Network


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

Hotspot


Dive into the research topics where Steven De Jonghe is active.

Publication


Featured researches published by Steven De Jonghe.


Bioorganic & Medicinal Chemistry Letters | 1999

Structure-activity relationship of short-chain sphingoid bases as inhibitors of sphingosine kinase.

Steven De Jonghe; Ilse Van Overmeire; Samantha Poulton; Chris Hendrix; Roger Busson; Serge Van Calenbergh; Denis De Keukeleire; Sarah Spiegel; Piet Herdewijn

Short-chain sphinganine analogues 8, 9, 18, and 19, as well as 3-fluoro-sphingosine analogues 25 and 26 were synthesized. Their potential as sphingosine kinase inhibitors was investigated, in combination with previously synthesized sphingosine and fluorinated sphinganine analogues.


Bioorganic & Medicinal Chemistry | 2011

Synthesis of novel 5-amino-thiazolo[4,5-d]pyrimidines as E. coli and S. aureus SecA inhibitors.

Miyeon Jang; Steven De Jonghe; Kenneth Segers; Jozef Anné; Piet Herdewijn

An efficient synthesis of a library of 5-amino-thiazolo[4,5-d]pyrimidines is reported. Regioselective displacements of chlorines, as well as regioselective diazotation reactions are described, which allow the introduction of structural diversity on the scaffold by consecutive reactions. Screening of this focused library led to the discovery of SecA inhibitors from Escherichia coli and Staphylococcus aureus.


World Journal of Biological Chemistry | 2011

Gelatin degradation assay reveals MMP-9 inhibitors and function of O-glycosylated domain.

Jennifer Vandooren; Nathalie Geurts; Erik Martens; Philippe E. Van den Steen; Steven De Jonghe; Piet Herdewijn; Ghislain Opdenakker

AIM To establish a novel, sensitive and high-throughput gelatinolytic assay to define new inhibitors and compare domain deletion mutants of gelatinase B/matrix metalloproteinase (MMP)-9. METHODS Fluorogenic Dye-quenched (DQ)™-gelatin was used as a substrate and biochemical parameters (substrate and enzyme concentrations, DMSO solvent concentrations) were optimized to establish a high-throughput assay system. Various small-sized libraries (ChemDiv, InterBioScreen and ChemBridge) of heterocyclic, drug-like substances were tested and compared with prototypic inhibitors. RESULTS First, we designed a test system with gelatin as a natural substrate. Second, the assay was validated by selecting a novel pyrimidine-2,4,6-trione (barbiturate) inhibitor. Third, and in line with present structural data on collagenolysis, it was found that deletion of the O-glycosylated region significantly decreased gelatinolytic activity (k(cat)/k(M) ± 40% less than full-length MMP-9). CONCLUSION The DQ™-gelatin assay is useful in high-throughput drug screening and exosite targeting. We demonstrate that flexibility between the catalytic and hemopexin domain is functionally critical for gelatinolysis.


Journal of Medicinal Chemistry | 2011

Discovery of 7-N-Piperazinylthiazolo[5,4-d]pyrimidine Analogues as a Novel Class of Immunosuppressive Agents with in Vivo Biological Activity

Mi-Yeon Jang; Yuan Lin; Steven De Jonghe; Ling-Jie Gao; Bart Vanderhoydonck; Mathy Froeyen; Jef Rozenski; Jean Herman; Thierry Louat; Kristien Van Belle; Mark Waer; Piet Herdewijn

Herein we describe the synthesis and in vitro and in vivo activity of thiazolo[5,4-d]pyrimidines as a novel class of immunosuppressive agents, useful for preventing graft rejection after organ transplantation. This research resulted in the discovery of a series of compounds with potent activity in the mixed lymphocyte reaction (MLR) assay, which is well-known as the in vitro model for in vivo rejection after organ transplantation. The most potent congeners displayed IC(50) values of less than 50 nM in this MLR assay and hence are equipotent to cyclosporin A, a clinically used immunosuppressive drug. One representative of this series was further evaluated in a preclinical animal model of organ transplantation and showed excellent in vivo efficacy. It validates these compounds as new promising immunosuppressive drugs.


European Journal of Organic Chemistry | 2000

Synthesis of fluorinated sphinganine and dihydroceramide analogues

Steven De Jonghe; Ilse Van Overmeire; Serge Van Calenbergh; Chris Hendrix; Roger Busson; Denis De Keukeleire; Piet Herdewijn

With the aim of uncovering inhibitors of dihydroceramide desaturase and ceramide synthase and studying their substrate specificity, the synthesis of short-chain 3-fluorosphinganine and 3-fluorodihydroceramide analogues was effected. The synthesis starts from the known alkynols 1 and 10, respectively, and from the Garner aldehyde 15. The key step is the introduction of a fluorine atom using diethylaminosulfur trifluoride, which proceeds efficiently for erythro-alcohols 2 and 16, but gives rise to cyclization reactions for threo compounds 11 and 17.


Bioorganic & Medicinal Chemistry Letters | 1999

Synthesis and apoptogenic activity of fluorinated ceramide and dihydroceramide analogues

Steven De Jonghe; Ilse Van Overmeire; Johan Gunst; André De Bruyn; Chris Hendrix; Serge Van Calenbergh; Roger Busson; Denis De Keukeleire; Jan Philippé; Piet Herdewijn

Short-chain 3-fluoro-(dihydro)ceramide analogues are synthesized from L-serine using diethylaminosulfur trifluoride (DAST) as fluorinating agent. The apoptogenic activity of these compounds was measured in three different cell lines and compared with their hydroxylated counterparts.


ChemMedChem | 2013

Discovery of an Acyclic Nucleoside Phosphonate that Inhibits Mycobacterium tuberculosis ThyX Based on the Binding Mode of a 5‐Alkynyl Substrate Analogue

Anastasia Parchina; Matheus Froeyen; Lia Margamuljana; Jef Rozenski; Steven De Jonghe; Yves Briers; Rob Lavigne; Piet Herdewijn; Eveline Lescrinier

The urgent need for new antibiotics poses a challenge to target un(der)exploited vital cellular processes. Thymidylate biosynthesis is one such process due to its crucial role in DNA replication and repair. Thymidylate synthases (TS) catalyze a crucial step in the biosynthesis of thymidine 5‐triphosphate (TTP), an elementary building block required for DNA synthesis and repair. To date, TS inhibitors have only been successfully applied in anticancer therapy due to their lack of specificity for antimicrobial versus human enzymes. However, the discovery of a new family of TS enzymes (ThyX) in a range of pathogenic bacteria that is structurally and biochemically different from the “classic” TS (ThyA) has opened the possibility to develop selective ThyX inhibitors as potent antimicrobial drugs. Here, the interaction of the known inhibitor 5‐(3‐octanamidoprop‐1yn‐1yl)‐2′‐deoxyuridine‐5′‐monophosphate (1) with Mycobacterium tuberculosis ThyX enzyme is explored using molecular modeling starting from published crystal structures, with further confirmation through NMR experiments. While the deoxyuridylate (dUMP) moiety of compound 1 occupies the cavity of the natural substrate in ThyX, the rest of the ligand (the “5‐alkynyl tail”) extends to the outside of the enzyme between two of its four subunits. The hydrophobic pocket that accommodates the alkyl part of the tail is formed by displacement of Tyr 44.C, Tyr 108.A and Lys 165.A. Changes to the resonance of the Lys 165 NH3 group upon ligand binding were monitored in a titration experiment by 2D HISQC NMR. Guided by the results of the modeling and NMR studies, and inspired by the success of acyclic antiviral nucleosides, compounds where a 5‐alkynyl uracyl moiety is coupled to an acyclic nucleoside phosphonate (ANP) were synthesized and evaluated. Of the compounds evaluated, sodium (6‐(5‐(3‐octanamidoprop‐1‐yn‐1‐yl)‐2,4‐dioxo‐3,4‐dihydropyrimidin‐1(2H)‐yl)hexyl)phosphonate (3 e) exhibited 43 % of inhibitory effect on ThyX at 50 μM. While only modest activity was achieved, this is the first example of an ANP inhibiting ThyX, and these results can be used to further guide structural modifications to this class to develop more potent compounds with potential application as antibacterial agents acting through a novel mechanism of action.


Journal of Medicinal Chemistry | 2015

Selective Inhibitors of Cyclin G Associated Kinase (GAK) as Anti-Hepatitis C Agents

Sona Kovackova; Lei Chang; Elena Bekerman; Gregory Neveu; Rina Barouch-Bentov; A. Chaikuad; Christina Heroven; Michal Šála; Steven De Jonghe; Stefan Knapp; Shirit Einav; Piet Herdewijn

Cyclin G associated kinase (GAK) emerged as a promising drug target for the treatment of viral infections. However, no potent and selective GAK inhibitors have been reported in the literature to date. This paper describes the discovery of isothiazolo[5,4-b]pyridines as selective GAK inhibitors, with the most potent congeners displaying low nanomolar binding affinity for GAK. Cocrystallization experiments revealed that these compounds behaved as classic type I ATP-competitive kinase inhibitors. In addition, we have demonstrated that these compounds exhibit a potent activity against hepatitis C virus (HCV) by inhibiting two temporally distinct steps in the HCV life cycle (i.e., viral entry and assembly). Hence, these GAK inhibitors represent chemical probes to study GAK function in different disease areas where GAK has been implicated (including viral infection, cancer, and Parkinsons disease).


Journal of Clinical Investigation | 2017

Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad-spectrum antiviral effects

Elena Bekerman; Gregory Neveu; Ana Shulla; Jennifer M. Brannan; Szu-Yuan Pu; Stanley Wang; Fei Xiao; Rina Barouch-Bentov; Russell R. Bakken; Roberto Mateo; Jennifer Govero; Claude M. Nagamine; Michael S. Diamond; Steven De Jonghe; Piet Herdewijn; John M. Dye; Glenn Randall; Shirit Einav

Global health is threatened by emerging viral infections, which largely lack effective vaccines or therapies. Targeting host pathways that are exploited by multiple viruses could offer broad-spectrum solutions. We previously reported that AAK1 and GAK, kinase regulators of the host adaptor proteins AP1 and AP2, are essential for hepatitis C virus (HCV) infection, but the underlying mechanism and relevance to other viruses or in vivo infections remained unknown. Here, we have discovered that AP1 and AP2 cotraffic with HCV particles in live cells. Moreover, we found that multiple viruses, including dengue and Ebola, exploit AAK1 and GAK during entry and infectious virus production. In cultured cells, treatment with sunitinib and erlotinib, approved anticancer drugs that inhibit AAK1 or GAK activity, or with more selective compounds inhibited intracellular trafficking of HCV and multiple unrelated RNA viruses with a high barrier to resistance. In murine models of dengue and Ebola infection, sunitinib/erlotinib combination protected against morbidity and mortality. We validated sunitinib- and erlotinib-mediated inhibition of AAK1 and GAK activity as an important mechanism of antiviral action. Additionally, we revealed potential roles for additional kinase targets. These findings advance our understanding of virus-host interactions and establish a proof of principle for a repurposed, host-targeted approach to combat emerging viruses.


Journal of Medicinal Chemistry | 2014

Discovery of Dual Death-Associated Protein Related Apoptosis Inducing Protein Kinase 1 and 2 Inhibitors by a Scaffold Hopping Approach

Ling-Jie Gao; Sona Kovackova; Michal Šála; Anna Teresa Ramadori; Steven De Jonghe; Piet Herdewijn

DRAK2 emerged as a promising drug target for the treatment of autoimmune diseases and to prevent graft rejection after organ transplantation. Screening of a compound library in a DRAK2 binding assay led to the identification of an isothiazolo[5,4-b]pyridine derivative as a novel ligand for DRAK2, displaying a Kd value of 1.6 μM. Subsequent medicinal chemistry work led to the discovery of a thieno[2,3-b]pyridine derivative with strong DRAK2 binding affinity (Kd = 9 nM). Moreover, this compound also behaves as a functional inhibitor of DRAK2 enzymatic activity, displaying an IC50 value of 0.82 μM, although lacking selectivity, when tested against DRAK1. This paper describes for the first time functionally active dual DRAK1 and DRAK2 inhibitors that can be used as starting point for the synthesis of chemical tool compounds to study DRAK1 and DRAK2 biology, or they can be considered as hit compounds for hit-to-lead optimization campaigns in drug discovery programs.

Collaboration


Dive into the Steven De Jonghe's collaboration.

Top Co-Authors

Avatar

Piet Herdewijn

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jef Rozenski

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Eveline Lescrinier

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elisabetta Groaz

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Graciela Andrei

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ling-Jie Gao

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bart Vanderhoydonck

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Chris Hendrix

Rega Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge