDLr(-/-) bone marrow showed only a few LDLr-positive foam cells. In vitro, peritoneal macrophages isolated from wild-type C57Bl/6 mice were, respectively, 4.7- and 10.7-fold more effective in cell association and degradation of atherogenic (125)I-beta-very low density lipoprotein than were LDLr(-/-) peritoneal macrophages, establishing that the LDLr on macrophages is important for the interaction of macrophages with beta-very low density lipoprotein. It is concluded that the LDLr on macrophages can facilitate the development of atherosclerosis, possibly by mediating the uptake of atherogenic lipoproteins.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1992

Characterization of the interaction of acetylated LDL and oxidatively modified LDL with human liver parenchymal and Kupffer cells in culture.

J. A. A. M. Kamps; Johan K. Kruijt; Johan Kuiper; T. J. C. Van Berkel

The interaction of acetylated low density lipoprotein (Ac-LDL) and oxidatively modified low density lipoprotein (Ox-LDL) with cultured human liver parenchymal cells and human Kupffer cells was investigated to define, for humans, the presence of scavenger receptors in the liver. A direct comparison of the capacity of Kupffer and parenchymal cells to interact with Ac-LDL and Ox-LDL indicated that the capacity of Kupffer cells per milligram of cell protein to degrade Ac-LDL and Ox-LDL is 14-fold and sixfold higher, respectively, than that of parenchymal cells. The degradation of both Ac-LDL and Ox-LDL by parenchymal cells and Kupffer cells could be inhibited by chloroquine and ammonium chloride, indicating that degradation occurs in the lysosomes. Competition studies showed that unlabeled Ox-LDL competed efficiently with the cell association and degradation of 125I-labeled Ac-LDL by human parenchymal cells and human Kupffer cells. However, unlabeled Ac-LDL did not compete (parenchymal cells) or only partially competed (40% in Kupffer cells) with the cell association and degradation of 125I-labeled Ox-LDL. Polyinosinic acid completely blocked the cell association and degradation of Ac-LDL and Ox-LDL with Kupffer cells while no significant effect on parenchymal cells was noted. It is concluded that human liver parenchymal cells contain a scavenger receptor that interacts with Ac-LDL and Ox-LDL and an additional recognition site that recognizes Ox-LDL specifically.(ABSTRACT TRUNCATED AT 250 WORDS)


Current Pharmaceutical Design | 2007

Chemokines and atherosclerotic plaque progression: towards therapeutic targeting?

A.O. Kraaijeveld; S.C.A. de Jager; T. J. C. Van Berkel; E.A.L. Biessen; J.W. Jukema

Atherosclerosis is currently viewed as an inflammatory disease in which the initiation and progression of the atherosclerotic plaque towards a rupture prone, unstable plaque is driven by leukocyte recruitment mediated by various inflammatory mediators. Recently, interest in chemotactic cytokines or chemokines with regard to atherosclerosis has been growing as chemokines mediate the influx of leukocytes that is typical of atherothrombosis. The activity of the majority of chemokines is overlapping and chemokines are not only produced by the various cellular constituents of the atherosclerotic plaque but also by activated platelets. Consequently, the direct influence of individual chemokines on plaque destabilisation and rupture is widespread and rather unclear. Experimental research has already established the role of a number of chemokines in advanced atherosclerosis. Nevertheless, given the complexity and size of the chemokine family, further screening of cardiovascular disease for chemokine level and genetic polymorphisms for chemokines will be warranted as the search for viable biomarkers of plaque destabilization as well as novel therapeutic targets for specific atheroregressive therapeutic compounds is ongoing. With regard to the latter, clinical trials with specific chemokine inhibitory strategies, like chemokine receptor antagonists, are already underway in other inflammatory disorders. Summarizing, chemokine inhibition likely constitutes an important therapeutic option next to already established drugs in the management of cardiovascular disease.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1990

Characterization in vitro of interaction of human apolipoprotein E-free high density lipoprotein with human hepatocytes.

Donald Schouten; M.F. Kleinherenbrink-Stins; A. Brouwer; D.L. Knook; J. A. A. M. Kamps; Johan Kuiper; T. J. C. Van Berkel

Characterization of the interaction of iodinated apolipoprotein (apo) E-free high density lipoprotein (HDL) with cultured human hepatocytes provides evidence for a saturable, Ca2(+)-independent, high affinity binding site with an apparent km value of 20 micrograms/ml of apolipoprotein. Nitrated HDL and low density lipoprotein (LDL) did not compete for the binding of HDL, in contrast to very low density lipoprotein (VLDL). It is suggested that VLDL competition is exerted by the presence of apo Cs. Degradation of HDL was relatively low and in some cases not detectable. In cases where degradation was found, inhibitors of the lysosomal pathway of protein degradation had no effect, while LDL degradation was inhibited more than 80%. In the presence of 10 microM of monensin, the cell-association of HDL was unaffected, but the degradation was inhibited by 30%. Under similar conditions, LDL association was inhibited by 40% and LDL degradation, by 90%. Incubation of human hepatocytes with fluorescently labeled HDL (Dil-HDL) revealed (in contrast to Dil-LDL) mainly strong membrane-bound fluorescence and hardly any labeling of small intracellular vesicles. It is concluded that human hepatocytes possess a specific high affinity site for human HDL with recognition properties similar to those described earlier on rat hepatocytes. No evidence that the binding of HDL is actively coupled to uptake and lysosomal degradation could be obtained, indicating that binding of LDL and HDL to human hepatocytes is coupled differently to intracellular pathways.


The FASEB Journal | 2000

Novel hepatotrophic prodrugs of the antiviral nucleoside 9-(2-phosphonylmethoxyethyl)adenine with improved pharmacokinetics and antiviral activity

E.A.L. Biessen; A.R.P.M. Valentijn; R. L. A. De Vrueh; E. Van De Bilt; Leo A. J. M. Sliedregt; Perry Prince; Martin K. Bijsterbosch; J. H. Van Boom; G.A. van der Marel; P. J. Abrahams; T. J. C. Van Berkel

The device of new hepatotrophic prodrugs of the antiviral nucleoside 9‐(2‐phosphonylme‐thoxyethyl)adenine (PMEA) with specificity for the asialoglycoprotein receptor on parenchymal liver cells is described. PMEA was conjugated to bi‐ and trivalent cluster glycosides (K(GN)2 and K2(GN)3, respectively) with nanomolar affinity for the asialoglycoprotein receptor. The liver uptake of the PMEA prodrugs was more than 10‐fold higher than that of the parent drug (52±6% and 62±3% vs. 4.8±0.7% of the injected dose for PMEA) and could be attributed for 90% to parenchymal cells. Accumulation of the PMEA prodrugs in extrahepatic tissue (e.g., kidney, skin) was substantially reduced. The ratio of parenchymal liver cell‐to‐kidney uptake—a measure of the prodrugs therapeutic window—was increased from 0.058 ± 0.01 for PMEA to 1.86 ± 0.57 for K(GN)2‐PMEA and even 2.69 ± 0.24 for K2(GN)3‐PMEA. Apparently both glycosides have a similar capacity to redirect (antiviral) drugs to the liver. After cellular uptake, both PMEA prodrugs were converted into the parent drug, PMEA, during acidification of the lysosomal milieu (t1/2≈100 min), and the released PMEA was rapidly translocated into the cytosol. The antiviral activity of the prodrugs in vitro was dramatically enhanced as compared to the parent drug (5‐ and 52‐fold for K(GN)2‐PMEA and K2(GN)3‐PMEA, respectively). Given the 15‐fold enhanced liver uptake of the prodrugs, we anticipate that the potency in vivo will be similarly increased. We conclude that PMEA prodrugs have been developed with greatly improved pharmacokinetics and therapeutic activity against viral infections that implicate the liver parenchyma (e.g., HBV). In addition, the significance of the above prodrug concept also extends to drugs that intervene in other liver disorders such as cholestasis and dyslipidemia.—Biessen, E. A. L., Valentijn, A. R. P. M., de Vrueh, R. L. A., van de Bilt, E., Sliedregt, L A. J. M., Prince, P., Bijsterbosch, M. K., van Boom, J. H., van der Marel, G. A., Abrahams, P. J., van Berkel, T. J. C. Novel hepatotrophic prodrugs of the antiviral nucleoside 9‐(2‐phosphonylmethoxyethyl)adenine with improved pharmacokinetics and antiviral activity. FASEB J. 14, 1784–1792 (2000)

Collaboration


Dive into the T. J. C. Van Berkel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Johan K. Kruijt

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

I Meurs

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge