Brochure symposium

Page 1

MS Research Days 2019 May 16 - 17, 2019 • Hotel Van der Valk, Groningen – Hoogkerk

Program



content Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 Short program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 Complete program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Poster walks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6 Location . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7 General information . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 Instructions for MS speed dates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 Keynote speakers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11 Abstracts oral presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 Abstracts poster presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43

1


Introduction MS Research Days 2019 Dear attendees, It is a pleasure and a privilege to welcome you to the MS research days 2019 here in Groningen. Sharing new information and strategies to improve treatment and directing basic science is essential to tackle multiple sclerosis. The friendly and open atmosphere here in the North of the Netherlands provides a perfect environment for enthusiastic discussions and information sharing. During the MS research days there is ample time to interact and inform people with MS and their caregivers, and to meet old and new colleagues who work in the field of MS. There is a special focus on young researchers who are encouraged to present their research work. MS is a complex disease and further steps to cure or prevent MS can only be made if scientists, clinicians and people with MS collaborate and share their thoughts and information. We hope that the program and the renowned invited speakers Tanja Kuhlmann (University Hospital Münster), Ulrich Dalgas (Aarhus University), and Bart Eggen (UMC Groningen) will create such a stimulating and encouraging environment and will enhance interactions between young and more senior scientists, between scientists and persons with MS and their care givers, between basic and more applied scientists, and between colleagues working within MS care and MS science. We are happy to welcome many Dutch and Belgian MS researchers. Thank you all for attending this meeting, we have laid the ground work and now rely on your enthusiasm and inspiration, and we encourage you to interact with many both old and new friends to make this an interesting and inspiring conference. On behalf of the ‘MS centrum Noord Nederland’ and ‘the Biomedical Research Institute (UHasselt)’.

2

Dr. Inge Zijdewind

Prof.dr. Piet Stinissen


Short program THURSDAY, 16 MAY 2019 Time

Program

Location

09:00 – 10:00

Registration and coffee

Tilburg bar

10:00 – 11:45

Opening

11:45 – 12:30

Scientific presentations 1 – Treatment

12:30 – 13:30

Lunch

Tilburg bar

13:30 – 14:00

MS speed dates

Utrecht room

14:00 – 15:00

Scientific presentations 2 – Causes of MS

Amsterdam & Rotterdam room

15:00 – 15:15

Battle of the labs finale

15:15 – 15:45

Break

Tilburg bar

15:45 – 16:45

Poster walks

Tilburg bar & Utrecht room

16:45 – 17:30

Keynote lecture prof.dr. Bart J.L. Eggen

17:30 – 18:00

Thesis award & presentation

18:00 – 18:30

Drinks

Maastricht bar

18:30 – 21:30

Meet & eat

Delft-Leiden-Groningen room

Amsterdam & Rotterdam room

Amsterdam & Rotterdam room

FRIDAY, 17 MAY 2019 Time

Program

Location

08:30 – 09:00

Welcome and coffee

Tilburg bar

09:00 – 09:45

Keynote lecture prof.dr. Tanja Kuhlmann

09:45 – 10:30

Scientific presentations 3 – Underlying processes

10:30 – 11:00

Break

11:00 – 11:45

Keynote lecture dr. Ulrike Dalgas

11:45 – 12:45

Scientific presentations 4 – Monitoring MS

12:45 – 12:50

Announcement nominees MS speed dates

12:50 – 14:00

Lunch

14:00 – 14:15

Presentations nominees MS speed dates

14:15 – 15:15

Scientific presentations 5 – Immune cells

15:15 – 15:30

Awards

15:30 – 15:35

Closure

Amsterdam & Rotterdam room

Tilburg bar

Amsterdam & Rotterdam room

Tilburg bar

Amsterdam & Rotterdam room

3


Complete program Thursday, may 16th 2019 9:00 – 10:00 10:00 – 11:45 11:45 – 12:30

Registration and coffee & cake Opening

A quick journey passing MS research institutes in The Netherlands and Belgium

Scientific presentations I – Treatment Moderators: prof.dr. Veerle Somers, dr. Joost Smolders • Clinical and immunological control of experimental autoimmune encephalomyelitis by multi-epitope-engineered tolerogenic dendritic cells - Inez Wens (page 13) • A transcranial stimulation protocol targeting processing speed in MS - Nena Lejko (page 15) • VISTA expression by microglia decreases during inflammation and is differentially regulated in MS - Malte Boggrewe (page 17)

12:30 – 13:30

Lunch Poster viewing

13:30 – 14:00

MS speed dates

14:00 – 15:00

Scientific presentations II – Causes of MS Moderators: prof.dr. Jack van Horssen, dr. Bieke Broux • Oncostatin M-induced astrocytic Tissue inhibitor of metalloproteinase-1 drives remyelination - Evelien Houben (page 18) • Cortical Atrophy Accelerates as Cognitive Decline Worsens in MS - Menno Schoonheim (page 19) • Post-mortem MS lesion pathology is influenced by single nucleotide polymorphisms Nina Fransen (page 20) • Specialized pro-resolving lipid mediator production in the cerebrospinal fluid is impaired in MS: implications for its pathogenesis and therapy - Gijs Kooij (page 22)

15:00 – 15:15

Battle of the labs finale

15:15 – 15:45

Break

15:45 – 16:45

Poster walks • Poster walk A – Monitoring (symptoms of) MS Moderators: dr. Thea Heersema, dr. Hugo Vrenken • Poster walk B – Underlying processes Moderators: drs. Nina Fransen, dr. Marvin van Luijn • Poster walk C – Treatment of MS Moderators: dr. Susanne Kooistra, dr.ir. Liesbet Peeters

16:45 – 17:30

Keynote lecture prof.dr. Bart J.L. Eggen (Epi-)genetic regulation of microglia innate immune memory

Moderator: prof.dr. Elga de Vries

4

17:30 – 18:00

Thesis award & presentation

18:00 – 18:30

Drinks Prepare proposal MS speed dates

18:30 – 21:30

Meet & eat


Complete program friday, may 17th 2019 8:30 – 9:00 9:00 – 9:45

Welcome and Coffee Prepare proposal MS speed dates Keynote lecture prof.dr. Tanja Kuhlmann How to wrap the myelin: What can we learn from stem cells?

Moderator: dr. Wia Baron 9:45 – 10:30

Scientific presentations III – Underlying processes Moderators: dr. Wia Baron, dr. Gijs Kooij • RNA-sequencing of laser-captured leukocortical lesions from MS patients - Thecla van Wageningen (page 24) • Transcriptional profiling of human microglia reveals grey-white matter heterogeneity and MS-associated changes - Marlijn van der Poel (page 26) • Cortical microglia in progressive MS acquire a unique phenotype in response to chronic meningeal inflammation - Lynn van Olst (page 27)

10:30 – 11:00 11:00 – 11:45

Break Keynote lecture dr. Ulrik Dalgas

Exercise and MS – a promising MS rehabilitation intervention

Moderator: dr. Inge Zijdewind 11:45 – 12:45

Scientific presentations IV – Monitoring MS Moderators: prof.dr.ir. Charlotte Teunissen, dr. Karim Kreft • Serum neurofilament light chain in pediatric MS and other acquired demyelinating syndromes - Arlette Bruijstens (page 28) • Functional imaging biomarkers of cognitive impairment in MS: a resting-state magnetoencephalography study - Shanna Kulik (page 30) • Deciphering morphology of Evoked Potentials - Jan Yperman (page 32) • Next generation microRNA sequencing and targeted proteomics to find novel subtype specific biomarkers for MS - Ineke Tan (page 34)

12:45 – 12:50

Announcement nominees MS speed dates

12:50 – 14:00

Lunch Meet the expert & poster viewing

14:00 – 14:15

Presentations nominees MS speed dates

14:15 – 15:15

Scientific presentations 5 – Immune cells Moderators: prof.dr. Piet Stinissen, prof.dr. Jon Laman • Regulatory T cells have a distinct migratory phenotype in relapsing-remitting MS patients - Paulien Baeten (page 36) • Inflammatory lesion activity in chronic progressive MS is related to the accumulation of tissue resident memory T cells in the perivascular space - Joost Smolders (page 37) • Human T-bet+ B cells: induction and effector functions from a MS perspective - Jamie van Langelaar (page 39) • Contribution of risk allele CLEC16A to the B cell receptor-mediated HLA class II pathway in MS - Marvin van Luijn (page 41)

15:15 – 15:30

Awards - Best proposal - Best oral presentation - Best poster presentation

15:30 - 15:35

Closure

5


Poster walks Poster walk A, B and C: Thursday May 16th 2019, 15:45 - 16:45 hours Poster walk A – Monitoring (symptoms of) MS Moderators: dr. Thea Heersema, dr. Hugo Vrenken • S pecific cerebellar atrophy patterns can distinguish physical disability and cognitive impairment in MS

Iris Dekker

page 43

• M yelin imaging with [11C]MeDAS PET

Kars van der Weijden

page 45

• Subjective visual complaints in patients with multiple sclerosis

Iris van der Lijn

page 46

• Outdoor mobility of persons with multiple sclerosis. A systematic review

Iris van der Lijn

page 47

• T halamic volume as a measure to discriminate between subjective and objective cognitive complaints in MS: towards clinical application?

Marijn Huiskamp

page 48

• P erceived Neuropsychological Impairment, Depression, Health and Employment status in Multiple Sclerosis

Marie D’hooghe

page 50

• B IA study: a prospective observational multi-centre study on alterations of gut microbiome and the brain-immune-intestine axis in patients with RRMS who start treatment with oral Cladribine

Jeske van Pamelen

page 51

• T he association of Epstein-Barr virus load with CNS-infiltrating B cells in MS: lessons learned from current treatments

Jamie van Langelaar

page 53

• I gD-CD27- double negative (DN) B cells of MS patients are mature memory cells that can migrate towards pro-inflammatory chemokines

Lien Beckers

page 55

• D ifferential responses of developing and mature grey and white matter oligodendrocytes to pro-inflammatory cytokines

Jacomien Jongsma

page 56

• T owards the suppression of glucocorticoid-resistant T cells driving multiple sclerosis disease activity

Steven Koetzier

page 57

• I mmunoglobulin repertoire analysis indicates a common origin of ageassociated IgD-CD27- double negative (DN) B cells in healthy individuals and MS patients

Judith Fraussen

page 59

• H eterogeneity in oligodendrocyte progenitor cells derived from cortex and corpus callosum

Dennis Lentferink

page 60

• C ytotoxic CD4 T cells escape suppression by regulatory T cells while promoting suppression of conventional helper T cells

Cindy Hoeks

page 61

• H igh-dose vitamin D3 supplementation in multiple sclerosis is not associated with lower plasma neurofilament light chain levels

Joost Smolders

page 62

• E xploring the neuroprotective function of extracellular vesicles containing small heat shock proteins (HSPB1 and HSPB8) upon neuroinflammation

Bram Van den Broek

page 63

• S etmelanotide, a novel, selective melanocortin receptor-4 agonist exerts anti-inflammatory actions in astrocytes and promotes an antiinflammatory macrophage phenotype

Merel Rijnsburger

page 64

• TNF-TNFRs signalling modulation in an MS mouse model

Valentina Pegoretti

page 65

• C ognitive rehabilitation in patients with advanced progressive multiple sclerosis: possible within limits?

Stefanos Prouskas

page 66

• D ifferent network functional connectivity characteristics of responders and non-responders to attention training in MS

Stefanos Prouskas

page 68

• M emory retraining in MS: lessons learned from healthy aging, mild cognitive impairment and Alzheimer’s disease

Piet Bouman

page 70

Poster walk B – Underlying processes Moderators: drs. Nina Fransen, dr. Marvin van Luijn

Poster walk C – TreatmenT of MS Moderators: dr. Susanne Kooistra, dr.ir. Liesbet Peeters

6


Location Congress venue Hotel Van der Valk Groningen – Hoogkerk Borchsingel 53 9766 PP Eelderwolde, The Netherlands Tel +31 50 820 05 10 Directions Van der Valk Hotel Groningen-Hoogkerk is easily accessible by car and public transport (P+R Hoogkerk). There is a spacious, free car park available for guests (with 4 electronic charge points). By car From Heerenveen • You drive on the A7 motorway in the direction of Groningen • Take exit number 35 Hoogkerk / Peize • At the bottom of the exit, take the first exit at the roundabout • At the next roundabout, take the second exit • After 200 meters you will find our parking area on your left From Assen • You drive on the A28 motorway in the direction of Groningen • At the Julianaplein, keep left and follow the signs for Ring-West / Zuidhorn • Continue on the A7 towards Amsterdam / Drachten • Take exit number 35 Hoogkerk / Peize / Roden • At the bottom of the exit, take the third exit under the viaduct at the roundabout and then take the first right at the next roundabout • At the next roundabout, take the second exit • After 200 meters you will find our parking area on your left By public transport The nearest NS station is Groningen. From the station you can take a bus (Qbuzz lines 3 or 4) that brings you in 10 minutes to P + R Hoogkerk. It is a few minutes walking from P + R Hoogkerk to Hotel Van der Valk Groningen – Hoogkerk. For more information consult www.ns.nl (train) or www.9292.nl (all public transport).

7


Location

8


General information Registration The registration desk is located near the entrance of the Amsterdam & Rotterdam room. Registered participants will receive the short program and a name batch. The program book, including abstracts, is only available electronically.

ORAL PRESENTATIONS Presenters are kindly requested to timely hand in their PowerPoint presentation at the Amsterdam & Rotterdam room. Technical support is present on Thursday May 16th from 9:00 till 10:00 hours and on Friday May 17th from 8:30 till 9:30 hours.

POSTER PRESENTATIONS Posters are on display during the entire meeting. Presenters are kindly requested to attach their poster on designated poster board on Thursday May 16th before the start of the plenary program at 10:00 hours. Poster walks are scheduled on Thursday May 16th from 15:45 till 16:45 hours.

WIFI Free wifi is available in the meeting room area.

FOOD & DRINKS Coffee and tea are available in the Tilburg bar during the breaks as indicated in the program. The Tilburg bar is available for networking and gathering throughout the Research days. Lunch (Thursday and Friday) and diner (Thursday) are included. The lunch is served in the Tilburg bar and dinner in the Delft-Leiden-Groningen room.

CHAIR Dr. Jan Meilof (University Medical Center Groningen, Groningen)

JURY THESIS AWARD Prof.dr. Jerome Hendriks (Hasselt University, Diepenbeek) Prof.dr. Inge Huitinga (Netherlands Institute of Neurosciences, Amsterdam) Prof.dr. Jon Laman (University Medical Center Groningen, Groningen)

9


Instructions for MS speed date session The aim of the MS speed dates is to stimulate the collaborations between different institutes and disciplines. Researchers (PhD students and junior postdocs) in the field of fundamental, translational and clinical MS research can participate. Participants will have three speed dates with three colleagues from other institutes. Partners with the best match write down their idea for a joined research proposal with a focus on MS. Only researchers that are present on both Thursday and Friday can participate.

PROGRAM Thursday 13:30 – 14:00: Speed dates After a short introduction, every participant is scheduled for 3 speed dates of each 7 minutes. Explain your field of research and discuss the possibilities for a joint research project with your speed-date partner. Thursday until Friday 9:00: write research proposal Write a short research proposal together with your best match. Please use the MS-speed date form (on the back) and remember to hand in your proposal by 9:00 h on Friday at the registration desk (one copy). Friday 12:45: announcement nominees Nominated top-3 research proposals are announced by the jury. Friday 14:00: presentations nominees The 3 nominated couples will have 5 minutes per team to present their research proposal (elevator pitch, no slides). The preferred language of speak is English. Friday 15:15: award ceremony The researchers with the best research proposal and presentation will be rewarded with a chance to apply for a € 17.000,- research grant!

GUIDELINES • Only one research proposal per person; • A multidisciplinary approach is preferred; • Feasibility and relevance for MS should be taken into account.

JURY Dr. Anne-Marie van Dam Dr. Inez Wens Dr. Inge Zijdewind

10


Keynote lectures KEYNOTE 1: Prof.dr. BART J.L. EGGEN Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (Epi-)genetic regulation of microglia innate immune memory Microglia, the innate immune cells of the central nervous system (CNS), surveil their microenvironment to maintain homeostasis and quickly respond to insults and infections. Their central nervous system (CNS)-tailored functions and ontogeny distinguish microglia from other tissue-resident macrophages and genome-wide expression and epigenetic profiling studies of human and mouse microglia have confirmed their uniqueness and CNS functions. Microglia are a self-sustained cell population with limited cellular turn-over. Due to this longevity, microglia can serve as a long-term memory for inflammatory or neurodegenerative events. Indeed, altered epigenetic signatures were detected in microglia after inflammatory of neurodegenerative challenges, inducing distinct functional microglia phenotypes.

KEYNOTE 2: Prof.dr. TANJA KUHLMANN Institute of Neuropathology, University Hospital MĂźnster, Germany How to wrap the myelin: What can we learn from stem cells? Multiple Sclerosis (MS) is the most frequent inflammatory demyelinating disease of the CNS and a leading cause for disability in young adults. Immunosuppressive or immunomodulatory therapies prevent new inflammatory lesions that underlie clinical relapses but do not arrest disease. Therefore, therapies promoting remyelination represent a promising new treatment approach. However, limited access to human oligodendrocytes represents a major obstacle for better understanding of myelin diseases and development of new treatment options. To circumvent this problem a number of protocols have been developed to generate myelinating oligodendrocytes from human induced pluripotent stem cells (iPSC) in 2D and 3D culture systems. In my presentation I will summarize the knowledge regarding endogenous remyelination in MS and discuss possibilities how iPSC derived oligodendrocytes from MS patients and healthy controls can be used for disease modeling and identification of new treatment options.

11


Keynote lectures KEYNOTE 3: DR. ULRIK DALGAS Section of Sport Science, Department of Public Health, Aarhus University, Denmark Exercise and MS – a promising MS rehabilitation intervention For years patients with multiple sclerosis (MS) were advised not to participate in physical exercise because it was reported to lead to deterioration of symptoms or fatigue. During the last decades, however, studies on exercise in MS have instead shown a number of beneficial effects, and today exercise is an important part of MS rehabilitation programs, with a number of recent advances. During the presentation an overview of the most recent advances within the field of exercise and multiple sclerosis will be provided. Moreover, the potential beneficial effects include improved functional capacity such as improved walking speed and distance. Additional effects include reduced fatigue, improved balance, improved health and ultimately improved quality of life. Currently, the effects of exercise therapy on brain health is attracting substantial attention. This include the effects on brain volume and cortical thickness but also on cognition. Cross-sectional studies of fitness and cognition indicate that better cardiorespiratory fitness (i.e. a surrogate for aerobic exercise training) is associated with faster processing speed, but studies are not consistent. Furthermore, promising longitudinal exercise studies have been conducted, which have further strengthened the interest in the effects of exercise on cognition. Finally, it is proposed that exercise therapy to a larger extend should also be used as a preventive (potentially disease-modifying) tool rather than as a symptomatic treatment alone.

12


Abstracts oral presentations SCIENTIFIC PRESENTATIONS 1 - TREATMENT Thursday 11:45 – 12:30 Moderators: prof.dr. Veerle Somers and dr. Joost Smolders Clinical and immunological control of experimental autoimmune encephalomyelitis by multi-epitope-engineered tolerogenic dendritic cells Author Judith Derdelinckx1,2, María Mansilla3,4, Maxime De Laere1, Wai-Ping Lee1,5, Juan Navarro-Barriuso3,4, Inez Wens1, Hans De Reu1, Aneta Keliris6, Johan Van Audekerke6, Verdi Vanreusel6, Zoë Pieters7,8, Annemie Van der Linden6, Marleen Verhoye6, Geert Molenberghs7,9, Niel Hens7,8, Herman Goossens10, Barbara Willekens1,2, Patrick Cras2,11, Peter Ponsaerts1, Zwi Berneman1,5, Eva Martínez-Cáceres3,4, Nathalie Cools1,5 Affiliation 1Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium, 2Division of Neurology, Antwerp University Hospital, Edegem, Belgium, 3Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain, 4Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain, 5Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium, 6Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium, 7Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium, 8Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium, 9L-BioStat, I-BioStat, KU Leuven, Leuven, Belgium, Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute

10

(VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium,

11

Born Bunge Institute, Translational Neurosciences, Faculty of Medicine and

Health Sciences, University of Antwerp, Antwerp, Belgium

Although effective in reducing relapse rate and delaying progression, current therapies for multiple sclerosis (MS) do not completely halt disease progression. T cell autoimmunity to myelin antigens is considered to be one of the main mechanisms driving MS. It is characterized by autoreactivity to disease-initiating myelin antigen epitope(s), followed by a cascade of epitope spreading, which are both strongly patient-dependent. Targeting a variety of MS-associated antigens by myelin antigen-presenting tolerogenic dendritic cells (tolDC) is a promising treatment strategy to re-establish tolerance in MS. Electroporation with mRNA encoding myelin proteins is an innovative technique to load tolDC with the full spectrum of naturally-processed myelin epitopes.

13


In this study, we assessed the efficacy of myelin oligodendrocyte glycoprotein (MOG) mRNA-electroporated tolDC in experimental autoimmune encephalomyelitis (EAE). Treatment of MOG35-55-immunized C57BL/6 mice with MOG mRNA-electroporated or MOG35-55-pulsed tolDC led to a stabilization of the EAE clinical score from the first administration onwards, whereas it worsened in mice treated with non-antigen-loaded tolDC or with vehicle only. In addition, MOG35-55-specific pro-inflammatory pathogenic T cell responses and myelin antigen epitope spreading were inhibited in the peripheral immune system of tolDCtreated mice. Finally, magnetic resonance imaging analysis of hyperintense spots along the spinal cord was in line with the clinical score. In conclusion, mRNA electroporation is an efficient and versatile tool to generate myelin-presenting tolDC that are capable to stabilize the clinical score in EAE. These results pave the way for further research into mRNA-electroporated tolDC treatment as a patient-tailored therapy for MS.

14


A transcranial stimulation protocol targeting processing speed in multiple sclerosis Author Nena Lejko1, André Aleman1, Thea Heersema2,4, Jan Meilof3,4, Joke Spikman2, Inge Zijdewind1, Christoph Herrmann5, Remco Renken1, Branislava Ćurčić-Blake1 1 Affiliation Biomedical Sciences of Cells & Systems, Cognitive Neuroscience Center, University

Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Neurology, University Medical Center Groningen, University of Groningen,

2

Groningen, The Netherlands, 3Department of Neurology, Martini Hospital, Groningen, The Netherlands, 4MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, 5Department for Psychology, University of Oldenburg, Oldenburg, Germany

Introduction Multiple sclerosis (MS) is often accompanied by progressive cognitive impairment. The most common cognitive symptom is the slowing of processing speed, which affects the accuracy and independence of performing everyday tasks. There are currently no effective interventions for improving processing speed in MS patients, but synchronisation of theta oscillations induced by transcranial alternating current stimulation (tACS) was recently found to improve processing speed in healthy participants. The aim of the present proofof-principle pilot study was to investigate the feasibility of a tACS simulation protocol in cognitively impaired MS patients and its effect on synchronisation and processing speed. Methods 12 cognitively impaired MS patients were included in the study. 9 received real and 3 sham stimulation. Electroencephalography (EEG) data was available for 11 participants (8 real, 3 sham). Participants received 18 minutes of synchronous 6 Hz or sham tACS combined with cognitive training on three consecutive days. Processing speed at baseline and after stimulation was measured with a battery of neuropsychological tests, and 10 minutes of eyes-open resting state EEG was recorded pre- and post-stimulation. Due to the small sample size, any results should be interpreted with caution. Results Following the stimulation protocol, participants displayed increased fronto-parietal synchronisation. There was a decrease in motor time and a non-significant decrease in reaction time on the VRTT. Performance on the digit symbol modalities test, a measure of processing speed, attention, and planning, also improved. However, time*stimulation type interactions were not significant.

15


Conclusions We found increased fronto-parietal synchronisation and processing speed following three sessions of 6 Hz and sham tACS in cognitively impaired MS patients. Increase in synchronisation following sham stimulation may be an after-effect of the working memory task performed during the stimulation. Based on the results of this pilot study, the link between tACS, synchronisation, and cognition in MS patients needs to be further investigated in larger, adequately powered randomised control trials.

16


VISTA expression by microglia decreases during inflammation and is differentially regulated in MS Author

Malte Borggrewe1, Corien Grit1, Tegan Otto1, Tiago Medeiros Furquim Mendonça1, Wilfred den Dunnen2, Randolph Noelle3, Bart Eggen1, Jon Laman1

1 Affiliation University Medical Center Groningen, Department of Biomedical Sciences of Cells

& Systems / Molecular Neurobiology, Groningen, Netherlands, 2University Medical Center Groningen, Department of Pathology, Groningen, Netherlands, 3Geisel School of Medicine at Dartmouth, Department of Microbiology and Immunology, Lebanon, New Hampshire, United States of America

V-type immunoglobulin domain-containing suppressor of T-cell activation (VISTA) is a negative checkpoint regulator (NCR) involved in inhibition of T cell-mediated immunity. VISTA is also involved in various myeloid cell functions including efferocytosis and cytokine response. Expression changes of other NCRs (PD-1, PD-L1/L2, CTLA-4) during inflammation of the central nervous system (CNS) were previously demonstrated, but VISTA expression in the CNS has not yet been explored. Here, we report that in the human and mouse CNS, VISTA is most abundantly expressed by microglia, and to lower levels by endothelial cells. Upon TLR stimulation, VISTA expression was reduced in primary neonatal mouse microglia in vitro. In mice, microglial VISTA expression was reduced after lipopolysaccharide (LPS) injection, during experimental autoimmune encephalomyelitis (EAE), and in the accelerated aging Ercc1Δ/- mouse model. After LPS injection, decreased VISTA expression in mouse microglia was accompanied by decreased acetylation of lysine residue 27 in histone 3 in both its promoter and enhancer region. ATAC-sequencing indicated a potential regulation of VISTA expression by Pu.1 and Mafb, two transcription factors crucial for microglia function. Furthermore, our data shows that VISTA expression varies in different types of multiple sclerosis (MS) lesions including preactive, active, chronic active, and inactive lesions. Finally, we observed a reduced ramification of microglia in VISTA knockout mice, concomitant with an increase in phagocytic ability of VISTA-deficient microglia in vitro. This study is the first to demonstrate that in the CNS, microglial VISTA expression is differentially regulated during MS, and that knockout of VISTA affects microglia biology.

17


SCIENTIFIC PRESENTATIONS 2 – CAUSES OF MS Thursday 14:00 – 15:00 Moderators: prof.dr. Jack van Horssen and dr. Bieke Broux Oncostatin M-induced astrocytic Tissue inhibitor of metalloproteinase-1 drives remyelination Author Evelien Houben1*, Kris Janssens1*, Doryssa Hermans1, Jennifer Vandooren3, Chris Van den Haute2, Melissa Schepers1, Tim Vanmierlo1, Ivo Lambrichts4, Veerle Baekelandt2, Ghislain Opdenakker3, Wia Baron5, Bieke Broux1*, Helena Slaets1*, Niels Hellings1* 1 Affiliation Department of Immunology, Biomedical Research Institute, Hasselt University, 3590

Diepenbeek, Belgium, 2Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium, 3Department of Microbiology and Immunology, Laboratory of Immunobiology, Rega Institute, KU Leuven, 3000 Leuven, Belgium, 4Department of Morphology, Biomedical Research Institute, Hasselt University, 3590 Diepenbeek, Belgium, 5Department of Cell Biology, University of Groningen, University Medical Center Groningen (UMCG) Netherlands. * These authors contributed equally to this work

The molecular framework that regulates remyelination is incompletely understood. In demyelinating disorders, oligodendrocyte precursor cells (OPCs) migrate to demyelinated lesions in the central nervous system (CNS), differentiate into mature oligodendrocytes and restore myelin sheaths. During disease, this remyelination process is gradually compromised, resulting in disturbed neuronal signaling and leaving denuded axons vulnerable for degeneration. To investigate potential remyelination boosters, the generally accepted cuprizoneinduced demyelination model was used. In this study, we identify oncostatin M (OSM), a member of the interleukin (IL)-6 cytokine family, as a crucial contributor that potentiates remyelination. Expression of the OSM receptor (OSMR) was strongly upregulated in demyelinated regions. While remyelination was completely abrogated in OSMR knock-out (KO) mice, lentivirus (LV) mediated-OSM overexpression in the CNS increased the number of newly-formed mature oligodendrocytes and induced remyelination. One of the determining factors involved in OSM-mediated remyelination was OSMinduced astrocytic expression of tissue inhibitor of metalloproteinase-1 (TIMP-1). TIMP-1 deficiency completely abolished spontaneous remyelination, phenocopying the results of the OSMR KO mice. In vitro experiments confirmed the involvement of OSM-induced astrocytic TIMP-1 in OPC differentiation into mature oligodendrocytes. Taken together, this study demonstrates a crucial role for OSMR signaling during remyelination and suggests that OSM and its downstream effector molecule TIMP-1 are promising therapeutic candidates for demyelinating disorders. 18


Cortical Atrophy Accelerates as Cognitive Decline Worsens in Multiple Sclerosis Author Anand Eijlers1, Iris Dekker2,3, Martijn Steenwijk1, Kim Meijer1, Hanneke Hulst1, Petra Pouwels2, Bernard Uitdehaag3, Frederik Barkhof2,4, Hugo Vrenken2, Menno Schoonheim1, Jeroen Geurts1 1 Affiliation Departments of Anatomy & Neurosciences, 2Radiology and Nuclear Medicine, 3

Neurology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC,

Location VUmc, Amsterdam, the Netherlands, 4Institutes of Neurology and Healthcare Engineering, UCL, London, UK

Background and objective Cognitive decline is common in multiple sclerosis and seems to accelerate over time, which is poorly understood. This study aimed to determine which pathological process could be responsible for the acceleration of cognitive decline during the course of multiple sclerosis, using longitudinal structural MR imaging, which was related to cognitive decline in relapsing-remitting and progressive patients. Methods A prospective cohort of 230 patients with multiple sclerosis (179 relapsing-remitting and 51 progressive) and 59 healthy controls was evaluated twice with a five-year interval (mean=4.9, SD=0.94). Annual rates of cortical and deep grey matter atrophy as well as lesion volume increase were computed on longitudinal MR imaging data (acquired at 3-Tesla) and correlated to the annual rate of cognitive decline as measured using an extensive cognitive evaluation at both time points. Results The deep grey matter atrophy rate did not differ between progressive and relapsingremitting patients (-0.82%/year versus -0.71%/year, P=0.11), while faster cortical atrophy was observed in progressive patients (-0.87%/year versus -0.48%/year, P<0.01). Similarly, faster cognitive decline was observed in progressive compared to relapsingremitting patients (P<0.01). Correlates of annual cognitive decline shifted from the rate of annual lesion volume increase in relapsing-remitting who did not convert to progressive MS during the follow-up period (r=-0.17, P=0.03) to the rate of annual deep grey matter atrophy in relapsing-remitting patients who did convert to progressive MS (r=0.50, P=0.02) and annual cortical atrophy in progressive patients (r=0.35, P=0.01). Conclusions These results indicate that cortical atrophy and cognitive decline accelerate together during the course of multiple sclerosis. In relapsing-remitting patients, cognitive decline was related to worsening lesional pathology and deep grey matter atrophy, whereas cognitive decline in progressive patients was related to the accelerated cortical atrophy only. 19


Post-mortem multiple sclerosis lesion pathology is influenced by single nucleotide polymorphisms Author

Nina Fransen1, Jakob Crusius2, Joost Smolders1,3, Mark Mizee1, Corbert van Eden1, Sabina Luchetti1, Ester Remmerswaal4,5, Jรถrg Hamann1,4, Matthew Mason1, Inge Huitinga1

1 Affiliation Department of Neuroimmunology, The Netherlands Institute for Neuroscience,

Amsterdam, The Netherlands, 2Laboratory for Immunogenetics, Department of Medical Microbiology and Infection Control, Amsterdam UMC, VU University, Amsterdam, The Netherlands, 3MS Center CWZ, Department of Neurology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands, 4Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands, 5Renal Transplant Unit, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands

Background Over the last few decades, several common single nucleotide polymorphisms (SNPs) have been identified that correlate with clinical outcome in multiple sclerosis (MS), but the pathogenic mechanisms underlying the clinical effects of these SNPs are unknown. This is in part due to the difficulty in the functional translation of genotype into diseaserelevant mechanisms. Building on our recent work showing the association of clinical disease course with post-mortem MS lesion characteristics, we hypothesized that SNPs that correlate with clinical disease course would also correlate with specific MS lesion characteristics in autopsy tissue. Methods 179 MS brain donors from the Netherlands Brain Bank MS autopsy cohort were genotyped for 102 SNPs, selected based on their reported associations with clinical outcome or their associations with genes that show differential gene expression in MS lesions. Results Three SNPs linked to MS clinical severity showed a significant association between the genotype and either the proportion of active lesions (rs2234978/FAS and rs11957313/ KCNIP1) or the proportion of chronic active lesions (rs8056098/CLEC16A). Three SNPs linked to MS pathology-associated genes showed a significant association with either proportion of active lesions (rs3130253/MOG), incidence of cortical grey matter lesions (rs1064395/NCAN) or the proportion of remyelinated lesions (rs5742909/CTLA4). In addition, rs2234978/FAS T-allele carriers showed increased FAS gene expression levels in perivascular T cells and perilesional oligodendrocytes, cell types that have been implicated in MS lesion formation. 20


Conclusion By combining pathological characterization of MS brain autopsy tissue with genetics, we now start to translate genotypes linked to clinical outcomes in MS into mechanisms involved in MS lesion pathogenesis.

21


Specialized pro-resolving lipid mediator production in the cerebrospinal fluid is impaired in multiple sclerosis: implications for its pathogenesis and therapy Author Gijs Kooij1,2, Valerio Chiurchiù3,4, Sabela Rodriquez1, Paul Norris2, Claudio Derada Troletti1, Iliyan Vlassakov2, Suzanne van der Pol1, Bert van het Hof1, Alessandro Leuti3,4, Mohsen Khameni5, Tomas Olsson5, Fredrik Piehl5, Lou Brundin5, Ellen Lacobeus5, Horst Schroten6, Hiroshi Ishikawa7, Charlotte Teunissen8, Bart Eggen9, Britta Engelhardt10, Elga de Vries1, Charles Serhan2 1 Affiliation Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell

Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, The Netherlands, 2Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA, 3Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy, 4European Center for Brain Research, Santa Lucia Foundation, Rome, Italy, 5Department of Clinical Neuroscience, Karolinska Institutet Stockholm, Sweden, 6Pediatric Infectious Diseases, University Children’s Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany, 7Amsterdam UMC, Vrije Universiteit Amsterdam, Neurochemistry lab, Department of Clinical Chemistry, Amsterdam Neuroscience, The Netherlands, 8

Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of

Medicine, University of Tsukuba, Japan, 9Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, University of Groningen, The Netherlands, Theodor Kocher Institute, University of Bern, Switzerland

10

Background and objective The acute inflammatory response is host protective and efficient resolution of inflammation is required to prevent excessive inflammation and restore tissue homeostasis. This protective process is orchestrated by specialized pro-resolving lipid mediators (SPMs) that are biosynthesized from omega-3 fatty acids. In the chronic neuro-inflammatory disease multiple sclerosis (MS), the abundant presence of pro-inflammatory cells and wide-spread microglial activation within the central nervous system (CNS) suggests that this resolution process is impaired. Consequently, the uncontrolled inflammatory response will acquire a chronic nature, leading to severe tissue damage (neurodegeneration) and disease progression. To date, fundamental insights into the regulation of CNS resolution processes and whether impairments in this system correlate with MS progression remain elusive. Methods We used liquid chromatography-tandem mass spectrometry (LC-MS-MS)-based metabololipidomics to reveal lipid mediator signatures in the cerebrospinal fluid (CSF) of RRMS (either in relapse or remission), SPMS and PPMS patients as well as age/sex matched controls. 22


Results We identified unique lipid mediator signatures associated with MS clinical forms and provide first evidence for an altered resolution-inflammation pathway in MS. Specifically, we observed a reduction of SPMs like lipoxin B4 and resolvin D3 in various clinical disease stages along with high levels of classical eicosanoids. Epithelial cells from the choroid plexus were found to contribute to the production of SPMs and when isolated from post-mortem brain of MS patients to contain defects in SPM biosynthesis. SPM administration potently reduced the inflammatory profile of both human innate (microglia) and adaptive (Th1 and Th17) immune cells under inflammatory conditions and ameliorated clinical signs in an MS mouse model. Conclusion By using metabololipidomic profiling of human CSF, we here provide critical evidence of a failed resolution pathway in MS, suggesting new insights into the pathogenesis and providing innovative diagnostic and therapeutic approaches for this neurodegenerative disease.

23


SCIENTIFIC PRESENTATIONS 3 – UNDERLYING PROCESSES Friday 09:45 – 10:30 Moderators: dr. Wia Baron and dr. Gijs Kooij RNA-sequencing of laser-captured leukocortical lesions from multiple sclerosis patients Author Thecla van Wageningen1, Emma Gerrits2, Nieske Brouwer2, Bart Eggen2, Jeroen Geurts1, Erik Boddeke2, Anne-Marie van Dam1 1 Affiliation Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. Anatomy & Neurosciences,

MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands, 2

Department of Biomedical Sciences of Cells and Systems, section Molecular

Neurobiology, University Medical Center Groningen

White-matter demyelination in multiple sclerosis (MS) is accompanied by a dysfunctional blood-brain-barrier leading to infiltration of leukocytes into the central nervous system (CNS). In post-mortem material of MS patients, these leukocytes and amoeboid, activated microglia are present in white-matter lesions (WMLs). However, grey-matter lesions (GMLs) are almost devoid of infiltrated leukocytes and only show modest microglial reaction. What underlies these differences in immune activation and pathology in WMLs and GMLs is still poorly understood. The contrast between WML pathology and GML pathology is especially visible in type I leukocortical lesions. Contrary to subpial lesions or intracortical lesions, these lesions show demyelination in an area encompassing both WM and GM where the WM lesioned area shows microglial/macrophage activation and infiltration of leukocytes which is largely absent in the nearby GM lesioned area. Studying leukocortical lesions could be of great interest, especially to elucidate possible differences in pathological processes contributing to WML and GML formation. The fact that both WM and GM demyelination is present in the same lesioned area the effect of possible confounding factors like lesion location and time of lesion development is extremely limited. In this study, we conducted RNA-sequencing of WM and GM areas of leukocortical lesions obtained from 8 MS patients. Using laser capture microdissection, normal appearing WM and normal appearing GM, demyelinated WM and demyelinated GM were collected from cryopreserved tissue slides featuring a leukocortical lesion. This method allowed us to directly compare gene expression in WMLs and GMLs between patients but also differences in gene expression within the leukocortical lesion of one patient. Initial data analysis revealed differential expression of genes between WML and normal appearing WM and between GML and normal appearing GM. As was expected, pathway analysis of differentially expressed genes between lesioned and non-lesioned areas 24


showed a statistical overrepresentation of genes affected by demyelination (e.g. “axon ensheatment” and “myelination”). Furthermore, GMLs show higher expression of glial related oxidative phosphorylation genes than in WMLs. Additional, more detailed analyses are ongoing to further delineate the differences in (lesioned) white and grey MS CNS tissue.

25


Transcriptional profiling of human microglia reveals grey-white matter heterogeneity and multiple sclerosis-associated changes Author

Marlijn van der Poel1, Thomas Ulas2, Mark Mizee1, Cheng-Chih Hsiao3, Suzanne Miedema1, Adelia4, Karianne Schuurman1, Boy Helder3,5, Sander Tas3,5, Joachim Schultze2,6, JĂśrg Hamann1,3, Inge Huitinga1

1 Affiliation Neuroimmunology Research Group, Netherlands Institute for Neuroscience,

Amsterdam, The Netherlands, 2Genomics and Immunoregulation, LIMES Institute, University of Bonn, Germany, 3Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam, The Netherlands, 4Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands, 5Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, The Netherlands, 6PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases, University of Bonn, Germany.

Microglia are resident phagocytes of the central nervous system involved in the maintenance of brain homeostasis, yet their role in multiple sclerosis (MS) lesion initiation is unclear. We isolated microglia from normal-appearing tissue of 10 MS and 11 non-neurological control brain donors from both grey (GM) and white matter (WM) regions and performed RNA-sequencing to find possible early changes related to MS pathology. Microglia showed a clear region-specific profile, indicated by higher expression of type-I interferon genes in GM and higher expression of NF-ÎşB pathway genes in WM. MS WM microglia showed an increased expression of lipid metabolism genes, which relates to MS pathology, since active MS lesion-derived microglial nuclei showed similar altered gene expression. Microglia from MS GM showed increased expression of genes associated with glycolysis and iron homeostasis, possibly reflecting microglia reacting to iron depositions. Except for ADGRG1/GPR56, expression of homeostatic genes, such as P2RY12 and TMEM119, was unaltered in normal-appearing MS tissue, demonstrating overall preservation of microglia homeostatic functions in the initiation phase of MS. Early changes in normal-appearing MS tissue include the clustering of microglia in so-called nodules. We currently characterize MS microglial nodules by immunohistochemistry for molecules that are potentially involved in lesion development and apply laser dissection for latter transcriptional profiling to define their role in MS lesion initiation.

26


Cortical microglia in progressive MS acquire a unique phenotype in response to chronic meningeal inflammation Author

Lynn van Olst1, Carla Rodriguez Mogeda1, Svenja Kiljan2, Iliana Michailidou3, Carmen Picon Munoz4, Evelien Drost1, Susanne Van der Pol1, Steffi Jonk1, Frank Baas3, Geert Schenk2, Jeroen Geurts2, Jack van Horssen1, Elga de Vries1, Richard Reynolds4, Maarten Witte1

1 Affiliation Department of Molecular Cell Biology & Immunology, Amsterdam UMC, The

Netherlands, 2Department of Anatomy & Neurosciences, Amsterdam UMC, The Netherlands, 3Department of Clinical Genetics, Leiden University Medical Center, The Netherlands, 4Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK

Background Cortical neurodegeneration, which encompasses the major loss of synapses, dendrites and neuronal cell bodies, is widely accepted as the main contributor to clinical disability in progressive MS patients. However, remarkably little is known about the mechanisms underlying this cortical neuronal pathology. It is currently unknown whether cortical microglia actively contribute to neurodegeneration in MS, protect neurons from further damage or switch between both phenotypes. One of the main reasons for this gap in our knowledge is the absence of a proper animal model for MS-related cortical pathology. Methods & Results Here we extensively characterized cortical microglia in a large cohort of human postmortem brain tissue to show that in progressive MS, cortical microglia adapt a more ramified morphology, lose expression of homeostatic markers and upregulate proteins related to activation. This altered microglial phenotype, which is different from other agerelated neurodegenerative diseases, associates with local inflammation of the meninges and correlates with the extent of neuronal loss. Remarkably, microglia appear to physically interact with damaged neurons in MS cortex, which was previously described as a neuroprotective act. Secondly, we performed similar experiments in a novel animal model for cortical MS pathology. In this model, we induce chronic inflammation of the meninges overlying the rat cortex, resulting in cortical demyelination and neuronal loss. Similar to progressive MS, we observe increased morphological complexity of cortical microglia, upregulation of microglial activation markers and a close interaction with neuronal cell bodies. Conclusion Taken together, we conclude that chronic meningeal inflammation induces a unique cortical microglial phenotype in progressive MS patients. Whether these microglia contribute or try to salvage ongoing neurodegeneration warrants further study. 27


SCIENTIFIC PRESENTATIONS 4 – MONITORING MS Friday 11:45 – 12:45 Moderators: prof.dr.ir. Charlotte Teunissen and dr. Karim Kreft Serum neurofilament light chain in pediatric MS and other acquired demyelinating syndromes Author Yu Yi Wong1, Arlette Bruijstens1, Christian Barro2, Zuzanna Michalak2, Marie-José Melief3, Annet Wierenga3, Danielle van Pelt1, Rinze Neuteboom4, Jens Kuhle2*, Rogier Hintzen1,3* 1 Affiliation Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, The

Netherlands, 2Neurologic Clinic and Policlinic, University hospital Basel, Basel, Switzerland, 3Department of Immunology, Erasmus MC, Rotterdam, The Netherlands, 4

Department of Paediatric Neurology, Erasmus MC, Rotterdam, The Netherlands.

*shared last authors

Background Neurofilament light chain (NfL) is a biomarker for neuro-axonal damage and is elevated in CSF and serum of multiple sclerosis (MS) patients. An ultrasensitive single-molecule array (Simoa) has been developed to reliably detect NfL levels in serum. In adult MS, CSF and serum NfL (sNfL) are highly correlated and the levels are associated with future disease activity. We aimed to explore these associations in pediatric patients with a fist attack of acquired demyelinating syndromes (ADS). Methods In total, 102 children <18 years with a first attack of CNS demyelination and 23 age matched controls were included. Clinically definite MS (CDMS) was set as an endpoint for analysis. CSF NfL was tested by the commercially available ELISA (Uman Diagnostics); sNfL was tested with a Simoa assay. Hazard ratios (HR) were calculated with Cox regression analysis. Results Of the 102 patients, 47 (46%) were tested for CSF NfL. CSF and serum NfL correlated significantly in the total group (ρ 0.532, p<0.001) and even more significant in the subgroup of patients with future CDMS diagnosis (ρ 0.773, p<0.001). sNfL was higher in patients than in controls (geometric mean 36.1 pg/ml vs 6.1 pg/mL, p<0.001), and was highest in ADS presenting with encephalopathy (ADEM, n=28; 100.4 pg/mL), followed by patients without encephalopathy (non-ADEM) with future CDMS diagnosis (n=40; 32.5 pg/mL), and non-ADEM patients who remained monophasic (n=34; 17.6 pg/mL). sNfL levels higher than a median of 26.7 pg/mL at baseline are associated with a shorter time to CDMS diagnosis in non-ADEM patients (p=0.045). HR for CDMS diagnosis was 1.09 28


for each 10 pg/mL increase of sNfL, after correction for age, OCB and MRI parameters (p=0.012). Conclusion The significant correlation between CSF and serum NfL strengthens its reliability as a peripheral marker of neuroaxonal damage. Higher sNfL levels at baseline were associated with higher probability of future CDMS diagnoses in non-ADEM patients.

29


Functional imaging biomarkers of cognitive impairment in multiple sclerosis: a resting-state magnetoencephalography study Author Shanna Kulik*1, Ilse Nauta*2, Bouke Lith1, Eva Strijbis2,3, Prejaas Tewarie2, Linda Douw1, Arjan Hillebrand3, Cornelis Stam3, Jeroen Geurts1, Brigit de Jong2, Menno Schoonheim1 (*Shared first authorship) 1 Affiliation Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and

Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam The Netherlands, 2Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurology, MS Center Amsterdam, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam The Netherlands, 3Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Neurophysiology and MEG Center, De Boelelaan 1117, Amsterdam The Netherlands

Background Previous studies have highlighted the importance of functional neuroimaging measures as biomarkers for cognitive impairment in MS. However, it remains unclear which of the previously identified measures are most promising. Objective To determine which magnetoencephalography (MEG) measures best predict cognitive performance in MS, and to analyse the importance of more advanced functional brain measures (functional connectivity (FC) and network topology) relative to more basic measures (oscillatory activity). Methods Resting-state MEG recordings, structural MRI and neuropsychological assessments were analysed in 275 MS patients. Computed MEG measures included: peak frequency and relative power, average FC (phase lag index), clustering coefficient, path length and eigenvector centrality. Linear regression was applied to determine which combination of measures best related to cognitive performance. Results Worse cognitive performance was related to increased theta power (β=-0.356, p<0.001), and decreased delta power (β=0.265, p=0.002), as well as increased beta (β=-0.234, p=0.027) but decreased gamma eigenvector centrality (β=0.240, p=0.028), and decreased alpha1 clustering (β=0.146, p=0.011). FC measures were only related to cognition at trend level (theta and gamma PLI, p<0.1). The multivariate model (R2=0.233, p<0.001) only showed significant relations for oscillatory activity and network topology measures and both remained unique correlates of cognition. FC did not add explanatory power. 30


Conclusions Oscillatory activity and network topological properties uniquely correlated with cognition, whereas FC did not add explanatory value. This indicates that especially the combination of measures of functional activation and functional network topology hold promise as potential biomarkers.

31


Deciphering morphology of Evoked Potentials Author Jan Yperman1, Thijs Becker2, Dominique Dive2, Melissa Cambron4,5, Guy Laureys6, Dirk Valkenborg2, Niels Hellings7, Bart Van Wijmeersch7,8, Veronica Popescu7,8, Liesbet Peeters7 1 Affiliation Theoretical Physics, Hasselt University, Diepenbeek, Belgium, 2CENSTAT, Hasselt

University, Diepenbeek, Belgium, 3Neurology, CHU Liège, Esneux, Belgium, 4Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium, 5

Departement of Neurology, AZ Sint-Jan, Brugge, Belgium, 6Neurology, University

Hospital Ghent, Ghent, Belgium, 7BIOMED, Hasselt University, Diepenbeek, Belgium, 8

Revalidation and MS Center Pelt, Pelt, Belgium

Background Evoked Potentials Time Series (EPTS) are one of the tools neurologists employ for monitoring disease progression in multiple sclerosis (MS). The specific modality we consider here, called Motor Evoked Potentials (MEP), is measured by exciting the motor cortex and subsequently measuring the electrical activity in the muscles of the hands or feet. EPTS are usually summarized by three values: latency, peak-to-peak amplitude and the signal “morphology”. But currently, there is no tangible definition of what constitutes the morphology of the muscle response. Therefore, this feature remains highly dependent on the neurologist’s individual interpretation. With this study, we aimed to find a concise definition of this feature, based on the interpretation of a panel of MS neurologists. Given such a definition, extracting morphology from EPTS may be automated, which will allow unbiased and reproducible evaluation of the prognostic value of morphology using large multicenter and retrospective datasets. Methods We extracted ~400 000 EPTS from the Rehabilitation & MS Center in Pelt, Belgium. Following 2 consensus workshops, 5 neurologists from different MS centers in Belgium independently assigned a binary label to the same set of 1000 EPTS (normal vs abnormal “morphology”). In order to find a suitable proxy for the morphology, we extract ~5800 time series features from the EPTS. Using cross-validation, we then employ logistic regression on each of the extracted features to find a feature that best corresponds to the morphology. For the final performance estimate we evaluate a logistic regression model on an independent test set (consisting of 450 EPTS), with labels created by a majority vote of the 5 neurologists. Results The intra- and inter-rater agreement of the independent labeling was 90 +- 6 % and 79 +- 7 % respectively. One feature appeared consistently and independently in the top-10 features of each neurologist, namely approximate entropy, which is a measure 32


of the regularity of the time series. Initial results show an AUC of 0.92 +- 0.01 on the independent test set. Conclusion Morphology can be consistently defined by pooling the interpretations from multiple neurologists. Approximate entropy seems to be a valid proxy for morphology.

33


Next generation microRNA sequencing and targeted proteomics to find novel subtype specific biomarkers for multiple sclerosis Author

Ineke Tan1,2, Ania Stachurska, Rodrigo Almeida3, Rutger Modderman1, Cisca Wijmenga1,4, Jan Meilof5,6, Sebo Withoff1

1 Affiliation Department of Genetics, University of Groningen and University Medical Center

Groningen, 2Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, 3Department of Biomedical Data sciences, section Molecular epidemiology, Leiden University Medical Center, Leiden, Department of Immunology, K.G. Jebsen Coeliac Disease Research Centre, University

4

of Oslo, Norway, 5Department of Neurology, Martini Hospital Groningen, 6Department of Neurology, University Medical Center Groningen, Groningen

Introduction and aim Biomarkers that can be used to determine the onset of the secondary progressive phase in MS at an early stage, and thus would help to optimize MS subtype dependent treatment decisions, are currently missing. We examined whether targeted proteomics and microRNA (miRNA) transcriptomics analysis could address this issue and hypothesize how these biomarkers play a role in disease mechanisms. Material & Methods Pre mortem cerebrospinal fluid samples (CSF) and serum samples of 30 disease controls and MS patients (30 relapsing-remitting (RR-MS), 30 secondary-progressive (SP-MS), 10 primary-progressive (PP-MS)) were obtained from the Dutch Brain Bank. For proteomics analysis we quantitated 92 proteins using the Olink Proseek multiplex Immuno-oncology panel. For extracellular miRNA transcriptomics we generated miRNAseq libraries which were sequenced on the Illumina HiSeq2500. Results 68 out of 92 proteins were detected in >25% of samples in CSF and in 84 out of 92 in serum. In CSF 13 proteins showed significantly higher (11) or lower (2) levels in MS compared to controls, including the previously described MS biomarkers CD27 (3.4-fold change) and CXCL13 (2.9-fold change). Gene Set Enrichment Analysis (GSEA) of these 13 markers using STRING yielded 177 significantly enriched Gene Ontology (GO) terms, most of which were immune-related. In serum, 3 proteins were significantly different between groups. In the post-hoc analyses, all 3 proteins were decreased in RR-MS versus controls, but not different between SP-MS and controls. GSEA analyses of these 3 serum markers resulted in 34 significantly enriched GO-terms which were different than those for the CSF proteins (e.g. angiogenesis and extracellular matrix). 34


miRNA transcriptome analysis yielded 165 miRNAs in CSF and 281 miRNAs in serum. In CSF no miRNAs were significantly differentially expressed between MS and controls, but 7 miRNAs were significantly differentially expressed between SP-MS and RR-MS. In serum, 51 miRNAs were significantly DE between MS and controls (22 up and 29 downregulated in MS). Additionally, 8 miRNAs seem to be subtype specific markers, 5 for RR-MS and 3 SP-MS. Conclusions The results of our combined proteomics and miRNA transcriptomics analysis suggest that these approaches can be used to find new and even MS subtype specific biomarker candidates for MS. Acknowledgements This work was supported by a Stichting MS Research grant (grant 11-752), an European Research Council advanced grant (FP7/2007-2013/ERC Advanced Grant Agreement 2012-322698), a NWO Spinoza Prize (NWO SPI 92-266), a NWO Gravitation Netherlands Organ-on-Chip Initiative (024.003.001) and a MD/PhD scholarship from the Junior Scientific Masterclass, Universitair Medisch Centrum Groningen to I. Tan.

35


SCIENTIFIC PRESENTATIONS 5 – IMMUNE CELLS Friday 14:15 – 15:15 Moderators: prof.dr. Piet Stinissen and prof.dr. Jon Laman Regulatory T cells have a distinct migratory phenotype in relapsing-remitting multiple sclerosis patients Author

Paulien Baeten1, Niels Hellings1, Bieke Broux1

1 Affiliation Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium

Peripheral tolerance mainly consists of regulatory T cells (Tregs) suppressing autoreactive effector T cells (Teff) which have escaped central tolerance mechanisms. In many autoimmune diseases, including multiple sclerosis (MS), Tregs display a defective suppressive capacity. In MS, where autoreactive Teff are reactivated in the central nervous system (CNS), Treg migration to these local sites of inflammation is desirable. To date, the migratory behaviour of Tregs in MS is unclear. Here, using an in vitro model of the human blood brain barrier (BBB), we found that Tregs (thymus-derived, CD25hiCD127loFoxp3+) of untreated relapsing-remitting (RR) MS patients have a migration frequency of 9.4 ± 2.2% (mean ± SEM) across an inflamed BBB. This is comparable to healthy donors (HD) who have a migratory frequency of 8.6 ± 1.9%. In addition, analysis of paired blood and cerebrospinal fluid (CSF) samples of untreated RRMS patients shows comparable percentages of Tregs (CSF: 1.7 ± 0.5%; blood: 2.5 ± 0.5%), indicating their capability of migrating to the CNS. In blood of HD, we found that Tregs have a distinct migratory phenotype, with higher expression of CCR6 and lower expression of CD49d compared to Teff. Interestingly, CCR6 and CCR8 are downregulated on Tregs in RRMS patients compared to HD, while the expression is restored in secondary-progressive (SP) MS patients. In CSF, we found enrichment of MCAM+, CD49d+, CCR5+, CXCR3+ and CCR6+ Tregs compared to paired blood samples of untreated RRMS patients. The ligands of CCR6, CD49d and CXCR3, CCL20, VCAM-1 and CXCL10 and CXCL11 respectively, are significantly upregulated by inflamed BBB endothelial cells. Taken together, these results suggest that Tregs use a unique set of adhesion molecules and chemokine receptors to migrate to the CNS during MS. Further results of ongoing analyses (including FlowSOM analysis) will be presented at the meeting.

36


Inflammatory lesion activity in chronic progressive MS is related to the accumulation of tissue resident memory T cells in the perivascular space Author Nina Fransen1, Cheng-Chih Hsiao1,3, Marlijn van der Poel1, Jeen Engelenburg1, Soraya van Etten1, Kim Verdaasdonk1, Ester Remmerswaal3,4, Jörg Hamann1,3, Joost Smolders1,2, Inge Huitinga1 1 Affiliation Department of Neuroimmunology, The Netherlands Institute for Neuroscience,

Amsterdam, The Netherlands, 2MS Center CWZ, Department of Neurology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands, 3Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands, Renal Transplant Unit, Department of Internal Medicine, Amsterdam UMC, University of

4

Amsterdam, Amsterdam, The Netherlands

Background Multiple Sclerosis (MS) is a chronic inflammatory demyelinating disease, however it has been suggested that in the progressive late-phase of the disease inflammatory lesion activity is no longer related to disease progression. We recently showed that in the Netherlands Brain Bank (NBB) MS autopsy cohort consisting of 182 MS cases with an average disease duration of 29 ± 13 years (mean ± SD) years, 57% of all dissected lesions are inflammatory active or chronic active. This is suggesting that there is ongoing inflammatory lesion activity at the end stage of the disease. We now study the relation between the innate inflammatory lesion activity and T cells in chronic progressive MS cases. Methods We performed histological and flow-cytometric analysis of T cells in MS lesions and normal- appearing white matter (NAWM). Results Quantitative histological analysis for CD3 shows that in MS NAWM, the number of T cells is increased compared to control WM (5.03 ± 6.01 vs. 1.89 ± 2.51, respectively). In active and chronic active lesions, the number of T cells is further increased (22.21 ± 22.06 and 13.24 ± 13.87, respectively). MS lesion T cells are enriched for CD8positive T cells. In control WM and NAWM, CD8+ T cells are mostly located within the perivascular space, while in chronic active MS lesions, CD8+ T cells are more often located in the brain parenchyma. T cells from chronic active MS lesions show a tissue resident memory (TRM) profile, with expression of CD69, CD103, CXCR6, CD49a and PD1. Furthermore, perivascular T cell clusters in MS lesions consist of TRM cells with expression of CD103. MS cases that showed perivascular clustering of TRM cells were associated with a secondary progressive disease course, a higher percentage of chronic active lesions and a higher lesion load. 37


Conclusions These data suggest that in chronic progressive MS cases, inflammatory lesion activity is related to clustering of TRM cells within the perivascular space. In chronic active MS lesions, CD8+ TRM cells are infiltrating the brain parenchyma.

38


Human T-bet+ B cells: induction and effector functions from a multiple sclerosis perspective Author Jamie van Langelaar1,*, Liza Rijvers1,*, Malou Janssen1,2, Annet Wierenga-Wolf1, Marie-José Melief1, Theodora Siepman2, Helga de Vries3, Peter-Paul Unger4, Marieke van Ham4, Rogier Hintzen1,2, Marvin van Luijn1 1 Affiliation Depts. Immunology and 2Neurology, MS Center ErasMS, Erasmus MC, University

Medical Center, Rotterdam, The Netherlands, 3Dept. Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam,

3

Amsterdam, The Netherlands, 4Dept. Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands. *shared first authors

Background In multiple sclerosis (MS), peripheral B-cell tolerance checkpoints are defective and ectopic B-cell follicles are found in the central nervous system (CNS). However, which and how human B cells are triggered in the periphery to infiltrate and mediate pathology in the CNS remains unknown. Methods Thirteen color flow cytometry was used to assess chemokine receptor profiles of B cell populations in single-cell suspensions from paired blood, cerebrospinal fluid (CSF), meninges and brain of MS patients (n=10). Similar analyses were performed for distinct memory subsets in untreated and natalizumab-treated MS patients (n=38). To elucidate how the peripheral differentiation and function of T-bet(CXCR3)+ memory B cells are regulated in MS, we used an in vitro culture system to stimulate naive B cells from MS patients (n=21) and healthy individuals (n=34) with 3T3-CD40L cells, IL-21, IFN-γ and TLR9 ligand CpG-ODN. The CNS transmigration capacity of memory subsets was confirmed using brain endothelial monolayers. Results In this study, we show that CXCR3-expressing B cells are selectively enriched in paired CSF, meningeal and brain versus blood compartments of MS patients. Treatment with clinically effective drug natalizumab mainly prevented the recruitment of CXCR3high IgG1+ subsets, corresponding to their increased ability to cross CNS barriers in vitro. These subsets positively correlated with Th17.1 (IFN-γhighIL-17low) and not Th17 (IFN-γneg) cells in MS blood. IFN-γ produced by Th cells increased the transmigration potential and the antigen-presenting capacity of human CXCR3+ B cells. Furthermore, IFN-γ-induced B cells from MS patients showed increased T-bet expression and plasmablast development. TLR9 triggering of B cells further induced T-bet and CXCR3 levels, and was essential for IgG1 switching during Tfh1-dependent B-cell responses. 39


Conclusion These findings indicate that human T-bet-expressing IgG1+ B cells are strongly induced by Tfh1 and TLR9 signals in the periphery, likely contributing to enhanced CXCR3-mediated transmigration and local reactivity in the CNS of MS patients.

40


Contribution of risk allele CLEC16A to the B cell receptor-mediated HLA class II pathway in MS Author Liza Rijvers1, Marie-JosĂŠ Melief1, Jamie van Langelaar1, Roos van der Vuurst de Vries2, Annet Wierenga-Wolf1, Steven Koetzier1, Marieke van Ham3, Rogier Hintzen1,2, Marvin van Luijn1 1 Affiliation Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical

Center, Rotterdam, 2Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, 3Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam

Background Results from anti-CD20 therapies and large genome-wide association studies imply that B cells serve as potent antigen-presenting cells during MS pathogenesis. This was further supported by a recent study in MS showing that B cells from HLA class II (HLA-II) risk carriers are major inducers of pro-inflammatory, brain-infiltrating T cells (Jelcic, Cell 2018). HLA-II is also crucial for maintaining human peripheral B-cell tolerance, which is defective in MS patients. Previously, we found that MS-associated gene CLEC16A is a critical regulator of the HLA-II pathway. The current study aimed to uncover how HLA-II expression in B cells is regulated during disease onset and influenced by CLEC16A. Methods Naive and memory B-cell subsets of clinically isolated syndrome (CIS) and relapsingremitting MS (RRMS) patients were assessed for expression of key members of the HLA-II pathway, i.e. the invariant chain (CD74), class II-associated invariant chain peptide (CLIP) and HLA-DR. Similar analyses were performed after stimulation with several germinal center-dependent and -independent stimuli in vitro. Surface CLIP and HLA-DR were correlated to expression levels of CLEC16A and other risk alleles in B cells. B cells were silenced for CLEC16A using distinct lentiviral shRNA constructs and analyzed for IgM-dependent antigen internalization and processing. Results B cells from high-risk CIS patients and RRMS patients showed increased surface levels of CLIP-containing HLA-DR molecules and reduced levels of CD74 as compared to lowrisk CIS patients and matched healthy controls (n=9-12 per group). CLEC16A was only triggered under CLIP-stimulating conditions in vitro and mainly expressed in CLIPhigh naive B cells. CLEC16A knockdown in B cells (Âą50%) resulted in upregulation of HLA-DR and CD74, but not CLIP on the plasma membrane. Moreover, CLEC16A-silenced human B cells revealed impaired IgM-mediated antigen uptake and an extensive cytoplasmic scattering of specialized antigen-loading compartments termed MIICs. This indicates that 41


CLEC16A is important for the processing of BCR/antigen and HLA-II/CD74 complexes in MIICs. Conclusion This work demonstrates that the HLA-II pathway in B cells is dysregulated in rapid-onset MS patients and is functionally controlled by CLEC16A. The abundance of CLIP-loaded HLA-II molecules already on naive B cells mechanistically links to their escape from peripheral tolerance checkpoints in MS.

42


Abstracts poster presentations POSTER WALK A – MONITORING (SYMPTOMS OF) MS Thursday 15:45 – 16:45 hours Moderators: dr. Thea Heersema and dr. Hugo Vrenken Specific cerebellar atrophy patterns can distinguish physical disability and cognitive impairment in MS Author Iris Dekker1,2, Aurélie Ruet3, Myrte Strik4,5, Hanneke Hulst4, Mike Wattjes1,6, Frederik Barkhof1,7, Bernard Uitdehaag2, Joep Killestein2, Jeroen Geurts4, Menno Schoonheim4 1 Affiliation Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear

Medicine, 2Neurology, 4Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, The Netherlands. 3University of Bordeaux, Bordeaux, France/Inserm U1215, Neurocentre Magendie, Bordeaux, France/ CHU Pellegrin, CHU de Bordeaux, Bordeaux, France, 5Department of Radiology and Medicine, University of Melbourne, Melbourne, Australia, 6Dept. of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany, 7Institutes of Neurology and Healthcare Engineering, UCL, London, UK.

Introduction Cerebellar atrophy is known to be related to clinical dysfunction in multiple sclerosis (MS), although it is unclear how cognitive or physical symptoms might be explained by specific cerebellar atrophy patterns. Aim To identify regional cerebellar atrophy patterns specific to physical disability and cognitive impairment. Methods Of the Amsterdam MS cohort, 331 patients (73.1% relapsing-remitting MS, disease duration 14.6±8.4 years) and 95 healthy controls (HCs) were included. Clinical assessments included an expanded BRB-N and the Expanded Disability Status Scale (EDSS). Cerebellar gray matter was parcellated with SUIT, clustered into anterior lobes (I-V) and posterior lobes (VI-X), dentate, interposited and fastigial nuclei. Backward linear regression models assessed the relations of atrophy with physical disability and cognition, controlling for age, sex and education. Post-hoc models explored relations with individual cerebellar regions. Results Cognitive impairment, Z=-1.5 on ≥2 domains, was seen in 42% of patients. All clustered regions and nuclei showed MS-related atrophy compared to HCs. Physical disability was associated with age and education, and volumes of the dentate nucleus, anterior and 43


posterior lobes (particularly lobule V and vermis) (adjusted R2=0.33). Cognition was associated with age, sex and education, and atrophy of the posterior lobe (particularly lobule VIIb) and interposited nucleus (adjusted R2=0.19). Atrophy of lobule VIIb most strongly correlated with working memory and information processing speed; interposited nucleus volume most strongly correlated with visuospatial memory, verbal memory and executive functioning. Conclusion Although cerebellar atrophy is generalized in MS patients, specific atrophy patterns of cerebellar subregions are related to either physical disability (anterior regions) or cognitive impairment (posterior regions).

44


Myelin imaging with [11C]MeDAS PET Author

Kars van der Weijden1, Jan Meilof2,3, Rudi Dierckx1, Erik de Vries1

1 Affiliation Department of Nuclear Medicine and Molecular Imaging, University of Groningen,

University Medical Center Groningen, Hanzeplein 1, Groningen, the Netherlands, Department of Neurology, University of Groningen, University Medical Center Groningen,

2

Hanzeplein 1, Groningen, the Netherlands, 3Department of Neurology, Martini Ziekenhuis, Groningen, the Netherlands

Introduction and aim Magnetic resonance imaging (MRI) is important for the diagnosis of Multiple Sclerosis (MS), but lacks the specificity needed for evaluating demyelination and remyelination. [11C]MeDAS Positron Emission Tomography (PET) scanning was found to be a promising method for in vivo myelin quantification in animal models, but has not been used in humans yet. Our study aims to evaluate the feasibility of myelin imaging with [11C] MeDAS PET in MS patients. Material & Method A total of 10 MS patients will be included in the study. Following intravenous injection of [11C]MeDAS, patients will receive a 60-min dynamic brain PET scan and subsequently a 10 min static spinal cord PET scan. Concomitant arterial blood sampling will be performed to obtain a metabolite-corrected plasma input function. Regional time–activity curves will be generated from the PET images using pMOD (version 4.002). These curves will be analysed using several pharmacokinetic models. In addition, lesion segmentation of [11C] MeDAS PET will be compared with MRI. Some preliminary results will be presented. Results Patient inclusion is still on-going. Visual assessment of tracer binding in the first patient indicates that the tracer primarily binds to white matter. This was confirmed by quantitative analysis, showing a higher volume of distribution in white than in grey matter. Furthermore, rapid metabolism of [11C]MeDAS was observed, with 73% of the tracer being metabolized after 10 min. The kinetics of the tracer was best described by a reversible two tissue compartment model (AIC 20.9). Lesion volumes detected with [11C] MeDAS PET were smaller than detected with MRI. Conclusions The first in human [11C]MeDAS scan was successfully performed. No adverse events occurred and both the scan protocol as the scan itself were well tolerated. Preliminary results indicate that [11C]MeDAS binds primarily to white matter and is rapidly metabolised, which leads to a fast clearance from the body. [11C]MeDAS kinetics are well described by a reversible two tissue compartment model. More precise data will follow after more inclusions.

45


Subjective visual complaints in patients with multiple sclerosis Author Fleur van der Feen1,2, Gera de Haan1,2, Iris van der Lijn1,2, Famke Huizinga1, Bart MelisDankers2, Anne Vrijling2, Nynke Stellingwerf2, Thea Heersema3, Jan Meilof4, Joost Heutink1,2 1 Affiliation Clinical and Developmental Neuropsychology, University of Groningen, Groningen,

Royal Dutch Visio, Centre of expertise for blind and partially sighted people, Huizen,

2

University Medical Centre Groningen, Groningen, 4Martini Hospital Groningen,

3

Groningen

Background and Objectives While care for patients with multiple sclerosis is improving, vision problems may be underestimated and patients do not always receive appropriate care for these complaints. This is striking as vision problems may have a vast impact on quality of life and as vision is perceived by multiple sclerosis patients as extremely valuable. The aim of this study is to map the prevalence, nature and severity of subjective visual complaints. A better understanding of these complaints will improve rehabilitation for people with multiple sclerosis. Method We developed a 19-item questionnaire to screen for subjective visual complaints, which was administered to 255 patients with multiple sclerosis in two Dutch hospitals. Results Almost 90% of patients reported to have one or more visual complaints. The most common complaints were blurry vision, having difficulty focusing and photophobia, which were all experienced by more than half of the patients. In addition, needing more time to perceive visual information, having difficulty with reading or adapting to light or darkness and a reduced contrast sensitivity were commonly experienced. The two least common complaints were having difficulty finding and searching and experiencing a distorted image, but were nevertheless experienced by at least 10% of the patients. Conclusions Visual complaints are very common among multiple sclerosis patients and a wide range of visual complaints is experienced. Recognition and knowledge of these visual complaints can facilitate high quality rehabilitation that can be specially designed for these patients.

46


Outdoor mobility of persons with multiple sclerosis. A systematic review Author Fleur van der Feen1,2, Gera de Haan1,2, Iris van der Lijn1,2, Joost Heutink1,2 Affiliation Clinical and Developmental Neuropsychology, University of Groningen, Groningen, Royal Dutch Visio, Centre of expertise for blind and partially sighted people, Huizen

Background and objectives Multiple sclerosis can manifest itself in many ways, all of which can affect the outdoor mobility of persons with multiple sclerosis (pwMS). In turn, a reduced mobility can have a great impact on quality of life. In most studies, the mobility of pwMS is defined by the ability to walk. However, other types of mobility are also important to explore. The aim of this review was to synthesize the existing literature on several types of mobility in pwMS. Methods Three data bases were used (PubMed, PsychInfo and Web of Science). Search terms such as ‘driving’, ‘wheelchair’, and ‘public transportation’ were combined with ‘multiple sclerosis’. Using the databases and search terms, systematic literature searches was performed on the 1st of September 2017 and the 3rd of December 2018. Studies were included if a group of patients with multiple sclerosis and some type of mobility was described. In total, 58 studies were included which in total described 9180 pwMS and their mobility. Results The literature showed that compared to the general population, pwMS have a decreased fitness to drive, make use of a wheelchair or mobility scooter more often and have more difficulties making use of public transport. Mobility problems may especially occur in patients with secondary progressive multiple sclerosis, high disability or cognitive problems, such as attention and speed of processing deficits. It was also found that a reduced mobility may have negative implications on daily life in numerous ways. Conclusions Few studies investigated interventions or rehabilitation options to improve mobility of pwMS. However, valuable insights that could help develop rehabilitation programs were extracted from the existing literature. In addition, longitudinal studies should be performed in order to predict which patients could benefit from which kind of mobility. Multiple sclerosis can affect the mobility of pwMS in different ways and the reduced mobility can stand in the way of participating in society. Interventions or rehabilitations options to improve mobility should be explored.

47


Thalamic volume as a measure to discriminate between subjective and objective cognitive complaints in MS: towards clinical application? Author

Marijn Huiskamp1, Brigit de Jong2, Eileen Stalman1, Martijn Steenwijk1, Ilse Nauta2, Sanne de Geus-Driessen3, Ilona Reuling3, Hugo Vrenken4, Vincent de Groot5, Martin Klein3, Bernard Uitdehaag2, Jeroen Geurts1, Menno Schoonheim1, Hanneke Hulst1

1 Affiliation Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. Anatomy and Neurosciences,

MS center Amsterdam, Amsterdam Neuroscience, the Netherlands, 2Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. of Neurology, MS center Amsterdam, Amsterdam Neuroscience, the Netherlands, 3Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. of Medical Psychology, Amsterdam Neuroscience, the Netherlands, 3Amsterdam UMC, Vrije Universiteit Amsterdam, 4Dept. of Radiology and Nuclear Medicine, MS center Amsterdam, Amsterdam Neuroscience, the Netherlands, 5Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. of Rehabilitation Medicine, the Netherlands

Introduction Thalamic atrophy occurs early in MS and relates strongly to cognitive impairment. However, it is unknown whether thalamic volume is able to predict the presence of cognitive disturbances in a clinical setting. Aim To determine whether thalamic volume can be used to distinguish cognitively impaired (CI) from preserved (CP) patients in a real-life sample presenting with subjective cognitive complaints and to define the cut-off value for thalamic volume with the highest discriminatory value. Methods 52 MS patients and 76 healthy controls (HC) underwent tests for verbal and visuospatial memory, information processing speed and verbal fluency. CI was defined as ≼2 tests with scores ≤-2 standard deviations below the scores of HCs. MRI scanning was performed at 3T. Thalamic volumes of MS patients were corrected for head size, adjusted for age and converted to z-scores. Receiver Operating Characteristic analysis was performed to determine optimal thalamic cut-offs. Results 23.1% of MS patients (12/52) were defined as CI. Thalamic volume was significantly lower in CI compared to CP patients (P<.001). The optimal thalamus volumetric cutoff was at -2.3 SD, at which 9/12 patients were correctly classified as CI and 28/40 were correctly classified as CP (i.e. sensitivity=75%, specificity=70%). Positive Predictive Value was 42%, Negative Predictive Value was 90% (NPV) and area under the curve was 77%. 48


Conclusion With a moderate sensitivity and specificity and a high NPV, thalamic volume serves as an indication for cognitive preservation rather than impairment. Consequently, preservation of thalamic volume might indicate that neuropsychological examination will have limited added value. In the future, the detection of cognitive impairment may be improved by taking into account more (sub)cortical volumes and other factors that are of influence, such as cognitive reserve.

49


Perceived Neuropsychological Impairment, Depression, Health and Employment status in Multiple Sclerosis Author Marie D’hooghe1-3, Alexander De Cock1-3, Ralph Benedict4, Jeroen Gielen1, Ann Van Remoortel1, Piet Eelen1, Annick Van Merhaegen3, Jacques De Keyser2,3, Miguel D’haeseleer1-3, Erika Peeters1, Guy Nagels1,2,3,5 1 Affiliation National MS Center, Neurology, Vanheylenstraat 16, 1820 Melsbroek, BE, 2Vrije

Universiteit Brussel (VUB), Center for Neuroscience (C4N), Laarbeeklaan 101, 1090 Brussel, BE Brussel, 3UZ Brussel (VUB), Neurology, Laarbeeklaan 101, 1090 Jette, BE, 4

SUNY Buffalo, Neurology, BGH 100 High Street Suite D6, Buffalo, NY, USA 14226,

5

Oxford University, St Edmund Hall, Queen’s Ln, Oxford, UK OX1 4AR

Background MS patients frequently report cognitive difficulties with impact on daily functioning. Objective Investigate the interrelation between self-assessed neuropsychological impairment and depressive symptoms, as well as their relationship with disease-related variables, quality of life, health promoting behaviors, social network satisfaction and employment. Method A large 2-center survey included the MS Neuropsychological Questionnaire (MSNQ), 2-question screening tool for depression, vitality, Health-Related Quality of Life, HealthPromoting Lifestyle Profile II and questions assessing social network satisfaction and employment status with MS variables being retrieved from the database. Results Two thirds of 751 respondents reported either perceived neuropsychological impairment or depressive symptoms, with an overlap in about one quarter. While depressive symptoms were related to higher MSNQ scores, the MSNQ was not a good predictor of depression. The MSNQ and depression screening did not correlate with EDSS scores. After correcting for age, disease duration, sex, education and EDSS, the MSNQ scores and depression response significantly contributed to health outcomes and a low social network satisfaction. In participants below retirement age, the MSNQ, EDSS, age and education significantly contributed to the probability of unemployment. Conclusion The unique explanatory power with regard to unemployment illustrates the relevance of the MSNQ when measuring the impact of MS.

50


BIA study: a prospective observational multi-centre study on alterations of gut microbiome and the brain-immune-intestine axis in patients with relapsing remitting Multiple Sclerosis who start treatment with oral Cladribine Author

Jeske van Pamelen1, Leo Visser1, on behalf of the BIA study group, Janet de Beukelaar2, Jiske Fermont3, Stefan Frequin4, Koen de Gans5, Raymond Hupperts6, Jop Mostert7, Bob van Oosten8, Wim Verhagen9, Judith van Vliet10

1 Affiliation Department of Neurology, Elisabeth-TweeSteden Ziekenhuis, Tilburg, 2Department of

Neurology, Albert Schweitzer Ziekenhuis, Dordrecht, 3Department of Neurology, Amphia Ziekenhuis, Breda, 4Department of Neurology, St. Antonius Ziekenhuis, Nieuwegein, Department of Neurology, Groene Hart Ziekenhuis, Gouda, 6Department of Neurology,

5

Zuyderland Ziekenhuis, Sittard, 7Department of Neurology, Rijnstate Ziekenhuis, Arnhem, 8Department of Neurology, VU Medisch Centrum, Amsterdam, 9Department of Neurology, Canisius Wilhelmina Ziekenhuis, Nijmegen,

Department of Neurology,

10

Jeroen Bosch Ziekenhuis, ‘s Hertogenbosch

Background & objective Environmental factors are known to be important in the pathogenesis of Multiple Sclerosis (MS). Recent findings show that the intestinal microbiota in MS patients is different from controls and different between patients with relapses and without relapses. The microbiota plays an important role in shaping the immune system and recent studies suggest an association between the gut microbiome and inflammatory pathways in the central nervous system. Substitution of certain microbial populations in the gut can lead to a pro-inflammatory state. However, the connection between microbiota, treatment and changes in immunity has not been examined well yet. Our goal is to examine the effect of oral Cladribine on the gut, oral microbiota and the immunological system and to determine if the change in gut and oral microbiome in the first 3 months after start of oral Cladribine is a predictor for treatment response. Methods This is a prospective, observational, multi-centre study. Eligible subjects are patients with relapsing remitting MS (RRMS) between the age of 18 and 55 years who will start treatment with oral Cladribine. Patients who used probiotics one month prior to start of oral Cladribine are excluded. At baseline (before start), after 3, 12 and 24 months the Expanded Disability Status Scale (EDSS) will be assessed and a fecal, oral and blood sample will be collected. Also, subjects will be asked to register their food intake for 7 consecutive days following the visits. After 24 months a MRI of the brain is performed. Responders are subjects without relapses, without progression on EDSS and without radiological progression on MRI.

51


Results Inclusion started from January 2019. Five patients are included at the moment. The aim is to include 80 patients in 10 participating centres during a period of approximately 24 months. Conclusion This study will show us if patients with active RRMS on oral Cladribine, with or without freedom of disease after two years, have a distinct microbial gut and oral profile and immune response.

52


POSTER WALK B – UNDERLYING PROCESSES Thursday 15:45 – 16:45 hours Moderators: drs. Nina Fransen and dr. Marvin van Luijn The association of Epstein-Barr virus load with CNS-infiltrating B cells in MS: lessons learned from current treatments Author Jamie van Langelaar1, Liza Rijvers1, Annet Wierenga-Wolf1, Marie-José Melief1, Andrew Bell3, Menno van Zelm4, Rogier Hintzen1,2, Marvin van Luijn1 1 Affiliation Departments of Immunology and 2Neurology, MS Center ErasMS, Erasmus MC,

University Medical Center, Rotterdam, The Netherlands, 3Institute of Cancer and Genomic Sciences, University of Birmingham, United Kingdom, 4Department of Immunology and Pathology, Monash University, Melbourne, Australia.

Background The Epstein-Barr virus (EBV) is considered as an important causative trigger in MS. It is proposed that certain EBV-infected B cells escape control by EBV-specific CD8+ T cells to infiltrate the MS brain. This role is exemplified by massive B-cell recruitment and EBV reactivation in the brain, contributing to severe MS rebounds after discontinuation of natalizumab (anti-VLA-4). Moreover, the novel MS drug anti-CD20 is used to successfully treat both EBV-related post-transplant lymphoproliferative disorders and MS. Our ongoing work reveals that blood CXCR3+IgG+ B cells accumulate 1 year after natalizumab treatment and have an enhanced capacity to recruit to the CNS of MS patients. How changes in EBV load associate with B-cell transmigration in MS remains unknown. Methods To elucidate this, we used peripheral blood of MS patients before and after natalizumab treatment (n=10) or hematopoietic stem cell transplantation (HSCT; n=10). Four patients showed EBV reactivation after HSCT. Thirteen-color flow cytometry was used to assess chemokine and macrophage migration inhibitory factor receptors on naive and memory B-cell subsets. In addition, total and memory B cells were sorted from these patients to determine EBV DNA copy numbers using a highly sensitive qPCR assay. Results CXCR3 expression further increased on IgG+ B cells that were trapped in the blood during long-term natalizumab therapy (2-4 years). This was not the case for other migration markers and particularly seen for patients who clinically responded. Similar results were obtained with respect to EBV load in B cells. Only in EBVhigh patients, the accumulation of EBV DNA correlated to CXCR3+ B-cell frequencies and was confined to memory B cells during treatment. In MS patients who received HSCT treatment, both EBV load 53


and CXCR3 surface expression were enhanced in switched memory B cells after EBV reactivation, which was not observed in patients without EBV reactivation. Conclusion This study implies that EBV DNA especially accumulates in memory B cells that are prone to migrate into the CNS of MS patients. Whether and how this accumulation of EBV actually contributes to the pathogenic role of CXCR3(T-bet)+ B cells in MS needs to be further explored.

54


IgD-CD27- double negative (DN) B cells of MS patients are mature memory cells that can migrate towards pro-inflammatory chemokines Author

Lien Beckers1, Gwendoline Montes Diaz1, Luisa Villar2, Bart Van Wijmeersch1,3, Veerle Somers1, Judith Fraussen1

1 Affiliation Hasselt University, Biomedical Research Institute and Transnationale Universiteit

Limburg, School of Life Sciences, Hasselt, Belgium, 2Department of Immunology, Hospital Universitario Ramรณn y Cajal, Madrid, Spain, 3Rehabilitation & MS-Center, Pelt, Belgium

Recently, we reported abnormal elevations of age-associated IgD-CD27- double negative (DN) B cells with pro-inflammatory characteristics in the peripheral blood of a proportion of MS patients (Claes N. et al. J Immunol 2016). This study aimed to further investigate the phenotype and pro-inflammatory function of DN B cells in MS. The distribution of immunoglobulin (Ig) isotypes, developmental markers (CD5, CD10, CD38, CD95) and chemokine receptors (CXCR3, CXCR5) was measured on peripheral blood DN, IgD-CD27+ class-switched memory (CSM) and IgD+CD27- naive B cells of healthy controls (HC, n = 48) and MS patients (n = 96) by flow cytometry. Expression of the transcription factor T-bet was measured in DN B cells of MS patients (n = 58). Using an in vitro chemotaxis assay, migration of peripheral blood CD27- or CD27+ B cells of MS patients (n = 8) towards chemokines CXCL10 or CXCL13 was measured. Most DN B cells were class-switched although IgA+ cells were less frequent in the DN versus CSM B cell population for HC and MS patients (p < 0.0001). DN B cells resembled CSM B cells in developmental marker expression, although the DN B cell population had a higher frequency of immature CD5+ (p = 0.007 HC, p = 0.0003 MS) and CD38+ (p = 0.0001 HC, p = 0.01 MS) cells and a lower frequency of activated CD95+ (p < 0.0001) cells. T-bet expression was found in 21.6 [3.3, 52.0] % of MS DN B cells. The pro-inflammatory chemokine receptors CXCR3 and CXCR5 were expressed in 20% and 70% of DN B cells, respectively. In addition, MS DN B cells showed high migration towards CXCL10 and CXCL13 (CXCR3 and CXCR5 receptor, respectively) that was comparable to that of CSM B cells. Thus, DN B cells resemble CSM B cells but are at an earlier maturation state. The potential importance of DN B cells in MS pathology was further underlined by their high migration capacity towards pro-inflammatory chemokines.

55


Differential responses of developing and mature grey and white matter oligodendrocytes to pro-inflammatory cytokines Author

Jacomien Jongsma, Wia Baron

Affiliation Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands

Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system characterized by inflammation and neurodegeneration. Remyelination is the regeneration of myelin membranes and is a natural response to demyelination that is more efficient in grey matter (GM) than white matter (WM) lesions. Also in MS demyelination is not always permanent, given the presence of remyelinated lesions. However, remyelination often fails at later stages of the disease. In previous work, we have shown that cultured neonatal GM oligodendrocyte progenitor cells (OPCs) are less mature and less sensitive to interferon-gamma (IFNγ) than WM OPCs, and therefore may be better equipped for remyelination. Here, we extended these studies to more mature oligodendrocyte stages and examined whether transient exposure to pro-inflammatory cytokines elicits also different responses in developing and mature GM and WM oligodendrocytes. Our preliminary findings revealed that IFNγ, decreased the metabolism both in developing and mature GM and WM oligodendrocytes, an effect that was potentiated by simultaneous exposure to tumour necrosis factor alpha (TNFα). In addition, IFNγ, but not TNFα perturbed the differentiation of developing GM and WM oligodendrocytes. TNFα potentiates the IFNγ-induced inhibition of developing GM, and to a lesser extent WM, oligodendrocytes. In more mature GM oligodendrocytes TNFα slightly, but significantly, reduces the number of MBP-positive cells, indicating decreased differentiation or dedifferentiation. These findings indicate that mature GM oligodendrocytes appear more sensitive to pro-inflammatory cytokines than WM oligodendrocytes, in contrast to what was observed in OPCs. We are currently investigating the underlying mechanism(s) of the distinct response of regional oligodendrocytes to pro-inflammatory cytokines. A better understanding of regional oligodendrocyte biology in a demyelinating and inflammatory environment will aid to the development of therapeutics that overcome remyelination failure and prevent demyelination.

56


Towards the suppression of glucocorticoid-resistant T cells driving multiple sclerosis disease activity Author

Steven Koetzier1,4, Jamie van Langelaar1,4, Katelijn Blok2,4, Annet Wierenga-Wolf1,4, MarieJosĂŠ Melief1,4, Erik Lubberts3, Rogier Hintzen1,2,4, Marvin van Luijn1,4

1 Affiliation Departments of Immunology, 2Neurology, 3Rheumatology and 4MS Center ErasMS at

Erasmus MC, University Medical Center, Rotterdam, The Netherlands

Backgrounds We recently demonstrated that Th17.1 cells are key drivers of early multiple sclerosis (MS) activity (van Langelaar et al, Brain 2018). Th17.1 cells express high levels of the multidrug resistance receptor 1 (MDR1[ABCB1]), which makes them resistant to glucocorticoids (GC). Interestingly, GC responses by immune cells are reduced in MS patients and can be enhanced by vitamin D in vitro. How vitamin D and GC sensitivity of pathogenic Th cells is regulated in MS remains elusive. Here, we assessed the expression of vitamin D- and GC-associated genes amongst distinct CCR6+ Th subsets and how this relates to MS disease activity. Methods Th17 (CCR6+CCR4+CXCR3-), Th17.1 (CCR6+CCR4-CXCR3+) and Th17 doublepositive (DP; CCR6+CCR4+CXCR3+) cells were isolated from blood of healthy donors and natalizumab(NTZ)-treated MS patients using FACS. By the use of NTZ-treated MS blood, we are able to capture pathogenic Th17.1 cells that normally infiltrate the CNS. Expression levels of key genes involved in the vitamin and GC pathway were determined by qPCR. MDR1+ and MDR1- Th17.1 cells were analyzed for their pro-inflammatory and migratory potential as well as their presence in MS CSF. Results The vitamin D receptor (VDR) and vitamin D-induced gene DDIT4 were lower expressed in Th17.1 compared to Th17 and Th17 DP cells from healthy individuals, while no differences were found for vitamin D metabolism gene CYP27B1. DDIT4 has been proposed to act as a positive regulator of the glucocorticoid receptor (GR[NR3C1]). Consistently, NR3C1 was downregulated in Th17.1 cells, which together with its high ABCB1 expression, reflects a coordinately regulated mechanism of GC resistance. In NTZtreated MS patients, Th17.1 cells revealed higher ABCB1 and lower NR3C1 expression than those from matched healthy controls. This abundance of ABCB1 was seen in clinical responders, but not in non-responders to NTZ treatment. Finally, MDR1+ Th17.1 cells showed increased IFN-Îł, GM-CSF, VLA-4 and CCR6 expression compared to MDR1counterparts and were enriched in MS CSF versus paired blood.

57


Conclusion The impaired vitamin D signaling pathway, selective targeting by NTZ and local enrichment of pro-inflammatory, GC-resistant (MDR1highGRlow) Th17.1 cells, further defines this subset as a key orchestrator of MS disease activity.

58


Immunoglobulin repertoire analysis indicates a common origin of age-associated IgD-CD27- double negative (DN) B cells in healthy individuals and MS patients Author

Judith Fraussen1, Susanna Marquez2, Kazushiro Takata3, Chryssa Zografou3, Bart Van Wijmeersch1,4, Kevin O’Connor3,5, Steven Kleinstein2,5,6, Veerle Somers1

1 Affiliation Biomedical Research Institute, Hasselt University and School of Life Sciences,

Transnationale Universiteit Limburg, Hasselt, Belgium, 2Department of Pathology, Yale School of Medicine, New Haven, CT, USA, 3Department of Neurology, Yale School of Medicine, New Haven, CT, USA, 4Rehabilitation & MS-Center, Pelt, Belgium, 5Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA, 6Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA

Immune aging occurs in the elderly and in a proportion of multiple sclerosis (MS) patients. We recently described age-associated IgD-CD27- (double negative, DN) B cells with pro-inflammatory characteristics to be abnormally elevated in a proportion of MS patients (Claes N. et al. J Immunol 2016). Here, the origin and selection characteristics of DN B cells were studied in MS patients and healthy controls (HC). High-throughput adaptive immune receptor repertoire sequencing (AIRR-seq) was performed on peripheral blood DN and IgD-CD27+ memory B cells of MS patients and HC (n = 3 each). A total of 287,944 unique error-corrected heavy (H) and light (L) chain sequences were analyzed for isotype usage, clonality, variable region segment usage, mutational profiles and complementarity determining region 3 (CDR3) physicochemical properties. DN B cells from HC and MS patients showed similar repertoire characteristics, which indicates a common developmental pathway. In comparison with IgD-CD27+ memory B cells, DN B cells class switched to IgA less often and showed modestly lower clonal diversity. Shared clones were found between DN and IgD-CD27+ memory B cells, although > 95 % of the clones were unique to each population. DN B cells harbored fewer somatic mutations than IgD-CD27+ memory B cells, with IgG+ cells being more mutated than IgM+ cells. Differences in V(D)J usage and CDR3 physicochemical properties between IgG+, IgM+ DN and IgD-CD27+ memory B cells were observed. Thus, DN B cells arise in HC and MS patients via a common developmental pathway that is probably linked to immune aging. Immunoglobulin repertoire differences suggested unique differentiation pathways and responsiveness to different antigenic stimuli for DN and IgD-CD27+ memory B cells.

59


Heterogeneity in oligodendrocyte progenitor cells derived from cortex and corpus callosum Author

Dennis Lentferink, Marissa Dubbelaar, Inge Werkman, Bart Eggen, Wia Baron

Affiliation Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands

In the chronic demyelinating disease multiple sclerosis (MS) remyelination ultimately fails despite the presence of oligodendrocyte progenitor cells (OPCs) in most lesions. Since remyelination is crucial for functional neurological recovery and axonal survival, a therapeutic treatment aimed at restoring remyelination is a prerequisite for halting disease progression. Interestingly, spontaneous remyelination is more pronounced in grey than in white matter lesions. We have previously shown that, even after prolonged culture in vitro, neonatal cortical OPCs are less mature than neonatal non-cortical OPCs, and show increased proliferation and decelerated differentiation. This indicates that differences in remyelination capacity may be attributed to intrinsic differences in regional OPCs. Here, we investigated inherent molecular differences between freshly isolated neonatal grey (cortex) and white (corpus callosum) matter OPCs. OPCs were isolated from cortex and corpus callosum of postnatal day 7 rats via anti-A2B5 magnetic bead isolation. 3’-RNA sequencing was performed and the transcriptomic profile of these cells was compared. Our results show that expression levels of oligodendrocyte differentiation markers were lower in freshly isolated cortical OPCs than in OPCs from the corpus callosum. In addition, P7 cortical OPCs show enriched levels of Wnt-pathway components, and corpus callosum derived OPCs show higher expression levels of gene clusters involved in primary cilia formation, a cell organelle involved in signal transduction. Next, we investigated the expression of these primary cilia in our well-defined P2 cortical and non-cortical shake-off OPCs during differentiation. We found that primary cilia expression differs between regional OPCs; cortical OPCs express primary cilia earlier and in higher numbers compared to their non-cortical counterparts. Regional OPCs display their own intrinsic identity. Cortical OPCs appear to be less mature and, since OPCs revert to a more immature state before remyelination, may therefore be better equipped for remyelination. Also, the expression of primary cilia differs between regional OPCs, the putative role of which in myelination is currently under investigation. Exploring the molecular processes that differ between regional OPCs, including their aging, will lead to a better understanding of the process of remyelination, and may open therapeutic avenues to enhance remyelination in MS lesions. 60


Cytotoxic CD4 T cells escape suppression by regulatory T cells while promoting suppression of conventional helper T cells Author Cindy Hoeks, Marjan Vanheusden, Liesbet Peeters, Piet Stinissen, Bieke Broux, Niels Hellings Affiliation Hasselt University, Biomedical Research Institute and Transnationale Universiteit Limburg, Diepenbeek, Belgium

A terminally differentiated subset of CD4 T helper cells, characterized by loss of the costimulatory molecule CD28 and gain of cytotoxic activity, arises during aging and chronic inflammation. An age-inappropriate expansion of these cytotoxic CD4 T cells (CD4 CTL) has been found in autoimmune diseases like multiple sclerosis (MS). Previously, we found that this expansion is directly linked to MS disease severity and that this holds value as a novel prognostic marker in MS. However, the mechanisms behind these findings remain unclear. To understand how CD4 CTL contribute to autoimmunity, we studied the interaction between CD4 CTL, conventional CD4 helper T cells (TH cells), and CD4 regulatory T cells (Tregs). Here, we show that while CD4 CTL are resistant to suppression by functional Tregs in vitro, they simultaneously promote the suppression of TH cell proliferation in a triple co-culture system. It remains to be determined whether this is due to enhanced Treg-mediated suppression, or that CD4 CTL themselves are able to suppress TH cells. Interestingly, we found that the secretome of CD4 CTL induced a suppressive phenotype in Tregs, shown by upregulation of IL-10, granzyme B, and CTLA-4 mRNA, while also IFN-gamma gene expression was increased. We furthermore found that CD4 CTL have a pro-inflammatory and pro-survival mRNA expression profile when compared to conventional TH cells. Taken together, our results suggest that when TH cells terminally differentiate and acquire cytotoxic properties, the persistence of these cells is favored by paralleled resistance to Treg-mediated suppression and enhanced suppression of naive TH cells, possibly contributing to memory inflation.

61


POSTER WALK C – TREATMENT OF MS Thursday 15:45 – 16:45 hours Moderators: dr. Susanne Kooistra and dr. Menno Schoonheim High-dose vitamin D3 supplementation in multiple sclerosis is not associated with lower plasma neurofilament light chain levels Author Joost Smolders1, Zuzanna MichalakI2, Jan Damoiseaux3, Jody van den Ouweland4, Jens Kühle2, Raymond Hupperts5 1 Affiliation Department of Neurology, Canisius Wilhelmina Ziekenhuis, Nijmegen, 2Department of

Biomedicine, University Hospital Basel, Basel, 3Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, 4Department of Clinical Chemistry, Canisius Wilhel­ mina Ziekenhuis, Nijmegen, 5Department of Neurology, Zuyderland Medical Center, Sittard

Background Vitamin D-insufficiency is associated with an increased risk of relapses and active MRI lesions in relapsing remitting multiple sclerosis (RRMS). Randomized controlled supplementation trials are however negative on their primary endpoints, while secondary MRI-endpoints suggest anti-inflammatory effects. Circulating levels of neurofilament light chain (NfL) are a biomarker of disease activity in RRMS. We explored whether 48 weeks high-dose vitamin D3 supplements were associated with lower plasma NfL levels. Methods Plasma samples at baseline and 48 weeks were available of N=40 Dutch interferon betatreated participants with RRMS of the SOLAR trial, of which N=24 were supplemented with 14000 IU/day vitamin D3 and N= 16 with placebo. NfL levels were measured in duplicates with single molecule array (Simoa), 25-hydroxyvitamin D3 (25(OH)D3) levels with liquid chromatography-mass spectrometry/ mass spectrometry (LC-MS/MS). Results Plasma 25(OH)D3 levels at 48 weeks were higher in the vitamin D3 when compared to placebo-arm (median level 281 [IQR 205-330] vs. 72[39-88] nM; P<0.001). NfL levels at 48 weeks were not different between the treatment arms (median level 25.4 [IQR 19.6-32.2] vs. 25.3 [17.9-30.1] pg/mL; P=0.738), but did also no differ between participants with and without combined unique active MRI lesions at 48 weeks (median level 28.2 [IQR 22.7-38.4] vs. 22.6 [18.3-28.0]; yes vs. no, resp; P=0.118).

62

Conclusion Supplementation of high dose vitamin D3 for 48 weeks was not associated with lower plasma NfL levels. Although the small sample size and short treatment duration warrant a careful interpretation of our data, this study does not support an effect of vitamin D3 on this biomarker of RRMS activity.


Exploring the neuroprotective function of extracellular vesicles containing small heat shock proteins (HSPB1 and HSPB8) upon neuroinflammation Author

Bram Van den Broek1, Joël Beaumont1, Sam Vanherle1, Vicky De Winter2, Vincent Timmerman2, Veerle Somers1, Luc Michiels1, Joy Irobi1

1 Affiliation Hasselt University, Biomedical research institute (BIOMED), Martelarenlaan 42, 3500

Hasselt, Belgium, 2Peripheral Neuropathy Group, VIB-Department of Molecular Genetics, University of Antwerp, Antwerpen, Belgium

Introduction Currently, the repair mechanisms of Multiple Sclerosis (MS) is still unknown. However, it is known that small heat-shock proteins (HSPBs), which have protective functions are upregulated in MS lesions. Furthermore, it is shown that mutations in HSPB1 and 8 causes peripheral neurodegeneration. Although the protective intracellular functions are known, the extracellular functions remains unclear. One way cells secrete HSPBs is by releasing extracellular vesicles (EV). In MS, HSPB1/8 is upregulated in astrocytes but downregulated in oligodendrocytes and microglia cells during MS lesion development. We hypothesize that extracellular HSPBs exhibit neuroprotective roles which are altered upon inflammation in oligodendrocytes. Method To determine the protective activity of intracellular and extracellular HSPBs in oligodendrocytes, we establish HSPB overexpressing cell-lines for the production and characterization of HSPB positive EV under normal and inflamed conditions. These purified HSPB-EV are further applied to measure their role in cell survival. Results Pilot data shows that in oligodendrocyte, upon heat shock, there is a slight increase in early apoptosis. Striking, when oligodendrocytes were stimulated with inflamed EV, they exhibited a similar level of apoptosis comparable to known inflammatory mediators. In addition, immunoblot analysis of oligodendrocyte showed low expression of monomeric HSPB1/8 in non-stressed cells. Summary We observed low monomeric HSPB1/8 protein expression in non-stressed oligoden­ drocytes. Interestingly, inflamed-EV isolated from TNFα stimulated oligodendrocytes, exhibited a similar rate of apoptosis as compared to proinflammatory mediators. Reduction in endogenous expression of HSPB1/8 in oligodendrocytes might impair their cytoprotective activity during early inflammation.

63


Setmelanotide, a novel, selective melanocortin receptor-4 agonist exerts anti-inflammatory actions in astrocytes and promotes an anti-inflammatory macrophage phenotype Author Alwin Kamermans, Tom Verhoeven, Bert van het Hof, Maarten Witte, Jack van Horssen, Elga de Vries, Merel Rijnsburger Affiliation Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam The Netherlands.

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 agonists. Melanocortins are highly conserved neuropeptides which exert their action through the activation of the family of melanocortin receptors (MC1R-5R). Among the MCR, MC4R is most abundantly expressed in the CNS and several studies have described that MC4R is – besides neurons - expressed by astrocytes. Activation of neuronal MC4R in the hypothalamus via the melanocortin alpha-melanocyte stimulating hormone (ι-MSH), generally leads to weight loss due to a reduction in caloric intake and an increase in energy expenditure. Interestingly, activation of MC4R in astrocytes has shown to have potent anti-inflammatory as well as neuroprotective effects, suggesting that this could be a potential target to ameliorate ongoing inflammation and neurodegeneration in MS. However, to date, the expression of MC4R in human astrocytes as well as their cellular distribution in MS brain tissue has not been studied in detail. Therefore, in this study, we set out to investigate MC4R expression and analyze its downstream effects. Based on our immunohistochemical analysis, we identified the MC4R to be the dominant melanocortin receptor expressed in the human brain, particularly on astrocytes. Increased astrocytic MC4R expression was observed in active MS lesions. We furthermore show that the novel and 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 towards 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. 64


TNF-TNFRs signalling modulation in an MS mouse model Author

Valentina Pegoretti1, Wia Baron2, Jon Laman3, Ulrich Eisel1

1 Affiliation Department of Neurobiology, GELIFES - University of Groningen, 2Department of Cell

Biology, University Medical Center Groningen, University of Groningen, 3Department of Neuroscience, University Medical Center Groningen, University Groningen

The pathology of several autoimmune diseases has been associated with impaired regulation of Tumor Necrosis Factor (TNF) alpha. TNF is a major cytokine involved in the induction and maintenance of inflammation and it determines its opposing actions via two receptors: TNFR1 and TNFR2. In both in vitro and in vivo studies, TNFR1 directly and indirectly induces neurotoxicity under chronic condition while TNFR2 showed neuroprotection. Anti-TNF therapies are successfully used to treat diseases such as rheumatoid arthritis, colitis and psoriasis. However, clinical studies with a non-selective inhibitor of TNF (Lenercept) in patients with multiple sclerosis (MS) had to be halted due to exacerbation of clinical symptoms. Although the opposing effects of TNF receptors were obscure at that time, TNF and TNFR levels were found elevated in cerebrospinal fluid (CSF) and serum and correlating with the severity of MS. Altogether, these findings suggest an ambiguous role of TNF in MS, worth to be investigated. Interestingly, a TNFR1 antagonist was highly protective in an experimental autoimmune encephalomyelitis (EAE) mouse model. Here, we hypothesize that an agonist of TNFR2 could be equally effective or even superior to blocking TNFR1 in various models of MS and demyelination, including MOG induced EAE and cuprizone mediated demyelination. In order to functionally evaluate compounds that are specific for human TNFRs, we have previously generated knock-in mice expressing TNFRs with a humanized extracellular domain (huTNFR-ki). These novel mouse lines will be used to test TNFR2 agonists in in vivo and ex vivo models for MS. There is evidence to believe that activating TNFR2 has beneficial functions such as neuroprotection, immune regulation and tissue regeneration. Recently, we could show neuroprotection against NMDA mediated Nucleus basalis lesion in huTNFR2ki mice by using TNFR2 agonist. Further, preliminary data of an in vivo model of MS will be shown which will reveal the potential protective effect of a TNFR2 agonist. In conclusion, unravel the difference between TNFRs and their downstream cascades might lead to new insights in MS pathology with major implications for its effective treatment.

65


Cognitive rehabilitation in patients with advanced progressive multiple sclerosis: possible within limits? Author

Stefanos Prouskas1,2, Nancy Chiaravalloti3, Neeltje Kant2, Karlene Ball4, Vincent de Groot5, Bernard Uitdehaag6, Jeroen Geurts1, Liesbeth Kooij2, Hanneke Hulst1

1 Affiliation Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center

Amsterdam, VU University Medical Center, Amsterdam, The Netherlands, 2Nieuw Unicum, Zandvoort, The Netherlands, 3Neuropsychology and Neuroscience Laboratory, Kessler Foundation, West Orange, NJ, USA; Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, NJ, USA, 4Department of Psychology, University of Alabama at Birmingham , Birmingham, Alabama, 5Department of Rehabilitation Medicine, Amsterdam Movement Sciences, MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands, 6Department of Neurology, Amsterdam Neuroscience, MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands

Background Progressive MS patients exhibit cognitive impairment with increased frequency and severity compared to RRMS patients. While cognitive rehabilitation seems to be a promising approach to treatment for patients with RRMS, patients with advanced progressive MS are underrepresented in most such studies. Therefore, the feasibility of cognitive rehabilitation in patients with progressive MS is unknown. Objective The feasibility of two cognitive interventions (functional training and compensatory strategies) was investigated in patients with progressive MS requiring assisted living. Methods Eighteen patients with progressive MS requiring assisted living [median EDSS=7.5, mean age 58.1 (5.0) years, 13 SPMS, mean disease duration 20.6 (6.2) years] were randomized into 1) computer-based speed of processing training (SPT, n=9) for 5 weeks; or 2) compensatory memory strategies training (CST, n=9, care-as-usual) for 9 weeks. Patients underwent neuropsychological testing at baseline and follow-up. Feasibility was determined by evaluating practical aspects of the training (e.g. fatigue, motivation, concentration, difficulty, duration), as well as recruitment rate, dropout rate, completion rate, adherence rate and patient satisfaction (CSQ-8). Results A number of patients contacted for participation deemed the proposed intervention too intensive time-wise, resulting in a 36% recruitment rate. For the SPT sessions, average self-reported energy level decreased minimally post-session (pre=6.9/10, post=6.4/10). 66


Training difficulty (4.6) and duration (5.7) were almost ideal (5=ideal, <5 too easy/short, >5 too difficult/long). Completion rates and adherence rates for both interventions were good (>70%). Increased fatigue and decreased adherence were observed on very warm (>30°C) days, for both interventions. CSQ-8 showed high satisfaction in both groups (SPT=28.1, CST=24). Conclusion Our study shows that cognitive rehabilitation is within reach for patients with advanced progressive MS. For future studies we recommend screening a large number of potential participants, constant monitoring of fatigue and motivation, avoidance of warm temperatures, reduced session duration, and increased training frequency.

67


Different network functional connectivity characteristics of responders and non-responders to attention training in MS Author Stefanos Prouskas1, Martijn Steenwijk1, Karin Gehring2,3, Marijn Huiskamp1, Brigit de Jong4, Jeroen Geurts1, Margriet Sitskoorn2, Menno Schoonheim1, Hanneke Hulst1 1 Affiliation Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center

Amsterdam, Amsterdam UMC, Location VU University Medical Center, Amsterdam, The Netherlands, 2Department of Cognitive Neuropsychology, Tilburg University, The Netherlands, 3Department of Neurosurgery, Elisabeth-TweeSteden Hospital, The Netherlands, 4Department of Neurology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, location VU University Medical Center, Amsterdam, The Netherlands

Background Cognitive rehabilitation has been suggested as a means to improve cognitive function. However, cognitive rehabilitation is only effective in a subset of MS patients. It is therefore imperative to identify characteristics that might influence the ability to respond to rehabilitation of cognitive impairment. While cognitive impairment in MS is related to changes in functional connectivity (FC) of cognitive brain networks such as the default mode network (DMN), it is unknown how functional network characteristics affect the brain’s ability to respond to cognitive training. Aim To investigate whether baseline resting-state FC within and between the DMN, dorsal attention network (DAN) and ventral attention network (VAN), can distinguish responders from non-responders to an attention training in MS. Methods Patients were randomized into an attention training (home-based computerized C-Car: 7-week, 45 min/week, N=58, age=48.4±10.2 years, 34 women, RRMS=42, median EDSS=4.0) or a waiting-list control group (CG, N=24, age=48.5±9.4 years, 19 women, RRMS=16, median EDSS=4.0). Neuropsychological assessment at baseline and followup included tests of attention, memory, information processing speed, and executive functioning. Based on the CG, a reliable change index (RCI) was calculated, adjusted for practice effects. Responders were defined as patients scoring RCI>1.64 (90% CI) on at least two tests. 3D T1 MRI and resting-state fMRI was obtained at baseline. After preprocessing and denoising using ICA-AROMA, a subject-wise fMRI correlation matrix was computed. Within- and between-network measures of FC of the DMN, DAN, and VAN were calculated using relative correlations and the Brainnetome atlas and compared between responders and non-responders. 68


Results Responders (N=22) and non-responders (N=36) did not differ significantly in age, sex, education, MS subtype, EDSS, disease duration, or baseline cognition. Responders, compared to non-responders, had lower average FC between DMN and DAN (0.85 vs 0.92 respectively; p=0.04) and DMN and VAN (0.90 vs 0.98 respectively; p=0.04). There were no significant group differences in within-network FC. Conclusion Lower FC between DMN and attention networks seems an indicator of response to attention training in MS, which might reflect a more intact network functioning at baseline.

69


Memory retraining in MS: lessons learned from healthy aging, mild cognitive impairment and Alzheimer’s disease Author

Piet Bouman, Marijn Huiskamp, Marit Jonker, Jeroen Geurts, Geert Schenk, Hanneke Hulst

Affiliation Department of Anatomy and Neurosciences, MS center Amsterdam, Amsterdam Neuroscience; the Netherlands, Amsterdam UMC, Vrije Universiteit Amsterdam

Introduction Memory-impairment occurs in 40-70% of MS patients and is associated with hippocampal deterioration. Memory decline affects quality of life, stressing the need for effective memory rehabilitation strategies. This study evaluates the effects of cognitive and exercise interventions on memory function in patients with MS and healthy aging (HA), mild cognitive impairment (MCI) and Alzheimer’s disease. Methods A meta-analysis was performed, evaluating cognitive and exercise interventions targeting visuospatial and verbal memory performance. Articles were selected using a priori criteria and consensus judgement for risk of bias was performed using Cochrane criteria and Review Manager software. Results 142 articles were included: 51 on MS, 25 on HA, 49 on MCI, 10 on AD and 7 combining these conditions. 68/142 studies examined exercise interventions, 21/142 examined cognitive functional training (e.g. remembering specific information), 53/142 examined compensatory strategies. Compared to other interventions, high-intensive memory-strategy training was most effective in all conditions: std. mean difference 1.03 (95% CI [0.681.38]) vs. std. mean difference 0.30 [0.16-0.44] (functional training) and std. mean difference 0.46 [0.16-0.66] (exercise). Overall, greatest improvements were obtained in HA (std. mean difference 0.48, [0.28-0.67]), followed by MCI (std. mean difference 0.46, [0.23-0.69], AD (std. mean difference 0.43, [0.16-0.70]) and MS (std. mean difference 0.34, [0.19-0.49]). Conclusions Memory-strategy interventions exerted the largest effects, especially in healthy elderly. MCI patients perform better than MS patients, which might be a consequence of the amount of hippocampal degeneration (i.e. more extensive in MS) or of methodological origin, since fewer memory-strategy interventions in MS have been studied.

70


The MS Research Days are made possible by the kind support of:

Leidseweg 557-1 2253 JJ Voorschoten Postbus 200 2250 AE Voorschoten T 071 - 5 600 500 www.msresearch.nl 71



Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.