Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Mult Scler ; 24(9): 1144-1150, 2018 08.
Article in English | MEDLINE | ID: mdl-29117778

ABSTRACT

BACKGROUND: The significance of the gut microbiome for the pathogenesis of multiple sclerosis (MS) has been established, although the underlying signaling mechanisms of this interaction have not been sufficiently explored. OBJECTIVES: We address this point and use serotonin (5-hydroxytryptamine (5-HT))-a microbial-modulated neurotransmitter (NT) as a showcase to demonstrate that NTs regulated by the gut microbiome are potent candidates for mediators of the gut-brain axis in demyelinating disorders. Methods, Results, and Conclusion: Our comprehensive overview of literature provides evidence that 5-HT levels in the gut are controlled by the microbiome, both via secretion and through regulation of metabolites. In addition, we demonstrate that the gut microbiome can influence the formation of the serotonergic system (SS) in the brain. We also show that SS alterations have been related to MS directly-altered expression of 5-HT transporters in central nervous system (CNS) and indirectly-beneficial effects of 5-HT modulating drugs on the course of the disease and higher prevalence of depression in patients with MS. Finally, we discuss briefly the role of other microbiome-modulated NTs such as γ-aminobutyric acid and dopamine in MS to highlight a new direction for future research aiming to relate microbiome-regulated NTs to demyelinating disorders.


Subject(s)
Gastrointestinal Microbiome/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/microbiology , Neuroimmunomodulation/physiology , Serotonin/metabolism , Animals , Humans , Multiple Sclerosis/metabolism
2.
J Neuroinflammation ; 14(1): 105, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28494768

ABSTRACT

BACKGROUND: The influx of leukocytes into the central nervous system (CNS) is a key hallmark of the chronic neuro-inflammatory disease multiple sclerosis (MS). Strategies that aim to inhibit leukocyte migration across the blood-brain barrier (BBB) are therefore regarded as promising therapeutic approaches to combat MS. As the CD40L-CD40 dyad signals via TNF receptor-associated factor 6 (TRAF6) in myeloid cells to induce inflammation and leukocyte trafficking, we explored the hypothesis that specific inhibition of CD40-TRAF6 interactions can ameliorate neuro-inflammation. METHODS: Human monocytes were treated with a small molecule inhibitor (SMI) of CD40-TRAF6 interactions (6877002), and migration capacity across human brain endothelial cells was measured. To test the therapeutic potential of the CD40-TRAF6-blocking SMI under neuro-inflammatory conditions in vivo, Lewis rats and C57BL/6J mice were subjected to acute experimental autoimmune encephalomyelitis (EAE) and treated with SMI 6877002 for 6 days (rats) or 3 weeks (mice). RESULTS: We here show that a SMI of CD40-TRAF6 interactions (6877002) strongly and dose-dependently reduces trans-endothelial migration of human monocytes. Moreover, upon SMI treatment, monocytes displayed a decreased production of ROS, tumor necrosis factor (TNF), and interleukin (IL)-6, whereas the production of the anti-inflammatory cytokine IL-10 was increased. Disease severity of EAE was reduced upon SMI treatment in rats, but not in mice. However, a significant reduction in monocyte-derived macrophages, but not in T cells, that had infiltrated the CNS was eminent in both models. CONCLUSIONS: Together, our results indicate that SMI-mediated inhibition of the CD40-TRAF6 pathway skews human monocytes towards anti-inflammatory cells with reduced trans-endothelial migration capacity, and is able to reduce CNS-infiltrated monocyte-derived macrophages during neuro-inflammation, but minimally ameliorates EAE disease severity. We therefore conclude that SMI-mediated inhibition of the CD40-TRAF6 pathway may represent a beneficial treatment strategy to reduce monocyte recruitment and macrophage activation in the CNS and has the potential to be used as a co-treatment to combat MS.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , CD40 Antigens/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Monocytes/drug effects , TNF Receptor-Associated Factor 6/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Cerebellum/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Monocytes/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Nitric Oxide Synthase Type I/metabolism , Peptide Fragments/toxicity , Rats , Rats, Inbred Lew , Reactive Oxygen Species/metabolism , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Front Immunol ; 8: 1791, 2017.
Article in English | MEDLINE | ID: mdl-29312317

ABSTRACT

The CD40-CD40L dyad is an immune checkpoint regulator that promotes both innate and adaptive immune responses and has therefore an essential role in the development of inflammatory diseases, including multiple sclerosis (MS). In MS, CD40 and CD40L are expressed on immune cells present in blood and lymphoid organs, affected resident central nervous system (CNS) cells, and inflammatory cells that have infiltrated the CNS. CD40-CD40L interactions fuel the inflammatory response underlying MS, and both genetic deficiency and antibody-mediated inhibition of the CD40-CD40L dyad reduce disease severity in experimental autoimmune encephalomyelitis (EAE). Both proteins are therefore attractive therapeutic candidates to modulate aberrant inflammatory responses in MS. Here, we discuss the genetic, experimental and clinical studies on the role of CD40 and CD40L interactions in EAE and MS and we explore novel approaches to therapeutically target this dyad to combat neuroinflammatory diseases.

4.
FASEB J ; 31(2): 719-731, 2017 02.
Article in English | MEDLINE | ID: mdl-27806992

ABSTRACT

Clinical trials have shown that administration of the nematode Trichuris suis can be beneficial in treating various immune disorders. To provide insight into the mechanisms by which this worm suppresses inflammatory responses, an active component was purified from T. suis soluble products (TsSPs) that suppress---- TNF and IL-12 secretion from LPS-activated human dendritic cells (DCs). Analysis by liquid chromatography tandem mass spectrometry identified this compound as prostaglandin (PG)E2. The purified compound showed similar properties compared with TsSPs and commercial PGE2 in modulating LPS-induced expression of many cytokines and chemokines and in modulating Rab7B and P2RX7 expression in human DCs. Furthermore, the TsSP-induced reduction of TNF secretion from DCs is reversed by receptor antagonists for EP2 and EP4, indicating PGE2 action. T. suis secretes extremely high amounts of PGE2 (45-90 ng/mg protein) within their excretory/secretory products but few related lipid mediators as established by metabololipidomic analysis. Culture of T. suis with several cyclooxygenase (COX) inhibitors that inhibit mammalian prostaglandin synthesis affected the worm's motility but did not inhibit PGE2 secretion, suggesting that the worms can synthesize PGE2 via a COX-independent pathway. We conclude that T. suis secretes PGE2 to suppress proinflammatory responses in human DCs, thereby modulating the host's immune response.-Laan, L. C., Williams, A. R., Stavenhagen, K., Giera, M., Kooij, G., Vlasakov, I., Kalay, H., Kringel, H., Nejsum, P., Thamsborg, S. M., Wuhrer, M., Dijkstra, C. D., Cummings, R. D., van Die, I. The whipworm (Trichuris suis) secretes prostaglandin E2 to suppress proinflammatory properties in human dendritic cells.


Subject(s)
Dendritic Cells/metabolism , Dinoprostone/metabolism , Dinoprostone/pharmacology , Inflammation/metabolism , Trichuris/metabolism , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/toxicity , Molecular Structure , Species Specificity
5.
FASEB J ; 30(8): 2826-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27095802

ABSTRACT

Helminths have strong immunoregulatory properties that may be exploited in treatment of chronic immune disorders, such as multiple sclerosis and inflammatory bowel disease. Essential players in the pathogenesis of these diseases are proinflammatory macrophages. We present evidence that helminths modulate the function and phenotype of these innate immune cells. We found that soluble products derived from the Trichuris suis (TsSP) significantly affect the differentiation of monocytes into macrophages and their subsequent polarization. TsSPs reduce the expression and production of inflammatory cytokines, including IL-6 and TNF, in human proinflammatory M1 macrophages. TsSPs induce a concomitant anti-inflammatory M2 signature, with increased IL-10 production. Furthermore, they suppress CHIT activity and enhance secretion of matrix metalloproteinase 9. Short-term triggering of monocytes with TsSPs early during monocyte-to-macrophage differentiation imprinted these phenotypic alterations, suggesting long-lasting epigenetic changes. The TsSP-induced effects in M1 macrophages were completely reversed by inhibiting histone deacetylases, which corresponded with decreased histone acetylation at the TNF and IL6 promoters. These results demonstrate that TsSPs have a potent and sustained immunomodulatory effect on human macrophage differentiation and polarization through epigenetic remodeling and provide new insights into the mechanisms by which helminths modulate human immune responses.-Hoeksema, M. A., Laan, L. C., Postma, J. J., Cummings, R. D., de Winther, M. P. J., Dijkstra, C. D., van Die, I., Kooij, G. Treatment with Trichuris suis soluble products during monocyte-to-macrophage differentiation reduces inflammatory responses through epigenetic remodeling.


Subject(s)
Cell Differentiation/drug effects , Epigenesis, Genetic/drug effects , Lipopolysaccharides/pharmacology , Macrophages/physiology , Monocytes/physiology , Trichuris/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Epigenesis, Genetic/physiology , Gene Expression Regulation/drug effects , Helminth Proteins , Humans , Inflammation , Lipopolysaccharides/chemistry , Trichuris/chemistry
6.
Acta Neuropathol Commun ; 3: 45, 2015 Jul 25.
Article in English | MEDLINE | ID: mdl-26205402

ABSTRACT

INTRODUCTION: The inverse correlation between prevalence of auto-immune disorders like the chronic neuro-inflammatory disease multiple sclerosis (MS) and the occurrence of helminth (worm) infections, suggests that the helminth-trained immune system is protective against auto-immunity. As monocytes are regarded as crucial players in the pathogenesis of auto-immune diseases, we explored the hypothesis that these innate effector cells are prime targets for helminths to exert their immunomodulatory effects. RESULTS: Here we show that soluble products of the porcine nematode Trichuris suis (TsSP) are potent in changing the phenotype and function of human monocytes by skewing classical monocytes into anti-inflammatory patrolling cells, which exhibit reduced trans-endothelial migration capacity in an in vitro model of the blood-brain barrier. Mechanistically, we identified the mannose receptor as the TsSP-interacting monocyte receptor and we revealed that specific downstream signalling occurs via protein kinase C (PKC), and in particular PKCδ. CONCLUSION: This study provides comprehensive mechanistic insight into helminth-induced immunomodulation, which can be therapeutically exploited to combat various auto-immune disorders.


Subject(s)
Inflammation/parasitology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Monocytes/physiology , Monocytes/parasitology , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism , Trichuris/physiology , Animals , Antigens, CD/metabolism , Cell Movement/physiology , Cytokines/metabolism , Flow Cytometry , Humans , Inflammation/pathology , Mannose Receptor
7.
PLoS One ; 10(4): e0124347, 2015.
Article in English | MEDLINE | ID: mdl-25884209

ABSTRACT

Galectin-2 is a monocyte-expressed carbohydrate-binding lectin, for which increased expression is genetically determined and associated with decreased collateral arteriogenesis in obstructive coronary artery disease patients. The inhibiting effect of galectin-2 on arteriogenesis was confirmed in vivo, but the mechanism is largely unknown. In this study we aimed to explore the effects of galectin-2 on monocyte/macrophage phenotype in vitro and vivo, and to identify the receptor by which galectin-2 exerts these effects. We now show that the binding of galectin-2 to different circulating human monocyte subsets is dependent on monocyte surface expression levels of CD14. The high affinity binding is blocked by an anti-CD14 antibody but not by carbohydrates, indicating a specific protein-protein interaction. Galectin-2 binding to human monocytes modulated their transcriptome by inducing proinflammatory cytokines and inhibiting pro-arteriogenic factors, while attenuating monocyte migration. Using specific knock-out mice, we show that galectin-2 acts through the CD14/toll-like receptor (TLR)-4 pathway. Furthermore, galectin-2 skews human macrophages to a M1-like proinflammatory phenotype, characterized by a reduced motility and expression of an anti-arteriogenic cytokine/growth factor repertoire. This is accompanied by a switch in surface protein expression to CD40-high and CD206-low (M1). In a murine model we show that galectin-2 administration, known to attenuate arteriogenesis, leads to increased numbers of CD40-positive (M1) and reduced numbers of CD206-positive (M2) macrophages surrounding actively remodeling collateral arteries. In conclusion galectin-2 is the first endogenous CD14/TLR4 ligand that induces a proinflammatory, non-arteriogenic phenotype in monocytes/macrophages. Interference with CD14-Galectin-2 interaction may provide a new intervention strategy to stimulate growth of collateral arteries in genetically compromised cardiovascular patients.


Subject(s)
Collateral Circulation/physiology , Galectin 2/physiology , Inflammation/physiopathology , Macrophages/physiology , Monocytes/physiology , Animals , CD40 Antigens/biosynthesis , Cell Differentiation , Cells, Cultured , Collateral Circulation/drug effects , Dendritic Cells/metabolism , Galectin 2/deficiency , Galectin 2/genetics , Galectin 2/pharmacology , Gene Expression Regulation , Humans , Lectins, C-Type/biosynthesis , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/physiology , Macrophages/classification , Macrophages/drug effects , Mannose Receptor , Mannose-Binding Lectins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Phenotype , Protein Binding/drug effects , RAW 264.7 Cells , Receptors, Cell Surface/biosynthesis , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction , T-Lymphocytes/metabolism , Toll-Like Receptor 4/metabolism
8.
Eur J Immunol ; 45(6): 1808-19, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25756873

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). Infiltration of monocytes into the CNS is crucial for disease onset and progression. Animal studies indicate that granulocyte-macrophages colony-stimulating factor (GM-CSF) may play an essential role in this process, possibly by acting on the migratory capacities of myeloid cells across the blood-brain barrier. This study describes the effect of GM-CSF on human monocytes, macrophages, and microglia. Furthermore, the expression of GM-CSF and its receptor was investigated in the CNS under healthy and pathological conditions. We show that GM-CSF enhances monocyte migration across human blood-brain barrier endothelial cells in vitro. Next, immunohistochemical analysis on human brain tissues revealed that GM-CSF is highly expressed by microglia and macrophages in MS lesions. The GM-CSF receptor is expressed by neurons in the rim of combined gray/white matter lesions and astrocytes. Finally, the effect of GM-CSF on human macrophages was determined, revealing an intermediate activation status, with a phenotype similar to that observed in active MS lesions. Together our data indicate that GM-CSF is a powerful stimulator of monocyte migration, and is abundantly present in the inflamed CNS where it may act as an activator of macrophages and microglia.


Subject(s)
Blood-Brain Barrier/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Monocytes/immunology , Monocytes/metabolism , Transendothelial and Transepithelial Migration/immunology , Adult , Aged , Aged, 80 and over , Blood-Brain Barrier/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , Cells, Cultured , Cytokines/metabolism , Endothelial Cells , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Microglia/immunology , Microglia/metabolism , Middle Aged , Monocytes/drug effects , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Reactive Oxygen Species/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Transendothelial and Transepithelial Migration/drug effects
9.
J Neuropathol Exp Neurol ; 74(1): 48-63, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25470347

ABSTRACT

Similar to macrophages, microglia adopt diverse activation states and contribute to repair and tissue damage in multiple sclerosis. Using reverse transcription-quantitative polymerase chain reaction and immunohistochemistry, we show that in vitro M1-polarized (proinflammatory) human adult microglia express the distinctive markers CD74, CD40, CD86, and CCR7, whereas M2 (anti-inflammatory) microglia express mannose receptor and the anti-inflammatory cytokine CCL22. The expression of these markers was assessed in clusters of activated microglia in normal-appearing white matter (preactive lesions) and areas of remyelination, representing reparative multiple sclerosis lesions. We show that activated microglia in preactive and remyelinating lesions express CD74, CD40, CD86, and the M2 markers CCL22 and CD209, but not mannose receptor. To examine whether this intermediate microglia profile is static or dynamic and thus susceptible to changes in the microenvironment, we polarized microglia into M1 or M2 phenotype in vitro and then subsequently treated them with the opposing polarization regimen. These studies revealed that expression of CD40, CXCL10, and mannose receptor is dynamic and that microglia, like macrophages, can switch between M1 and M2 phenotypic profiles. Taken together, our data define the differential activation states of microglia during lesion development in multiple sclerosis-affected CNS tissues and underscore the plasticity of human adult microglia in vitro.


Subject(s)
Brain/pathology , Histocompatibility Antigens Class II/metabolism , Microglia/pathology , Multiple Sclerosis/pathology , Myelin Proteolipid Protein/metabolism , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Flow Cytometry , Humans , Macrophages/pathology , Male , Microglia/metabolism , Middle Aged , Myelin Proteolipid Protein/genetics , RNA, Messenger/metabolism , Statistics, Nonparametric , Transcriptome
10.
PLoS One ; 9(10): e109995, 2014.
Article in English | MEDLINE | ID: mdl-25334032

ABSTRACT

AIM: Thymic epithelial cells (TECs) are thought to play an essential role in T cell development and have been detected mainly in mice using lectin binding and antibodies to keratins. Our aim in the present study was to create a precise map of rat TECs using antibodies to putative markers and novel monoclonal antibodies (i.e., ED 18/19/21 and anti-CD205 antibodies) and compare it with a map from mouse counterparts and that of rat thymic dendritic cells. RESULTS: Rat TECs were subdivided on the basis of phenotype into three subsets; ED18+ED19+/-keratin 5 (K5)+K8+CD205+ class II MHC (MHCII)+ cortical TECs (cTECs), ED18+ED21-K5-K8+Ulex europaeus lectin 1 (UEA-1)+CD205- medullary TECs (mTEC1s), and ED18+ED21+K5+K8dullUEA-1-CD205- medullary TECs (mTEC2s). Thymic nurse cells were defined in cytosmears as an ED18+ED19+/-K5+K8+ subset of cTECs. mTEC1s preferentially expressed MHCII, claudin-3, claudin-4, and autoimmune regulator (AIRE). Use of ED18 and ED21 antibodies revealed three subsets of TECs in mice as well. We also detected two distinct TEC-free areas in the subcapsular cortex and in the medulla. Rat dendritic cells in the cortex were MHCII+CD103+ but negative for TEC markers, including CD205. Those in the medulla were MHCII+CD103+ and CD205+ cells were found only in the TEC-free area. CONCLUSION: Both rats and mice have three TEC subsets with similar phenotypes that can be identified using known markers and new monoclonal antibodies. These findings will facilitate further analysis of TEC subsets and DCs and help to define their roles in thymic selection and in pathological states such as autoimmune disorders.


Subject(s)
Antibodies, Monoclonal/immunology , Epithelial Cells/metabolism , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cells, Cultured , Claudin-3/immunology , Claudin-3/metabolism , Claudin-4/immunology , Claudin-4/metabolism , Epithelial Cells/cytology , Epithelial Cells/pathology , Female , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Keratin-5/immunology , Keratin-5/metabolism , Keratin-8/immunology , Keratin-8/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Male , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Phenotype , Plant Lectins/immunology , Plant Lectins/metabolism , Rats , Rats, Inbred Lew , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Thymus Gland/cytology
11.
Immunobiology ; 219(9): 695-703, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24916404

ABSTRACT

Macrophages form a heterogeneous cell population displaying multiple functions, and can be polarized into pro- (M1) or anti-inflammatory (M2) macrophages, by environmental factors. Their activation status reflects a beneficial or detrimental role in various diseases. Currently several in vitro maturation and activation protocols are used to induce an M1 or M2 phenotype. Here, the impact of different maturation factors (NHS, M-CSF, or GM-CSF) and activation methods (IFN-γ/LPS, IL-4, dexamethason, IL-10) on the macrophage phenotype was determined. Regarding macrophage morphology, pro-inflammatory (M1) activation stimulated cell elongation, and anti-inflammatory (M2) activation induced a circular appearance. Activation with pro-inflammatory mediators led to increased CD40 and CD64 expression, whereas activation with anti-inflammatory factors resulted in increased levels of MR and CD163. Production of pro-inflammatory cytokines was induced by activation with IFN-γ/LPS, and TGF-ß production was enhanced by the maturation factors M-CSF and GM-CSF. Our data demonstrate that macrophage marker expression and cytokine production in vitro is highly dependent on both maturation and activation methods. In vivo macrophage activation is far more complex, since a plethora of stimuli are present. Hence, defining the macrophage activation status ex vivo on a limited number of markers could be indecisive. From this study we conclude that maturation with M-CSF or GM-CSF induces a moderate anti- or pro-inflammatory state respectively, compared to maturation with NHS. CD40 and CD64 are the most distinctive makers for human M1 and CD163 and MR for M2 macrophage activation and therefore can be helpful in determining the activation status of human macrophages ex vivo.


Subject(s)
Immunologic Techniques , Macrophage Activation/immunology , Macrophages/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Macrophage Activation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Serum
12.
J Neuroinflammation ; 11: 23, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24485070

ABSTRACT

BACKGROUND: In neuroinflammatory diseases, macrophages can play a dual role in the process of tissue damage, depending on their activation status (M1 / M2). M1 macrophages are considered to exert damaging effects to neurons, whereas M2 macrophages are reported to aid regeneration and repair of neurons. Their migration within the central nervous system may be of critical importance in the final outcome of neurodegeneration in neuroinflammatory diseases e.g. multiple sclerosis (MS). To provide insight into this process, we examined the migratory capacity of human monocyte-derived M1 and M2 polarised macrophages towards chemoattractants, relevant for neuroinflammatory diseases like MS. METHODS: Primary cultures of human monocyte-derived macrophages were exposed to interferon gamma and lipopolysaccharide (LPS) to evoke proinflammatory (M1) activation or IL-4 to evoke anti-inflammatory (M2) activation. In a TAXIScan assay, migration of M0, M1 and M2 towards chemoattractants was measured and quantified. Furthermore the adhesion capacity and the expression levels of integrins as well as chemokine receptors of M0, M1 and M2 were assessed. Alterations in cell morphology were analysed using fluorescent labelling of the cytoskeleton. RESULTS: Significant differences were observed between M1 and M2 macrophages in the migration towards chemoattractants. We show that M2 macrophages migrated over longer distances towards CCL2, CCL5, CXCL10, CXCL12 and C1q compared to non-activated (M0) and M1 macrophages. No differences were observed in the adhesion of M0, M1 and M2 macrophages to multiple matrix components, nor in the expression of integrins and chemokine receptors. Significant changes were observed in the cytoskeleton organization upon stimulation with CCL2, M0, M1 and M2 macrophages adopt a spherical morphology and the cytoskeleton is rapidly rearranged. M0 and M2 macrophages are able to form filopodia, whereas M1 macrophages only adapt a spherical morphology. CONCLUSIONS: Together our results indicate that the alternative activation status of macrophages promotes their migratory properties to chemoattractants relevant for neuroinflammatory diseases like MS. Conversely, classically activated, proinflammatory macrophages have reduced migratory properties. Based on our results, we postulate that the activation status of the macrophage influences the capacity of the macrophages to rearrange their cytoskeleton. This is the first step in understanding how modulation of macrophage activation affects macrophage migration in neuroinflammatory diseases like MS.


Subject(s)
Cell Movement/physiology , Cytokines/metabolism , Cytoskeleton/metabolism , Gene Expression Regulation/physiology , Macrophages/physiology , Cell Adhesion , Cells, Cultured , Complement C1q/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Receptors, Chemokine/metabolism
13.
J Neuroinflammation ; 10: 35, 2013 Mar 04.
Article in English | MEDLINE | ID: mdl-23452918

ABSTRACT

BACKGROUND: Macrophages play a dual role in multiple sclerosis (MS) pathology. They can exert neuroprotective and growth promoting effects but also contribute to tissue damage by production of inflammatory mediators. The effector function of macrophages is determined by the way they are activated. Stimulation of monocyte-derived macrophages in vitro with interferon-γ and lipopolysaccharide results in classically activated (CA/M1) macrophages, and activation with interleukin 4 induces alternatively activated (AA/M2) macrophages. METHODS: For this study, the expression of a panel of typical M1 and M2 markers on human monocyte derived M1 and M2 macrophages was analyzed using flow cytometry. This revealed that CD40 and mannose receptor (MR) were the most distinctive markers for human M1 and M2 macrophages, respectively. Using a panel of M1 and M2 markers we next examined the activation status of macrophages/microglia in MS lesions, normal appearing white matter and healthy control samples. RESULTS: Our data show that M1 markers, including CD40, CD86, CD64 and CD32 were abundantly expressed by microglia in normal appearing white matter and by activated microglia and macrophages throughout active demyelinating MS lesions. M2 markers, such as MR and CD163 were expressed by myelin-laden macrophages in active lesions and perivascular macrophages. Double staining with anti-CD40 and anti-MR revealed that approximately 70% of the CD40-positive macrophages in MS lesions also expressed MR, indicating that the majority of infiltrating macrophages and activated microglial cells display an intermediate activation status. CONCLUSIONS: Our findings show that, although macrophages in active MS lesions predominantly display M1 characteristics, a major subset of macrophages have an intermediate activation status.


Subject(s)
Brain/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Macrophages/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Adult , Aged , Brain/pathology , CD40 Antigens/metabolism , Cells, Cultured , Female , Humans , Inflammation Mediators/physiology , Macrophage Activation/physiology , Male , Middle Aged
14.
Mol Immunol ; 51(2): 210-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22482518

ABSTRACT

The increased incidence of auto-inflammatory and autoimmune diseases in the developed countries seems to be caused by an imbalance of the immune system due to the lack of proper regulation. Helminth parasites are well known modulators of the immune system and as such are of great interest for the treatment of these disorders. Clinical studies showed that administration of eggs of the pig nematode Trichuris suis to patients with inflammatory bowel disease reduces the disease severity. Here we demonstrate that treatment with soluble products from the nematodes T. suis and Trichinella spiralis induces significant suppression of symptoms in murine experimental autoimmune encephalomyelitis, a validated animal model for multiple sclerosis. These data show that infection with live nematodes is not a prerequisite for suppression of inflammation. To translate these results to the human system, the effects of soluble products of T. suis, T. spiralis and Schistosoma mansoni on the phenotype and function of human dendritic cells (DCs) were compared. Our data show that soluble products of T. suis, S. mansoni and T. spiralis suppress TNF-α and IL-12 secretion by TLR-activated human DCs, and that T. suis and S. mansoni, but not T. spiralis, strongly enhance expression of OX40L. Furthermore, helminth-primed human DCs differentially suppress the development of Th1 and/or Th17 cells. In conclusion, our data demonstrate that soluble helminth products have strong immunomodulatory capacities, but might exert their effects through different mechanisms. The suppressed secretion of pro-inflammatory cytokines together with an upregulation of OX40L expression on human DCs might contribute to achieve this modulation.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Helminth Proteins/immunology , Immunomodulation/immunology , Animals , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
15.
Glia ; 60(3): 422-31, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22161990

ABSTRACT

Neuroaxonal degeneration is a pathological hallmark of multiple sclerosis (MS) contributing to irreversible neurological disability. Pathological mechanisms leading to axonal damage include autoimmunity to neuronal antigens. In actively demyelinating lesions, myelin is phagocytosed by microglia and blood-borne macrophages, whereas the fate of degenerating or damaged axons is unclear. Phagocytosis is essential for clearing neuronal debris to allow repair and regeneration. However, phagocytosis may lead to antigen presentation and autoimmunity, as has been described for neuroaxonal antigens. Despite this notion, it is unknown whether phagocytosis of neuronal antigens occurs in MS. Here, we show using novel, well-characterized antibodies to axonal antigens, that axonal damage is associated with HLA-DR expressing microglia/macrophages engulfing axonal bulbs, indicative of axonal damage. Neuronal proteins were frequently observed inside HLA-DR(+) cells in areas of axonal damage. In vitro, phagocytosis of neurofilament light (NF-L), present in white and gray matter, was observed in human microglia. The number of NF-L or myelin basic protein (MBP) positive cells was quantified using the mouse macrophage cell line J774.2. Intracellular colocalization of NF-L with the lysosomal membrane protein LAMP1 was observed using confocal microscopy confirming that NF-L is taken up and degraded by the cell. In vivo, NF-L and MBP was observed in cerebrospinal fluid cells from patients with MS, suggesting neuronal debris is drained by this route after axonal damage. In summary, neuroaxonal debris is engulfed, phagocytosed, and degraded by HLA-DR(+) cells. Although uptake is essential for clearing neuronal debris, phagocytic cells could also play a role in augmenting autoimmunity to neuronal antigens.


Subject(s)
Microglia/physiology , Multiple Sclerosis/pathology , Neurons/pathology , Phagocytosis/physiology , Adult , Aged , Aged, 80 and over , Animals , Cathepsin D/pharmacology , Cathepsins/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Female , HLA-DR Antigens/metabolism , Humans , Male , Mice , Microglia/drug effects , Microscopy, Confocal , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Myelin Basic Protein/cerebrospinal fluid , Myelin Basic Protein/metabolism , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Neurofilament Proteins/cerebrospinal fluid , Neurofilament Proteins/drug effects , Neurofilament Proteins/metabolism , Neurons/drug effects , Phagocytosis/drug effects , Time Factors
16.
J Neuroinflammation ; 8: 58, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21615896

ABSTRACT

BACKGROUND: Macrophages play an important role in neuroinflammatory diseases such as multiple sclerosis (MS) and spinal cord injury (SCI), being involved in both damage and repair. The divergent effects of macrophages might be explained by their different activation status: classically activated (CA/M1), pro-inflammatory, macrophages and alternatively activated (AA/M2), growth promoting, macrophages. Little is known about the effect of macrophages with these phenotypes in the central nervous system (CNS) and how they influence pathogenesis. The aim of this study was therefore to determine the characteristics of these phenotypically different macrophages in the context of the CNS in an in vitro setting. RESULTS: Here we show that bone marrow derived CA and AA macrophages have a distinct migratory capacity towards medium conditioned by various cell types of the CNS. AA macrophages were preferentially attracted by the low weight (< 10 kD) fraction of neuronal conditioned medium, while CA macrophages were attracted in higher numbers by astrocyte- and oligodendrocyte conditioned medium. Intrinsic motility was twice as high in AA macrophages compared to CA macrophages. The adhesion to extracellular matrix molecules (ECM) was significantly enhanced in CA macrophages compared to control and AA macrophages. The actin cytoskeleton was differentially organized between CA and AA macrophages, possibly due to greater activity of the GTPases RhoA and Rac in CA macrophages. Phagocytosis of myelin and neuronal fragments was increased in CA macrophages compared to AA macrophages. The increase in myelin phagocytosis was associated with higher expression of CR3/MAC-1 in CA macrophages. CONCLUSION: In conclusion, since AA macrophages are more motile and are attracted by NCM, they are prone to migrate towards neurons in the CNS. CA macrophages have a lower motility and a stronger adhesion to ECM. In neuroinflammatory diseases the restricted migration and motility of CA macrophages might limit lesion size due to bystander damage.


Subject(s)
Cell Movement/physiology , Central Nervous System/cytology , Cytoskeleton/metabolism , Macrophages/cytology , Macrophages/physiology , Neuroglia/metabolism , Neurons/metabolism , Actins/metabolism , Animals , Cells, Cultured , Culture Media, Conditioned/chemistry , Flavonoids/pharmacology , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Neuroglia/cytology , Neurons/cytology , Phagocytosis/drug effects , Phagocytosis/physiology , Phenotype
17.
Brain ; 134(Pt 2): 555-70, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21183485

ABSTRACT

Adenosine triphosphate-binding cassette efflux transporters are highly expressed at the blood-brain barrier and actively hinder passage of harmful compounds, thereby maintaining brain homoeostasis. Since, adenosine triphosphate-binding cassette transporters drive cellular exclusion of potential neurotoxic compounds or inflammatory molecules, alterations in their expression and function at the blood-brain barrier may contribute to the pathogenesis of neuroinflammatory disorders, such as multiple sclerosis. Therefore, we investigated the expression pattern of different adenosine triphosphate-binding cassette efflux transporters, including P-glycoprotein, multidrug resistance-associated proteins-1 and -2 and breast cancer resistance protein in various well-characterized human multiple sclerosis lesions. Cerebrovascular expression of P-glycoprotein was decreased in both active and chronic inactive multiple sclerosis lesions. Interestingly, foamy macrophages in active multiple sclerosis lesions showed enhanced expression of multidrug resistance-associated protein-1 and breast cancer resistance protein, which coincided with their increased function of cultured foamy macrophages. Strikingly, reactive astrocytes display an increased expression of P-glycoprotein and multidrug resistance-associated protein-1 in both active and inactive multiple sclerosis lesions, which correlated with their enhanced in vitro activity on astrocytes derived from multiple sclerosis lesions. To investigate whether adenosine triphosphate-binding cassette transporters on reactive astrocytes can contribute to the inflammatory process, primary cultures of reactive human astrocytes were generated through activation of Toll-like receptor-3 to mimic the astrocytic phenotype as observed in multiple sclerosis lesions. Notably, blocking adenosine triphosphate-binding cassette transporter activity on reactive astrocytes inhibited immune cell migration across a blood-brain barrier model in vitro, which was due to the reduction of astrocytic release of the chemokine (C-C motif) ligand 2. Our data point towards a novel (patho)physiological role for adenosine triphosphate-binding cassette transporters, suggesting that limiting their activity by dampening astrocyte activation may open therapeutic avenues to diminish tissue damage during multiple sclerosis pathogenesis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Chemokine CCL2/metabolism , Multiple Sclerosis/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Blood-Brain Barrier/physiology , Brain/metabolism , Brain/physiopathology , Cell Culture Techniques , Cell Movement/physiology , Female , Humans , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Monocytes/physiology , Multiple Sclerosis/physiopathology
18.
Mol Imaging ; 9(5): 268-77, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20868627

ABSTRACT

AbstractInfiltrated monocytes play a crucial role in the demyelination process during multiple sclerosis (MS), an inflammatory disease of the central nervous system (CNS). Still, methods to monitor their infiltration pattern over time are lacking. In this study, magnetoelectroporation (MEP) was used to label rat monocytes with the superparamagnetic iron oxide particles Sinerem, Endorem, and Supravist. Supravist-labeled monocytes were injected in rats that we induced with experimental autoimmune encephalomyelitis, a model for MS. Imaging at 4.7 and 9.4 T revealed multiple foci of decreased signal intensity predominantly located in the cerebellum. Immunohistochemical evaluation confirmed the presence of intracellular iron in infiltrated cells, indicating the suitability of MEP to specifically follow labeled monocytes in vivo in this disease model. This technique may be further optimized and potentially used in MS patients to assess monocyte migration into the brain and to monitor the efficacy of therapeutic agents aimed at blocking cellular migration into the CNS.


Subject(s)
Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Monocytes/chemistry , Monocytes/cytology , Multiple Sclerosis/pathology , Animals , Cells, Cultured , Dextrans/chemistry , Rats
19.
NMR Biomed ; 23(9): 1087-96, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20665906

ABSTRACT

Signal loss observed in the brain by MRI following the administration of ultrasmall superparamagnetic particles of iron oxide (USPIO) has been correlated with immune cell activity in inflammatory areas during multiple sclerosis. Uptake of USPIO by circulating monocytes and their migration towards inflammatory areas have been considered as the most important mechanism for USPIO uptake by the brain parenchyma. However, the involvement of a damaged blood-brain barrier is also debated as a possible mechanism for cerebral USPIO uptake. Compared with these uptake-associated issues, little is known about the clearance of USPIO from the brain. The acute uptake and chronic clearance of USPIO in the brain were therefore studied with MRI in an animal model of multiple sclerosis. Lewis Hannover rats with acute experimental autoimmune encephalomyelitis received a single intravenous injection of USPIO (300 µmol Fe/kg), and repetitive MRI of the brain and cervical lymph nodes, a possible drainage pathway, was performed. USPIO were detected in the brain within 1 h after injection independent of the severity of experimental autoimmune encephalomyelitis, and histological analysis revealed extracellular iron clusters colocalising with a leaky blood-brain barrier. Loss of signal was not present 72 h after USPIO injection, irrespective of the disease state. MR images of cervical lymph nodes showed USPIO accumulation at 24 h after administration, which stabilised at 72 h. Histological analyses revealed that USPIO accumulated in infiltrated macrophages in the medulla and subcapsular sinus. The current study demonstrates that USPIO enter the central nervous system directly after administration, pointing to the involvement of a damaged blood-brain barrier in the appearance of USPIO-associated MR abnormalities. Furthermore, a possible role of the cervical lymph nodes as a drainage pathway of USPIO is suggested. These data shed new light on the use of USPIO in neuroinflammatory diseases, identifying USPIO as a marker for both cellular infiltration and blood-brain barrier damage.


Subject(s)
Central Nervous System , Contrast Media/metabolism , Dextrans/metabolism , Encephalomyelitis, Autoimmune, Experimental , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Dextrans/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/administration & dosage , Male , Particle Size , Rats , Rats, Inbred Lew
20.
Glia ; 58(12): 1465-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20648639

ABSTRACT

Sphingolipids are a class of biologically active lipids that have a role in multiple biological processes including inflammation. Sphingolipids exert their functions by direct signaling or through signaling by their specific receptors. Phosphorylated FTY720 (FTY720P) is a sphingosine 1-phosphate (S1P) analogue that is currently in trial for treatment of multiple sclerosis (MS), which targets all S1P receptors but S1P(2). To date, however, it remains unknown whether FTY720P may exert direct anti-inflammatory effects within the central nervous system (CNS), because data concerning S1P receptor expression and regulation under pathological conditions in the human brain are lacking. To investigate potential regulation of S1P receptors in the human brain during MS, we performed immunohistochemical analysis of S1P receptor 1 and 3 expression in well-characterized MS lesions. A strong increase in S1P receptor 1 and 3 expression on reactive astrocytes was detected in active and chronic inactive MS lesions. In addition, we treated primary cultures of human astrocytes with the proinflammatory cytokine tumor necrosis factor-alpha to identify the regulation of S1P(1/3) on astrocytes under pathological conditions. Importantly, we demonstrate that FTY720P exerts an anti-inflammatory action on human astrocytes by limiting secretion of proinflammatory cytokines. Our data demonstrate that reactive astrocytes in MS lesions and cultured under proinflammatory conditions strongly enhance expression of S1P receptors 1 and 3. Results from this study indicate that astrocytes may act as a yet-unknown target within the CNS for the anti-inflammatory effects observed after FTY720P administration in the treatment of MS.


Subject(s)
Multiple Sclerosis/physiopathology , Receptors, Lysosphingolipid/metabolism , Up-Regulation/physiology , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Brain/cytology , Cells, Cultured , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacology , Male , Middle Aged , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...