Network


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

Hotspot


Dive into the research topics where Marie-José Melief is active.

Publication


Featured researches published by Marie-José Melief.


Journal of Immunology | 2005

Proinflammatory bacterial peptidoglycan as a cofactor for the development of central nervous system autoimmune disease

Lizette Visser; Hendrik Jan de Heer; Leonie A. Boven; Debby van Riel; Marjan van Meurs; Marie-José Melief; Ulrich Zähringer; Jos A. G. van Strijp; Bart N. Lambrecht; Edward E. S. Nieuwenhuis; Jon D. Laman

Upon stimulation by microbial products through TLR, dendritic cells (DC) acquire the capacity to prime naive T cells and to initiate a proinflammatory immune response. Recently, we have shown that APC within the CNS of multiple sclerosis (MS) patients contain peptidoglycan (PGN), a major cell wall component of Gram-positive bacteria, which signals through TLR and NOD. In this study, we report that Staphylococcus aureus PGN as a single component can support the induction of experimental autoimmune encephalomyelitis (EAE) in mice, an animal model for MS. Mice immunized with an encephalitogenic myelin oligodendrocyte glycoprotein peptide in IFA did not develop EAE. In contrast, addition of PGN to the emulsion was sufficient for priming of autoreactive Th1 cells and development of EAE. In vitro studies demonstrate that PGN stimulates DC-mediated processes, reflected by increased Ag uptake, DC maturation, Th1 cell expansion, activation, and proinflammatory cytokine production. These data indicate that PGN-mediated interactions result in proinflammatory stimulation of Ag-specific effector functions, which are important in the development of EAE. These PGN-mediated processes may occur both within the peripheral lymph nodes as well as in the CNS and likely involve recognition by TLR on DC. Thus, PGN may provide a physiological trigger of DC maturation, and in this way disrupt the normal tolerance to self Ag. As such, PGN signaling pathways may serve as novel targets for the treatment of MS.


Journal of Molecular Medicine | 2009

Brain antigens in functionally distinct antigen-presenting cell populations in cervical lymph nodes in MS and EAE

Marloes van Zwam; Ruth Huizinga; Marie-José Melief; Annet F. Wierenga-Wolf; Marjan van Meurs; Jane S. A. Voerman; Knut Biber; Hendrikus Boddeke; Uta E. Höpken; Christian Meisel; Andreas Meisel; Ingo Bechmann; Rogier Q. Hintzen; Bert A. 't Hart; Sandra Amor; Jon D. Laman; Leonie A. Boven

Drainage of central nervous system (CNS) antigens to the brain-draining cervical lymph nodes (CLN) is likely crucial in the initiation and control of autoimmune responses during multiple sclerosis (MS). We demonstrate neuronal antigens within CLN of MS patients. In monkeys and mice with experimental autoimmune encephalomyelitis (EAE) and in mouse models with non-inflammatory CNS damage, the type and extent of CNS damage was associated with the frequencies of CNS antigens within the cervical lymph nodes. In addition, CNS antigens drained to the spinal-cord-draining lumbar lymph nodes. In human MS CLN, neuronal antigens were present in pro-inflammatory antigen-presenting cells (APC), whereas the majority of myelin-containing cells were anti-inflammatory. This may reflect a different origin of the cells or different drainage mechanisms. Indeed, neuronal antigen-containing cells in human CLN did not express the lymph node homing receptor CCR7, whereas myelin antigen-containing cells in situ and in vitro did. Nevertheless, CLN from EAE-affected CCR7-deficient mice contained equal amounts of myelin and neuronal antigens as wild-type mice. We conclude that the type and frequencies of CNS antigens within the CLN are determined by the type and extent of CNS damage. Furthermore, the presence of myelin and neuronal antigens in functionally distinct APC populations within MS CLN suggests that differential immune responses can be evoked.


Arthritis & Rheumatism | 2000

Antigen-presenting cells containing bacterial peptidoglycan in synovial tissues of rheumatoid arthritis patients coexpress costimulatory molecules and cytokines

Ingrid A. Schrijver; Marie-José Melief; Paul P. Tak; Maarten P. Hazenberg; J. O. N. D. Laman

OBJECTIVE Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by intimal lining hyperplasia and massive infiltration of the synovial sublining by antigen-presenting cells (APCs), lymphocytes, and plasma cells. Peptidoglycan (PG), a major cell wall component of gram-positive bacteria, which is abundantly expressed in all mucosa, is believed to be involved in the pathogenesis of RA because of its ability to induce the production of proinflammatory cytokines as well as to induce arthritis in rodents. While PG has been detected in APCs in RA joints, little is known about the role of these cells in RA. In this study, the presence and immune competence of PG-containing cells in synovial tissues from 14 RA and 14 osteoarthritis (OA) patients were analyzed in situ. METHODS Using immunohistochemistry, we examined the coexpression of phenotypic markers, costimulatory molecules, and cytokines by PG-containing cells. RESULTS PG was present in higher numbers in RA than in OA synovial tissues, although the difference was not significant. PG-containing cells were mainly macrophages, but some mature dendritic cells also contained PG. A high percentage of PG-containing cells in both RA and OA synovial tissues coexpressed HLA-DR. CD40, CD80, and CD86 expression by PG-containing cells was higher in RA than in OA tissues. Furthermore, PG-containing cells coexpressed cytokines, which modulate inflammatory reactions, in particular, tumor necrosis factor alpha and interleukins 6 and 10. CONCLUSION The results suggest that PG-containing cells may contribute to inflammation within the microenvironment of the joint in RA patients.


The Journal of Pathology | 2009

Surgical excision of CNS-draining lymph nodes reduces relapse severity in chronic-relapsing experimental autoimmune encephalomyelitis

Marloes van Zwam; Ruth Huizinga; Nicole Heijmans; Marjan van Meurs; Annet F. Wierenga-Wolf; Marie-José Melief; Rogier Q. Hintzen; Bert A. 't Hart; Sandra Amor; Leonie A. Boven; Jon D. Laman

Despite lack of classical lymphatic vessels in the central nervous system (CNS), cells and antigens do reach the CNS‐draining lymph nodes. These lymph nodes are specialized to mediate mucosal immune tolerance, but can also generate T‐ and B‐cell immunity. Their role in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE) therefore remains elusive. We hypothesized that drainage of CNS antigens to the CNS‐draining lymph nodes is vital for the recurrent episodes of CNS inflammation. To test this, we surgically removed the superficial cervical lymph nodes, deep cervical lymph nodes, and the lumbar lymph nodes prior to disease induction in three mouse EAE models, representing acute, chronic, and chronic‐relapsing EAE. Excision of the CNS‐draining lymph nodes in chronic‐relapsing EAE reduced and delayed the relapse burden and EAE pathology within the spinal cord, which suggests initiation of CNS antigen‐specific immune responses within the CNS‐draining lymph nodes. Indeed, superficial cervical lymph nodes from EAE‐affected mice demonstrated proliferation against the immunizing peptide, and the deep cervical lymph nodes, lumbar lymph nodes, and spleen demonstrated additional proliferation against other myelin antigen epitopes. This indicates that intermolecular epitope spreading occurs and that CNS antigen‐specific immune responses are differentially generated within the different CNS‐draining lymphoid organs. Proliferation of splenocytes from lymphadenectomized and sham‐operated mice against the immunizing peptide was similar. These data suggest a role for CNS‐draining lymph nodes in the induction of detrimental immune responses in EAE relapses, and conclusively demonstrate that the tolerance‐inducing capability of cervical lymph nodes is not involved in EAE. Copyright


Journal of Neuroimmunology | 2002

Expression of the EGF-TM7 receptor CD97 and its ligand CD55 (DAF) in multiple sclerosis

Lizette Visser; Alex F. de Vos; Jörg Hamann; Marie-José Melief; Marjan van Meurs; René A. W. van Lier; Jon D. Laman; Rogier Q. Hintzen

CD97 is a recently identified seven-span transmembrane (7-TM) protein that is expressed by leukocytes early after activation. CD97 binds to its cellular ligand CD55 (decay accelerating factor), which protects several cell types from complement-mediated damage. The functional consequences of CD97-CD55 binding are largely unknown, but previous data imply that CD97-CD55 interactions play a role in cellular activation, migration, and adhesion under inflammatory conditions. Here we examined the expression of CD97 and CD55 by immunohistochemistry in multiple sclerosis (MS). On the basis of established criteria for inflammation and demyelination, different lesion stages were distinguished in MS post-mortem brain tissue. In normal white matter, CD97 expression was not found, but CD55 was expressed with weak staining intensity on endothelial cells. In pre-active lesions, defined by abnormalities of the white matter, many infiltrating T cells, macrophages (MPhi) and microglia expressed CD97. CD55 was highly expressed by endothelial cells. In active lesions with myelin degradation, MPhi and microglia expressed both CD55 and CD97. Furthermore, a sandwich ELISA showed significantly (p<0.05) elevated levels of soluble CD97 in serum but not in cerebrospinal fluid of MS patients (37%) compared to healthy controls (8%).Collectively, these data suggest that CD97-CD55 interactions are involved in the inflammatory processes in MS. CD55, which is expressed in lesions by vessels to protect against complement-mediated damage, might bind to CD97 on infiltrating leukocytes. This interaction may facilitate cell activation and migration through the blood-brain barrier. In addition, CD97-CD55 interactions in the parenchyma of the brain may contribute to the inflammation.


The Journal of Infectious Diseases | 1999

Bacterial peptidoglycan polysaccharides in sterile human spleen induce proinflammatory cytokine production by human blood cells

Ingrid A. Schrijver; Marie-José Melief; Frits Eulderink; Maarten P. Hazenberg; Jon D. Laman

Peptidoglycan (PG) is the major component of the cell wall of gram-positive bacteria. In vitro, PG isolated from conventional bacterial cultures can induce secretion of proinflammatory cytokines by human monocytes, indicating that PG may be involved in immune responses against infections by gram-positive bacteria. To investigate the biologic activity of PG in human tissues, an improved method was developed to isolate significant amounts of PG from sterile human spleen tissue. Biochemical analysis demonstrated that PG isolated from human spleen is largely intact. Human whole blood cell cultures were able to produce the proinflammatory cytokines tumor necrosis factor-alpha and interleukin-1 and -6 after stimulation with PG isolated from human spleen. Cytokine induction was not sensitive to inhibition by polymyxin B, in contrast to lipopolysaccharide. Collectively, the data show that intact PG in sterile human tissue is biologically active and may induce local proinflammatory cytokine production.


Biochimica et Biophysica Acta | 1996

Purification and characterization of N-acetylmuramyl-L-alanine amidase from human plasma using monoclonal antibodies

Maarten A. Hoijer; Marie-José Melief; Wolfgang Keck; Maarten P. Hazenberg

N-Acetylmuramyl-L-alanine amidase (EC 3.5.1.28) cleaves the amide bond between N-acetyl muramic acid and L-alanine in the peptide side chain of different peptidoglycan products. The enzyme was purified from human plasma using a three-step column chromatography procedure. Monoclonal antibodies were produced against the purified human enzyme. By coupling of a high affinity monoclonal antibody to sepharose beads an immunoadsorbent column was prepared. Using this second purification method it was possible to purify large amounts of the amidase from human plasma in a single step. SDS-PAGE showed one single band of 70 kDa and two-dimensional electrophoresis showed the presence of multiple isomeric forms of the protein with pI between 6.5 and 7.9. Two different methods were used for determination of substrate specificity, a HPLC method separating peptidoglycan monomers from the reaction products after incubation with amidase and a colorimetric method when high molecular weight peptidoglycan was used as a substrate for amidase. It is shown that the disaccharide tetra peptide, disaccharide penta peptide and the anhydro disaccharide tetrapeptide are good substrates for the amidase and that muramyl dipeptide and disaccharide dipeptide are not a substrate for the amidase. Using one of the monoclonal antibodies against the amidase it was shown in FACScan analysis that N-acetylmuramyl-L-alanine amidase is present in granulocytes but not in monocytes from unstimulated peripheral blood of a healthy donor. The presence of N-acetylmuramyl-L-alanine amidase in granulocytes is a novel finding and perhaps important for the inactivation of biologically active peptidoglycan products still present after hydrolysis by lysozyme.


Brain | 2015

Multiple sclerosis-associated CLEC16A controls HLA class II expression via late endosome biogenesis

Karim L. Kreft; Marlieke L.M. Jongsma; Steven W. Mes; Annet F. Wierenga-Wolf; Marjan van Meurs; Marie-José Melief; Rik van der Kant; Lennert Janssen; Hans Janssen; Rusung Tan; John J. Priatel; Jacques Neefjes; Jon D. Laman; Rogier Q. Hintzen

C-type lectins are key players in immune regulation by driving distinct functions of antigen-presenting cells. The C-type lectin CLEC16A gene is located at 16p13, a susceptibility locus for several autoimmune diseases, including multiple sclerosis. However, the function of this gene and its potential contribution to these diseases in humans are poorly understood. In this study, we found a strong upregulation of CLEC16A expression in the white matter of multiple sclerosis patients (n = 14) compared to non-demented controls (n = 11), mainly in perivascular leukocyte infiltrates. Moreover, CLEC16A levels were significantly enhanced in peripheral blood mononuclear cells of multiple sclerosis patients (n = 69) versus healthy controls (n = 46). In peripheral blood mononuclear cells, CLEC16A was most abundant in monocyte-derived dendritic cells, in which it strongly co-localized with human leukocyte antigen class II. Treatment of these professional antigen-presenting cells with vitamin D, a key protective environmental factor in multiple sclerosis, downmodulated CLEC16A in parallel with human leukocyte antigen class II. Knockdown of CLEC16A in distinct types of model and primary antigen-presenting cells resulted in severely impaired cytoplasmic distribution and formation of human leucocyte antigen class II-positive late endosomes, as determined by immunofluorescence and electron microscopy. Mechanistically, CLEC16A participated in the molecular machinery of human leukocyte antigen class II-positive late endosome formation and trafficking to perinuclear regions, involving the dynein motor complex. By performing co-immunoprecipitations, we found that CLEC16A directly binds to two critical members of this complex, RILP and the HOPS complex. CLEC16A silencing in antigen-presenting cells disturbed RILP-mediated recruitment of human leukocyte antigen class II-positive late endosomes to perinuclear regions. Together, we identify CLEC16A as a pivotal gene in multiple sclerosis that serves as a direct regulator of the human leukocyte antigen class II pathway in antigen-presenting cells. These findings are a first step in coupling multiple sclerosis-associated genes to the regulation of the strongest genetic factor in multiple sclerosis, human leukocyte antigen class II.


Journal of Histochemistry and Cytochemistry | 1994

Detection of intestinal flora-derived bacterial antigen complexes in splenic macrophages of rats.

Jeanette Kool; H De Visser; M Y Gerrits-Boeye; Ina S. Klasen; Marie-José Melief; C G Van Helden-Meeuwsen; L. M. C. Van Lieshout; J.G.H. Ruseler-van Embden; W. B. Van Den Berg; G M Bahr

We studied the presence of bacterial antigens in rat tissues. We produced a monoclonal antibody (MAb 2E9) directed against intestinal flora-derived peptidoglycan-polysaccharide complexes from human and rat feces. With several immunological techniques, the specificity of 2E9 for this bacterial product was demonstrated. Using 2E9 in an immunohistological assay, we were able to show the presence of bacterial products in macrophages in the red pulp of spleens of conventional Lewis rats. However, we found no correlation between the development of the intestinal flora and positive spleen staining with MAb 2E9. The results were confirmed by immunohistology with a previously described MAb 2-4 directed to muramyl dipeptide. Other lymphoid organs did not stain positively with 2E9 and 2-4. Neonatal and young rats showed no staining of the spleen, but positivity could be induced by injecting peptidoglycan-polysaccharide complexes systemically. We conclude that bacterial fragments are present in splenic macrophages of conventional rats.


European Journal of Immunology | 2004

Antagonist anti‐human CD40 antibody inhibits germinal center formation in cynomolgus monkeys

Alex F. de Vos; Marie-José Melief; Debby van Riel; Louis Boon; Marco van Eijk; Mark de Boer; Jon D. Laman

Interactions between CD40 on APC and CD154 (CD40L) expressed by activated CD4+ T cells are crucially involved in formation and function of germinal centers (GC), but mechanistic insight into these interactions remains limited. Functional studies have mostly been restricted to experimental immunization of young‐adult inbred SPF rodents that are often genetically manipulated, while studies in humans disallow in vivo manipulation. Therefore, we asked whether a functional antagonist of CD40 interferes with natural GC formation in adult cynomolgus monkeys (Macaca fascicularis) exposed to the environmental antigens of their conventional housing in captivity. Animals were treated with different doses of a unique chimeric antagonist anti‐CD40 mAb (ch5D12) and analyzed 1 week or 7 weeks after last injection. Detailed in situ analysis showed that high‐dose anti‐CD40 treatment increased the ratio of primary over secondary follicles compared to PBS or low‐dose treatment, indicative of impairment of the CG reaction. This impairment was reversible since recovery animals, except those with residual anti‐CD40 levels, had normalized ratios. Anti‐CD40 treatment was associated with decreased antibody production and increased numbers of apoptotic cells in GC. These data demonstrate that CD40‐CD154 interactions are pivotal in physiological GC formation in primates responding to environmental antigens, and they support immunotherapeutic strategies using antagonist anti‐CD40.

Collaboration


Dive into the Marie-José Melief's collaboration.

Top Co-Authors

Avatar

Jon D. Laman

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marjan van Meurs

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Rogier Q. Hintzen

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maarten P. Hazenberg

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Leonie A. Boven

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Bert A. 't Hart

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Ingrid A. Schrijver

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Debby van Riel

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Karim L. Kreft

Erasmus University Rotterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge