Network


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

Hotspot


Dive into the research topics where Roel P. Gazendam is active.

Publication


Featured researches published by Roel P. Gazendam.


Blood | 2013

Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency

Agata Drewniak; Roel P. Gazendam; Anton Tool; Michel van Houdt; Machiel H. Jansen; John L. van Hamme; Ester M. M. van Leeuwen; Dirk Roos; Emmanuel Scalais; Carine De Beaufort; Hans Janssen; Timo K. van den Berg; Taco W. Kuijpers

Caspase recruitment domain-containing protein 9 (CARD9) is an adaptor molecule in the cytosol of myeloid cells, required for induction of T-helper cells producing interleukin-17 (Th17 cells) and important in antifungal immunity. In a patient suffering from Candida dubliniensis meningoencephalitis, mutations in the CARD9 gene were found to result in the loss of protein expression. Apart from the reduced numbers of CD4(+) Th17 lymphocytes, we identified a lack of monocyte-derived cytokines in response to Candida strains. Importantly, CARD9-deficient neutrophils showed a selective Candida albicans killing defect with abnormal ultrastructural phagolysosomes and outgrowth of hyphae. The neutrophil killing defect was independent of the generation of reactive oxygen species by the reduced NAD phosphate oxidase system. Taken together, this demonstrates that human CARD9 deficiency results in selective defect in the host defense against invasive fungal infection, caused by an impaired phagocyte killing.


Blood | 2014

Two independent killing mechanisms of Candida albicans by human neutrophils: evidence from innate immunity defects

Roel P. Gazendam; J. L. van Hamme; Anton Tool; M. van Houdt; Paul Verkuijlen; M. Herbst; J. G. Liese; F.L. van de Veerdonk; Dirk Roos; T. K. van den Berg; Taco W. Kuijpers

Invasive fungal infections, accompanied by high rates of mortality, represent an increasing problem in medicine. Neutrophils are the major effector immune cells in fungal killing. Based on studies with neutrophils from patients with defined genetic defects, we provide evidence that human neutrophils use 2 distinct and independent phagolysosomal mechanisms to kill Candida albicans. The first mechanism for the killing of unopsonized C albicans was found to be dependent on complement receptor 3 (CR3) and the signaling proteins phosphatidylinositol-3-kinase and caspase recruitment domain-containing protein 9 (CARD9), but was independent of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. The second mechanism for the killing of opsonized C albicans was strictly dependent on Fcγ receptors, protein kinase C (PKC), and reactive oxygen species production by the NADPH oxidase system. Each of the 2 pathways of Candida killing required Syk tyrosine kinase activity, but dectin-1 was dispensable for both of them. These data provide an explanation for the variable clinical presentation of fungal infection in patients suffering from different immune defects, including dectin-1 deficiency, CARD9 deficiency, or chronic granulomatous disease.


JCI insight | 2016

Extrapulmonary Aspergillus infection in patients with CARD9 deficiency

Nikolaus Rieber; Roel P. Gazendam; Alexandra F. Freeman; Amy P. Hsu; Amanda L. Collar; Janyce A. Sugui; Rebecca A. Drummond; Chokechai Rongkavilit; Kevin W. Hoffman; Carolyn Henderson; Lily P. Clark; Markus Mezger; Muthulekha Swamydas; Maik Engeholm; Rebecca Schüle; Bettina Neumayer; Frank Ebel; Constantinos M. Mikelis; Stefania Pittaluga; Vinod K. Prasad; Anurag Singh; Joshua D. Milner; Kelli W. Williams; Jean K. Lim; Kyung J. Kwon-Chung; Steven M. Holland; Dominik Hartl; Taco W. Kuijpers; Michail S. Lionakis

Invasive pulmonary aspergillosis is a life-threatening mycosis that only affects patients with immunosuppression, chemotherapy-induced neutropenia, transplantation, or congenital immunodeficiency. We studied the clinical, genetic, histological, and immunological features of 2 unrelated patients without known immunodeficiency who developed extrapulmonary invasive aspergillosis at the ages of 8 and 18. One patient died at age 12 with progressive intra-abdominal aspergillosis. The other patient had presented with intra-abdominal candidiasis at age 9, and developed central nervous system aspergillosis at age 18 and intra-abdominal aspergillosis at age 25. Neither patient developed Aspergillus infection of the lungs. One patient had homozygous M1I CARD9 (caspase recruitment domain family member 9) mutation, while the other had homozygous Q295X CARD9 mutation; both patients lacked CARD9 protein expression. The patients had normal monocyte and Th17 cell numbers in peripheral blood, but their mononuclear cells exhibited impaired production of proinflammatory cytokines upon fungus-specific stimulation. Neutrophil phagocytosis, killing, and oxidative burst against Aspergillus fumigatus were intact, but neither patient accumulated neutrophils in infected tissue despite normal neutrophil numbers in peripheral blood. The neutrophil tissue accumulation defect was not caused by defective neutrophil-intrinsic chemotaxis, indicating that production of neutrophil chemoattractants in extrapulmonary tissue is impaired in CARD9 deficiency. Taken together, our results show that CARD9 deficiency is the first known inherited or acquired condition that predisposes to extrapulmonary Aspergillus infection with sparing of the lungs, associated with impaired neutrophil recruitment to the site of infection.


Pediatric Infectious Disease Journal | 2015

Chronic Candida albicans Meningitis in a 4-Year-Old Girl with a Homozygous Mutation in the CARD9 Gene (Q295X)

Martin Herbst; Roel P. Gazendam; Denise Reimnitz; Julie Sawalle-Belohradsky; Andreas H. Groll; Paul-Gerhardt Schlegel; Bernd H. Belohradsky; Ellen D. Renner; Jörg Klepper; Bodo Grimbacher; Taco W. Kuijpers; Johannes G. Liese

A 4-year-old Turkish girl of consanguineous parents was hospitalized for the evaluation of headaches and recurrent febrile episodes of unknown origin. Her medical history was unremarkable except for a few episodes of uncomplicated oral thrush. Meningitis was diagnosed, and Candida albicans was the only pathogen identified by polymerase chain reaction and culture. Despite systemic antifungal multidrug therapy, a prolonged course of 16 months of therapy was necessary to clear C. albicans from the cerebrospinal fluid. Molecular genetic analysis revealed a homozygous caspase recruitment domain 9 (CARD9) mutation (Q295X), which was reported to predispose to chronic mucocutaneous candidiasis. Immunologic workup excluded predisposing B-cell and T-cell defects. In addition, T cells producing interleukin-17 were repeatedly measured within the normal range. Analyses of neutrophils demonstrated normal nicotinamide adenine dinucleotide phosphate oxidase activity in response to various stimuli including Staphylococcus aureus and C. albicans. Additional neutrophilic functional testing, however, showed a decreased cytotoxicity to nonopsonized C. albicans, indicating an impaired killing mechanism against Candida spp. independent from the production of reactive oxygen species by the nicotinamide adenine dinucleotide phosphate oxidase system. Because this defect was only demonstrated in the absence of opsonins, it might especially predispose to chronic C. albicans infections in the central nervous system where opsonin concentrations are usually low. We, therefore, suggest that due to an additional neutrophil dependent defect CARD9 deficiency predisposes not only to chronic mucocutaneous candidiasis, but also to invasive chronic Candida infections, especially of the central nervous system.


Blood | 2013

Defects in neutrophil granule mobilization and bactericidal activity in familial hemophagocytic lymphohistiocytosis type 5 (FHL-5) syndrome caused by STXBP2/Munc18-2 mutations

Xi Wen Zhao; Roel P. Gazendam; Agata Drewniak; Michel van Houdt; Anton Tool; John L. van Hamme; Iwan Kustiawan; Alexander B. Meijer; Hans Janssen; David G. Russell; Lisette van de Corput; Kiki Tesselaar; Jaap Jan Boelens; Ingrid Kuhnle; Jutte van der Werff ten Bosch; Taco W. Kuijpers; Timo K. van den Berg

Familial hemophagocytic lymphohistiocytosis (FHL) is caused by genetic defects in cytotoxic granule components or their fusion machinery, leading to impaired natural killer cell and/or T lymphocyte degranulation and/or cytotoxicity. This may accumulate into a life-threatening condition known as macrophage activation syndrome. STXBP2, also known as MUNC18-2, has recently been identified as the disease-causing gene in FHL type 5 (FHL-5). A role for STXBP2 in neutrophils, and for neutrophils in FHL in general, has not been documented thus far. Here, we report that FHL-5 neutrophils have a profound defect in granule mobilization, resulting in inadequate bacterial killing, in particular, of gram-negative Escherichia coli, but not of Staphylococcus aureus, which rather depends on intact reduced NAD phosphate oxidase activity. This impairment of bacterial killing may contribute to the apparent susceptibility to gastrointestinal tract inflammation in patients with FHL-5.


Immunological Reviews | 2016

How neutrophils kill fungi.

Roel P. Gazendam; Annemarie van de Geer; Dirk Roos; Timo K. van den Berg; Taco W. Kuijpers

Neutrophils play a critical role in the prevention of invasive fungal infections. Whereas mouse studies have demonstrated the role of various neutrophil pathogen recognition receptors (PRRs), signal transduction pathways, and cytotoxicity in the murine antifungal immune response, much less is known about the killing of fungi by human neutrophils. Recently, novel primary immunodeficiencies have been identified in patients with a susceptibility to fungal infections. These human ‘knock‐out’ neutrophils expand our knowledge to understand the role of PRRs and signaling in human fungal killing. From the studies with these patients it is becoming clear that neutrophils employ fundamentally distinct mechanisms to kill Candida albicans or Aspergillus fumigatus.


Haematologica | 2016

Impaired killing of Candida albicans by granulocytes mobilized for transfusion purposes: a role for granule components

Roel P. Gazendam; Annemarie van de Geer; John L. van Hamme; Anton Tool; Dieke J. van Rees; Cathelijn Aarts; Maartje van den Biggelaar; Floris van Alphen; Paul Verkuijlen; Alexander B. Meijer; Hans Janssen; Dirk Roos; Timo K. van den Berg; Taco W. Kuijpers

Granulocyte transfusions are used to treat neutropenic patients with life-threatening bacterial or fungal infections that do not respond to anti-microbial drugs. Donor neutrophils that have been mobilized with granulocyte-colony stimulating factor (G-CSF) and dexamethasone are functional in terms of antibacterial activity, but less is known about their fungal killing capacity. We investigated the neutrophil-mediated cytotoxic response against C. albicans and A. fumigatus in detail. Whereas G-CSF/dexamethasone-mobilized neutrophils appeared less mature as compared to neutrophils from untreated controls, these cells exhibited normal ROS production by the NADPH oxidase system and an unaltered granule mobilization capacity upon stimulation. G-CSF/dexamethasone-mobilized neutrophils efficiently inhibited A. fumigatus germination and killed Aspergillus and Candida hyphae, but the killing of C. albicans yeasts was distinctly impaired. Following normal Candida phagocytosis, analysis by mass spectrometry of purified phagosomes after fusion with granules demonstrated that major constituents of the antimicrobial granule components, including major basic protein (MBP), were reduced. Purified MBP showed candidacidal activity, and neutrophil-like Crisp-Cas9 NB4-KO-MBP differentiated into phagocytes were impaired in Candida killing. Together, these findings indicate that G-CSF/dexamethasone-mobilized neutrophils for transfusion purposes have a selectively impaired capacity to kill Candida yeasts, as a consequence of an altered neutrophil granular content.


Mbio | 2016

Aspergillus Cell Wall Chitin Induces Anti- and Proinflammatory Cytokines in Human PBMCs via the Fc-γ Receptor/Syk/PI3K Pathway

Katharina L. Becker; Vishukumar Aimanianda; X. Wang; Mark S. Gresnigt; Anne Ammerdorffer; Cor W. M. Jacobs; Roel P. Gazendam; Leo A. B. Joosten; Mihai G. Netea; Jean Paul Latgé; F.L. van de Veerdonk

ABSTRACT Chitin is an important cell wall component of Aspergillus fumigatus conidia, of which hundreds are inhaled on a daily basis. Previous studies have shown that chitin has both anti- and proinflammatory properties; however the exact mechanisms determining the inflammatory signature of chitin are poorly understood, especially in human immune cells. Human peripheral blood mononuclear cells were isolated from healthy volunteers and stimulated with chitin from Aspergillus fumigatus. Transcription and production of the proinflammatory cytokine interleukin-1β (IL-1β) and the anti-inflammatory cytokine IL-1 receptor antagonist (IL-1Ra) were measured from the cell culture supernatant by quantitative PCR (qPCR) or enzyme-linked immunosorbent assay (ELISA), respectively. Chitin induced an anti-inflammatory signature characterized by the production of IL-1Ra in the presence of human serum, which was abrogated in immunoglobulin-depleted serum. Fc-γ-receptor-dependent recognition and phagocytosis of IgG-opsonized chitin was identified as a novel IL-1Ra-inducing mechanism by chitin. IL-1Ra production induced by chitin was dependent on Syk kinase and phosphatidylinositol 3-kinase (PI3K) activation. In contrast, costimulation of chitin with the pattern recognition receptor (PRR) ligands lipopolysaccharide, Pam3Cys, or muramyl dipeptide, but not β-glucan, had synergistic effects on the induction of proinflammatory cytokines by human peripheral blood mononuclear cells (PBMCs). In conclusion, chitin can have both pro- and anti-inflammatory properties, depending on the presence of pathogen-associated molecular patterns and immunoglobulins, thus explaining the various inflammatory signatures reported for chitin. IMPORTANCE Invasive aspergillosis and allergic aspergillosis are increasing health care problems. Patients get infected by inhalation of the airborne spores of Aspergillus fumigatus. A profound knowledge of how Aspergillus and its cell wall components are recognized by the host cell and which type of immune response it induces is necessary to develop target-specific treatment options with less severe side effects than the treatment options to date. There is controversy in the literature about the receptor for chitin in human cells. We identified the Fc-γ receptor and Syk/PI3K pathway via which chitin can induce anti-inflammatory immune responses by inducing IL-1 receptor antagonist in the presence of human immunoglobulins but also proinflammatory responses in the presence of bacterial components. This explains why Aspergillus does not induce strong inflammation just by inhalation and rather fulfills an immune-dampening function. While in a lung coinfected with bacteria, Aspergillus augments immune responses by shifting toward a proinflammatory reaction. Invasive aspergillosis and allergic aspergillosis are increasing health care problems. Patients get infected by inhalation of the airborne spores of Aspergillus fumigatus. A profound knowledge of how Aspergillus and its cell wall components are recognized by the host cell and which type of immune response it induces is necessary to develop target-specific treatment options with less severe side effects than the treatment options to date. There is controversy in the literature about the receptor for chitin in human cells. We identified the Fc-γ receptor and Syk/PI3K pathway via which chitin can induce anti-inflammatory immune responses by inducing IL-1 receptor antagonist in the presence of human immunoglobulins but also proinflammatory responses in the presence of bacterial components. This explains why Aspergillus does not induce strong inflammation just by inhalation and rather fulfills an immune-dampening function. While in a lung coinfected with bacteria, Aspergillus augments immune responses by shifting toward a proinflammatory reaction.


The Journal of Allergy and Clinical Immunology | 2015

Impaired microbial killing by neutrophils from patients with protein kinase C delta deficiency

Katka Szilagyi; Roel P. Gazendam; John L. van Hamme; Anton Tool; Michel van Houdt; Wilhelm A.J.W. Vos; Paul Verkuijlen; Hans Janssen; Alexandre Belot; Laurent Juillard; Elisabeth Förster-Waldl; Kaan Boztug; Georg Kraal; Menno P.J. de Winther; Taco W. Kuijpers; Timo K. van den Berg

Health, University of Genoa, Genoa, Italy; the Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Md; ‘‘Angelo Nocivelli’’ Institute for Molecular Medicine, University of Brescia, Brescia, Italy; the Pediatric Hematology Oncology Unit, Spedali Civili, Brescia, Italy; the Division of Allergy and Clinical Immunology, Rebagliati Martins National Hospital, Lima, Peru; the Division of Pediatric Hematology, Children’s Hospital Orange County, University of California at Irvine, Irvine, Calif; the Department of Immunology, ‘‘Aghia Sophia’’ Children’s Hospital, Athens, Greece; the Division of Pediatric Immunology, Hospital Luis CalvoMackenna, Santiago, Chile; the Clinic of Pediatric Hematology-Oncology, Department for Woman and Child Health, University Hospital, Padua, Italy; and the Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon. E-mail: luigi. [email protected]. *These authors contributed equally to this work. Supported by a grant from the National Heart, Lung, and Blood Institute/National Institutes of Health (grant 5P01HL059561-13 to L.D.N.); an educational grant (5T32AI007512) from the National Institute of Allergy and Infectious Diseases (to E.C. [Dr Raif S. Geha, principal investigator]); an educational grant from the National Heart, Lung and Blood Institute/National Institutes of Health (grant 5T32HL00757433 to J.C.); and a grant from the UNIL-CHUV (CGRB 29583 to F.C.). Disclosure of potential conflict of interest: F. Candotti has received a grant from University of Lausanne-Centre hospitalier universitaire vaudois and is employed by Centre hospitalier universitaire vaudois. J. Chu has received a grant from the National Institutes of Health (NIH). J. Chou is employed by Boston Children’s Hospital and has received grants from the NIH and the JeffreyModell Foundation. F. Porta has received payment for lectures from Pfizer. S.-Y. Pai has received a grant from Translational Investigator Service, is employed by Boston Children’s Hospital, and has a grant pending from the National Heart, Lung, and Blood Institute. L. D. Notarangelo has received grants from the NIH and the March of Dimes; is an Associate Editor for the Journal of Allergy and Clinical Immunology and the Journal of Clinical Immunology; has consultant arrangements with Novimmune and Sigma-Tau; is employed by Children’s Hospital Pediatric Associates; and has received royalties from UpToDate. The rest of the authors declare that they have no relevant conflicts of interest.


Vox Sanguinis | 2017

Characterization of buffy coat-derived granulocytes for clinical use: a comparison with granulocyte colony-stimulating factor/dexamethasone-pretreated donor-derived products.

A. van de Geer; Roel P. Gazendam; A. T. J. Tool; J. L. van Hamme; D. de Korte; T. K. van den Berg; Sacha Zeerleder; Ton Kuijpers

Buffy coat‐derived granulocytes have been described as an alternative to the apheresis product from donors pretreated with dexamethasone and granulocyte colony‐stimulating factor (G‐CSF). The latter is – dependent on the local and national settings – obtained following a demanding and time‐consuming procedure, which is undesirable in critically ill septic patients. In contrast, buffy coat‐derived products have a large volume and are often heavily contaminated with red cells and platelets. We developed a new pooled buffy coat‐derived product with high purity and small volume, and performed a comprehensive functional characterization of these granulocytes.

Collaboration


Dive into the Roel P. Gazendam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anton Tool

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Dirk Roos

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans Janssen

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge