Network


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

Hotspot


Dive into the research topics where J.F. Dekkers is active.

Publication


Featured researches published by J.F. Dekkers.


Nature Chemical Biology | 2013

Mechanism-based corrector combination restores ΔF508-CFTR folding and function

Tsukasa Okiyoneda; Guido Veit; J.F. Dekkers; Miklos Bagdany; Naoto Soya; Haijin Xu; Ariel Roldan; A. S. Verkman; Mark J. Kurth; Ágnes Simon; Tamás Hegedüs; Jeffrey M. Beekman; Gergely L. Lukacs

The most common cystic fibrosis mutation, ΔF508 in nucleotide binding domain 1 (NBD1), impairs cystic fibrosis transmembrane conductance regulator (CFTR)-coupled domain folding, plasma membrane expression, function and stability. VX-809, a promising investigational corrector of ΔF508-CFTR misprocessing, has limited clinical benefit and an incompletely understood mechanism, hampering drug development. Given the effect of second-site suppressor mutations, robust ΔF508-CFTR correction most likely requires stabilization of NBD1 energetics and the interface between membrane-spanning domains (MSDs) and NBD1, which are both established primary conformational defects. Here we elucidate the molecular targets of available correctors: class I stabilizes the NBD1-MSD1 and NBD1-MSD2 interfaces, and class II targets NBD2. Only chemical chaperones, surrogates of class III correctors, stabilize human ΔF508-NBD1. Although VX-809 can correct missense mutations primarily destabilizing the NBD1-MSD1/2 interface, functional plasma membrane expression of ΔF508-CFTR also requires compounds that counteract the NBD1 and NBD2 stability defects in cystic fibrosis bronchial epithelial cells and intestinal organoids. Thus, the combination of structure-guided correctors represents an effective approach for cystic fibrosis therapy.


Science Translational Medicine | 2016

Characterizing responses to CFTR-modulating drugs using rectal organoids derived from subjects with cystic fibrosis

J.F. Dekkers; G. Berkers; Evelien Kruisselbrink; Annelotte Vonk; Hugo R. de Jonge; H.M. Janssens; I. Bronsveld; Eduard A. van de Graaf; Edward E. S. Nieuwenhuis; Roderick H. J. Houwen; Frank P. Vleggaar; Johanna C. Escher; Yolanda B. de Rijke; Christof J. Majoor; Harry Heijerman; Karin M. de Winter-de Groot; Hans Clevers; Cornelis K. van der Ent; Jeffrey M. Beekman

Rectal organoids from subjects with cystic fibrosis can be used to assess responses to drugs that modulate CFTR. Mini-guts for personalized cystic fibrosis therapy Cystic fibrosis is caused by mutations in the CFTR gene that severely reduce the function of the CFTR protein. New drugs for treating cystic fibrosis modulate CFTR protein function, but drug efficacy is dependent on which CFTR mutation a patient carries. Dekkers et al. now show that the efficacy of these drugs can be individually assessed in a laboratory test using epithelial cells cultured as mini-guts from rectal biopsies from subjects with cystic fibrosis. The authors show that the drug responses observed in mini-guts or rectal organoids can be used to predict which patients may be potential responders to the drug. This preclinical test may help to quickly identify responders to CFTR-modulating drug therapy even when patients carry very rare CFTR mutations. Identifying subjects with cystic fibrosis (CF) who may benefit from cystic fibrosis transmembrane conductance regulator (CFTR)–modulating drugs is time-consuming, costly, and especially challenging for individuals with rare uncharacterized CFTR mutations. We studied CFTR function and responses to two drugs—the prototypical CFTR potentiator VX-770 (ivacaftor/KALYDECO) and the CFTR corrector VX-809 (lumacaftor)—in organoid cultures derived from the rectal epithelia of subjects with CF, who expressed a broad range of CFTR mutations. We observed that CFTR residual function and responses to drug therapy depended on both the CFTR mutation and the genetic background of the subjects. In vitro drug responses in rectal organoids positively correlated with published outcome data from clinical trials with VX-809 and VX-770, allowing us to predict from preclinical data the potential for CF patients carrying rare CFTR mutations to respond to drug therapy. We demonstrated proof of principle by selecting two subjects expressing an uncharacterized rare CFTR genotype (G1249R/F508del) who showed clinical responses to treatment with ivacaftor and one subject (F508del/R347P) who showed a limited response to drug therapy both in vitro and in vivo. These data suggest that in vitro measurements of CFTR function in patient-derived rectal organoids may be useful for identifying subjects who would benefit from CFTR-correcting treatment, independent of their CFTR mutation.


Cellular Microbiology | 2013

Distinct localization of the complement C5b‐9 complex on Gram‐positive bacteria

Evelien T.M. Berends; J.F. Dekkers; Reindert Nijland; Annemarie Kuipers; Jasper A. Soppe; Jos A. G. van Strijp; Suzan H.M. Rooijakkers

The plasma proteins of the complement system fulfil important immune defence functions, including opsonization of bacteria for phagocytosis, generation of chemo‐attractants and direct bacterial killing via the Membrane Attack Complex (MAC or C5b‐9). The MAC is comprised of C5b, C6, C7, C8, and multiple copies of C9 that generate lytic pores in cellular membranes. Gram‐positive bacteria are protected from MAC‐dependent lysis by their thick peptidoglycan layer. Paradoxically, several Gram‐positive pathogens secrete small proteins that inhibit C5b‐9 formation. In this study, we found that complement activation on Gram‐positive bacteria in serum results in specific surface deposition of C5b‐9 complexes. Immunoblotting revealed that C9 occurs in both monomeric and polymeric (SDS‐stable) forms, indicating the presence of ring‐structured C5b‐9. Surprisingly, confocal microscopy demonstrated that C5b‐9 deposition occurs at specialized regions on the bacterial cell. On Streptococcus pyogenes, C5b‐9 deposits near the division septum whereas on Bacillus subtilis the complex is located at the poles. This is in contrast to C3b deposition, which occurs randomly on the bacterial surface. Altogether, these results show a previously unrecognized interaction between the C5b‐9 complex and Gram‐positive bacteria, whichmight ultimately lead to a new model of MAC assembly and functioning.


Rare diseases (Austin, Tex.) | 2013

Novel opportunities for CFTR-targeting drug development using organoids

J.F. Dekkers; Cornelis K. van der Ent; Jeffrey M. Beekman

Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CFTR mutations lead to production of non-functional CFTR, reduced amounts of normal functioning CFTR or misfolded CFTR with defects in trafficking or function. For decades, CF treatment has been focused on the symptoms of CF, but pharmacotherapy using small molecules that target the basic defect of CF, the mutant CFTR protein, is now possible for a limited amount of subjects with CF. This raises the exciting possibility that the majority of people with CF may receive effective treatment targeting the different CFTR mutants in the future. We recently described a functional CFTR assay using rectal biopsies from subjects with CF that were cultured in vitro into self-organizing mini-guts or organoids. We here describe how this model may assist in the discovery of new CFTR-targeting drugs, the subjects that may benefit from these drugs, and the mechanisms underlying variability in CFTR genotype-phenotype relations.


Journal of Cystic Fibrosis | 2016

Limited premature termination codon suppression by read-through agents in cystic fibrosis intestinal organoids

D.D. Zomer-van Ommen; L.A.W. Vijftigschild; E. Kruisselbrink; A.M. Vonk; J.F. Dekkers; Hettie M. Janssens; K.M. de Winter-de Groot; C.K. van der Ent; Jeffrey M. Beekman

Premature termination codon read-through drugs offer opportunities for treatment of multiple rare genetic diseases including cystic fibrosis. We here analyzed the read-through efficacy of PTC124 and G418 using human cystic fibrosis intestinal organoids (E60X/4015delATTT, E60X/F508del, G542X/F508del, R1162X/F508del, W1282X/F508del and F508del/F508del). G418-mediated read-through induced only limited CFTR function, but functional restoration of CFTR by PTC124 could not be confirmed. These studies suggest that better read-through agents are needed for robust treatment of nonsense mutations in cystic fibrosis.


