Network


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

Hotspot


Dive into the research topics where Ronnie G. Wismans is active.

Publication


Featured researches published by Ronnie G. Wismans.


Journal of The American Society of Nephrology | 2007

In Vivo Degradation of Heparan Sulfates in the Glomerular Basement Membrane Does Not Result in Proteinuria

Tessa J.M. Wijnhoven; Joost F.M. Lensen; Ronnie G. Wismans; Mohammed Lamrani; L.A.H. Monnens; Ron A. Wevers; Angelique L. Rops; Johan van der Vlag; J.H.M. Berden; Lambert P. van den Heuvel; Toin H. van Kuppevelt

Heparan sulfates (HS) are long, unbranched, negatively charged polysaccharides that are bound to core proteins. HS in the glomerular basement membrane (GBM) is reported to be important for charge-selective permeability. Aberrant GBM HS expression has been observed in several glomerular diseases, such as diabetic nephropathy and membranous glomerulopathy, and a decrease in HS generally is associated with proteinuria. This study, with the use of a controlled in vivo approach, evaluated whether degradation of HS in rat GBM resulted in acute proteinuria. Rats received two intravenous injections of either heparinase III to digest HS or neuraminidase to remove neuraminic acids (positive control). Urine samples were taken at various time points, and at the end of the experiment, kidneys were removed and analyzed. Injection with heparinase III resulted in a complete loss of glomerular HS as demonstrated by immunofluorescence staining using anti-HS antibodies and by electron microscopy using cupromeronic blue in a critical electrolyte concentration mode. In the urine, a strong increase in HS was found within 2 h after the first injection. Staining for agrin, the major HS proteoglycan core protein in the GBM, was unaltered. No urinary albumin or other proteins were detected at any time point, and no changes in glomerular morphology were noticed. Injection of rats with neuraminidase, however, resulted in a major increase of urinary albumin and was associated with an increase in urinary free neuraminic acid. An increased glomerular staining with Peanut agglutinin lectin, indicative of removal of neuraminic acid, was noted. In conclusion, removal of HS from the GBM does not result in acute albuminuria, whereas removal of neuraminic acid does.


Journal of Biological Chemistry | 2012

Organ-specific Sulfation Patterns of Heparan Sulfate Generated by Extracellular Sulfatases Sulf1 and Sulf2 in Mice

Satoshi Nagamine; Michiko Tamba; Hisako Ishimine; Kota Araki; Kensuke Shiomi; Takuya Okada; Tatsuyuki Ohto; Satoshi Kunita; Satoru Takahashi; Ronnie G. Wismans; Toin H. van Kuppevelt; Masayuki Masu; Kazuko Keino-Masu

Background: Extracellular endosulfatases Sulf1 and Sulf2 hydrolyze 6-O-sulfate in heparan sulfate. Results: Disaccharide analysis showed that 2-O-, 6-O-, and N-trisulfated disaccharide units in heparan sulfate were increased to different degrees in different organs in Sulf1 and Sulf2 knock-out mice. Conclusion: Sulfs generate organ-specific sulfation patterns of heparan sulfate. Significance: This may indicate differences in activity between Sulf1 and Sulf2 in vivo. Heparan sulfate endosulfatases Sulf1 and Sulf2 hydrolyze 6-O-sulfate in heparan sulfate, thereby regulating cellular signaling. Previous studies have revealed that Sulfs act predominantly on UA2S-GlcNS6S disaccharides and weakly on UA-GlcNS6S disaccharides. However, the specificity of Sulfs and their role in sulfation patterning of heparan sulfate in vivo remained unknown. Here, we performed disaccharide analysis of heparan sulfate in Sulf1 and Sulf2 knock-out mice. Significant increases in ΔUA2S-GlcNS6S were observed in the brain, small intestine, lung, spleen, testis, and skeletal muscle of adult Sulf1−/− mice and in the brain, liver, kidney, spleen, and testis of adult Sulf2−/− mice. In addition, increases in ΔUA-GlcNS6S were seen in the Sulf1−/− lung and small intestine. In contrast, the disaccharide compositions of chondroitin sulfate were not primarily altered, indicating specificity of Sulfs for heparan sulfate. For Sulf1, but not for Sulf2, mRNA expression levels in eight organs of wild-type mice were highly correlated with increases in ΔUA2S-GlcNS6S in the corresponding organs of knock-out mice. Moreover, overall changes in heparan sulfate compositions were greater in Sulf1−/− mice than in Sulf2−/− mice despite lower levels of Sulf1 mRNA expression, suggesting predominant roles of Sulf1 in heparan sulfate desulfation and distinct regulation of Sulf activities in vivo. Sulf1 and Sulf2 mRNAs were differentially expressed in restricted types of cells in organs, and consequently, the sulfation patterns of heparan sulfate were locally and distinctly altered in Sulf1 and Sulf2 knock-out mice. These findings indicate that Sulf1 and Sulf2 differentially contribute to the generation of organ-specific sulfation patterns of heparan sulfate.


Tissue Engineering Part C-methods | 2009

Micro-computed tomographical imaging of soft biological materials using contrast techniques.

Kaeuis A. Faraj; Vincent M.J.I. Cuijpers; Ronnie G. Wismans; X. Frank Walboomers; John A. Jansen; Toin H. van Kuppevelt; Willeke F. Daamen

The aim of this work was to introduce high-resolution computed tomography (micro-CT) for scaffolds made from soft natural biomaterials, and to compare these data with the conventional techniques scanning electron microscopy and light microscopy. Collagen-based scaffolds were used as examples. Unlike mineralized tissues, collagen scaffolds do not provide enough X-ray attenuation for micro-CT imaging. Therefore, various metal-based contrast agents were applied and evaluated using two structurally distinct scaffolds, one with round pores and one with unidirectional lamellae. The optimal contrast techniques for obtaining high-resolution three-dimensional images were either a combination of osmium tetroxide and uranyl acetate, or a combination of uranyl acetate and lead citrate. The data obtained by micro-CT analysis were in line with data obtained by light and electron microscopy. However, small structures (less than a few mum) could not be visualized due to limitation of the spot size of the micro-CT apparatus. In conclusion, reliable three-dimensional images of scaffolds prepared from soft natural biomaterials can be obtained using appropriate contrast protocols. This extends the use of micro-CT analysis to soft materials, such as protein-based biomaterials.


Acta Biomaterialia | 2010

Controlled fabrication of triple layered and molecularly defined collagen/elastin vascular grafts resembling the native blood vessel.

Martin J.W. Koens; Kaeuis A. Faraj; Ronnie G. Wismans; J.A. van der Vliet; A.G. Krasznai; Vincent M.J.I. Cuijpers; John A. Jansen; Willeke F. Daamen; T.H. van Kuppevelt

There is a consistent need for a suitable natural biomaterial to function as an arterial prosthesis in achieving arterial regeneration. Natural grafts are generally obtained by decellularization of native blood vessels, but batch to batch variations may occur and the nature/content of remaining contaminants is generally unknown. In this study we fabricated a molecularly defined natural arterial graft from scratch resembling the native three layered architecture from the fibrillar extracellular matrix components collagen and elastin. Using casting, moulding, freezing and lyophilization techniques, a triple layered construct was prepared consisting of an inner layer of elastin fibres, a middle (porous) film layer of collagen fibrils and an outer scaffold layer of collagen fibrils. The construct was carbodiimide cross-linked and heparinized. Characterization included biochemical/biophysical analyses, scanning electron microscopy, micro-computed tomography, (immuno)histology and haemocompatibility. Burst pressures were up to 400mm Hg and largely conferred by the intermediate porous collagen film layer. The highly purified type I collagen fibrils and elastin fibres used did not evoke platelet aggregation in vitro. Suturability of the graft in end to side anastomosis was successful and considered adequate for in vivo application.


Biomaterials | 2009

Microscale mechanical properties of single elastic fibers: The role of fibrillin-microfibrils

Mieke Mjf Koenders; Lanti Yang; Ronnie G. Wismans; Kees van der Werf; Dieter P. Reinhardt; Willeke F. Daamen; Martin L. Bennink; Pieter J. Dijkstra; Toin H. van Kuppevelt; Jan Feijen

Micromechanical properties of single elastic fibers and fibrillin-microfibrils, isolated from equine ligamentum nuchae using chemical and enzymatic methods, were determined with atomic force microscopy (AFM). Youngs moduli of single elastic fibers immersed in water, devoid of or containing fibrillin-microfibrils, were determined using bending tests. Bending freely suspended elastic fibers on a micro-channeled substrate by a tip-less AFM cantilever generated a force versus displacement curve from which Youngs moduli were calculated. For single elastic fibers, Youngs moduli in the range of 0.3-1.5 MPa were determined, values not significantly affected by the presence of fibrillin-microfibrils. To further understand the role of fibrillin-microfibrils in vertebrate elastic fibers, layers of fibrillin-microfibrils were subjected to nano-indentation tests. From the slope of the force versus indentation curves, Youngs moduli ranging between 0.56 and 0.74 MPa were calculated. The results suggest that fibrillin-microfibrils are not essential for the mechanical properties of single vertebrate elastic fibers.


Biomaterials | 2010

High density gene expression microarrays and gene ontology analysis for identifying processes in implanted tissue engineering constructs.

Gerwen Lammers; Christian Gilissen; Suzan T.M. Nillesen; Peter J.E. Uijtdewilligen; Ronnie G. Wismans; Joris A. Veltman; Willeke F. Daamen; Toin H. van Kuppevelt

The in vivo performance of tissue-engineered constructs is often based on generally accepted read-out parameters, like (immuno)histology. In this study, high-density gene expression microarrays and gene ontology (GO) analysis were used as a read-out tool to identify the biological processes occurring after implantation of an acellular collagen-based skin construct using a rat full-thickness wound model. A freely-available program (DAVID) was used to identify up/downregulated biological processes (GO-terms) and results were compared to wound healing/regeneration without a construct. The entire process from RNA isolation to biological interpretation is explained step-by-step. Conventional (immuno)histology was used to validate the biological processes identified and indicate that microarray analysis may provide a valuable, fast and unbiased tool to evaluate the in vivo performance of tissue-engineered constructs. However, challenges remain e.g. with regards to the development of specific GO-terms and annotation of the (rat) genome.


Acta Biomaterialia | 2011

Design and in vivo evaluation of a molecularly defined acellular skin construct: reduction of early contraction and increase in early blood vessel formation.

S.T.M. Nillesen; Gerwen Lammers; Ronnie G. Wismans; M.M. Ulrich; E. Middelkoop; P.H.M. Spauwen; Kaeuis A. Faraj; Joost Schalkwijk; Willeke F. Daamen; A.H.M.S.M. van Kuppevelt

Skin substitutes are of great benefit in the treatment of patients with full thickness wounds, but there is a need for improvement with respect to wound closure with minimal contraction, early vascularisation, and elastin formation. In this study we designed and developed an acellular double-layered skin construct, using matrix molecules and growth factors to target specific biological processes. The epidermal layer was prepared using type I collagen, heparin and fibroblast growth factor 7 (FGF7), while the porous dermal layer was prepared using type I collagen, solubilised elastin, dermatan sulfate, heparin, fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor (VEGF). The construct was biochemically and morphologically characterised and evaluated in vivo using a rat full thickness wound model. The results were compared with the commercial skin substitute IntegraDRT and untreated wounds. The double-layered construct was prepared according to the design specifications. The epidermal layer was about 40 μm thick, containing 9% heparin and 0.2 μg FGF7 mg per layer, localised at the periphery. The dermal layer was 2.5 mm thick, had rounded pores and contained 10% dermatan sulfate+heparin, and 0.7 μg FGF2+VEGF mg per layer. The double-layered skin construct was implanted in a skin defect and on day 7, 14, 28 and 112 the (remaining) wound area was photographed, excised and (immuno) histologically evaluated. The double-layered skin construct showed more cell influx, significantly less contraction and increased blood vessel formation at early time points in comparison with IntegraDRT and/or the untreated wound. On day 14 the double-layered skin construct also had the fewest myofibroblasts present. On day 112 the double-layered skin construct contained more elastic fibres than IntegraDRT and the untreated wound. Structures resembling hair follicles and sebaceous glands were found in the double-layered skin construct and the untreated wound, but hardly any were found in IntegraDRT. The results provide new opportunities for the application of acellular skin constructs in the treatment of surgical wounds.


Macromolecular Bioscience | 2012

Construction of a Microstructured Collagen Membrane Mimicking the Papillary Dermis Architecture and Guiding Keratinocyte Morphology and Gene Expression

Gerwen Lammers; Günter Roth; Mathias Heck; Roland Zengerle; G. Sandra Tjabringa; Elly M. M. Versteeg; Theo Hafmans; Ronnie G. Wismans; Dieter P. Reinhardt; Eugène T P Verwiel; Patrick L.J.M. Zeeuwen; Joost Schalkwijk; Roland Brock; Willeke F. Daamen; Toin H. van Kuppevelt

A papillary-structured collagen fibril membrane is created, mimicking the 3D-architecture of the human papillary dermis. Primary human keratinocytes cultured to confluency on papillar-structured films are compared to keratinocytes cultured on flat membranes. Microscopical evaluation reveals the presence of morphologically distinct cells at the base of the papillar structures that are not observed on flat membranes. Gene expression microarrays and RT-qPCR indicate that these cells are in a more proliferative/migrational state, whereas cells on flat membranes have a more differentiated expression profile. Immunohistochemical stainings confirm these results. In conclusion, specific collagen architecture can direct keratinocyte behavior, and this may be used to further improve skin regeneration.


International Journal of Pharmaceutics | 2012

Lyophilisomes as a new generation of drug delivery capsules

Etienne van Bracht; René Raavé; Wouter P. R. Verdurmen; Ronnie G. Wismans; Paul J. Geutjes; Roland Brock; Egbert Oosterwijk; Toin H. van Kuppevelt; Willeke F. Daamen

Nanoparticulate drug delivery systems are currently explored to overcome critical challenges associated with classical administration forms. In this study, we present a drug delivery system based on a novel class of proteinaceous biodegradable nano/micro capsules, lyophilisomes. Lyophilisomes can be prepared from biomolecules without the need for amphiphilicity. Albumin-based lyophilisomes were prepared by freezing, annealing and lyophilizing, resulting in capsules ranging from 100 to 3000 nm. Lyophilisomes were loaded with the anti-tumor drugs doxorubicin and curcumin using different concentrations and time/temperature regimes. Incubation in 0.1 mg/ml doxorubicin or 1.0 mg/ml curcumin resulted in an entrapment efficiency of 95±1% and 4±1%, respectively. This corresponds to a drug loading of 0.24 mg doxorubicin per milligram albumin and 0.10 mg curcumin per milligram albumin. Drug release profiles from doxorubicin and curcumin-loaded lyophilisomes were studied in culture medium and showed slow release for doxorubicin (2.7% after 72 h), and rapid release for curcumin (55% after 72 h). When applied to cells, non-loaded lyophilisomes did not influence cell viability, even at high concentrations (1 mg/ml). Lyophilisomes were internalized by cells. When loaded with doxorubicin and curcumin, lyophilisomes strongly reduced cell proliferation and viability of SKOV-3 and HeLa cells, respectively, to a level similar or better compared to an equal amount of free drugs. In conclusion, albumin lyophilisomes show potential as (nano)carriers of drugs for tumor cell elimination.


Journal of Tissue Engineering and Regenerative Medicine | 2013

Repair of surgically created diaphragmatic defect in rat with use of a crosslinked porous collagen scaffold.

Katrien M. Brouwer; Willeke F. Daamen; Daphne Reijnen; Ruud H. Verstegen; Gerwen Lammers; Theo Hafmans; Ronnie G. Wismans; Toin H. van Kuppevelt; Rene Wijnen

Large defects in congenital diaphragmatic hernia are closed by patch repair, which is associated with a high complication risk and reherniation rate. New treatment modalities are warranted. We evaluated the feasibility of using an acellular biodegradable collagen bioscaffold for a regenerative medicine approach to close a surgically created diaphragmatic defect in a rat model. Scaffold degradation, cellular ingrowth and regeneration of the diaphragm were studied. In 25 rats, a subcostal incision was made and one third of the right hemidiaphragm was resected. Crosslinked porous type I collagen scaffolds (Ø ~ 14 mm) were sutured into the lesion. Rats were sacrificed at 2, 4, 8, 12 or 24 weeks after scaffold implantation. Implants were evaluated macroscopically and (immuno)histologically. Survival after surgery was 88% with no evidence of reherniation. Histological examination showed that the collagen scaffold degraded slowly and new collagen, elastin and mesothelium were deposited. Blood vessels were observed primarily at the outer borders of the scaffold; their number gradually increased in time. Muscle fibres were found on the scaffold covering up to 10% of the defect. Macroscopically, adhesion of the scaffold to the liver was observed. Use of a collagen scaffold to close a surgically created diaphragmatic defect is feasible, with evidence of new tissue formation. The use of crosslinked collagen scaffolds allows targeted modification; e.g. addition of growth factors to further stimulate growth of muscle cells. Copyright

Collaboration


Dive into the Ronnie G. Wismans's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Willeke F. Daamen

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Kaeuis A. Faraj

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Paul J. Geutjes

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Angelique L. Rops

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Gerwen Lammers

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

J.H.M. Berden

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Joost Schalkwijk

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Suzan T.M. Nillesen

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

T.G.M. Hafmans

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge