67 results on '"van het Hof B"'
Search Results
2. Increased matrix metalloproteinases expression in tuberous sclerosis complex: modulation by microRNA 146a and 147b in vitro
- Author
-
ZL Kinder Ner en Nec Medisch, Brain, Pathologie mortuarium en obductie, Pathologie Pathologen staf, Cancer, Zorglijn FNE Medisch, Broekaart, D. W.M., van Scheppingen, J., Anink, J. J., Wierts, L., van het Hof, B., Jansen, F. E., Spliet, W. G., van Rijen, P. C., Kamphuis, W. W., de Vries, H. E., Aronica, E., van Vliet, E. A., ZL Kinder Ner en Nec Medisch, Brain, Pathologie mortuarium en obductie, Pathologie Pathologen staf, Cancer, Zorglijn FNE Medisch, Broekaart, D. W.M., van Scheppingen, J., Anink, J. J., Wierts, L., van het Hof, B., Jansen, F. E., Spliet, W. G., van Rijen, P. C., Kamphuis, W. W., de Vries, H. E., Aronica, E., and van Vliet, E. A.
- Published
- 2020
3. Approach for defining endogenous reference genes in gene expression experiments
- Author
-
Garcı́a-Vallejo, J.J, Van het Hof, B, Robben, J, Van Wijk, J.A.E, Van Die, I, Joziasse, D.H, and Van Dijk, W
- Published
- 2004
- Full Text
- View/download PDF
4. Specific glycosylation of α1-acid glycoprotein characterises patients with familial Mediterranean fever and obligatory carriers of MEFV
- Author
-
Poland, D C W, Drenth, J P H, Rabinovitz, E, Livneh, A, Bijzet, J, van het Hof, B, and van Dijk, W
- Published
- 2001
5. Purinergic receptors P2Y12R and P2X7R: potential targets for PET imaging of microglia phenotypes in multiple sclerosis
- Author
-
Beaino, Wissam, primary, Janssen, Bieneke, additional, Kooij, Gijs, additional, van der Pol, Susanne M. A., additional, van Het Hof, B., additional, van Horssen, Jack, additional, Windhorst, Albert D., additional, and de Vries, Helga E., additional
- Published
- 2017
- Full Text
- View/download PDF
6. Geen nieuwe 'Betuwelijn' in het IJmeer
- Author
-
Koopmans, C.C., van het Hof, B., and Spatial Economics
- Published
- 2012
7. Specific glycosylation of [[Alpha].sub.1]-acid glycoprotein characterises patients with familial Mediterranean fever and obligatory carriers of MEFV
- Author
-
Poland, D C W, Drenth, J P H, Rabinovitz, E, Livneh, A, Bijzet, J, van het Hof, B, and van Dijk, W
- Subjects
Inflammation -- Mediators ,Acute phase reaction -- Research -- Physiological aspects ,Familial Mediterranean fever -- Physiological aspects -- Research ,Health ,Physiological aspects ,Research - Abstract
Abstract Background--Familial Mediterranean fever (FMF) is a periodic febrile disorder, characterised by fever and serositis. The acute phase response during attacks of FMF results from the release of cytokines, which [...]
- Published
- 2001
8. Effecten van kilometerbeprijzing op het bbp
- Author
-
Poort, J., van het Hof, B., Koopmans, C.C., Blom, M., Bruyn, S., Schroten, A., Marlet, G., Woerkens, C., SEO Economisch Onderzoek, and Spatial Economics
- Abstract
Wat zijn de effecten van kilometerbeprijzing op het bruto binnenlands product? Ondanks het vele onderzoek dat de afgelopen jaren is verricht naar Anders Betalen voor Mobiliteit, is deze vraag vooralsnog onderbelicht gebleven. Deze studie geeft antwoord op deze vraag, en sluit daarbij nauw aan bij eerder onderzoek naar de welvaartseffecten van kilometerbeprijzing: welke welvaartseffecten zijn van invloed op het bbp, welke niet, en in welke mate?
- Published
- 2010
9. Effecten van kilometerbeprijzing op het bbp
- Author
-
Poort, J., van het Hof, B., Koopmans, C.C., Blom, M., Bruyn, S., Schroten, A., Marlet, G., and Woerkens, C.
- Published
- 2009
10. Convergent actions of I kappa B kinase beta and protein kinase C delta modulate mRNA stability through phosphorylation of 14-3-3 beta complexed with tristetraprolin
- Author
-
Gringhuis, S.I., Garcia Vallejo, J.J., Van Het Hof, B, van Dijk, W., and VU University medical center
- Published
- 2005
- Full Text
- View/download PDF
11. Retinoic Acid Induces Blood-Brain Barrier Development
- Author
-
Mizee, M. R., primary, Wooldrik, D., additional, Lakeman, K. A. M., additional, van het Hof, B., additional, Drexhage, J. A. R., additional, Geerts, D., additional, Bugiani, M., additional, Aronica, E., additional, Mebius, R. E., additional, Prat, A., additional, de Vries, H. E., additional, and Reijerkerk, A., additional
- Published
- 2013
- Full Text
- View/download PDF
12. Interferons overrule the TNF-induced proliferation of rheumatoid fibroblast-like synoviocytes
- Author
-
Blits, M., primary, de Ridder, S., additional, Timmer, T. C. G., additional, Kasperkovitz, P. V., additional, Trujillo, A. M., additional, van het Hof, B., additional, van der Pouw Kraan, T. C. T. M., additional, Vosslamber, S., additional, de Jong, T., additional, Wesseling, J., additional, Gringhuis, S. I., additional, and Verweij, C. L., additional
- Published
- 2011
- Full Text
- View/download PDF
13. Approach for defining endogenous reference genes in gene expression experiments
- Author
-
García-Vallejo, J.J., Van het Hof, B., Robben, J., Van Wijk, J.A.E., Van Die, I., Joziasse, D.H., and Van Dijk, W.
- Subjects
- *
GENE expression , *POLYMERASE chain reaction , *B cells , *BLOOD vessels - Abstract
The quantification of gene expression by real-time polymerase chain reaction (PCR) has revolutionized the field of gene expression analysis. Due to its sensitivity and flexibility it is becoming the method of choice for many investigators. However, good normalization protocols still have to be implemented to facilitate data exchange and comparison. We have designed primers for 10 unrelated genes and developed a simple protocol to detect genes with stable expression that are suitable for use as endogenous reference genes for further use in the normalization of gene expression data obtained by real-time PCR. Using this protocol, we were able to identify human proteosome subunit Y as a reliable endogenous reference gene for human umbilical vein endothelial cells treated for up to 18 h with TNFα, IL-4, or IFNγ and for B cells isolated from healthy controls and patients suffering from IgA nephropathy. Other optional endogenous reference genes that can be considered are phosphomannomutase (PPMM) and actin for endothelial cells and glyceraldehyde-3-phosphate dehydrogenase and PPMM for B cells. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
14. Specific glycosylation of α1-acid glycoprotein characterises patients with familial Mediterranean fever and obligatory carriers of MEFV.
- Author
-
Poland, D. C. W., Drenth, J. P. H., Rabinovitz, E., Livneh, A., Bijzet, J., van het Hof, B., and van Dijk, W.
- Published
- 2001
- Full Text
- View/download PDF
15. Sphingosine 1-phosphate receptor 5 mediates the immune quiescence of the human brain endothelial barrier
- Author
-
van Doorn Ruben, Lopes Pinheiro Melissa A, Kooij Gijs, Lakeman Kim, van het Hof Bert, van der Pol Susanne MA, Geerts Dirk, van Horssen Jack, van der Valk Paul, van der Kam Elizabeth, Ronken Eric, Reijerkerk Arie, and de Vries Helga E
- Subjects
Sphingosine 1-phosphate (S1P) receptor ,FTY720P ,Blood–brain barrier ,Neuroinflammation ,Monocyte ,Multiple sclerosis ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The sphingosine 1-phosphate (S1P) receptor modulator FTY720P (Gilenya®) potently reduces relapse rate and lesion activity in the neuroinflammatory disorder multiple sclerosis. Although most of its efficacy has been shown to be related to immunosuppression through the induction of lymphopenia, it has been suggested that a number of its beneficial effects are related to altered endothelial and blood–brain barrier (BBB) functionality. However, to date it remains unknown whether brain endothelial S1P receptors are involved in the maintenance of the function of the BBB thereby mediating immune quiescence of the brain. Here we demonstrate that the brain endothelial receptor S1P5 largely contributes to the maintenance of brain endothelial barrier function. Methods We analyzed the expression of S1P5 in human post-mortem tissues using immunohistochemistry. The function of S1P5 at the BBB was assessed in cultured human brain endothelial cells (ECs) using agonists and lentivirus-mediated knockdown of S1P5. Subsequent analyses of different aspects of the brain EC barrier included the formation of a tight barrier, the expression of BBB proteins and markers of inflammation and monocyte transmigration. Results We show that activation of S1P5 on cultured human brain ECs by a selective agonist elicits enhanced barrier integrity and reduced transendothelial migration of monocytes in vitro. These results were corroborated by genetically silencing S1P5 in brain ECs. Interestingly, functional studies with these cells revealed that S1P5 strongly contributes to brain EC barrier function and underlies the expression of specific BBB endothelial characteristics such as tight junctions and permeability. In addition, S1P5 maintains the immunoquiescent state of brain ECs with low expression levels of leukocyte adhesion molecules and inflammatory chemokines and cytokines through lowering the activation of the transcription factor NFκB. Conclusion Our findings demonstrate that S1P5 in brain ECs contributes to optimal barrier formation and maintenance of immune quiescence of the barrier endothelium.
- Published
- 2012
- Full Text
- View/download PDF
16. Inflammation-induced TRPV4 channels exacerbate blood-brain barrier dysfunction in multiple sclerosis.
- Author
-
Hansen CE, Kamermans A, Mol K, Berve K, Rodriguez-Mogeda C, Fung WK, van Het Hof B, Fontijn RD, van der Pol SMA, Michalick L, Kuebler WM, Kenkhuis B, van Roon-Mom W, Liedtke W, Engelhardt B, Kooij G, Witte ME, and de Vries HE
- Subjects
- Humans, Central Nervous System metabolism, Inflammation metabolism, Blood-Brain Barrier metabolism, Multiple Sclerosis pathology, TRPV Cation Channels metabolism
- Abstract
Background: Blood-brain barrier (BBB) dysfunction and immune cell migration into the central nervous system (CNS) are pathogenic drivers of multiple sclerosis (MS). Ways to reinstate BBB function and subsequently limit neuroinflammation present promising strategies to restrict disease progression. However, to date, the molecular players directing BBB impairment in MS remain poorly understood. One suggested candidate to impact BBB function is the transient receptor potential vanilloid-type 4 ion channel (TRPV4), but its specific role in MS pathogenesis remains unclear. Here, we investigated the role of TRPV4 in BBB dysfunction in MS., Main Text: In human post-mortem MS brain tissue, we observed a region-specific increase in endothelial TRPV4 expression around mixed active/inactive lesions, which coincided with perivascular microglia enrichment in the same area. Using in vitro models, we identified that microglia-derived tumor necrosis factor-α (TNFα) induced brain endothelial TRPV4 expression. Also, we found that TRPV4 levels influenced brain endothelial barrier formation via expression of the brain endothelial tight junction molecule claudin-5. In contrast, during an inflammatory insult, TRPV4 promoted a pathological endothelial molecular signature, as evidenced by enhanced expression of inflammatory mediators and cell adhesion molecules. Moreover, TRPV4 activity mediated T cell extravasation across the brain endothelium., Conclusion: Collectively, our findings suggest a novel role for endothelial TRPV4 in MS, in which enhanced expression contributes to MS pathogenesis by driving BBB dysfunction and immune cell migration., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. PET imaging of P2X 7 R in the experimental autoimmune encephalomyelitis model of multiple sclerosis using [ 11 C]SMW139.
- Author
-
Beaino W, Janssen B, Kooijman E, Vos R, Schuit RC, O'Brien-Brown J, Kassiou M, van Het Hof B, Vugts DJ, de Vries HE, and Windhorst AD
- Subjects
- Animals, Brain diagnostic imaging, Encephalomyelitis, Autoimmune, Experimental diagnostic imaging, Female, HEK293 Cells, Humans, Male, Multiple Sclerosis chemically induced, Multiple Sclerosis diagnostic imaging, Purinergic P2X Receptor Agonists chemistry, Purinergic P2X Receptor Agonists metabolism, Rats, Rats, Inbred Lew, Rats, Wistar, Brain metabolism, Carbon Radioisotopes metabolism, Encephalomyelitis, Autoimmune, Experimental metabolism, Multiple Sclerosis metabolism, Positron-Emission Tomography methods, Receptors, Purinergic P2X7 metabolism
- Abstract
Background: Non-invasive imaging of the activation status of microglia and the ability to identify a pro- or anti-inflammatory environment can provide valuable insights not only into pathogenesis of neuro-inflammatory and neurodegenerative diseases but also the monitoring of the efficacy of immunomodulatory therapies. P2X
7 R is highly expressed on pro-inflammatory microglia and [11 C]SMW139, a specific P2X7 R tracer for positron emission tomography imaging, showed good pharmacokinetics, stability, and brain permeability in vivo. Our objective was to evaluate the potential of [11 C]SMW139 for PET imaging of neuroinflammation in vivo in the experimental autoimmune encephalomyelitis (EAE) model., Methods: We induced EAE in Lewis rats by immunization with MBP 69-88 in complete Freund's adjuvant (CFA). We determined the affinity of [11 C]SMW139 to human and rat P2X7 R using saturation binding assay. Using this tracer, PET imaging was performed at the peak of disease and in the recovery phase. In vivo blocking experiments were conducted to validate the specific brain uptake of the tracer. Immunohistochemistry staining and autoradiography were performed to evaluate the level of neuroinflammation and validate the specific binding of [11 C]SMW139., Results: [11 C]SMW139 showed good affinity for the rat P2X7 R with a Kd of 20.6 ± 1.7 nM. The uptake of [11 C]SMW139 was significantly higher in EAE animals at the peak of disease compared to the recovery phase but not in CFA control animals. The amplitude of increase of [11 C]SMW139 uptake showed significant positive correlation with clinical scores mainly in the spinal cord (Pearson = 0.75, Spearman = 0.76; p < 0.0001). Treating EAE animals with P2X7 R antagonist JNJ-47965567 blocked the uptake of [11 C]SMW139 in the spinal cord, cerebellum, and brain stem, demonstrating specific accumulation of the tracer. P-glycoprotein blocking with tariquidar (30 mg/kg) did not affect tracer penetration in the brain showing that [11 C]SMW139 is not a Pgp substrate., Conclusion: Our data shows that [11 C]SMW139 is a promising PET tracer for imaging neuroinflammation and evaluating the dynamics of pro-inflammatory microglia in the brain. This can provide crucial insights into the role of microglia in disease progression and enables the development of novel treatment strategies aimed at modulating the immune response in order to promote neuroprotection.- Published
- 2020
- Full Text
- View/download PDF
18. Specialized pro-resolving lipid mediators are differentially altered in peripheral blood of patients with multiple sclerosis and attenuate monocyte and blood-brain barrier dysfunction.
- Author
-
Kooij G, Troletti CD, Leuti A, Norris PC, Riley I, Albanese M, Ruggieri S, Libreros S, van der Pol SMA, van Het Hof B, Schell Y, Guerrera G, Buttari F, Mercuri NB, Centonze D, Gasperini C, Battistini L, de Vries HE, Serhan CN, and Chiurchiù V
- Subjects
- Blood-Brain Barrier, Eicosanoids, Humans, Inflammation, Inflammation Mediators, Monocytes, Multiple Sclerosis drug therapy
- Abstract
Chronic inflammation is a key pathological hallmark of multiple sclerosis (MS) and suggests that resolution of inflammation, orchestrated by specialized pro-resolving lipid mediators (LM), is impaired. Here, through targeted-metabololipidomics in peripheral blood of patients with MS, we revealed that each disease form was associated with distinct LM profiles that significantly correlated with disease severity. In particular, relapsing and progressive MS patients were associated with high eicosanoids levels, whereas the majority of pro-resolving LM were significantly reduced or below limits of detection and correlated with disease progression. Furthermore, we found impaired expression of several pro-resolving LM biosynthetic enzymes and receptors in blood-derived leukocytes of MS patients. Mechanistically, differentially expressed mediators like LXA
4 , LXB4 , RvD1 and PD1 reduced MS-derived monocyte activation and cytokine production, and inhibited inflammation-induced blood-brain barrier dysfunction and monocyte transendothelial migration. Altogether, these findings reveal peripheral defects in the resolution pathway in MS, suggesting pro-resolving LM as novel diagnostic biomarkers and potentially safe therapeutics., (Copyright© 2020 Ferrata Storti Foundation.)- Published
- 2020
- Full Text
- View/download PDF
19. Altered secretory and neuroprotective function of the choroid plexus in progressive multiple sclerosis.
- Author
-
Rodríguez-Lorenzo S, Ferreira Francisco DM, Vos R, van Het Hof B, Rijnsburger M, Schroten H, Ishikawa H, Beaino W, Bruggmann R, Kooij G, and de Vries HE
- Subjects
- Adrenomedullin cerebrospinal fluid, Adrenomedullin genetics, Adult, Aged, Case-Control Studies, Female, Gene Expression Profiling, Gene Ontology, Glycoproteins cerebrospinal fluid, Glycoproteins genetics, Humans, Hypoxia-Inducible Factor 1, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Intercellular Signaling Peptides and Proteins cerebrospinal fluid, Intercellular Signaling Peptides and Proteins genetics, Lateral Ventricles, Male, Metallothionein genetics, Middle Aged, Multiple Sclerosis, Chronic Progressive cerebrospinal fluid, Multiple Sclerosis, Relapsing-Remitting cerebrospinal fluid, Plasminogen Activator Inhibitor 1 cerebrospinal fluid, Plasminogen Activator Inhibitor 1 genetics, RNA, Antisense genetics, RNA, Long Noncoding, RNA-Seq, Choroid Plexus metabolism, Hypoxia genetics, Multiple Sclerosis, Chronic Progressive genetics, Multiple Sclerosis, Relapsing-Remitting genetics, Neuroprotection genetics, Neurosecretion genetics
- Abstract
The choroid plexus (CP) is a key regulator of the central nervous system (CNS) homeostasis through its secretory, immunological and barrier properties. Accumulating evidence suggests that the CP plays a pivotal role in the pathogenesis of multiple sclerosis (MS), but the underlying mechanisms remain largely elusive. To get a comprehensive view on the role of the CP in MS, we studied transcriptomic alterations of the human CP in progressive MS and non-neurological disease controls using RNA sequencing. We identified 17 genes with significantly higher expression in progressive MS patients relative to that in controls. Among them is the newly described long non-coding RNA HIF1A-AS3. Next to that, we uncovered disease-affected pathways related to hypoxia, secretion and neuroprotection, while only subtle immunological and no barrier alterations were observed. In an ex vivo CP explant model, a subset of the upregulated genes responded in a similar way to hypoxic conditions. Our results suggest a deregulation of the Hypoxia-Inducible Factor (HIF)-1 pathway in progressive MS CP. Importantly, cerebrospinal fluid levels of the hypoxia-responsive secreted peptide PAI-1 were higher in MS patients with high disability relative to those with low disability. These findings provide for the first time a complete overview of the CP transcriptome in health and disease, and suggest that the CP environment becomes hypoxic in progressive MS patients, highlighting the altered secretory and neuroprotective properties of the CP under neuropathological conditions. Together, these findings provide novel insights to target the CP and promote the secretion of neuroprotective factors into the CNS of progressive MS patients.
- Published
- 2020
- Full Text
- View/download PDF
20. Increased matrix metalloproteinases expression in tuberous sclerosis complex: modulation by microRNA 146a and 147b in vitro.
- Author
-
Broekaart DWM, van Scheppingen J, Anink JJ, Wierts L, van Het Hof B, Jansen FE, Spliet WG, van Rijen PC, Kamphuis WW, de Vries HE, Aronica E, and van Vliet EA
- Subjects
- Brain metabolism, Brain pathology, Child, Preschool, Humans, Male, Tissue Inhibitor of Metalloproteinases metabolism, Tuberous Sclerosis pathology, Tumor Cells, Cultured, Matrix Metalloproteinases metabolism, MicroRNAs metabolism, Tuberous Sclerosis metabolism
- Abstract
Aim: Matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors (TIMPs) control proteolysis within the extracellular matrix (ECM) of the brain. Dysfunction of this enzymatic system due to brain inflammation can disrupt the blood-brain barrier (BBB) and has been implicated in the pathogenesis of epilepsy. However, this has not been extensively studied in the epileptogenic human brain., Methods: We investigated the expression and cellular localization of major MMPs (MMP2, MMP3, MMP9 and MMP14) and TIMPs (TIMP1, TIMP2, TIMP3 and TIMP4) using quantitative real-time polymerase chain reaction (RT-PCR) and immunohistochemistry in resected epileptogenic brain tissue from patients with tuberous sclerosis complex (TSC), a severe neurodevelopmental disorder characterized by intractable epilepsy and prominent neuroinflammation. Furthermore, we determined whether anti-inflammatory microRNAs, miR146a and miR147b, which can regulate gene expression at the transcriptional level, could attenuate dysregulated MMP and TIMP expression in TSC tuber-derived astroglial cultures., Results: We demonstrated higher mRNA and protein expression of MMPs and TIMPs in TSC tubers compared to control and perituberal brain tissue, particularly in dysmorphic neurons and giant cells, as well as in reactive astrocytes, which was associated with BBB dysfunction. More importantly, IL-1β-induced dysregulation of MMP3, TIMP2, TIMP3 and TIMP4 could be rescued by miR146a and miR147b in tuber-derived TSC cultures., Conclusions: This study provides evidence of dysregulation of the MMP/TIMP proteolytic system in TSC, which is associated with BBB dysfunction. As dysregulated MMP and TIMP expression can be ameliorated in vitro by miR146a and miR147b, these miRNAs deserve further investigation as a novel therapeutic approach., (© 2019 The Authors. Neuropathology and Applied Neurobiology published by John Wiley & Sons Ltd on behalf of British Neuropathological Society.)
- Published
- 2020
- Full Text
- View/download PDF
21. Setmelanotide, a Novel, Selective Melanocortin Receptor-4 Agonist Exerts Anti-inflammatory Actions in Astrocytes and Promotes an Anti-inflammatory Macrophage Phenotype.
- Author
-
Kamermans A, Verhoeven T, van Het Hof B, Koning JJ, Borghuis L, Witte M, van Horssen J, de Vries HE, and Rijnsburger M
- Subjects
- Adult, Aged, Cells, Cultured, Cyclic AMP Response Element-Binding Protein metabolism, Female, Humans, Interleukin-11 biosynthesis, Interleukin-6 biosynthesis, Male, Middle Aged, Multiple Sclerosis drug therapy, Phenotype, Phosphorylation, Receptor, Melanocortin, Type 4 drug effects, Receptor, Melanocortin, Type 4 genetics, alpha-MSH pharmacology, Anti-Inflammatory Agents pharmacology, Astrocytes drug effects, Macrophages drug effects, Receptor, Melanocortin, Type 4 agonists, alpha-MSH analogs & derivatives
- Abstract
To date, available treatment strategies for multiple sclerosis (MS) are ineffective in preventing or reversing progressive neurologic deterioration, creating a high, and unmet medical need. One potential way to fight MS may be by limiting the detrimental effects of reactive astrocytes, a key pathological hallmark for disease progression. One class of compounds that may exert beneficial effects via astrocytes are melanocortin receptor (MCR) agonists. Among the MCR, MC4R is most abundantly expressed in the CNS and several rodent studies have described that MC4R is-besides neurons-expressed by astrocytes. Activation of MC4R in astrocytes has shown to have potent anti-inflammatory as well as neuroprotective effects in vitro , suggesting that this could be a potential target to ameliorate ongoing inflammation, and neurodegeneration in MS. In this study, we set out to investigate human MC4R expression and analyze its downstream effects. We identified MC4R mRNA and protein to be expressed on astrocytes and observed increased astrocytic MC4R expression in active MS lesions. Furthermore, we show that the novel, highly selective MC4R agonist setmelanotide ameliorates the reactive phenotype in astrocytes in vitro and markedly induced interleukin-6 and -11 production, possibly through enhanced cAMP response element-binding protein (CREB) phosphorylation. Notably, stimulation of human macrophages with medium from astrocytes that were exposed to setmelanotide, skewed macrophages toward an anti-inflammatory phenotype. Taken together, these findings suggest that targeting MC4R on astrocytes might be a novel therapeutic strategy to halt inflammation-associated neurodegeneration in MS., (Copyright © 2019 Kamermans, Verhoeven, van het Hof, Koning, Borghuis, Witte, van Horssen, de Vries and Rijnsburger.)
- Published
- 2019
- Full Text
- View/download PDF
22. Glycan-Modified Apoptotic Melanoma-Derived Extracellular Vesicles as Antigen Source for Anti-Tumor Vaccination
- Author
-
Horrevorts SK, Stolk DA, van de Ven R, Hulst M, van Het Hof B, Duinkerken S, Heineke MH, Ma W, Dusoswa SA, Nieuwland R, Garcia-Vallejo JJ, van de Loosdrecht AA, de Gruijl TD, van Vliet SJ, and van Kooyk Y
- Abstract
Tumors that lack T cell infiltration are less likely to respond to immune checkpoint inhibition and could benefit from cancer vaccination for the initiation of anti-tumor T cell responses. An attractive vaccine strategy is in vivo targeting of dendritic cells (DCs), key initiators of antigen-specific T cell responses. In this study we generated tumor-derived apoptotic extracellular vesicles (ApoEVs), which are potentially an abundant source of tumor-specific neo-antigens and other tumor-associated antigens (TAAs), and which can be manipulated to express DC-targeting ligands for efficient antigen delivery. Our data demonstrates that by specifically modifying the glycocalyx of tumor cells, high-mannose glycans can be expressed on their cell surface and on extracellular vesicles derived after the induction of apoptosis. High-mannose glycans are the natural ligands of dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), a dendritic cell associated C-type lectin receptor (CLR), which has the ability to efficiently internalize its cargo and direct it to both major histocompatibility complex (MHC)-I and MHC-II pathways for the induction of CD8
+ and CD4+ T cell responses, respectively. Compared to unmodified ApoEVs, ApoEVs carrying DC-SIGN ligands are internalized to a higher extent, resulting in enhanced priming of tumor-specific CD8+ T cells. This approach thus presents a promising vaccination strategy in support of T cell-based immunotherapy of cancer., Competing Interests: The authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
23. Liver X Receptor Alpha Is Important in Maintaining Blood-Brain Barrier Function.
- Author
-
Wouters E, de Wit NM, Vanmol J, van der Pol SMA, van Het Hof B, Sommer D, Loix M, Geerts D, Gustafsson JA, Steffensen KR, Vanmierlo T, Bogie JFJ, Hendriks JJA, and de Vries HE
- Subjects
- Animals, Blood-Brain Barrier pathology, Cell Line, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental pathology, Endothelial Cells pathology, Gene Knockdown Techniques, Humans, Liver X Receptors genetics, Mice, Mice, Knockout, Vascular Cell Adhesion Molecule-1 genetics, Vascular Cell Adhesion Molecule-1 immunology, Blood-Brain Barrier immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Endothelial Cells immunology, Liver X Receptors immunology
- Abstract
Dysfunction of the blood-brain barrier (BBB) contributes significantly to the pathogenesis of several neuroinflammatory diseases, including multiple sclerosis (MS). Potential players that regulate BBB function are the liver X receptors (LXRs), which are ligand activated transcription factors comprising two isoforms, LXRα, and LXRβ. However, the role of LXRα and LXRβ in regulating BBB (dys)function during neuroinflammation remains unclear, as well as their individual involvement. Therefore, the goal of the present study is to unravel whether LXR isoforms have different roles in regulating BBB function under neuroinflammatory conditions. We demonstrate that LXRα, and not LXRβ, is essential to maintain barrier integrity in vitro . Specific knockout of LXRα in brain endothelial cells resulted in a more permeable barrier with reduced expression of tight junctions. Additionally, the observed dysfunction was accompanied by increased endothelial inflammation, as detected by enhanced expression of vascular cell adhesion molecule (VCAM-1) and increased transendothelial migration of monocytes toward inflammatory stimuli. To unravel the importance of LXRα in BBB function in vivo , we made use of the experimental autoimmune encephalomyelitis (EAE) MS mouse model. Induction of EAE in a constitutive LXRα knockout mouse and in an endothelial specific LXRα knockout mouse resulted in a more severe disease score in these animals. This was accompanied by higher numbers of infiltrating leukocytes, increased endothelial VCAM-1 expression, and decreased expression of the tight junction molecule claudin-5. Together, this study reveals that LXRα is indispensable for maintaining BBB integrity and its immune quiescence. Targeting the LXRα isoform may help in the development of novel therapeutic strategies to prevent BBB dysfunction, and thereby neuroinflammatory disorders.
- Published
- 2019
- Full Text
- View/download PDF
24. Inflammation-induced endothelial to mesenchymal transition promotes brain endothelial cell dysfunction and occurs during multiple sclerosis pathophysiology.
- Author
-
Derada Troletti C, Fontijn RD, Gowing E, Charabati M, van Het Hof B, Didouh I, van der Pol SMA, Geerts D, Prat A, van Horssen J, Kooij G, and de Vries HE
- Subjects
- Cells, Cultured, Endothelial Cells metabolism, Epithelial-Mesenchymal Transition, Humans, Brain physiopathology, Inflammation complications, Multiple Sclerosis genetics, Multiple Sclerosis physiopathology
- Abstract
The blood-brain barrier (BBB) has a major role in maintaining brain homeostasis through the specialized function of brain endothelial cells (BECs). Inflammation of the BECs and loss of their neuroprotective properties is associated with several neurological disorders, including the chronic neuro-inflammatory disorder multiple sclerosis (MS). Yet, the underlying mechanisms of a defective BBB in MS remain largely unknown. Endothelial to mesenchymal transition (EndoMT) is a pathophysiological process in which endothelial cells lose their specialized function and de-differentiate into mesenchymal cells. This transition is characterized by an increase in EndoMT-related transcription factors (TFs), a downregulation of brain endothelial markers, and an upregulation of mesenchymal markers accompanied by morphological changes associated with cytoskeleton reorganization. Here, we postulate that EndoMT drives BEC de-differentiation, mediates inflammation-induced human BECs dysfunction, and may play a role in MS pathophysiology. We provide evidence that stimulation of human BECs with transforming growth factor (TGF)-β1 and interleukin (IL)-1β promotes EndoMT, a process in which the TF SNAI1, a master regulator of EndoMT, plays a crucial role. We demonstrate the involvement of TGF-β activated kinase 1 (TAK1) in EndoMT induction in BECs. Finally, immunohistochemical analysis revealed EndoMT-associated alterations in the brain vasculature of human post-mortem MS brain tissues. Taken together, our novel findings provide a better understanding of the molecular mechanisms underlying BECs dysfunction during MS pathology and can be used to develop new potential therapeutic strategies to restore BBB function.
- Published
- 2019
- Full Text
- View/download PDF
25. Angiopoietin like-4 as a novel vascular mediator in capillary cerebral amyloid angiopathy.
- Author
-
Chakraborty A, Kamermans A, van Het Hof B, Castricum K, Aanhane E, van Horssen J, Thijssen VL, Scheltens P, Teunissen CE, Fontijn RD, van der Flier WM, and de Vries HE
- Subjects
- Aged, Aged, 80 and over, Brain blood supply, Brain metabolism, Brain pathology, Cell Hypoxia, Cell Movement, Endothelial Cells metabolism, Female, Humans, Male, Microvessels pathology, Vascular Remodeling, Angiopoietin-Like Protein 4 metabolism, Astrocytes metabolism, Cerebral Amyloid Angiopathy metabolism
- Abstract
Increasing evidence suggests that vascular dysfunction in the brain is associated with early stages of Alzheimer's disease. Amyloid-β deposition in the microvasculature of the brain, a process referred to as capillary cerebral amyloid angiopathy (capillary CAA), propagates vascular remodelling, which results in impaired function of the blood-brain barrier, reduced cerebral perfusion and increased hypoxia. While improving vascular function may be an attractive new way to fight capillary CAA, the underlying factors that mediate vascular alterations in Alzheimer's disease and capillary CAA pathogenesis remain largely unknown. Here we provide first evidence that angiopoietin like-4 (ANGPTL4), a hypoxia-induced factor, is highly expressed by reactive astrocytes in well characterized post-mortem tissues of patients with capillary CAA. Our in vitro studies reveal that ANGPTL4 is upregulated and secreted by human cortical astrocytes under hypoxic conditions and in turn stimulates endothelial cell migration and sprouting in a 3D spheroid model of human brain endothelial cells. Interestingly, plasma levels of ANGPTL4 are significantly increased in patients with vascular dementia compared to patients with subjective memory complaints. Overall, our data suggest that ANGPTL4 contributes to pathological vascular remodelling in capillary CAA and that detection of ANGPTL4 levels may improve current diagnostics. Ways of counteracting the detrimental effects of ANGPTL4 and thus promoting cerebral vascular function may provide novel treatment regimens to halt the progression of Alzheimer's disease.
- Published
- 2018
- Full Text
- View/download PDF
26. Notch signaling is impaired during inflammation in a Lunatic Fringe-dependent manner.
- Author
-
Derada Troletti C, Lopes Pinheiro MA, Charabati M, Gowing E, van Het Hof B, van der Pol SMA, Geerts D, Prat A, Fontijn RD, Unger WW, and de Vries HE
- Subjects
- Brain metabolism, Cell Line, Cell Survival physiology, Glycosylation, Humans, Permeability, Blood-Brain Barrier metabolism, Endothelial Cells metabolism, Glycosyltransferases metabolism, Inflammation metabolism, Receptors, Notch metabolism, Signal Transduction physiology
- Abstract
The blood-brain barrier (BBB) assures brain homeostasis through the specialized function of brain endothelial cells (BECs). Dysfunction of the BBB due to inflammatory processes is associated with several neurological disorders, including multiple sclerosis (MS). Understanding the mechanisms that underlie these processes may ultimately lead to new therapeutic strategies to restore BBB function, thereby fighting disease progression. In this study, we demonstrate for the first time a critical role of the Notch signaling pathway in the function of the BBB under resting and inflammatory conditions. Inhibition of the Notch signaling, either by a γ-secretase inhibitor or by genetic ablation of endothelial NOTCH, led to BBB dysfunction in vitro as evidenced by reduced transendothelial electrical resistance (TEER), altered localization and loss of endothelial junction molecules and enhanced macromolecular permeability. Inflamed BECs showed impaired Notch signaling as indicated by reduced level of the downstream targets HES-1 and HES-5. Notably, barrier function was further reduced when the Notch signaling was inhibited under inflammatory conditions, suggesting an additive effect of the Notch signaling and inflammation in BECs. In contrast, inducible overexpression of Notch-intracellular domain 1 (NICD1) rescued the detrimental effect caused by inflammation. Furthermore, we provide evidence that inflammation reduced the expression of the glycosyltransferase Lunatic Fringe (LFNG), a known positive regulator of Notch glycosylation and signaling, thereby leading to disrupted barrier function of BECs. Together, our data demonstrate the functional importance of the conserved Notch signaling pathway in control of the brain endothelial barrier and shed light on the role of LFNG in the regulation of Notch glycosylation and signaling in the adult brain vasculature in both health and disease., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Internalization and presentation of myelin antigens by the brain endothelium guides antigen-specific T cell migration.
- Author
-
Lopes Pinheiro MA, Kamermans A, Garcia-Vallejo JJ, van Het Hof B, Wierts L, O'Toole T, Boeve D, Verstege M, van der Pol SM, van Kooyk Y, de Vries HE, and Unger WW
- Subjects
- CD4-Positive T-Lymphocytes physiology, Cells, Cultured, Endocytosis, Endothelium metabolism, Histocompatibility Antigens Class II metabolism, Humans, Antigen Presentation, Antigens immunology, Brain pathology, CD4-Positive T-Lymphocytes immunology, Cell Movement, Endothelium immunology, Myelin Sheath immunology
- Abstract
Trafficking of myelin-reactive CD4(+) T-cells across the brain endothelium, an essential step in the pathogenesis of multiple sclerosis (MS), is suggested to be an antigen-specific process, yet which cells provide this signal is unknown. Here we provide direct evidence that under inflammatory conditions, brain endothelial cells (BECs) stimulate the migration of myelin-reactive CD4(+) T-cells by acting as non-professional antigen presenting cells through the processing and presentation of myelin-derived antigens in MHC-II. Inflamed BECs internalized myelin, which was routed to endo-lysosomal compartment for processing in a time-dependent manner. Moreover, myelin/MHC-II complexes on inflamed BECs stimulated the trans-endothelial migration of myelin-reactive Th1 and Th17 2D2 cells, while control antigen loaded BECs did not stimulate T-cell migration. Furthermore, blocking the interaction between myelin/MHC-II complexes and myelin-reactive T-cells prevented T-cell transmigration. These results demonstrate that endothelial cells derived from the brain are capable of enhancing antigen-specific T cell recruitment.
- Published
- 2016
- Full Text
- View/download PDF
28. Protective effects of monomethyl fumarate at the inflamed blood-brain barrier.
- Author
-
Lim JL, van der Pol SM, Di Dio F, van Het Hof B, Kooij G, de Vries HE, and van Horssen J
- Subjects
- Blood-Brain Barrier metabolism, Blood-Brain Barrier pathology, Cell Adhesion drug effects, Cell Adhesion Molecules metabolism, Cells, Cultured, Coculture Techniques, Cytoprotection, Endothelial Cells metabolism, Endothelial Cells pathology, Heme Oxygenase-1 metabolism, Humans, Leukocytes metabolism, Leukocytes pathology, Multiple Sclerosis metabolism, Multiple Sclerosis pathology, NAD(P)H Dehydrogenase (Quinone) metabolism, NF-E2-Related Factor 2 metabolism, NF-kappa B metabolism, Anti-Inflammatory Agents pharmacology, Antioxidants pharmacology, Blood-Brain Barrier drug effects, Endothelial Cells drug effects, Fumarates pharmacology, Leukocytes drug effects, Maleates pharmacology, Multiple Sclerosis drug therapy, Transendothelial and Transepithelial Migration drug effects
- Abstract
Background: Reactive oxygen species play a key role in the pathogenesis of multiple sclerosis as they induce blood-brain barrier disruption and enhance transendothelial leukocyte migration. Thus, therapeutic compounds with antioxidant and anti-inflammatory potential could have clinical value in multiple sclerosis. The aim of the current study was to elucidate the therapeutic effects of monomethyl fumarate on inflammatory-mediated changes in blood-brain barrier function and gain insight into the underlying mechanism., Methods: The effects of monomethyl fumarate on monocyte transendothelial migration across and adhesion to inflamed human brain endothelial cells (hCMEC/D3) were quantified using standardized in vitro migration and adhesion assays. Flow cytometry analysis and qPCR were used to measure the concomitant effects of monomethyl fumarate treatment on protein expression of cell adhesion molecules. Furthermore, the effects of monomethyl fumarate on the expression and nuclear localization of proteins involved in the activation of antioxidant and inflammatory pathways in human brain endothelial cells were elucidated using nuclear fractionation and Western blotting. Statistical analysis was performed using one-way ANOVA followed by the Bonferroni post-hoc test., Results: Our results show that monomethyl fumarate induced nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 and concomitant production of the antioxidant enzymes heme oxygenase-1 and NADPH:quinone oxidoreductase-1 in brain endothelial cells. Importantly, monomethyl fumarate treatment markedly decreased monocyte transendothelial migration across and adhesion to inflamed human brain endothelial cells. Treatment of brain endothelial cells with monomethyl fumarate resulted in a striking reduction of vascular cell adhesion molecule expression. Surprisingly, monomethyl fumarate did not affect nuclear translocation of nuclear factor-кB suggesting that monomethyl fumarate potentially affects activity of nuclear factor-ĸB downstream of nuclear translocation., Conclusions: Taken together, we show that monomethyl fumarate, the primary metabolite of dimethyl fumarate, which is currently used in the clinics for the treatment of relapsing-remitting multiple sclerosis, demonstrates beneficial therapeutic effects at the inflamed blood-brain barrier., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Acid Sphingomyelinase-Derived Ceramide Regulates ICAM-1 Function during T Cell Transmigration across Brain Endothelial Cells.
- Author
-
Lopes Pinheiro MA, Kroon J, Hoogenboezem M, Geerts D, van Het Hof B, van der Pol SM, van Buul JD, and de Vries HE
- Subjects
- Adult, Aged, Aged, 80 and over, Brain cytology, Brain immunology, Cell Adhesion genetics, Cell Adhesion immunology, Cell Line, Ceramides metabolism, Cytoskeletal Proteins metabolism, Endothelial Cells immunology, Endothelial Cells metabolism, Female, Filamins metabolism, Humans, Intercellular Adhesion Molecule-1 biosynthesis, Intercellular Adhesion Molecule-1 immunology, Male, Middle Aged, Multiple Sclerosis immunology, Phosphorylation genetics, Sphingomyelin Phosphodiesterase genetics, Sphingomyelin Phosphodiesterase immunology, Brain metabolism, Intercellular Adhesion Molecule-1 metabolism, Sphingomyelin Phosphodiesterase metabolism, T-Lymphocytes immunology, Transendothelial and Transepithelial Migration immunology
- Abstract
Multiple sclerosis (MS) is a chronic demyelinating disorder of the CNS characterized by immune cell infiltration across the brain vasculature into the brain, a process not yet fully understood. We previously demonstrated that the sphingolipid metabolism is altered in MS lesions. In particular, acid sphingomyelinase (ASM), a critical enzyme in the production of the bioactive lipid ceramide, is involved in the pathogenesis of MS; however, its role in the brain vasculature remains unknown. Transmigration of T lymphocytes is highly dependent on adhesion molecules in the vasculature such as intercellular adhesion molecule-1 (ICAM-1). In this article, we hypothesize that ASM controls T cell migration by regulating ICAM-1 function. To study the role of endothelial ASM in transmigration, we generated brain endothelial cells lacking ASM activity using a lentiviral shRNA approach. Interestingly, although ICAM-1 expression was increased in cells lacking ASM activity, we measured a significant decrease in T lymphocyte adhesion and consequently transmigration both in static and under flow conditions. As an underlying mechanism, we revealed that upon lack of endothelial ASM activity, the phosphorylation of ezrin was perturbed as well as the interaction between filamin and ICAM-1 upon ICAM-1 clustering. Functionally this resulted in reduced microvilli formation and impaired transendothelial migration of T cells. In conclusion, in this article, we show that ASM coordinates ICAM-1 function in brain endothelial cells by regulating its interaction with filamin and phosphorylation of ezrin. The understanding of these underlying mechanisms of T lymphocyte transmigration is of great value to develop new strategies against MS lesion formation., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
30. Astroglial PGC-1alpha increases mitochondrial antioxidant capacity and suppresses inflammation: implications for multiple sclerosis.
- Author
-
Nijland PG, Witte ME, van het Hof B, van der Pol S, Bauer J, Lassmann H, van der Valk P, de Vries HE, and van Horssen J
- Subjects
- Adult, Aged, Antioxidants metabolism, Case-Control Studies, Cells, Cultured, Cytokines genetics, Cytokines metabolism, Female, Gene Expression Regulation, Humans, Male, Middle Aged, Multiple Sclerosis pathology, Myelin Proteolipid Protein metabolism, Oxidative Stress physiology, Peroxiredoxin III genetics, Peroxiredoxin III metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Reactive Oxygen Species, Thioredoxins genetics, Thioredoxins metabolism, Transcription Factors genetics, White Matter metabolism, White Matter pathology, Astrocytes metabolism, Brain metabolism, Encephalitis etiology, Encephalitis pathology, Multiple Sclerosis complications, Transcription Factors metabolism
- Abstract
Recent evidence suggests that reactive oxygen species (ROS) produced by inflammatory cells drive axonal degeneration in active multiple sclerosis (MS) lesions by inducing mitochondrial dysfunction. Mitochondria are endowed with a variety of antioxidant enzymes, including peroxiredoxin-3 and thioredoxin-2, which are involved in limiting ROS-induced damage. In this study, we explored the distribution and role of the mitochondrial antioxidants peroxiredoxin-3 and thioredoxin-2 as well as their regulator peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α) in MS pathogenesis. Immunohistochemical analysis of a large cohort of MS patients revealed a striking upregulation of PGC-1α and downstream mitochondrial antioxidants in active demyelinating MS lesions. Enhanced expression was predominantly observed in reactive astrocytes. To elucidate the functional role of astrocytic PGC-1α in MS pathology, we generated human primary astrocytes that genetically overexpressed PGC-1α. Upon an oxidative insult, these cells were shown to produce less ROS and were found to be more resistant to ROS-induced cell death compared to control cells. Intriguingly, also neuronal cells co-cultured with PGC-1α-overexpressing astrocytes were protected against an exogenous oxidative attack compared to neuronal cells co-cultured with control astrocytes. Finally, enhanced astrocytic PGC-1α levels markedly reduced the production and secretion of the pro-inflammatory mediators interleukin-6 and chemokine (C-C motif) ligand 2. Our findings suggest that increased astrocytic PGC-1α in active MS lesions might initially function as an endogenous protective mechanism to dampen oxidative damage and inflammation thereby reducing neurodegeneration. Activation of PGC-1α therefore represents a promising therapeutic strategy to improve mitochondrial function and repress inflammation.
- Published
- 2014
- Full Text
- View/download PDF
31. Astrocyte-derived retinoic acid: a novel regulator of blood-brain barrier function in multiple sclerosis.
- Author
-
Mizee MR, Nijland PG, van der Pol SM, Drexhage JA, van Het Hof B, Mebius R, van der Valk P, van Horssen J, Reijerkerk A, and de Vries HE
- Subjects
- Adult, Aged, Aged, 80 and over, Aldehyde Dehydrogenase genetics, Aldehyde Dehydrogenase metabolism, Aldehyde Dehydrogenase 1 Family, Astrocytes metabolism, Autopsy, Cells, Cultured, Cytokines metabolism, Endothelial Cells drug effects, Endothelial Cells physiology, Female, Glial Fibrillary Acidic Protein metabolism, HEK293 Cells, Humans, Male, Middle Aged, NF-E2-Related Factor 2 genetics, NF-E2-Related Factor 2 metabolism, Reactive Oxygen Species metabolism, Retinal Dehydrogenase genetics, Retinal Dehydrogenase metabolism, Time Factors, Astrocytes drug effects, Blood-Brain Barrier physiopathology, Brain pathology, Multiple Sclerosis pathology, Tretinoin pharmacology
- Abstract
Multiple sclerosis (MS) lesions are characterized by the presence of activated astrocytes, which are thought to actively take part in propagating lesion progression by secreting pro-inflammatory mediators. Conversely, reactive astrocytes may exert disease-dampening effects through the production of trophic factors and anti-inflammatory mediators. Astrocytic control of the blood-brain barrier (BBB) is crucial for normal brain homeostasis and BBB disruption is a well-established early event in MS lesion development. Here, we set out to unravel potential protective effects of reactive astrocytes on BBB function under neuroinflammatory conditions as seen in MS, where we focus on the role of the brain morphogen retinoic acid (RA). Immunohistochemical analysis revealed that retinaldehyde dehydrogenase 2 (RALDH2), a key enzyme for RA synthesis, is highly expressed by reactive astrocytes throughout white matter lesions compared to control and normal appearing white matter. In vitro modeling of reactive astrocytes resulted in increased expression of RALDH2, enhanced RA synthesis, and a protective role for astrocyte-derived RA on BBB function during inflammation-induced barrier loss. Furthermore, RA induces endothelial immune quiescence and decreases monocyte adhesion under inflammatory conditions. Finally, we demonstrated that RA attenuated oxidative stress in inflamed endothelial cells, through activation of the antioxidant transcription factor nuclear factor E2 related factor 2. In summary, RA synthesis by reactive astrocytes represents an endogenous protective response to neuroinflammation, possibly aimed at protecting the BBB against inflammatory insult. A better understanding of RA signaling in MS pathophysiology may lead to the discovery of novel targets to halt disease progression.
- Published
- 2014
- Full Text
- View/download PDF
32. Disturbed function of the blood-cerebrospinal fluid barrier aggravates neuro-inflammation.
- Author
-
Kooij G, Kopplin K, Blasig R, Stuiver M, Koning N, Goverse G, van der Pol SM, van Het Hof B, Gollasch M, Drexhage JA, Reijerkerk A, Meij IC, Mebius R, Willnow TE, Müller D, Blasig IE, and de Vries HE
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Brain blood supply, Brain pathology, Brain physiopathology, Choroid Plexus pathology, Claudin-3 genetics, Disease Progression, Encephalomyelitis, Autoimmune, Experimental pathology, Female, Humans, Male, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Microvessels pathology, Microvessels physiopathology, Middle Aged, Multiple Sclerosis pathology, Myelin-Oligodendrocyte Glycoprotein, Peptide Fragments, Severity of Illness Index, Choroid Plexus physiopathology, Claudin-3 metabolism, Encephalomyelitis, Autoimmune, Experimental physiopathology, Multiple Sclerosis physiopathology
- Abstract
Multiple sclerosis (MS) is a chronic neuro-inflammatory disorder, which is marked by the invasion of the central nervous system by monocyte-derived macrophages and autoreactive T cells across the brain vasculature. Data from experimental animal models recently implied that the passage of leukocytes across the brain vasculature is preceded by their traversal across the blood-cerebrospinal fluid barrier (BCSFB) of the choroid plexus. The correlation between the presence of leukocytes in the CSF of patients suffering from MS and the number of inflammatory lesions as detected by magnetic resonance imaging suggests that inflammation at the choroid plexus contributes to the disease, although in a yet unknown fashion. We here provide first insights into the involvement of the choroid plexus in the onset and severity of the disease and in particular address the role of the tight junction protein claudin-3 (CLDN3) in this process. Detailed analysis of human post-mortem brain tissue revealed a selective loss of CLDN3 at the choroid plexus in MS patients compared to control tissues. Importantly, mice that lack CLDN3 have an impaired BCSFB and experience a more rapid onset and exacerbated clinical signs of experimental autoimmune encephalomyelitis, which coincides with enhanced levels of infiltrated leukocytes in their CSF. Together, this study highlights a profound role for the choroid plexus in the pathogenesis of multiple sclerosis, and implies that CLDN3 may be regarded as a crucial and novel determinant of BCSFB integrity.
- Published
- 2014
- Full Text
- View/download PDF
33. Cellular distribution of glucose and monocarboxylate transporters in human brain white matter and multiple sclerosis lesions.
- Author
-
Nijland PG, Michailidou I, Witte ME, Mizee MR, van der Pol SM, van Het Hof B, Reijerkerk A, Pellerin L, van der Valk P, de Vries HE, and van Horssen J
- Subjects
- Adult, Aged, Aged, 80 and over, Astrocytes metabolism, Astrocytes pathology, Axons metabolism, Axons pathology, Brain blood supply, Brain pathology, Cell Line, Endothelial Cells metabolism, Endothelial Cells pathology, Female, Glucose Transporter Type 3 metabolism, Humans, Leukocytes metabolism, Leukocytes pathology, Male, Microglia metabolism, Microglia pathology, Middle Aged, Multiple Sclerosis pathology, Multiple Sclerosis, Chronic Progressive metabolism, Multiple Sclerosis, Chronic Progressive pathology, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Transcription Factors metabolism, White Matter blood supply, White Matter pathology, Brain metabolism, Glutamate Plasma Membrane Transport Proteins metabolism, Monocarboxylic Acid Transporters metabolism, Multiple Sclerosis metabolism, White Matter metabolism
- Abstract
To ensure efficient energy supply to the high demanding brain, nutrients are transported into brain cells via specific glucose (GLUT) and monocarboxylate transporters (MCT). Mitochondrial dysfunction and altered glucose metabolism are thought to play an important role in the progression of neurodegenerative diseases, including multiple sclerosis (MS). Here, we investigated the cellular localization of key GLUT and MCT proteins in human brain tissue of non-neurological controls and MS patients. We show that in control brain tissue GLUT and MCT proteins were abundantly expressed in a variety of central nervous system cells, particularly in microglia and endothelial cells. In active MS lesions, GLUTs and MCTs were highly expressed in infiltrating leukocytes and reactive astrocytes. Astrocytes manifest increased MCT1 staining and maintain GLUT expression in inactive lesions, whereas demyelinated axons exhibit significantly reduced GLUT3 and MCT2 immunoreactivity in inactive lesions. Finally, we demonstrated that the co-transcription factor peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α), an important protein involved in energy metabolism, is highly expressed in reactive astrocytes in active MS lesions. Overexpression of PGC-1α in astrocyte-like cells resulted in increased production of several GLUT and MCT proteins. In conclusion, we provide for the first time a comprehensive overview of key nutrient transporters in white matter brain samples. Moreover, our data demonstrate an altered expression of these nutrient transporters in MS brain tissue, including a marked reduction of axonal GLUT3 and MCT2 expression in chronic lesions, which may impede efficient nutrient supply to the hypoxic demyelinated axons thereby contributing to the ongoing neurodegeneration in MS., (Copyright © 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
34. P-glycoprotein regulates trafficking of CD8(+) T cells to the brain parenchyma.
- Author
-
Kooij G, Kroon J, Paul D, Reijerkerk A, Geerts D, van der Pol SM, van Het Hof B, Drexhage JA, van Vliet SJ, Hekking LH, van Buul JD, Pachter JS, and de Vries HE
- Subjects
- ATP Binding Cassette Transporter, Subfamily B genetics, Animals, Blood-Brain Barrier physiology, Brain blood supply, Brain pathology, CD4-Positive T-Lymphocytes physiology, Cell Line, Chemokine CCL2 genetics, Chemokine CCL2 metabolism, Encephalomyelitis, Autoimmune, Experimental pathology, Female, Humans, Mice, Inbred C57BL, Mice, Knockout, Microvessels pathology, Microvessels physiopathology, Multiple Sclerosis pathology, ATP-Binding Cassette Sub-Family B Member 4, ATP Binding Cassette Transporter, Subfamily B metabolism, Brain immunology, CD8-Positive T-Lymphocytes physiology, Encephalomyelitis, Autoimmune, Experimental immunology, Multiple Sclerosis immunology, Transendothelial and Transepithelial Migration physiology
- Abstract
The trafficking of cytotoxic CD8(+) T lymphocytes across the lining of the cerebral vasculature is key to the onset of the chronic neuro-inflammatory disorder multiple sclerosis. However, the mechanisms controlling their final transmigration across the brain endothelium remain unknown. Here, we describe that CD8(+) T lymphocyte trafficking into the brain is dependent on the activity of the brain endothelial adenosine triphosphate-binding cassette transporter P-glycoprotein. Silencing P-glycoprotein activity selectively reduced the trafficking of CD8(+) T cells across the brain endothelium in vitro as well as in vivo. In response to formation of the T cell-endothelial synapse, P-glycoprotein was found to regulate secretion of endothelial (C-C motif) ligand 2 (CCL2), a chemokine that mediates CD8(+) T cell migration in vitro. Notably, CCL2 levels were significantly enhanced in microvessels isolated from human multiple sclerosis lesions in comparison with non-neurological controls. Endothelial cell-specific elimination of CCL2 in mice subjected to experimental autoimmune encephalomyelitis also significantly diminished the accumulation of CD8(+) T cells compared to wild-type animals. Collectively, these results highlight a novel (patho)physiological role for P-glycoprotein in CD8(+) T cell trafficking into the central nervous system during neuro-inflammation and illustrate CCL2 secretion as a potential link in this mechanism.
- Published
- 2014
- Full Text
- View/download PDF
35. MicroRNAs regulate human brain endothelial cell-barrier function in inflammation: implications for multiple sclerosis.
- Author
-
Reijerkerk A, Lopez-Ramirez MA, van Het Hof B, Drexhage JA, Kamphuis WW, Kooij G, Vos JB, van der Pouw Kraan TC, van Zonneveld AJ, Horrevoets AJ, Prat A, Romero IA, and de Vries HE
- Subjects
- Blood-Brain Barrier drug effects, Cell Line, Transformed, Cytokines metabolism, Endothelial Cells drug effects, Endothelial Cells metabolism, Gene Expression Regulation drug effects, Genetic Vectors physiology, Humans, MicroRNAs genetics, RNA, Small Interfering pharmacology, Transendothelial and Transepithelial Migration drug effects, Transfection, Blood-Brain Barrier physiopathology, Brain pathology, Endothelial Cells physiology, Inflammation pathology, MicroRNAs metabolism, Multiple Sclerosis pathology
- Abstract
Blood-brain barrier (BBB) dysfunction is a major hallmark of many neurological diseases, including multiple sclerosis (MS). Using a genomics approach, we defined a microRNA signature that is diminished at the BBB of MS patients. In particular, miR-125a-5p is a key regulator of brain endothelial tightness and immune cell efflux. Our findings suggest that repair of a disturbed BBB through microRNAs may represent a novel avenue for effective treatment of MS.
- Published
- 2013
- Full Text
- View/download PDF
36. Reduced expression of PGC-1α partly underlies mitochondrial changes and correlates with neuronal loss in multiple sclerosis cortex.
- Author
-
Witte ME, Nijland PG, Drexhage JA, Gerritsen W, Geerts D, van Het Hof B, Reijerkerk A, de Vries HE, van der Valk P, and van Horssen J
- Subjects
- Adult, Aged, Aged, 80 and over, Blotting, Western, Cell Count, Down-Regulation, Female, Genetic Vectors, Gyrus Cinguli pathology, Heat-Shock Proteins biosynthesis, Heat-Shock Proteins genetics, Humans, Immunohistochemistry, Lentivirus genetics, Male, Middle Aged, Oxidation-Reduction, Oxidative Phosphorylation, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Pyramidal Cells pathology, RNA, Small Interfering genetics, Reactive Oxygen Species metabolism, Real-Time Polymerase Chain Reaction, Tissue Banks, Transcription Factors biosynthesis, Transcription Factors genetics, Cerebral Cortex pathology, Heat-Shock Proteins physiology, Mitochondria pathology, Multiple Sclerosis genetics, Multiple Sclerosis pathology, Neurons pathology, Transcription Factors physiology
- Abstract
There is growing evidence that mitochondrial dysfunction and associated reactive oxygen species (ROS) formation contribute to neurodegenerative processes in multiple sclerosis (MS). Here, we investigated whether alterations in transcriptional regulators of key mitochondrial proteins underlie mitochondrial dysfunction in MS cortex and contribute to neuronal loss. Hereto, we analyzed the expression of mitochondrial transcriptional (co-)factors and proteins involved in mitochondrial redox balance regulation in normal-appearing grey matter (NAGM) samples of cingulate gyrus and/or frontal cortex from 15 MS patients and nine controls matched for age, gender and post-mortem interval. PGC-1α, a transcriptional co-activator and master regulator of mitochondrial function, was consistently and significantly decreased in pyramidal neurons in the deeper layers of MS cortex. Reduced PGC-1α levels coincided with reduced expression of oxidative phosphorylation subunits and a decrease in gene and protein expression of various mitochondrial antioxidants and uncoupling proteins (UCPs) 4 and 5. Short-hairpin RNA-mediated silencing of PGC-1α in a neuronal cell line confirmed that reduced levels of PGC-1α resulted in a decrease in transcription of OxPhos subunits, mitochondrial antioxidants and UCPs. Moreover, PGC-1α silencing resulted in a decreased mitochondrial membrane potential, increased ROS formation and enhanced susceptibility to ROS-induced cell death. Importantly, we found extensive neuronal loss in NAGM from cingulate gyrus and frontal cortex of MS patients, which significantly correlated with the extent of PGC-1α decrease. Taken together, our data indicate that reduced neuronal PGC-1α expression in MS cortex partly underlies mitochondrial dysfunction in MS grey matter and thereby contributes to neurodegeneration in MS cortex.
- Published
- 2013
- Full Text
- View/download PDF
37. Fingolimod attenuates ceramide-induced blood-brain barrier dysfunction in multiple sclerosis by targeting reactive astrocytes.
- Author
-
van Doorn R, Nijland PG, Dekker N, Witte ME, Lopes-Pinheiro MA, van het Hof B, Kooij G, Reijerkerk A, Dijkstra C, van van der Valk P, van Horssen J, and de Vries HE
- Subjects
- Adult, Aged, Aged, 80 and over, Astrocytes metabolism, Astrocytes pathology, Blood-Brain Barrier pathology, Blood-Brain Barrier physiopathology, Cell Movement drug effects, Cells, Cultured, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelial Cells pathology, Female, Fingolimod Hydrochloride, Humans, Male, Middle Aged, Monocytes drug effects, Monocytes metabolism, Monocytes pathology, Multiple Sclerosis metabolism, Multiple Sclerosis pathology, Sphingomyelins metabolism, Sphingosine pharmacology, Astrocytes drug effects, Blood-Brain Barrier drug effects, Ceramides metabolism, Immunosuppressive Agents pharmacology, Multiple Sclerosis physiopathology, Propylene Glycols pharmacology, Sphingosine analogs & derivatives
- Abstract
Alterations in sphingolipid metabolism are described to contribute to various neurological disorders. We here determined the expression of enzymes involved in the sphingomyelin cycle and their products in postmortem brain tissue of multiple sclerosis (MS) patients. In parallel, we investigated the effect of the sphingosine-1 receptor agonist Fingolimod (Gilenya(®)) on sphingomyelin metabolism in reactive astrocytes and determined its functional consequences for the process of neuro-inflammation. Our results demonstrate that in active MS lesions, marked by large number of infiltrated immune cells, an altered expression of enzymes involved in the sphingomyelin cycle favors enhanced ceramide production. We identified reactive astrocytes as the primary cellular source of enhanced ceramide production in MS brain samples. Astrocytes isolated from MS lesions expressed enhanced mRNA levels of the ceramide-producing enzyme acid sphingomyelinase (ASM) compared to astrocytes isolated from control white matter. In addition, TNF-α treatment induced ASM mRNA and ceramide levels in astrocytes isolated from control white matter. Incubation of astrocytes with Fingolimod prior to TNF-α treatment reduced ceramide production and mRNA expression of ASM to control levels in astrocytes. Importantly, supernatants derived from reactive astrocytes treated with Fingolimod significantly reduced transendothelial monocyte migration. Overall, the present study demonstrates that reactive astrocytes represent a possible additional cellular target for Fingolimod in MS by directly reducing the production of pro-inflammatory lipids and limiting subsequent transendothelial leukocyte migration.
- Published
- 2012
- Full Text
- View/download PDF
38. Myc-associated zinc finger protein (MAZ) is regulated by miR-125b and mediates VEGF-induced angiogenesis in glioblastoma.
- Author
-
Smits M, Wurdinger T, van het Hof B, Drexhage JA, Geerts D, Wesseling P, Noske DP, Vandertop WP, de Vries HE, and Reijerkerk A
- Subjects
- Coculture Techniques, DNA-Binding Proteins biosynthesis, Down-Regulation, Endothelial Cells metabolism, Glioblastoma metabolism, HEK293 Cells, Humans, Transcription Factors biosynthesis, Tumor Cells, Cultured, DNA-Binding Proteins genetics, Glioblastoma blood supply, MicroRNAs physiology, Neovascularization, Pathologic pathology, Transcription Factors genetics, Vascular Endothelial Growth Factor A physiology
- Abstract
In patients with glioblastomas, vascular endothelial growth factor (VEGF) is a key mediator of tumor-associated angiogenesis. Glioblastomas are notorious for their capacity to induce neovascularization, driving continued tumor growth. Here we report that miR-125b is down-regulated in glioblastoma-associated endothelial cells, resulting in increased expression of its target, myc-associated zinc finger protein (MAZ), a transcription factor that regulates VEGF. The down-regulation of miR-125b was also observed on exposure of endothelial cells to glioblastoma-conditioned medium or VEGF, resulting in increased MAZ expression. Further analysis revealed that inhibition of MAZ accumulation by miR-125b, or by MAZ-specific shRNAs, attenuated primary human brain endothelial cell migration and tubule formation in vitro, phenomena considered to mimick angiogenic processes in vitro. Moreover, MAZ expression was elevated in brain blood vessels of glioblastoma patients. Altogether these results demonstrate a functional feed-forward loop in glioblastoma-related angiogenesis, in which VEGF inhibits the expression of miR-125b, resulting in increased expression of MAZ, which in its turn causes transcriptional activation of VEGF. This loop is functionally impeded by the VEGF receptor inhibitor vandetanib, and our results may contribute to the further development of inhibitors of tumor-angiogenesis.
- Published
- 2012
- Full Text
- View/download PDF
39. Brain endothelial barrier passage by monocytes is controlled by the endothelin system.
- Author
-
Reijerkerk A, Lakeman KA, Drexhage JA, van Het Hof B, van Wijck Y, van der Pol SM, Kooij G, Geerts D, and de Vries HE
- Subjects
- Aspartic Acid Endopeptidases metabolism, Blotting, Western, Cell Line, Endothelin-Converting Enzymes, Gene Knockdown Techniques, Humans, Immunohistochemistry, Metalloendopeptidases metabolism, Receptors, Endothelin metabolism, Blood-Brain Barrier metabolism, Endothelial Cells metabolism, Endothelins metabolism, Monocytes cytology, Transendothelial and Transepithelial Migration physiology
- Abstract
Homeostasis of the brain is dependent on the blood-brain barrier (BBB). This barrier tightly regulates the exchange of essential nutrients and limits the free flow of immune cells into the CNS. Perturbations of BBB function and the loss of its immune quiescence are hallmarks of a variety of brain diseases, including multiple sclerosis (MS), vascular dementia, and stroke. In particular, diapedesis of monocytes and subsequent trafficking of monocyte-derived macrophages into the brain are key mediators of demyelination and axonal damage in MS. Endothelin-1 (ET-1) is considered as a potent pro-inflammatory peptide and has been implicated in the development of cardiovascular diseases. Here, we studied the role of different components of the endothelin system, i.e., ET-1, its type B receptor (ET(B)) and endothelin-converting enzyme-1 (ECE-1) in monocyte diapedesis of a human brain endothelial cell barrier. Our pharmacological inhibitory and specific gene knockdown studies point to a regulatory function of these proteins in transendothelial passage of monocytes. Results from this study suggest that the endothelin system is a putative target within the brain for anti-inflammatory treatment in neurological diseases., (© 2011 The Authors. Journal of Neurochemistry © 2011 International Society for Neurochemistry.)
- Published
- 2012
- Full Text
- View/download PDF
40. Adenosine triphosphate-binding cassette transporters mediate chemokine (C-C motif) ligand 2 secretion from reactive astrocytes: relevance to multiple sclerosis pathogenesis.
- Author
-
Kooij G, Mizee MR, van Horssen J, Reijerkerk A, Witte ME, Drexhage JA, van der Pol SM, van Het Hof B, Scheffer G, Scheper R, Dijkstra CD, van der Valk P, and de Vries HE
- Subjects
- 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, ATP-Binding Cassette Transporters metabolism, Astrocytes metabolism, Blood-Brain Barrier metabolism, Chemokine CCL2 metabolism, Multiple Sclerosis metabolism
- 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.
- Published
- 2011
- Full Text
- View/download PDF
41. Sphingosine 1-phosphate receptor 1 and 3 are upregulated in multiple sclerosis lesions.
- Author
-
Van Doorn R, Van Horssen J, Verzijl D, Witte M, Ronken E, Van Het Hof B, Lakeman K, Dijkstra CD, Van Der Valk P, Reijerkerk A, Alewijnse AE, Peters SL, and De Vries HE
- Subjects
- 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, Multiple Sclerosis physiopathology, Receptors, Lysosphingolipid metabolism, Up-Regulation physiology
- 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., ((c) 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
42. T lymphocytes impair P-glycoprotein function during neuroinflammation.
- Author
-
Kooij G, van Horssen J, de Lange EC, Reijerkerk A, van der Pol SM, van Het Hof B, Drexhage J, Vennegoor A, Killestein J, Scheffer G, Oerlemans R, Scheper R, van der Valk P, Dijkstra CD, and de Vries HE
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Adult, Aged, Aged, 80 and over, Animals, Brain Chemistry, CD4-Positive T-Lymphocytes pathology, Encephalomyelitis, Autoimmune, Experimental pathology, Endothelial Cells metabolism, Endothelium, Vascular metabolism, Female, Gene Expression, Humans, Male, Middle Aged, Rats, Rats, Inbred Lew, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, Brain pathology, Inflammation pathology, Neuroimmunomodulation, T-Lymphocytes pathology
- Abstract
The ATP-binding cassette (ABC) transporter P-glycoprotein (P-gp; ABCB1) is highly expressed at the blood-brain barrier (BBB). P-gp actively secretes and keeps the central nervous system (CNS) safe from body-born metabolites, but also from drugs and food components, emphasising the importance of its optimal function to maintain brain homeostasis. Here we demonstrate that vascular P-gp expression and function are strongly decreased during neuroinflammation. In vivo, the expression and function of brain endothelial P-gp in experimental allergic encephalomyelitis (EAE), an animal model for multiple sclerosis (MS), were significantly impaired. Strikingly, vascular P-gp expression was decreased in both MS and EAE lesions and its disappearance coincided with the presence of perivascular infiltrates consisting of lymphocytes. Our data strongly suggest that activated CD4(+) T cells induce impaired function of brain endothelial P-gp. Notably, lymphocyte interaction through endothelial intracellular adhesion molecule -1 (ICAM-1) resulted in activation of a nuclear factor kappa B (NF-kappaB) signaling pathway, which resulted in endothelial P-gp malfunction. Our study provides first evidence that CD4(+) T cells are able to affect endogenous molecular protection mechanisms of brain endothelium. Loss of vascular P-gp function during neuroinflammation may disturb brain homeostasis and thereby aggravate disease progression via exposure of vulnerable CNS cells to detrimental compounds.
- Published
- 2010
- Full Text
- View/download PDF
43. The NR1 subunit of NMDA receptor regulates monocyte transmigration through the brain endothelial cell barrier.
- Author
-
Reijerkerk A, Kooij G, van der Pol SM, Leyen T, Lakeman K, van Het Hof B, Vivien D, and de Vries HE
- Subjects
- Animals, Blood-Brain Barrier drug effects, Cell Movement drug effects, Cells, Cultured, Chemokine CCL2 metabolism, Disease Models, Animal, Dizocilpine Maleate pharmacology, Encephalomyelitis, Autoimmune, Experimental etiology, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental pathology, Endothelial Cells drug effects, Endothelial Cells physiology, Enzyme Activation drug effects, Excitatory Amino Acid Antagonists pharmacology, Female, Humans, Immunoprecipitation methods, Leukocyte Common Antigens metabolism, Membrane Proteins metabolism, Mice, Monocytes drug effects, Myelin Proteins, Myelin-Associated Glycoprotein immunology, Myelin-Oligodendrocyte Glycoprotein, Phosphoproteins metabolism, Signal Transduction drug effects, Tissue Plasminogen Activator pharmacology, Zonula Occludens-1 Protein, Blood-Brain Barrier physiology, Brain cytology, Cell Movement physiology, Monocytes physiology, Receptors, N-Methyl-D-Aspartate physiology
- Abstract
Normal neuronal functioning is dependent on the blood-brain barrier. This barrier is confined to specialized brain endothelial cells lining the inner vessel wall, and tightly controlling transport of nutrients, efflux of potentially harmful molecules and entry of immune cells into the brain. Loss of blood-brain barrier function is an early and significant event which contributes to inflammation in the brain and subsequent progression of neuronal deficits in a number of brain disorders and has been well-documented for the auto-immune disease multiple sclerosis. Extravasation of cells happens by paracellular transport across the endothelial junctions, transcellularly across the endothelial cells, or both, and requires the active participation of endothelial cells. We and others have shown that this process requires the activity of proteases, including tissue-type plasminogen activator. We here describe a novel role for NMDA receptor, a potential cellular target of tissue-type plasminogen activator, in human brain endothelial cells. Our results show that the NMDA receptor subunit 1 (NR1) is expressed in brain endothelial cells, regulates tissue-type plasminogen activator-induced signal transduction and controls the passage of monocytes through the brain endothelial cell barrier. Together, our results hold significant promise for the treatment of chronic inflammation in the brain.
- Published
- 2010
- Full Text
- View/download PDF
44. Tissue-type plasminogen activator is a regulator of monocyte diapedesis through the brain endothelial barrier.
- Author
-
Reijerkerk A, Kooij G, van der Pol SM, Leyen T, van Het Hof B, Couraud PO, Vivien D, Dijkstra CD, and de Vries HE
- Subjects
- Animals, Cell Line, Chemotaxis, Coculture Techniques, Encephalomyelitis, Autoimmune, Experimental, Endothelial Cells cytology, Humans, Male, Membrane Proteins metabolism, Occludin, Rats, Rats, Inbred Lew, Rats, Wistar, Signal Transduction, Tissue Plasminogen Activator metabolism, Blood-Brain Barrier metabolism, Cell Communication, Endothelial Cells metabolism, Monocytes cytology, Tissue Plasminogen Activator physiology
- Abstract
Inflammatory cell trafficking into the brain complicates several neurological disorders including multiple sclerosis. Normally, reliable brain functioning is maintained and controlled by the blood-brain barrier (BBB), which is essential to restrict the entry of potentially harmful molecules and cells from the blood into the brain. The BBB is a selective barrier formed by dedicated brain endothelial cells and dependent on the presence of intracellular tight junctions. In multiple sclerosis, a severe dysfunction of the BBB is observed, which is key to monocyte infiltration and inflammation in the brain. Proteolytic activity has been associated with these inflammatory processes in the brain. Our studies in plasma of rats indicated that the extracellular protease tissue-type plasminogen activator (tPA) correlates with the clinical signs of experimental allergic encephalomyelitis, a rat model of multiple sclerosis. In this study, we studied the function of the tPA during diapedesis of monocytes through a rat and human brain endothelial barrier. Monocyte-brain endothelial cell coculture experiments showed that monocytes induce the release of tPA by brain endothelial cells, which subsequently activates the signal transduction protein extracellular signal related kinase (ERK1/2), both involved in monocyte diapedesis. Importantly, live imaging and immunoblot analyses of rat brain endothelial cells revealed that tPA and ERK1/2 control the breakdown of the tight junction protein occludin. These studies identify tPA as a novel and relevant pathological mediator of neuroinflammation and provide a potential mechanism for this.
- Published
- 2008
- Full Text
- View/download PDF
45. DC-SIGN mediates adhesion and rolling of dendritic cells on primary human umbilical vein endothelial cells through LewisY antigen expressed on ICAM-2.
- Author
-
García-Vallejo JJ, van Liempt E, da Costa Martins P, Beckers C, van het Hof B, Gringhuis SI, Zwaginga JJ, van Dijk W, Geijtenbeek TB, van Kooyk Y, and van Die I
- Subjects
- Animals, CHO Cells, Carbohydrate Metabolism drug effects, Cell Adhesion drug effects, Cell Line, Concanavalin A pharmacology, Cricetinae, Cricetulus, Dendritic Cells drug effects, Dendritic Cells immunology, Endothelial Cells drug effects, Endothelial Cells enzymology, Epitopes, Fucosyltransferases genetics, Fucosyltransferases metabolism, Gene Expression Regulation, Enzymologic drug effects, Humans, Lewis X Antigen, Ligands, RNA Interference, Umbilical Veins drug effects, Umbilical Veins enzymology, Galactoside 2-alpha-L-fucosyltransferase, Antigens, CD immunology, Cell Adhesion Molecules immunology, Cell Movement drug effects, Dendritic Cells cytology, Endothelial Cells cytology, Lectins, C-Type immunology, Lewis Blood Group Antigens immunology, Receptors, Cell Surface immunology, Umbilical Veins cytology
- Abstract
Immature dendritic cells (DCs) are recruited from blood into tissues to patrol for foreign antigens. After antigen uptake and processing, DCs mature and migrate to the secondary lymphoid organs where they initiate immune responses. DC-SIGN is a DC-specific C-type lectin that acts both as a pattern recognition receptor and as an adhesion molecule. As an adhesion molecule, DC-SIGN is able to mediate rolling and adhesion over endothelial cells under shear flow. In this study, we show that the binding partner of DC-SIGN on endothelial cells is the glycan epitope Lewis(Y) (Le(Y)), expressed on ICAM-2. The interaction between DC-SIGN on dendritic cells and ICAM-2 on endothelial cells is strictly glycan-specific. ICAM-2 expressed on CHO cells only served as a ligand for DC-SIGN when properly glycosylated, underscoring its function as a scaffolding protein. The expression of Le(Y) in endothelial cells is directed by the enzyme FUT1. Silencing of FUT1 results in a decrease in the rolling and adhesion of immature DCs over endothelial cells. The identification of Le(Y) as the carbohydrate ligand of DC-SIGN in endothelial cells opens new possibilities for the manipulation of DC migration.
- Published
- 2008
- Full Text
- View/download PDF
46. C-type lectin DC-SIGN modulates Toll-like receptor signaling via Raf-1 kinase-dependent acetylation of transcription factor NF-kappaB.
- Author
-
Gringhuis SI, den Dunnen J, Litjens M, van Het Hof B, van Kooyk Y, and Geijtenbeek TB
- Subjects
- Acetylation, Amino Acid Motifs, Cell Adhesion Molecules genetics, Cells, Cultured, DNA metabolism, Enzyme Activation, Humans, Interleukin-10 biosynthesis, Interleukin-10 genetics, Lectins, C-Type genetics, Phosphoserine metabolism, Protein Binding, Protein Serine-Threonine Kinases metabolism, Receptors, Antigen, T-Cell metabolism, Receptors, Cell Surface genetics, Toll-Like Receptor 3 metabolism, Toll-Like Receptor 4 metabolism, Toll-Like Receptor 5 metabolism, Transcription, Genetic genetics, ras Proteins metabolism, Cell Adhesion Molecules metabolism, Lectins, C-Type metabolism, NF-kappa B metabolism, Proto-Oncogene Proteins c-raf metabolism, Receptors, Cell Surface metabolism, Signal Transduction, Toll-Like Receptors metabolism
- Abstract
Adaptive immune responses by dendritic cells (DCs) are critically controlled by Toll-like receptor (TLR) function. Little is known about modulation of TLR-specific signaling by other pathogen receptors. Here, we have identified a molecular signaling pathway induced by the C-type lectin DC-SIGN that modulates TLR signaling at the level of the transcription factor NF-kappaB. We demonstrated that pathogens trigger DC-SIGN on human DCs to activate the serine and threonine kinase Raf-1, which subsequently leads to acetylation of the NF-kappaB subunit p65, but only after TLR-induced activation of NF-kappaB. Acetylation of p65 both prolonged and increased IL10 transcription to enhance anti-inflammatory cytokine responses. We demonstrated that different pathogens such as Mycobacterium tuberculosis, M. leprae, Candida albicans, measles virus, and human immunodeficiency virus-1 interacted with DC-SIGN to activate the Raf-1-acetylation-dependent signaling pathway to modulate signaling by different TLRs. Thus, this pathway is involved in regulation of adaptive immunity by DCs to bacterial, fungal, and viral pathogens.
- Published
- 2007
- Full Text
- View/download PDF
47. Activation of human endothelial cells by tumor necrosis factor-alpha results in profound changes in the expression of glycosylation-related genes.
- Author
-
García-Vallejo JJ, Van Dijk W, Van Het Hof B, Van Die I, Engelse MA, Van Hinsbergh VW, and Gringhuis SI
- Subjects
- Carbohydrate Conformation, Carbohydrate Sequence, Cells, Cultured, Endothelial Cells cytology, Endothelium, Vascular cytology, Epitopes, Gene Expression Profiling, Glycosylation, Humans, Leukocytes cytology, Leukocytes immunology, Molecular Sequence Data, Protein Binding, Cell Membrane chemistry, Endothelial Cells immunology, Endothelial Cells physiology, Gene Expression Regulation, Tumor Necrosis Factor-alpha immunology
- Abstract
The endothelium plays a central role in the logistics of the immune system by allowing the selective transmigration of leukocytes, as well as the maintenance of the circulation and coagulation homeostasis. Evidence is increasing that the carbohydrate composition of the endothelial cell surface is critical for the cells to exert their physiological function. The major aim of this study is to unravel the mechanisms underlying the expression of carbohydrate structures by endothelial cells, which are involved in leukocyte adhesion and migration. Using quantitative real-time PCR, the expression profile of a selected group of 74 glycosylation-related genes has been determined in human umbilical vein endothelial cells (HUVEC) and human foreskin microvascular endothelial cells (FMVEC) under control and TNFalpha-induced conditions. The set of genes comprised 59 glycosyltransferases, 6 mannosidases and 9 sulfotransferases. In parallel, the overall cell surface glycan profile has been assessed by the use of glycan-specific lectins and monoclonal antibodies. The results demonstrate that HUVEC and FMVEC differ substantially in the expression of glycosylation-related genes and, accordingly, also in the presence of different glycan epitopes on the cell membrane. Induction of an inflamed phenotype of the cells by treatment with TNFalpha differentially modulates a set of these genes in HUVEC and FMVEC resulting in a change in the cell membrane associated glycans that are of importance in inflammation-related endothelial cell-surface processes., (Copyright 2005 Wiley-Liss, Inc.)
- Published
- 2006
- Full Text
- View/download PDF
48. Activated human PMN synthesize and release a strongly fucosylated glycoform of alpha1-acid glycoprotein, which is transiently deposited in human myocardial infarction.
- Author
-
Poland DC, García Vallejo JJ, Niessen HW, Nijmeyer R, Calafat J, Hack CE, Van het Hof B, and Van Dijk W
- Subjects
- Complement Activation, Complement C3a metabolism, E-Selectin metabolism, Female, Fucose biosynthesis, Glycosylation, Humans, Inflammation metabolism, Male, Myocardial Infarction pathology, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Neutrophils pathology, Protein Binding, Cytoplasmic Granules metabolism, Myocardial Infarction metabolism, Neutrophils metabolism, Orosomucoid biosynthesis, Protein Processing, Post-Translational
- Abstract
Alpha1-acid glycoprotein (AGP) is a major acute-phase protein present in human plasma as well as in polymorphonuclear leukocytes (PMN). In this report, we show that PMN synthesize a specific glycoform of AGP, which is stored in the specific and azurophilic granules. Activation of PMN results in the rapid release of soluble AGP. PMN AGP exhibits a substantially higher apparent molecular weight than plasma AGP (50-60 kD vs. 40-43 kD), owing to the presence of strongly fucosylated and sialylated polylactosamine units on its five N-linked glycans. PMN AGP is also released in vivo from activated PMN, as appeared from studies using well-characterized myocard slices of patients that had died within 2 weeks after an acute myocardial infarction. AGP was found deposited transiently on damaged cardiomyocytes in areas with infiltrating PMN only. It is interesting that this was inversely related to the deposition of activated complement C3. Strongly fucosylated and sialylated AGP glycoforms have the ability to bind to E-selectin and to inhibit complement activation. We suggest that AGP glycoforms in PMN provide an endogenous feedback-inhibitory response to excessive inflammation.
- Published
- 2005
- Full Text
- View/download PDF
49. Differences in sulfation patterns of heparan sulfate derived from human bone marrow and umbilical vein endothelial cells.
- Author
-
Netelenbos T, Dräger AM, van het Hof B, Kessler FL, Delouis C, Huijgens PC, van den Born J, and van Dijk W
- Subjects
- Heparitin Sulfate chemistry, Heparitin Sulfate metabolism, Humans, Organ Specificity, Veins metabolism, Bone Marrow metabolism, Endothelium, Vascular metabolism, Heparitin Sulfate analysis
- Abstract
Objective: Heparan sulfates (HS), the polysaccharide side chains of HS proteoglycans, differ in structure and composition of sulfated domains among various tissue types, resulting in selective protein binding. HS proteoglycans on bone marrow endothelial cells (BMEC) could contribute to tissue specificity of the bone marrow endothelium and play a role in the presentation of chemokines such as stromal cell-derived factor-1 (SDF-1) and adhesion of hematopoietic progenitor cells after stem cell transplantations. We characterized differences in HS structure and SDF-1 binding between BMEC and human umbilical vein endothelial cells (HUVEC)., Materials and Methods: Expression of HS proteoglycans on human bone marrow microvessels was investigated by immunohistochemical staining. Comparison of three human BMEC cell lines with HUVEC and an HUVEC cell line was studied by flow cytometry using antibodies against different epitopes of the HS polysaccharide chain. HS proteoglycans were biochemically characterized after isolation from metabolically labeled cultures of the BMEC cell line 4LHBMEC and HUVEC. Binding of radiolabeled SDF-1 to 4LHBMEC and HUVEC and competition with heparins were investigated., Results: Bone marrow microvessels constitutively expressed HS proteoglycans. Flow cytometric experiments showed differences in HS chain composition between BMEC and HUVEC. Biochemical characterization revealed more O-sulfation of the N-sulfated domains present in cell-associated HS glycosaminoglycans in 4LHBMEC compared to HUVEC. Binding experiments showed that 4LHBMEC bound more 125[I]-SDF-1 per cell than HUVEC. This could be inhibited largely by heparin and O-sulfated heparin and to a lesser extent by N-sulfated heparin., Conclusions: Cellular HS from BMEC differs in composition from HUVEC. We postulate that the presence of highly sulfated domains in the HS chains from BMEC contributes to tissue specificity of bone marrow endothelium in which HS may be involved in SDF-1 presentation and adhesion of hematopoietic progenitor cells.
- Published
- 2001
- Full Text
- View/download PDF
50. Increased alpha3-fucosylation of alpha(1)-acid glycoprotein in patients with congenital disorder of glycosylation type IA (CDG-Ia).
- Author
-
Van Dijk W, Koeleman C, Van het Hof B, Poland D, Jakobs C, and Jaeken J
- Subjects
- Amidohydrolases metabolism, Carbohydrate Metabolism, Inborn Errors blood, Carbohydrate Metabolism, Inborn Errors classification, Carbohydrate Metabolism, Inborn Errors enzymology, Fucose analogs & derivatives, Fucosyltransferases blood, Fucosyltransferases metabolism, Glycosylation, Humans, Lectins metabolism, Molecular Weight, Neuraminidase metabolism, Orosomucoid chemistry, Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase, Polysaccharides chemistry, Polysaccharides metabolism, Carbohydrate Metabolism, Inborn Errors metabolism, Fucose metabolism, Orosomucoid metabolism
- Abstract
Increased fucosylation of the type (sialyl) Lewis(x) was detected on the acute-phase plasma protein alpha(1)-acid glycoprotein (AGP) in patients with the congenital disorder of glycosylation type IA. This is remarkable, because in these patients the biosynthesis of guanosine 5'-diphosphate (GDP)-D-mannose is strongly decreased, and GDP-D-mannose is the direct precursor for GDP-L-fucose, the substrate for fucosyltransferases. The concomitantly occurring increased branching of the glycans of AGP and the increased fucosyltransferase activity in plasma suggest that a chronic hepatic inflammatory reaction has induced the increase in fucosylation.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.