Trends in Molecular Medicine | 2012

PPARγ as a therapeutic target in cystic fibrosis

J.F. Dekkers; Cornelis K. van der Ent; Eric Kalkhoven; Jeffrey M. Beekman

Cystic fibrosis (CF) is characterized by a proinflammatory pulmonary condition that may result from increased infections and altered intracellular metabolism in CFTR-deficient cells. The lipid-activated transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) has well-established roles in immune cell function and inflammatory modulation and has been demonstrated to play an important role in the heightened inflammatory response in CF cells. Here, we summarize current literature describing PPARγ-dependent alterations of CF cells and discuss the potential of PPARγ ligands for treating CF.


European Respiratory Journal | 2016

β2-Adrenergic receptor agonists activate CFTR in intestinal organoids and subjects with cystic fibrosis

L.A.W. Vijftigschild; G. Berkers; J.F. Dekkers; Domenique D. Zomer-van Ommen; Elizabeth Matthes; Evelien Kruisselbrink; Annelotte Vonk; Chantal E Hensen; Sabine Heida-Michel; Margot Geerdink; Hettie M. Janssens; Eduard A. van de Graaf; I. Bronsveld; Karin M. de Winter-de Groot; Christof J. Majoor; Harry Heijerman; Hugo R. de Jonge; John W. Hanrahan; Cornelis K. van der Ent; Jeffrey M. Beekman

We hypothesized that people with cystic fibrosis (CF) who express CFTR (cystic fibrosis transmembrane conductance regulator) gene mutations associated with residual function may benefit from G-protein coupled receptor (GPCR)-targeting drugs that can activate and enhance CFTR function. We used intestinal organoids to screen a GPCR-modulating compound library and identified β2-adrenergic receptor agonists as the most potent inducers of CFTR function. β2-Agonist-induced organoid swelling correlated with the CFTR genotype, and could be induced in homozygous CFTR-F508del organoids and highly differentiated primary CF airway epithelial cells after rescue of CFTR trafficking by small molecules. The in vivo response to treatment with an oral or inhaled β2-agonist (salbutamol) in CF patients with residual CFTR function was evaluated in a pilot study. 10 subjects with a R117H or A455E mutation were included and showed changes in the nasal potential difference measurement after treatment with oral salbutamol, including a significant improvement of the baseline potential difference of the nasal mucosa (+6.35 mV, p<0.05), suggesting that this treatment might be effective in vivo. Furthermore, plasma that was collected after oral salbutamol treatment induced CFTR activation when administered ex vivo to organoids. This proof-of-concept study suggests that organoids can be used to identify drugs that activate CFTR function in vivo and to select route of administration. β2-Adrenergic receptor agonists as CFTR activating drugs in subjects with CFTR residual function http://ow.ly/Dm44300wjuw


Journal of Cystic Fibrosis | 2013

CFTR-mutation specific applications of CFTR-directed monoclonal antibodies.

M.A. van Meegen; S.W.J. Terheggen; K.J. Koymans; L.A.W. Vijftigschild; J.F. Dekkers; C.K. van der Ent; Jeffrey M. Beekman

BACKGROUND Over the last decade novel monoclonal CFTR-specific antibodies have been developed. We here present a paired analysis to detect wild-type and mutant CFTR using Western blot analysis, flow cytometry and confocal microscopy in several cellular expression systems. METHODS The following CFTR-specific antibodies were used; 217, 432, 450, 570, 769, 596, 660, L12B4 and 24.1. Mutant CFTR was detected in HEK293 cells transiently expressing the mutations; G542X, R1162X, F508del, N1303K, G551D, R117H, A455E. RESULTS The majority of these antibodies are suitable for most applications tested. Using immunofluorescence, some antibodies can better detect mutant forms of CFTR (F508del and N1303K by mAbs 596 and 769), or display lower aspecific detection by Western blot analysis (mAbs 432, 450, 769 and 596) or immunofluorescence (mAbs 432, 450, 570 and 769). CONCLUSION Optimal detection of CFTR by monoclonal antibodies depends on CFTR mutation and the specific research application.


Journal of Cystic Fibrosis | 2015

WS01.1 rAAV2/5 encoding a truncated CFTR rescues the CF phenotype in intestinal organoids and a CF mouse model

Marianne Carlon; Dragana Vidovic; J.F. Dekkers; M. Faria da Cunha; M.I. Hollenhorst; M. Bijvelds; C. Van den Haute; Veerle Baekelandt; H.R. de Jonge; K. De Boeck; Rik Gijsbers; Isabelle Sermet-Gaudelus; Aleksander Edelman; Jeffrey M. Beekman; Zeger Debyser

To date, the majority of CF patients receive symptomatic treatment. Gene therapy offers the potential to cure CF in mutation-independent manner. Here, we evaluate rAAV2/5 gene therapy for CF in human intestinal organoids and a ΔF508 mouse model. First, we studied the longevity of gene expression after a single administration of a rAAV2/5-Fluc to mouse airways. This resulted in relatively stable gene expression up to 15 months with only a 4-fold decrease in bioluminescent signal in lungs. Due to the rAAV size limit, we used a truncated CFTR missing a portion of the R-domain, CFTRΔR (Ostedgaard et al., PNAS, 2005). First, we evaluated its functionality in a physiological relevant and highly translational organoid model. These data demonstrate that the therapeutic vector rAAV-CFTRΔR is functional and that the ion transport-induced organoid swelling is CFTR-specific. Indeed, despite the expression cassette being at the limit of the rAAV packaging capacity, intact genomes are incorporated into vector particles, that allow efficient second strand DNA synthesis and transgene expression. Finally, we administered rAAV2/5-CFTRΔR to ΔF508 mice by nasal instillation. 2–4 weeks later, we demonstrated a response to low-chloride and forskolin perfusion in 6 out of 8 mice by nasal potential differences, indicating restoration of chloride transport across nasal mucosa. In conclusion, we obtained sustained reporter gene expression in murine airways and demonstrated restoration of the CF phenotype in CF organoids and a CF mouse model. Our results underscore the therapeutic potential of rAAV2/5-CFTRΔR for CF gene therapy opening new avenues towards a definitive cure for all CF patients.


European Respiratory Journal | 2018

Stratifying infants with cystic fibrosis for disease severity using intestinal organoid swelling as a biomarker of CFTR function

Karin M. de Winter-de Groot; Hettie M. Janssens; Rick T. van Uum; J.F. Dekkers; G. Berkers; Annelotte Vonk; Evelien Kruisselbrink; Hugo Oppelaar; Robert P. de Vries; Hans Clevers; Roderick H. J. Houwen; Johanna C. Escher; Sjoerd G. Elias; Hugo R. de Jonge; Yolanda B. de Rijke; Harm A.W.M. Tiddens; Cornelis K. van der Ent; Jeffrey M. Beekman

Forskolin-induced swelling (FIS) of intestinal organoids from individuals with cystic fibrosis (CF) measures function of the cystic fibrosis transmembrane conductance regulator (CFTR), the protein mutated in CF. We investigated whether FIS corresponds with clinical outcome parameters and biomarkers of CFTR function in 34 infants diagnosed with CF. Relationships with FIS were studied for indicators of pulmonary and gastrointestinal disease. Children with low FIS had higher levels of immunoreactive trypsinogen (p=0.030) and pancreatitis-associated protein (p=0.039), more often had pancreatic insufficiency (p<0.001), had more abnormalities on chest computed tomography (p=0.049), and had lower z-scores for maximal expiratory flow at functional residual capacity (p=0.033) when compared to children with high FIS values. FIS significantly correlated with sweat chloride concentration (SCC) and intestinal current measurement (ICM) (r= −0.82 and r=0.70, respectively; both p<0.001). Individual assessment of SCC, ICM and FIS suggested that FIS can help to classify individual disease severity. Thus, stratification by FIS identified subgroups that differed in pulmonary and gastrointestinal outcome parameters. FIS of intestinal organoids correlated well with established CFTR-dependent biomarkers such as SCC and ICM, and performed adequately at group and individual level in this proof-of-concept study. Laboratory-grown mini-guts inform on individual disease characteristics of infants with cystic fibrosis http://ow.ly/J19W30ldzTH

Collaboration


Dive into the J.F. Dekkers's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C.K. van der Ent

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans Clevers

Royal Netherlands Academy of Arts and Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge