Network


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

Hotspot


Dive into the research topics where Bas Heinhuis is active.

Publication


Featured researches published by Bas Heinhuis.


Blood | 2009

Differential requirement for the activation of the inflammasome for processing and release of IL-1beta in monocytes and macrophages.

Mihea G Netea; Claudia A. Nold-Petry; Marcel F. Nold; Leo A. B. Joosten; Bastian Opitz; Jonathan H M van der Meer; Frank L. van de Veerdonk; Gerben Ferwerda; Bas Heinhuis; Isabel Devesa; C. Joel Funk; Robert J. Mason; Bart Jan Kullberg; Anna Rubartelli; Jos W. M. van der Meer; Charles A. Dinarello

The processing of pro-interleukin-1beta depends on activation of caspase-1. Controversy has arisen whether Toll-like receptor (TLR) ligands alone can activate caspase-1 for release of interleukin-1beta (IL-1beta). Here we demonstrate that human blood monocytes release processed IL-1beta after a one-time stimulation with either TLR2 or TLR4 ligands, resulting from constitutively activated caspase-1 and release of endogenous adenosine triphosphate. The constitutive activation of caspase-1 depends on the inflammasome components, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and NALP3, but in monocytes caspase-1 activation is uncoupled from pathogen-associated molecular pattern recognition. In contrast, macrophages are unable to process and release IL-1beta solely by TLR ligands and require a second adenosine triphosphate stimulation. We conclude that IL-1beta production is differentially regulated in monocytes and macrophages, and this reflects their separate functions in host defense and inflammation.


PLOS ONE | 2011

STAT1 hyperphosphorylation and defective IL12R/IL23R signaling underlie defective immunity in autosomal dominant chronic mucocutaneous candidiasis.

Sanne P. Smeekens; Theo S. Plantinga; Frank L. van de Veerdonk; Bas Heinhuis; Alexander Hoischen; Leo A. B. Joosten; Peter D. Arkwright; Andrew R. Gennery; Bart Jan Kullberg; Joris A. Veltman; Desa Lilic; Jos W. M. van der Meer; Mihai G. Netea

We recently reported the genetic cause of autosomal dominant chronic mucocutaneous candidiasis (AD-CMC) as a mutation in the STAT1 gene. In the present study we show that STAT1 Arg274Trp mutations in the coiled-coil (CC) domain is the genetic cause of AD-CMC in three families of patients. Cloning and transfection experiments demonstrate that mutated STAT1 inhibits IL12R/IL-23R signaling, with hyperphosphorylation of STAT1 as the likely underlying molecular mechanism. Inhibition of signaling through the receptors for IL-12 and IL-23 leads to strongly diminished Th1/Th17 responses and hence to increased susceptibility to fungal infections. The challenge for the future is to translate this knowledge into novel strategies for the treatment of this severe immunodeficiency.


Annals of the Rheumatic Diseases | 2011

Tumour necrosis factor alpha-driven IL-32 expression in rheumatoid arthritis synovial tissue amplifies an inflammatory cascade

Bas Heinhuis; M.M.J.F. Koenders; P.L.C.M. van Riel; F.A.J. van de Loo; Charles A. Dinarello; Mihai G. Netea; W.B. van den Berg; Leo A. B. Joosten

Objective To investigate the interplay between IL-32 and tumour necrosis factor alpha (TNFα) during the chronic inflammation of rheumatoid arthritis (RA) and to assess whether anti-TNFα treatment of RA patients modulates synovial IL-32 expression. Methods Induction of IL-32γ by Pam3Cys, lipopolysaccharide, IL-1β or TNFα was investigated in human fibroblast-like synoviocytes (FLS). Stimulation of TNFα production by IL-32γ was studied by adenoviral overexpression of IL-32γ (AdIL-32γ) and lipopolysaccharide stimulation of THP1 cells. Silencing of endogenous IL-32 was employed to study cytokine regulation in FLS. AdIL-32γ followed by TNFα stimulation was performed in FLS to investigate cytokine induction. Immunohistochemistry was applied to study IL-32 expression in synovial biopsies from RA patients. Results TNFα potently induced IL-32γ expression in FLS. Increased TNFα, IL-1β, IL-6 and CXCL8 production was observed after IL-32γ overexpression and lipopolysaccharide stimulation of THP1 cells. TNFα stimulation of FLS after silencing IL-32γ resulted in diminished IL-6 and CXCL8 production, whereas IL-32γ overexpression resulted in enhanced IL-6 and CXCL8 levels. Remarkably, the mechanism through which IL-32γ overexpression induced TNFα, IL-1β and CXCL8 was by counteracting messenger RNA decay. Importantly, treatment of RA patients with anti-TNFα resulted in significant reduction of IL-32 protein in synovial tissue. Conclusions TNFα is a potent inducer of endogenous IL-32 expression and IL-32 itself contributes to prolonged TNFα production, thus inducing an important auto-inflammatory loop. Treatment of RA patients with anti-TNFα antibodies diminished IL-32 expression in synovial tissue. The potent anti-inflammatory effect of TNFα blockade in RA patients may be partly due to the reduction of synovial IL-32 expression.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Inflammation-dependent secretion and splicing of IL-32{gamma} in rheumatoid arthritis

Bas Heinhuis; Marije I. Koenders; Fons A. J. van de Loo; Mihai G. Netea; Wim B. van den Berg; Leo A. B. Joosten

Different splice variants of the proinflammatory cytokine IL-32 are found in various tissues; their putative differences in biological function remain unknown. In the present study, we report that IL-32γ is the most active isoform of the cytokine. Splicing to one less active IL-32β appears to be a salvage mechanism to reduce inflammation. Adenoviral overexpression of IL-32γ (AdIL-32γ) resulted in exclusion of the IL-32γ–specific exon in vitro as well as in vivo, primarily leading to expression of IL-32β mRNA and protein. Splicing of the IL-32γ–specific exon was prevented by single-nucleotide mutation, which blocked recognition of the splice site by the spliceosome. Overexpression of splice-resistant IL-32γ in THP1 cells or rheumatoid arthritis (RA) synovial fibroblasts resulted in a greater induction of proinflammatory cytokines such as IL-1β, compared with IL-32β. Intraarticular introduction of IL-32γ in mice resulted in joint inflammation and induction of several mediators associated with joint destruction. In RA synovial fibroblasts, overexpression of primarily IL-32β showed minimal secretion and reduced cytokine production. In contrast, overexpression of splice-resistant IL-32γ in RA synovial fibroblasts exhibited marked secretion of IL-32γ. In RA, we observed increased IL-32γ expression compared with osteoarthritis synovial tissue. Furthermore, expression of TNFα and IL-6 correlated significantly with IL-32γ expression in RA, whereas this was not observed for IL-32β. These data reveal that naturally occurring IL-32γ can be spliced into IL-32β, which is a less potent proinflammatory mediator. Splicing of IL-32γ into IL-32β is a safety switch in controlling the effects of IL-32γ and thereby reduces chronic inflammation.


Cellular and Molecular Life Sciences | 2013

Novel insights into the biology of interleukin-32

Leo A. B. Joosten; Bas Heinhuis; Mihai G. Netea; Charles A. Dinarello

Interleukin (IL)-32 is known as a proinflammatory cytokine that is likely involved in several diseases, including infections, chronic inflammation, and cancer. Since the first report in 2005, IL-32 has been the subject of numerous studies to unravel the biological function of this molecule. For example, silencing of endogenous IL-32 in primary or cell lines of human origin consistently suppressed responses to Toll-like receptors. The protein folding structure of the six isoforms of IL-32 does not resemble that of any classical cytokine and as of this writing, a specific IL-32 receptor has not been identified. Instead, we propose a mechanism by which exposure to extracellular IL-32 or overexpression of the molecule results in binding to intracellular partners that influences functions such as gene expression, cell death, or survival. As such, this review offers insights into the role of IL-32 in several diseases, host defense, inflammation, immune function, and cancer. Finally, possibilities to target IL-32 in several diseases are proposed.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Differential function of the NACHT-LRR (NLR) members Nod1 and Nod2 in arthritis

Leo A. B. Joosten; Bas Heinhuis; Shahla Abdollahi-Roodsaz; Gerben Ferwerda; Lionel LeBourhis; Dana J. Philpott; Marie-Anne Nahori; C. Popa; Servaas A. Morré; Jos W. M. van der Meer; Stephen E. Girardin; Mihai G. Netea; Wim B. van den Berg

The pathogenesis of chronic joint inflammation remains unclear, although the involvement of pathogen recognition receptors has been suggested recently. In the present article, we describe the role of two members of the NACHT-LRR (NLR) family, Nod1 (nucleotide-binding oligomerization domain) and Nod2 in a model of acute joint inflammation induced by intraarticular injection of Streptococcus pyogenes cell wall fragments. Here, we show that Nod2 deficiency resulted in reduced joint inflammation and protection against early cartilage damage. In contrast, Nod1 gene-deficient mice developed enhanced joint inflammation with concomitant elevated levels of proinflammatory cytokines and cartilage damage, consistent with a model in which Nod1 controls the inflammatory reaction. To explore whether the different function of Nod1 and Nod2 occurs also in humans, we exposed peripheral blood mononuclear cells (PBMCs) carrying either Nod1ins/del or Nod2fs mutation with SCW fragments in vitro. Production of both TNFα and IL-1β was clearly impaired in PBMCs carrying the Nod2fs compared with PBMCs isolated from healthy controls. In line with results in Nod1 gene-deficient mice, PBMCs from individuals bearing a newly described Nod1 mutation produced enhanced levels of proinflammatory cytokines after 24-h stimulation with SCW fragments. These data indicate that the NLR family members Nod1 and Nod2 have different functions in controlling inflammation, and that intracellular Nod1–Nod2 interactions may determine the severity of arthritis in this experimental model. Whether a distorted balance between the function of Nod1 and/or Nod2 is involved in the pathogenesis of human autoinflammatory or autoimmune disease, such as rheumatoid arthritis, remains to be elucidated.


Journal of Biological Chemistry | 2012

Interleukin 32 (IL-32) Contains a Typical α-Helix Bundle Structure That Resembles Focal Adhesion Targeting Region of Focal Adhesion Kinase-1

Bas Heinhuis; Marije I. Koenders; Wim B. van den Berg; Mihai G. Netea; Charles A. Dinarello; Leo A. B. Joosten

Background: IL-32 is involved in several cell processes, most likely through integrin signaling. Results: Modeling of IL-32 revealed a structure similar to FAT and binds to integrins, paxillin, and FAK-1, all members of the integrin-signaling pathway. Conclusion: IL-32 interacts with members of the focal adhesion protein complex. Significance: IL-32 might be a key protein in integrin signaling and downstream processes. IL-32 can be expressed in several isoforms. The amino acid sequences of the major IL-32 isoforms were used to predict the secondary and tertiary protein structure by I-TASSER software. The secondary protein structure revealed coils and α-helixes, but no β sheets. Furthermore, IL-32 contains an RGD motif, which potentially activates procaspase-3 intracellular and or binds to integrins. Mutation of the RGD motif did not result in inhibition of the IL-32β- or IL-32γ-induced cytotoxicity mediated through caspase-3. Although IL-32α interacted with the extracellular part of αVβ3 and αVβ6 integrins, only the αVβ3 binding was inhibited by small RGD peptides. Additionally, IL-32β was able to bind to αVβ3 integrins, whereas this binding was not inhibited by small RGD peptides. In addition to the IL-32/integrin interactions, we observed that IL-32 is also able to interact with intracellular proteins that are involved in integrin and focal adhesion signaling. Modeling of IL-32 revealed a distinct α-helix protein resembling the focal adhesion targeting region of focal adhesion kinase (FAK). Inhibition of FAK resulted in modulation of the IL-32β- or IL-32γ-induced cytotoxicity. Interestingly, IL-32α binds to paxillin without the RGD motif being involved. Finally, FAK inhibited IL-32α/paxillin binding, whereas FAK also could interact with IL-32α, demonstrating that IL-32 is a member of the focal adhesion protein complex. This study demonstrates for the first time that IL-32 binds to the extracellular domain of integrins and to intracellular proteins like paxillin and FAK, suggesting a dual role for IL-32 in integrin signaling.


Journal of Immunology | 2014

IL-32 Promotes Angiogenesis

Claudia A. Nold-Petry; Ina Rudloff; Yvonne Baumer; Menotti Ruvo; Daniela Marasco; Paolo Botti; Laszlo Farkas; Steven X. Cho; Jarod A. Zepp; Tania Azam; Hannah Dinkel; Brent E. Palmer; William A. Boisvert; Carlyne D. Cool; Laima Taraseviciene-Stewart; Bas Heinhuis; Leo A. B. Joosten; Charles A. Dinarello; Norbert F. Voelkel; Marcel F. Nold

IL-32 is a multifaceted cytokine with a role in infections, autoimmune diseases, and cancer, and it exerts diverse functions, including aggravation of inflammation and inhibition of virus propagation. We previously identified IL-32 as a critical regulator of endothelial cell (EC) functions, and we now reveal that IL-32 also possesses angiogenic properties. The hyperproliferative ECs of human pulmonary arterial hypertension and glioblastoma multiforme exhibited a markedly increased abundance of IL-32, and, significantly, the cytokine colocalized with integrin αVβ3. Vascular endothelial growth factor (VEGF) receptor blockade, which resulted in EC hyperproliferation, increased IL-32 three-fold. Small interfering RNA–mediated silencing of IL-32 negated the 58% proliferation of ECs that occurred within 24 h in scrambled-transfected controls. Reduction of IL-32 neither affected apoptosis (insignificant changes in Bak-1, Bcl-2, Bcl-xL, lactate dehydrogenase, annexin V, and propidium iodide) nor VEGF or TGF-β levels, but siIL-32–transfected adult and neonatal ECs produced up to 61% less NO, IL-8, and matrix metalloproteinase-9, and up to 3-fold more activin A and endostatin. In coculture-based angiogenesis assays, IL-32γ dose-dependently increased tube formation up to 3-fold; an αVβ3 inhibitor prevented this activity and reduced IL-32γ–induced IL-8 by 85%. In matrigel plugs loaded with IL-32γ, VEGF, or vehicle and injected into live mice, we observed the anticipated VEGF-induced increase in neocapillarization (8-fold versus vehicle), but unexpectedly, IL-32γ was equally angiogenic. A second signal such as IFN-γ was required to render cells responsive to exogenous IL-32γ; importantly, this was confirmed using a completely synthetic preparation of IL-32γ. In summary, we add angiogenic properties that are mediated by integrin αVβ3 but VEGF-independent to the portfolio of IL-32, implicating a role for this versatile cytokine in pulmonary arterial hypertension and neoplastic diseases.


Cytokine | 2012

Interleukin-32: A predominantly intracellular proinflammatory mediator that controls cell activation and cell death

Bas Heinhuis; Mihai G. Netea; Wim B. van den Berg; Charles A. Dinarello; Leo A. B. Joosten

In this review, we will discuss the current knowledge on IL-32 and provide new insights regarding the biological function of IL-32. IL-32 is seen as a cytokine that can induce a range of proinflammatory mediators and contribute to autoimmune diseases, such as rheumatoid arthritis, however present knowledge demonstrates that IL-32 is not a classical cytokine. We present the history of this cytokine, the role of IL-32 in several diseases and discuss a possible novel role of intracellular IL-32 in cell homeostasis. Taken into account the observed biological functions of IL-32, it may belong to a class of cytokines, like IL-1α, IL-33, and IL-37, with both intracellular and extracellular functions.


Annals of the Rheumatic Diseases | 2010

IL-32gamma and Streptococcus pyogenes cell wall fragments synergise for IL-1-dependent destructive arthritis via upregulation of TLR-2 and NOD2.

Bas Heinhuis; M.M.J.F. Koenders; F.A.J. van de Loo; P.L.E.M. van Lent; Soo Hyun Kim; Charles A. Dinarello; Leo A. B. Joosten; W.B. van den Berg

Objective To investigate the potential synergism between interleukin (IL) 32γ and Streptococcus pyogenes cell wall (SCW) fragments in the development of destructive arthritis. Methods An adenoviral vector encoding human IL-32γ (AdIL-32γ) was constructed and validated in HeLa cells. Fibroblast-like synoviocytes (FLS) were transduced with AdIL-32γ and stimulated with Toll-like receptor 2 (TLR-2) and nucleotide oligomerisation domain (NOD) 2 ligands. Expression levels of several proinflammatory cytokines, chemokines, matrix degrading enzymes, TLR-2 and NOD2 were measured by quantitative real-time PCR. Furthermore, IL-6 and CXCL8 protein levels were determined. In-vivo synergy between IL-32γ and SCW was studied by intra-articular injection of AdIL-32γ in C57Bl/6 mice followed by SCW injection. The contribution of endogenous IL-1 was assessed in mice deficient for both IL-1α and IL-1β. Results IL-32γ synergise with TLR-2/NOD2 ligands to induce proinflammatory cytokines, chemokines and matrix degrading enzymes in AdIL-32γ-transduced FLS. In mice, AdIL-32γ transduction followed by the injection of SCW displayed aggravated joint inflammation and cartilage destruction. However, IL-1-deficient mice were protected against IL-32γ/SCW-induced joint changes, indicating a requirement for IL-1 in downstream events triggered by IL-32γ plus SCW. To elucidate the synergistic mechanism, the authors investigated the expression of two pattern recognition receptors involved in sensing SCW fragments. TLR-2 and NOD2 receptor expression was enhanced by IL-32γ and Pam3Cys/muramyl dipeptide stimulation in FLS. Conclusions Here the authors show that IL-32γ aggravates SCW-induced arthritis by the upregulation of TLR-2/NOD2 expression and promotes severe joint erosion in an IL-1-dependent fashion. Targeting of IL-32γ may provide a novel therapy to prevent destructive arthritis.

Collaboration


Dive into the Bas Heinhuis's collaboration.

Top Co-Authors

Avatar

Leo A. B. Joosten

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Mihai G. Netea

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Charles A. Dinarello

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Theo S. Plantinga

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

C. Popa

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J.C. Dos Santos

Universidade Federal de Goiás

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerben Ferwerda

Radboud University Nijmegen Medical Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge