Network


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

Hotspot


Dive into the research topics where Sanne P. Smeekens is active.

Publication


Featured researches published by Sanne P. Smeekens.


The New England Journal of Medicine | 2011

STAT1 Mutations in Autosomal Dominant Chronic Mucocutaneous Candidiasis

Frank L. van de Veerdonk; Theo S. Plantinga; Alexander Hoischen; Sanne P. Smeekens; Leo A. B. Joosten; Christian Gilissen; Peer Arts; Diana C. Rosentul; Andrew J. Carmichael; Chantal A.A. Smits-van der Graaf; Bart Jan Kullberg; Jos W. M. van der Meer; Desa Lilic; Joris A. Veltman; Mihai G. Netea

BACKGROUND Chronic mucocutaneous candidiasis (CMC) is characterized by susceptibility to candida infection of skin, nails, and mucous membranes. Patients with recessive CMC and autoimmunity have mutations in the autoimmune regulator AIRE. The cause of autosomal dominant CMC is unknown. METHODS We evaluated 14 patients from five families with autosomal dominant CMC. We incubated their peripheral-blood mononuclear cells with different combinations of stimuli to test the integrity of pathways that mediate immunity, which led to the selection of 100 genes that were most likely to contain the genetic defect. We used an array-based sequence-capture assay, followed by next-generation sequencing, to identify mutations. RESULTS The mononuclear cells from the affected patients were characterized by poor production of interferon-γ, interleukin-17, and interleukin-22, suggesting that the defect lay within the interleukin-12 receptor and interleukin-23 receptor signaling pathways. We identified heterozygous missense mutations in the DNA sequence encoding the coiled-coil (CC) domain of signal transducer and activator of transcription 1 (STAT1) in the patients. These mutations lead to defective responses in type 1 and type 17 helper T cells (Th1 and Th17). The interferon-γ receptor pathway was intact in these patients. CONCLUSIONS Mutations in the CC domain of STAT1 underlie autosomal dominant CMC and lead to defective Th1 and Th17 responses, which may explain the increased susceptibility to fungal infection. (Funded by the Netherlands Organization for Scientific Research and others.).


Proceedings of the National Academy of Sciences of the United States of America | 2010

Reactive oxygen species–independent activation of the IL-1β inflammasome in cells from patients with chronic granulomatous disease

Frank L. van de Veerdonk; Sanne P. Smeekens; Leo A. B. Joosten; Bart Jan Kullberg; Charles A. Dinarello; Jos W. M. van der Meer; Mihai G. Netea

Humans with chronic granulomatous diseases (CGDs) due to mutations in p47-phox have defective NADPH activity and thus cannot generate NADPH-dependent reactive oxygen species (ROS). The role of ROS in inflammation is controversial; some in vitro studies suggest that ROS are crucial for secretion of IL-1β via inflammasome activation, whereas mice defective for ROS and patients with CGD have a proinflammatory phenotype. In this study, we evaluated activation of the IL-1β inflammasome in cells from CGD patients. In contrast to previous studies using the small molecule diphenylene iodonium (DPI) as a ROS inhibitor, we found no decrease in either caspase-1 activation or secretion of IL-1β and IL-18 in primary CGD monocytes. Moreover, activation of CGD monocytes by uric acid crystals induced a 4-fold higher level of IL-1β secretion compared with that seen in monocytes from unaffected subjects, and this increase was not due to increased synthesis of the IL-1β precursor. In addition, Western blot analysis of CGD cells revealed that caspase-1 activation was not decreased, but rather was increased compared with control cells. Examination of the effects exerted by the inhibition of ROS activity by DPI revealed that the decrease in IL-1β secretion by DPI was actually due to inhibition of IL-1β gene expression. Thus, inconsistent with the proinflammatory role of ROS, the present findings support the concept that ROS likely dampen inflammasome activation. The absence of ROS in CGD monocytes may explain the presence of an inflammatory phenotype characterized by granulomas and inflammatory bowel disease occurring in CGD patients.


Proceedings of the National Academy of Sciences of the United States of America | 2014

IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans

Antonella De Luca; Sanne P. Smeekens; Andrea Casagrande; Rossana G. Iannitti; Kara L. Conway; Mark S. Gresnigt; Jakob Begun; Theo S. Plantinga; Leo A. B. Joosten; Jos W. M. van der Meer; Georgios Chamilos; Mihai G. Netea; Ramnik J. Xavier; Charles A. Dinarello; Luigina Romani; Frank L. van de Veerdonk

Significance Chronic granulomatous disease (CGD) has an immunodeficiency component and, in addition, an autoinflammatory component in which autophagy and inflammasome activation are linked and amenable to IL-1 blockade. This study provides a rationale to perform clinical trials to investigate the efficacy of blocking IL-1 in CGD colitis and expands the therapeutic potential of IL-1 antagonists to inflammatory diseases with defective autophagy. Patients with chronic granulomatous disease (CGD) have a mutated NADPH complex resulting in defective production of reactive oxygen species; these patients can develop severe colitis and are highly susceptible to invasive fungal infection. In NADPH oxidase-deficient mice, autophagy is defective but inflammasome activation is present despite lack of reactive oxygen species production. However, whether these processes are mutually regulated in CGD and whether defective autophagy is clinically relevant in patients with CGD is unknown. Here, we demonstrate that macrophages from CGD mice and blood monocytes from CGD patients display minimal recruitment of microtubule-associated protein 1 light chain 3 (LC3) to phagosomes. This defect in autophagy results in increased IL-1β release. Blocking IL-1 with the receptor antagonist (anakinra) decreases neutrophil recruitment and T helper 17 responses and protects CGD mice from colitis and also from invasive aspergillosis. In addition to decreased inflammasome activation, anakinra restored autophagy in CGD mice in vivo, with increased Aspergillus-induced LC3 recruitment and increased expression of autophagy genes. Anakinra also increased Aspergillus-induced LC3 recruitment from 23% to 51% (P < 0.01) in vitro in monocytes from CGD patients. The clinical relevance of these findings was assessed by treating CGD patients who had severe colitis with IL-1 receptor blockade using anakinra. Anakinra treatment resulted in a rapid and sustained improvement in colitis. Thus, inflammation in CGD is due to IL-1–dependent mechanisms, such as decreased autophagy and increased inflammasome activation, which are linked pathological conditions in CGD that can be restored by IL-1 receptor blockade.


Journal of Leukocyte Biology | 2011

The dectin-1/inflammasome pathway is responsible for the induction of protective T-helper 17 responses that discriminate between yeasts and hyphae of Candida albicans

Shih-Chin Cheng; Frank L. van de Veerdonk; Megan D. Lenardon; Monique Stoffels; Theo S. Plantinga; Sanne P. Smeekens; Lisa Rizzetto; Liliane Mukaremera; Kanya Preechasuth; Duccio Cavalieri; Thirumala-Devi Kanneganti; Jos W. M. van der Meer; Bart Jan Kullberg; Leo A. B. Joosten; Neil A. R. Gow; Mihai G. Netea

In the mucosa, the immune pathways discriminating between colonizing and invasive Candida, thus inducing tolerance or inflammation, are poorly understood. Th17 responses induced by Candida albicans hyphae are central for the activation of mucosal antifungal immunity. An essential step for the discrimination between yeasts and hyphae and induction of Th17 responses is the activation of the inflammasome by C. albicans hyphae and the subsequent release of active IL‐1β in macrophages. Inflammasome activation in macrophages results from differences in cell‐wall architecture between yeasts and hyphae and is partly mediated by the dectin‐1/Syk pathway. These results define the dectin‐1/inflammasome pathway as the mechanism that enables the host immune system to mount a protective Th17 response and distinguish between colonization and tissue invasion by C. albicans.


Nature Genetics | 2016

The effect of host genetics on the gut microbiome

Marc Jan Bonder; Alexander Kurilshikov; Ettje F. Tigchelaar; Zlatan Mujagic; Floris Imhann; Arnau Vich Vila; Patrick Deelen; Tommi Vatanen; Melanie Schirmer; Sanne P. Smeekens; Daria V. Zhernakova; Soesma A. Jankipersadsing; Martin Jaeger; Marije Oosting; Maria Carmen Cenit; Ad Masclee; Morris A. Swertz; Yang Li; Vinod Kumar; Leo A. B. Joosten; Hermie J. M. Harmsen; Rinse K. Weersma; Lude Franke; Marten H. Hofker; Ramnik J. Xavier; Daisy Jonkers; Mihai G. Netea; Cisca Wijmenga; Jingyuan Fu; Alexandra Zhernakova

The gut microbiome is affected by multiple factors, including genetics. In this study, we assessed the influence of host genetics on microbial species, pathways and gene ontology categories, on the basis of metagenomic sequencing in 1,514 subjects. In a genome-wide analysis, we identified associations of 9 loci with microbial taxonomies and 33 loci with microbial pathways and gene ontology terms at P < 5 × 10−8. Additionally, in a targeted analysis of regions involved in complex diseases, innate and adaptive immunity, or food preferences, 32 loci were identified at the suggestive level of P < 5 × 10−6. Most of our reported associations are new, including genome-wide significance for the C-type lectin molecules CLEC4F–CD207 at 2p13.3 and CLEC4A–FAM90A1 at 12p13. We also identified association of a functional LCT SNP with the Bifidobacterium genus (P = 3.45 × 10−8) and provide evidence of a gene–diet interaction in the regulation of Bifidobacterium abundance. Our results demonstrate the importance of understanding host–microbe interactions to gain better insight into human health.


Cell | 2016

Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity

Melanie Schirmer; Sanne P. Smeekens; Hera Vlamakis; Martin Jaeger; Marije Oosting; Eric A. Franzosa; Rob ter Horst; Trees Jansen; Liesbeth Jacobs; Marc Jan Bonder; Alexander Kurilshikov; Jingyuan Fu; Leo A. B. Joosten; Alexandra Zhernakova; Curtis Huttenhower; Cisca Wijmenga; Mihai G. Netea; Ramnik J. Xavier

Gut microbial dysbioses are linked to aberrant immune responses, which are often accompanied by abnormal production of inflammatory cytokines. As part of the Human Functional Genomics Project (HFGP), we investigate how differences in composition and function of gut microbial communities may contribute to inter-individual variation in cytokine responses to microbial stimulations in healthy humans. We observe microbiome-cytokine interaction patterns that are stimulus specific, cytokine specific, and cytokine and stimulus specific. Validation of two predicted host-microbial interactions reveal that TNFα and IFNγ production are associated with specific microbial metabolic pathways: palmitoleic acid metabolism and tryptophan degradation to tryptophol. Besides providing a resource of predicted microbially derived mediators that influence immune phenotypes in response to common microorganisms, these data can help to define principles for understanding disease susceptibility. The three HFGP studies presented in this issue lay the groundwork for further studies aimed at understanding the interplay between microbial, genetic, and environmental factors in the regulation of the immune response in humans. PAPERCLIP.


European Journal of Immunology | 2011

The inflammasome drives protective Th1 and Th17 cellular responses in disseminated candidiasis

Frank L. van de Veerdonk; Leo A. B. Joosten; Patrick J. Shaw; Sanne P. Smeekens; R. K. Subbarao Malireddi; Jos W. M. van der Meer; Bart Jan Kullberg; Mihai G. Netea; Thirumala-Devi Kanneganti

The Nlrp3 inflammasome has been proposed to play an important role in antifungal host defense. However, studies exploring the role of the inflammasome in antifungal host defense have been limited to the direct effects on IL‐1β processing. Although IL‐1β has important direct effects on the innate immune response, important effects of the caspase‐1‐dependent cytokines IL‐1β and IL‐18 are exerted on the initiation of the adaptive Th1 and Th17 cellular responses. No studies have been employed to assess the impact of the inflammasome on the Th1/Th17 defense mechanisms in vivo during candidiasis. In the present study, we demonstrate an essential role for caspase‐1 and ASC (apoptosis‐associated speck‐like protein containing a caspase recruitment domain) in disseminated candidiasis through regulating antifungal Th1 and Th17 responses. Caspase‐1−/− and ASC−/− mice display diminished Th1/Th17 responses, followed by increased fungal outgrowth and lower survival. These observations identify a critical role for the inflammasome in controlling protective adaptive immune responses during invasive fungal infection.


Nature Communications | 2013

Functional genomics identifies type I interferon pathway as central for host defense against Candida albicans

Sanne P. Smeekens; Aylwin Ng; Vinod Kumar; Melissa D. Johnson; Theo S. Plantinga; Cleo C. van Diemen; Peer Arts; Eugène T P Verwiel; Mark S. Gresnigt; Karin Fransen; Suzanne van Sommeren; Marije Oosting; Shih-Chin Cheng; Leo A. B. Joosten; Alexander Hoischen; Bart Jan Kullberg; William K. Scott; John R. Perfect; Jos W. M. van der Meer; Cisca Wijmenga; Mihai G. Netea; Ramnik J. Xavier

Candida albicans is the most common human fungal pathogen causing mucosal and systemic infections. However, human antifungal immunity remains poorly defined. Here, by integrating transcriptional analysis and functional genomics, we identified Candida-specific host defense mechanisms in humans. Candida induced significant expression of genes from the type I interferon (IFN) pathway in human peripheral blood mononuclear cells. This unexpectedly prominent role of type I IFN pathway in anti-Candida host defense was supported by additional evidence. Polymorphisms in type I IFN genes modulated Candida-induced cytokine production and were correlated with susceptibility to systemic candidiasis. In in-vitro experiments, type I IFNs skewed Candida-induced inflammation from a Th17-response toward a Th1-response. Patients with chronic mucocutaneaous candidiasis displayed defective expression of genes in the type I IFN pathway. These findings indicate that the type I IFN pathway is a main signature of Candida-induced inflammation and plays a crucial role in anti-Candida host defense in humans.


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.


Embo Molecular Medicine | 2013

Genetic susceptibility to Candida infections

Sanne P. Smeekens; Frank L. van de Veerdonk; Bart Jan Kullberg; Mihai G. Netea

Candida spp. are medically important fungi causing severe mucosal and life‐threatening invasive infections, especially in immunocompromised hosts. However, not all individuals at risk develop Candida infections, and it is believed that genetic variation plays an important role in host susceptibility. On the one hand, severe fungal infections are associated with monogenic primary immunodeficiencies such as defects in STAT1, STAT3 or CARD9, recently discovered as novel clinical entities. On the other hand, more common polymorphisms in genes of the immune system have also been associated with fungal infections such as recurrent vulvovaginal candidiasis and candidemia. The discovery of the genetic susceptibility to Candida infections can lead to a better understanding of the pathogenesis of the disease, as well as to the design of novel immunotherapeutic strategies. This review is part of the review series on host‐pathogen interactions. See more reviews from this series.

Collaboration


Dive into the Sanne P. Smeekens's collaboration.

Top Co-Authors

Avatar

Mihai G. Netea

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Leo A. B. Joosten

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bart Jan Kullberg

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Marije Oosting

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cisca Wijmenga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Martin Jaeger

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Vinod Kumar

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge