Network


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

Hotspot


Dive into the research topics where Y.M. Bastiaansen-Jenniskens is active.

Publication


Featured researches published by Y.M. Bastiaansen-Jenniskens.


Osteoarthritis and Cartilage | 2010

The infrapatellar fat pad should be considered as an active osteoarthritic joint tissue: a narrative review

S. Clockaerts; Y.M. Bastiaansen-Jenniskens; J. Runhaar; G.J. van Osch; J. F. Van Offel; J.A.N. Verhaar; L. S. De Clerck; J. Somville

INTRODUCTION Osteoarthritis (OA) of the knee joint is caused by genetic and hormonal factors and by inflammation, in combination with biomechanical alterations. It is characterized by loss of articular cartilage, synovial inflammation and subchondral bone sclerosis. Considerable evidence indicates that the menisci, ligaments, periarticular muscles and the joint capsule are also involved in the OA process. This paper will outline the theoretical framework for investigating the infrapatellar fat pad (IPFP) as an additional joint tissue involved in the development and progression of knee-OA. METHODS A literature search was performed in Pubmed from 1948 until October 2009 with keywords InFrapatellar fat pad, Hoffa fat pad, intraarticular adipose tissue, knee, cartilage, bone, cytokine, adipokine, inflammation, growth factor, arthritis, and OA. RESULTS The IPFP is situated intracapsularly and extrasynovially in the knee joint. Besides adipocytes, the IPFP from patients with knee-OA contains macrophages, lymphocytes and granulocytes, which are able to contribute to the disease process of knee-OA. Furthermore, the IPFP contains nociceptive nerve fibers that could in part be responsible for anterior pain in knee-OA. These nerve fibers secrete substance P, which is able to induce inflammatory responses and cause vasodilation, which may lead to IPFP edema and extravasation of the immune cells. The IPFP secretes cytokines, interleukins, growth factors and adipokines that influence cartilage by upregulating the production of matrix metalloproteinases (MMPs), stimulating the expression of pro-inflammatory cytokines and inhibiting the production of cartilage matrix proteins. They may also stimulate the production of pro-inflammatory mediators, growth factors and MMPs in synovium. CONCLUSION These data are consistent with the hypothesis that the IPFP is an osteoarthritic joint tissue capable of modulating inflammatory and destructive responses in knee-OA.


Annals of the Rheumatic Diseases | 2011

The infrapatellar fat pad of patients with osteoarthritis has an inflammatory phenotype

I.R. Klein-Wieringa; Margreet Kloppenburg; Y.M. Bastiaansen-Jenniskens; E. Yusuf; J.C. Kwekkeboom; H. El-Bannoudi; Rob G. H. H. Nelissen; Anne-Marie Zuurmond; V. Stojanovic-Susulic; G.J. van Osch; René E. M. Toes; A. Ioan-Facsinay

Objectives Obesity is a risk factor for the development of osteoarthritis (OA) in hands and knees. Adipose tissue can secrete different adipokines with powerful immunomodulatory effects. The infrapatellar fat pad (IFP) is an intra-articular organ in the vicinity of the synovium and cartilage. It is hypothesised that IFP-derived soluble factors could contribute to pathological processes in the knee joint. A study was therefore undertaken to compare the release of inflammatory mediators in the IFP and subcutaneous adipose tissue (ScAT) and to characterise the adipocytes and immune cell infiltrate in these tissues. Methods Paired IFP and ScAT samples were obtained from 27 patients with primary OA. The stromal vascular cell fraction (SVF) was isolated and characterised by fluorescence activated cell sorting. Cytokine and adipokine release in fat- and adipocyte-conditioned media was measured by luminex. Results IFP secreted higher levels of inflammatory mediators such as interleukin 6 (IL-6), adipsin, adiponectin and visfatin than ScAT. This could be due to differences in the phenotype of adipocytes and/or in the composition and phenotype of the SVF cells. IFP adipocyte-conditioned media showed a trend towards more IL-6 and adipsin than ScAT. Moreover, the SVF fraction of IFP contained more cells/g tissue, a lower percentage of T cells and a higher percentage of mast cells than ScAT. In addition, T cells had a predominantly pro-inflammatory phenotype while macrophages had a mixed pro- and anti-inflammatory phenotype in the IFP. Conclusion There are profound differences in secreted inflammatory factors and immune cell composition between the IFP and ScAT. These data indicate that IFP-derived soluble mediators could contribute to pathophysiological processes in the OA knee joint.


Journal of Cellular and Molecular Medicine | 2009

Cartilage repair: past and future--lessons for regenerative medicine.

Gerjo J.V.M. van Osch; Mats Brittberg; James E. Dennis; Y.M. Bastiaansen-Jenniskens; Reinhold G. Erben; Yrjö T. Konttinen; Frank P. Luyten

•  Introduction: the impaired repair capacity of cartilage and the beginning of cell therapy ‐  Autologous chondrocyte implantation ‐  Combination products ‐  Randomized controlled studies •  Choice of cell types •  Regulating cellular activities ‐  Influence of growth factors on chondrogenic differentiation ‐  Influence of growth factors on matrix deposition ‐  Supportive effect of biomaterials •  Interference of inflammation with cartilage repair •  Tracking of transplanted cells in vivo •  Conclusion and future directions


Annals of the Rheumatic Diseases | 2012

Statin use is associated with reduced incidence and progression of knee osteoarthritis in the Rotterdam study

S. Clockaerts; G.J. van Osch; Y.M. Bastiaansen-Jenniskens; J.A.N. Verhaar; F. Van Glabbeek; J.B. van Meurs; Hanneke J. M. Kerkhof; A. Hofman; B.H.Ch. Stricker; S.M. Bierma-Zeinstra

Background Osteoarthritis is the most frequent chronic joint disease causing pain and disability. Besides biomechanical mechanisms, the pathogenesis of osteoarthritis may involve inflammation, vascular alterations and dysregulation of lipid metabolism. As statins are able to modulate many of these processes, this study examines whether statin use is associated with a decreased incidence and/or progression of osteoarthritis. Methods Participants in a prospective population-based cohort study aged 55 years and older (n=2921) were included. x-Rays of the knee/hip were obtained at baseline and after on average 6.5 years, and scored using the Kellgren and Lawrence score for osteoarthritis. Any increase in score was defined as overall progression (incidence and progression). Data on covariables were collected at baseline. Information on statin use during follow-up was obtained from computerised pharmacy databases. The overall progression of osteoarthritis was compared between users and non-users of statins. Using a multivariate logistic regression model with generalised estimating equation, OR and 95% CI were calculated after adjusting for confounding variables. Results Overall progression of knee and hip osteoarthritis occurred in 6.9% and 4.7% of cases, respectively. The adjusted OR for overall progression of knee osteoarthritis in statin users was 0.43 (95% CI 0.25 to 0.77, p=0.01). The use of statins was not associated with overall progression of hip osteoarthritis. Conclusions Statin use is associated with more than a 50% reduction in overall progression of osteoarthritis of the knee, but not of the hip.


Annals of the Rheumatic Diseases | 2012

Cytokine production by infrapatellar fat pad can be stimulated by interleukin 1β and inhibited by peroxisome proliferator activated receptor α agonist

S. Clockaerts; Y.M. Bastiaansen-Jenniskens; C. Feijt; L.S. De Clerck; J.A.N. Verhaar; A.-M. Zuurmond; V. Stojanovic-Susulic; J. Somville; M. Kloppenburg; G.J. van Osch

Background Infrapatellar fat pad (IPFP) might be involved in osteoarthritis (OA) by production of cytokines. It was hypothesised that production of cytokines is sensitive to environmental conditions. Objectives To evaluate cytokine production by IPFP in response to interleukin (IL)1β and investigate the ability to modulate this response with an agonist for peroxisome proliferator activated receptor α (PPARα), which is also activated by lipid-lowering drugs such as fibrates. Methods Cytokine secretion of IPFP was analysed in the medium of explant cultures of 29 osteoarthritic patients. IPFP (five donors) and synovium (six donors) were cultured with IL-1β and PPARα agonist Wy14643. Gene expression of IL-1β, monocyte chemoattractant protein (MCP1), (IL-6, tumour necrosis factor (TNF)α, leptin, vascular endothelial growth factor (VEGF), IL-10, prostaglandin-endoperoxide synthase (PTGS)2 and release of TNFα, MCP1 and prostaglandin E2 were compared with unstimulated IPFP and synovium explants. Results IPFP released large amounts of inflammatory cytokines, adipokines and growth factors. IL-1β increased gene expression of PTGS2, TNFα, IL-1β, IL-6 and VEGF and increased TNFα release in IPFP. MCP1, leptin, IL-10 gene expression and MCP1, leptin and PGE2 release did not increase significantly. Synovium responded to IL-1β similarly to IPFP, except for VEGF gene expression. Wy14643 decreased gene expression of PTGS2, IL-1β, TNFα, MCP1, VEGF and leptin in IPFP explants and IL-1β, TNFα, IL-6, IL-10 and VEGF in synovium that responded to IL-1β. Conclusion IPFP is an active tissue within the joint. IPFP cytokine production is increased by IL-1β and decreased by a PPARα agonist. The effects were similar to effects seen in synovium. Fibrates may represent a potential disease-modifying drug for OA by modulating inflammatory properties of IPFP and synovium.


Biochemical and Biophysical Research Communications | 2013

A culture model to analyze the acute biomaterial-dependent reaction of human primary macrophages

N. Grotenhuis; Yves Bayon; Johan F. Lange; G.J. van Osch; Y.M. Bastiaansen-Jenniskens

Macrophages are important in foreign body reactions. We devised a culture model with human primary macrophages to evaluate the acute response of macrophages to biomaterials. First we selected proteins representative for pro-inflammatory (M1) or anti-inflammatory/repair (M2) response of monocytes isolated from blood of healthy human donors by exposing them to LPS+IFNγ or IL-4. A relative M1/M2 index was calculated using IL-1β, IL-6, tumor necrosis factor (TNF)α, monocyte chemotactic protein (MCP)-3 and macrophage inflammatory protein (MIP)-1α as M1 markers, and IL-1 receptor antagonist (IL-1RA), CCL18, regulated and normal T-cell expressed and secreted (RANTES), and macrophage-derived chemokine (MDC) as M2 markers. Then monocytes were cultured for 3days on 4 materials selected for known different foreign body reactions: Permacol™, Parietex™ Composite, multifilament polyethylene terephthalate and multifilament polypropylene. Macrophages on polypropylene produced high levels of anti-inflammatory proteins with a low M1/M2 index. Macrophages on Parietex™ Composite produced high levels of inflammatory and anti-inflammatory proteins, with a high M1/M2 index. Macrophages on polyethylene terephthalate also resulted in a high M1/M2 index. Macrophages on Permacol™ produced a low amount of all proteins, with a low M1/M2 index. This model with human primary macrophages and the panel of read-out parameters can be used to evaluate the acute reaction of macrophages to biomaterials in vitro to get more insight in the foreign body reaction.


Arthritis & Rheumatism | 2013

Stimulation of Fibrotic Processes by the Infrapatellar Fat Pad in Cultured Synoviocytes From Patients With Osteoarthritis: A Possible Role for Prostaglandin F2α

Y.M. Bastiaansen-Jenniskens; W. Wei; Carola Feijt; J.H. Waarsing; J.A.N. Verhaar; Anne-Marie Zuurmond; Roeland Hanemaaijer; Reinout Stoop; Gerjo J.V.M. van Osch

OBJECTIVE Stiffening of the joint is a feature of knee osteoarthritis (OA) that can be caused by fibrosis of the synovium. The infrapatellar fat pad (IPFP) present in the knee joint produces immune-modulatory and angiogenic factors. The goal of the present study was to investigate whether the IPFP can influence fibrotic processes in synovial fibroblasts, and to determine the role of transforming growth factor β (TGFβ) and prostaglandin F2α (PGF2α ) in these processes. METHODS Batches of fat-conditioned medium (FCM) were made by culturing pieces of IPFP obtained from the knees of 13 patients with OA. Human OA fibroblast-like synoviocytes (FLS) (from passage 3) were cultured in FCM with or without inhibitors of TGFβ/activin receptor-like kinase 5 or PGF2α for 4 days. The FLS were analyzed for production of collagen and expression of the gene for procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2; encoding lysyl hydroxylase 2b, an enzyme involved in collagen crosslinking) as well as the genes encoding α-smooth muscle actin and type I collagen α1 chain. In parallel, proliferation and migration of the synoviocytes were analyzed. RESULTS Collagen production and PLOD2 gene expression by the FLS were increased 1.8-fold (P < 0.05) and 6.0-fold (P < 0.01), respectively, in the presence of FCM, relative to control cultures without FCM. Moreover, the migration and proliferation of synoviocytes were stimulated by FCM. Collagen production was positively associated with PGF2α levels in the FCM (R = 0.89, P < 0.05), and inhibition of PGF2α levels reduced the extent of FCM-induced collagen production and PLOD2 expression. Inhibition of TGFβ signaling had no effect on the profibrotic changes. CONCLUSION These results indicate that the IPFP can contribute to the development of synovial fibrosis in the knee joint by increasing collagen production, PLOD2 expression, cell proliferation, and cell migration. In addition, whereas the findings showed that TGFβ is not involved, the more recently discovered profibrotic factor PGF2α appears to be partially involved in the regulation of profibrotic changes.


Arthritis & Rheumatism | 2011

Osteoarthritic synovial tissue inhibition of proteoglycan production in human osteoarthritic knee cartilage: Establishment and characterization of a long‐term cartilage–synovium coculture

M. Beekhuizen; Y.M. Bastiaansen-Jenniskens; Wendy Koevoet; Daniël B.F. Saris; Wouter J.A. Dhert; Laura B. Creemers; Gerjo J.V.M. van Osch

OBJECTIVE Although both cartilage and synovium are affected in osteoarthritis (OA), no in vitro coculture models of human OA tissue have been described. The aim of this study was to develop an in vitro model that includes both the synovium and cartilage of patients with knee OA. METHODS Explants of human OA cartilage and synovium were cultured alone or in coculture for 21 days. Histologic evaluation and analyses of lactate dehydrogenase release, matrix metalloproteinase (MMP) activity, content, release, and synthesis of glycosaminoglycan (GAG), and cytokine production were used to evaluate synovial tissue functionality and its effect on cartilage metabolism. To assess the possibility of intervention in the model system, the effect of triamcinolone was studied. RESULTS Throughout the entire culture period, OA synovial tissue remained viable and produced cytokines. Monocultures of synovial and cartilage explants produced different cytokine subsets, with the subsets found in coculture being most similar to those previously described in OA synovial fluid. MMP activity was detectable only in the synovial explant monoculture and in coculture. Cocultures showed a reduction in final GAG content (P < 0.02), attributable to an inhibition of GAG production (P < 0.001) rather than an increase in GAG release. Addition of triamcinolone inhibited cytokine production and MMP activity in coculture and synovial tissue monoculture and counteracted the inhibition of GAG production induced by coculture. In cartilage monoculture, however, triamcinolone reduced GAG production. CONCLUSION OA synovium affects cartilage metabolism by reducting GAG production. Triamcinolone can relieve this effect of synovial tissue, while being inhibitory when added to cartilage monoculture. These results clearly indicate the importance of tissue coculture as a promising tool for studying OA pathophysiology and for development of possible interventions.


BioResearch Open Access | 2016

The Effect of Biomaterials Used for Tissue Regeneration Purposes on Polarization of Macrophages

Geesien S. A. Boersema; N. Grotenhuis; Yves Bayon; Johan F. Lange; Y.M. Bastiaansen-Jenniskens

Abstract Activation of macrophages is critical in the acute phase of wound healing after implantation of surgical biomaterials. To understand the response of macrophages, they are often cultured in vitro on biomaterials. Since a wide range of biomaterials is currently used in the clinics, we undertook a systematic review of the macrophage polarization in response to these different surgical biomaterials in vitro. Beside the chemistry, material characteristics such as dimension, pore size, and surface topography are of great influence on the response of macrophages. The macrophage response also appears to depend on the differences in sterilization techniques that induce lasting biochemical changes or residues of chemicals and their byproducts used for sterilization. Regarding tissue-based biomaterials, macrophages on human or porcine dermis, strongly cross-linked by chemicals elicit in general a proinflammatory response with higher amounts of proinflammatory cytokines. Synthetic biomaterials such as polyethylene, polyethylene terephthalate (PET) + polyacrylamide (PAAm), PET + sodium salt of poly(acrylic acid) (PAANa), perfluoropolyether (PFPE) with large posts, PEG-g-PA, and polydioxanone (PDO) always appear to elicit an anti-inflammatory response in macrophages, irrespective of origin of the macrophages, for example, buffy coats or full blood. In conclusion, in general in vitro models contribute to evaluate the foreign body reaction on surgical biomaterials. Although it is difficult to simulate complexity of host response elicited by biomaterials, after their surgical implantation, an in vitro model gives indications of the initial foreign body response and allows the comparison of this response between biomaterials.


Osteoarthritis and Cartilage | 2011

Peroxisome proliferator activated receptor alpha activation decreases inflammatory and destructive responses in osteoarthritic cartilage

S. Clockaerts; Y.M. Bastiaansen-Jenniskens; C. Feijt; J.A.N. Verhaar; J. Somville; L. S. De Clerck; G.J. van Osch

OBJECTIVE Peroxisome proliferator activated receptor α (PPARα) agonists are used in clinical practice as lipid-lowering drugs and are also known to exert anti-inflammatory effects on various tissues. We hypothesized that PPARα activation leads to anti-inflammatory and anti-destructive effects in human OA cartilage. METHODS Cartilage explants obtained from six OA patients were cultured for 48 h with 10 ng/ml interleukin (IL)1β as a pro-inflammatory stimulus. 100 μM Wy-14643, a potent and selective PPARα agonist, was added to the cultures and gene expression of matrix metalloproteinase (MMP)1, MMP3, MMP13, collagen type II (COL2A1), aggrecan and PPARα in cartilage explants and the release of glycosaminoglycans (GAGs), nitric oxide (NO) and prostaglandin E(2) (PGE(2)) in the culture media were analyzed and compared to the control without Wy-14643. RESULTS Addition of Wy-14643 decreased mRNA expression of MMP1, MMP3 and MMP13 in cartilage explants that responded to IL1β, whereas Wy-14643 did not affect gene expression of COL2A1 and aggrecan. Wy-14643 also decreased secretion of inflammatory marker NO in the culture medium of cartilage explants responding to IL1β. Wy-14643 inhibited the release of GAGs by cartilage explants in culture media. CONCLUSION PPARα agonist Wy-14643 inhibited the inflammatory and destructive responses in human OA cartilage explants and did not have an effect on COL2A1 or aggrecan mRNA expression. These effects of PPARα agonists on osteoarthritic cartilage warrant further investigation of these drugs as a potential therapeutic strategy for osteoarthritis (OA).

Collaboration


Dive into the Y.M. Bastiaansen-Jenniskens's collaboration.

Top Co-Authors

Avatar

G.J. van Osch

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

J.A.N. Verhaar

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

S. Clockaerts

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

W. Wei

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

L. Utomo

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Anne-Marie Zuurmond

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

C. Feijt

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Johan F. Lange

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nicole Kops

Erasmus University Rotterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge