Network


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

Hotspot


Dive into the research topics where Nathalie Arbour is active.

Publication


Featured researches published by Nathalie Arbour.


Nature Medicine | 2007

Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation

Hania Kebir; Katharina Kreymborg; Igal Ifergan; Aurore Dodelet-Devillers; Romain Cayrol; Monique Bernard; Fabrizio Giuliani; Nathalie Arbour; Burkhard Becher; Alexandre Prat

TH17 lymphocytes appear to be essential in the pathogenesis of numerous inflammatory diseases. We demonstrate here the expression of IL-17 and IL-22 receptors on blood-brain barrier endothelial cells (BBB-ECs) in multiple sclerosis lesions, and show that IL-17 and IL-22 disrupt BBB tight junctions in vitro and in vivo. Furthermore, TH17 lymphocytes transmigrate efficiently across BBB-ECs, highly express granzyme B, kill human neurons and promote central nervous system inflammation through CD4+ lymphocyte recruitment.


Nature Immunology | 2008

Activated leukocyte cell adhesion molecule promotes leukocyte trafficking into the central nervous system

Romain Cayrol; Karolina Wosik; Jennifer L. Berard; Aurore Dodelet-Devillers; Igal Ifergan; Hania Kebir; Arsalan S. Haqqani; Katharina Kreymborg; Sebastian Krug; Robert Moumdjian; Alain Bouthillier; Burkhard Becher; Nathalie Arbour; Samuel David; Danica Stanimirovic; Alexandre Prat

Adhesion molecules of the immunoglobulin superfamily are crucial effectors of leukocyte trafficking into the central nervous system. Using a lipid raft-based proteomic approach, we identified ALCAM as an adhesion molecule involved in leukocyte migration across the blood-brain barrier (BBB). ALCAM expressed on BBB endothelium localized together with CD6 on leukocytes and with BBB endothelium transmigratory cups. ALCAM expression on BBB cells was upregulated in active multiple sclerosis and experimental autoimmune encephalomyelitis lesions. Moreover, ALCAM blockade restricted the transmigration of CD4+ lymphocytes and monocytes across BBB endothelium in vitro and in vivo and reduced the severity and delayed the time of onset of experimental autoimmune encephalomyelitis. Our findings indicate an important function for ALCAM in the recruitment of leukocytes into the brain and identify ALCAM as a potential target for the therapeutic dampening of neuroinflammation.


Journal of Virology | 2000

Neuroinvasion by Human Respiratory Coronaviruses

Nathalie Arbour; Robert Day; Jia Newcombe; Pierre J. Talbot

ABSTRACT Human coronaviruses (HCoV) cause common colds but can also infect neural cell cultures. To provide definitive experimental evidence for the neurotropism and neuroinvasion of HCoV and its possible association with multiple sclerosis (MS), we have performed an extensive search and characterization of HCoV RNA in a large panel of human brain autopsy samples. Very stringent reverse transcription-PCR with two primer pairs for both viral strains (229E and OC43), combined with Southern hybridization, was performed on samples from 90 coded donors with various neurological diseases (39 with MS and 26 with other neurological diseases) or normal controls (25 patients). We report that 44% (40 of 90) of donors were positive for 229E and that 23% (21 of 90) were positive for OC43. A statistically significant higher prevalence of OC43 in MS patients (35.9%; 14 of 39) than in controls (13.7%; 7 of 51) was observed. Sequencing of nucleocapsid protein (N) gene amplicons revealed point mutations in OC43, some consistently found in three MS patient brains and one normal control but never observed in laboratory viruses. In situ hybridization confirmed the presence of viral RNA in brain parenchyma, outside blood vessels. The presence of HCoV in human brains is consistent with neuroinvasion by these respiratory pathogens. Further studies are needed to distinguish between opportunistic and disease-associated viral presence in human brains.


The Journal of Neuroscience | 2007

NKG2D-Mediated Cytotoxicity toward Oligodendrocytes Suggests a Mechanism for Tissue Injury in Multiple Sclerosis

Philippe Saikali; Jack P. Antel; Jia Newcombe; Zhihong Chen; Mark S. Freedman; Manon Blain; Romain Cayrol; Alexandre Prat; Jeffery A. Hall; Nathalie Arbour

NKG2D is an activating or coactivating receptor expressed on human natural killer (NK) cells, CD8+ T cells, and γ/δ T cells. NKG2D ligands have been detected on many tumor cell types and can be induced on nontransformed cells by environmental signals including DNA damage and inflammation. We investigated the contribution of NKG2D–NKG2D ligand interaction on CNS-directed immune responses. We observed that primary cultures of human adult oligodendrocytes and fetal astrocytes expressed ligands for NKG2D in vitro whereas neurons, microglia, and adult astrocytes did not. Disruption of the NKG2D–NKG2D ligand interaction using blocking antibodies significantly inhibited killing of primary human oligodendrocytes mediated by activated human NK cells, γ/δ T cells, and allo-reactive CD8+ T cells. NKG2D ligands [major histocompatibility complex class I chain-related molecules A and B (MICA/B)] were detected in groups of cells and colocalized with an oligodendrocyte marker (adenomatous polyposis coli) in white matter sections obtained from multiple sclerosis lesions but not in normal control samples. CD8+ T cells could be detected in close proximity to MICA/B+ cells within multiple sclerosis lesions, supporting an in vivo interaction between these immune effectors and stressed MICA/B-expressing oligodendrocytes. These results imply that NKG2D–NKG2D ligand interaction can potentially contribute to cytotoxic responses mediated by activated immune effector cells in the inflamed CNS, as observed in multiple sclerosis.


Annals of Neurology | 2013

Diminished Th17 (not Th1) responses underlie multiple sclerosis disease abrogation after hematopoietic stem cell transplantation

Peter J. Darlington; Tarik Touil; Jean Sebastien Doucet; Denis Gaucher; Joumana Zeidan; Dominique Gauchat; Rachel Corsini; Ho Jin Kim; Martin Duddy; Farzaneh Jalili; Nathalie Arbour; Hania Kebir; Jacqueline T. Chen; Douglas L. Arnold; Marjorie Bowman; Jack P. Antel; Alexandre Prat; Mark S. Freedman; Harold Atkins; Rafick Pierre Sekaly; Rémi Cheynier; Amit Bar-Or

To define changes in phenotype and functional responses of reconstituting T cells in patients with aggressive multiple sclerosis (MS) treated with ablative chemotherapy and autologous hematopoietic stem cell transplantation (HSCT).


European Journal of Immunology | 2011

IL‐27 increases the proliferation and effector functions of human naïve CD8+ T lymphocytes and promotes their development into Tc1 cells

Raphael Schneider; Teodora Yaneva; Diane Beauseigle; Lama El-Khoury; Nathalie Arbour

IL‐27 has been shown to exhibit both pro‐ and anti‐inflammatory properties; it favors mouse naïve CD4+T‐cell differentiation into Th1 cells to the detriment of Th17 and Th2 skewing and regulates IL‐10 and IL‐17 production by human CD4+ T cells. Moreover, IL‐27 promotes proliferation and cytotoxic functions of mouse CD8+ T lymphocytes, but no data are available on human CD8+ T cells. We investigated the impact of IL‐27 on human CD8+T cells. In contrast to mouse T cells, the IL‐27 receptor (IL‐27R), composed of T cell cytokine receptor (TCCR) and gp130, was detected on a greater percentage of human CD8+ than CD4+ T cells and these proportions increased upon polyclonal activation. IL‐27 induced rapid STAT1 and STAT3 signaling, enhanced STAT1 protein levels, and induced SOCS1 and SOCS3 expression in a STAT1‐dependent manner by human CD8+ T cells. Addition of IL‐27 to α‐CD3‐activated naïve CD8+ T cells significantly increased T‐box transcription factor expression levels, cell proliferation, and IFN‐γ and granzyme B production leading to increased CD8+ T‐cell‐mediated cytotoxicity. These results demonstrate that IL‐27, a rapidly produced cytokine by activated APC, has a profound impact on human naïve CD8+T cells, driving them to become highly efficient Tc1 cells.


Brain | 2012

Melanoma cell adhesion molecule identifies encephalitogenic T lymphocytes and promotes their recruitment to the central nervous system.

Catherine Larochelle; Romain Cayrol; Hania Kebir; Jorge Ivan Alvarez; Marc André Lécuyer; Igal Ifergan; Emilie Viel; Lyne Bourbonnière; Diane Beauseigle; Simone Terouz; Lamia Hachehouche; Steve Gendron; Josée Poirier; Céline Jobin; Pierre Duquette; Ken Flanagan; Ted Yednock; Nathalie Arbour; Alexandre Prat

In multiple sclerosis, encephalitogenic CD4(+) lymphocytes require adhesion molecules to accumulate into central nervous system inflammatory lesions. Using proteomic techniques, we identified expression of melanoma cell adhesion molecule (MCAM) on a subset of human effector memory CD4(+) lymphocytes and on human blood-brain barrier endothelium. Herein, we demonstrate that MCAM is a stable surface marker that refines the identification of interleukin 17(+), interleukin 22(+), RAR-related orphan receptor γ and interleukin 23 receptor(+) cells within the CD161(+)CCR6(+) subset of memory CD4(+) lymphocytes. We also show that MCAM(+) lymphocytes express significantly more granulocyte/macrophage colony stimulating factor and granzyme B than MCAM(-) lymphocytes. Furthermore, the proportion of MCAM(+) CD4(+) lymphocytes is significantly increased in the blood and in the central nervous system of patients with multiple sclerosis and experimental autoimmune encephalomyelitis animals compared with healthy controls or other neurological diseases, and MCAM expression is upregulated at the blood-brain barrier within inflammatory lesions. Moreover, blockade of MCAM or depletion of MCAM(+) CD4(+) T lymphocytes both restrict the migration of T(H)17 lymphocytes across blood-brain barrier endothelial cells and decrease the severity of experimental autoimmune encephalomyelitis. Our findings indicate that MCAM could serve as a potential biomarker for multiple sclerosis and represents a valuable target for the treatment of neuroinflammatory conditions.


The New England Journal of Medicine | 2014

Maraviroc and JC Virus–Associated Immune Reconstitution Inflammatory Syndrome

Paul S. Giacomini; Ayal Rozenberg; Imke Metz; David Araujo; Nathalie Arbour; Amit Bar-Or

The immune reconstitution inflammatory syndrome can be a serious complication of immune reversal in the treatment of progressive multifocal leukoencephalopathy. A new potential treatment for IRIS is described in this letter.


Glia | 2012

Lipocalin 2 is a novel immune mediator of experimental autoimmune encephalomyelitis pathogenesis and is modulated in multiple sclerosis.

Jennifer L. Berard; Juan G. Zarruk; Nathalie Arbour; Alexandre Prat; V. Wee Yong; Francois H. Jacques; Shizuo Akira; Samuel David

Experimental autoimmune encephalomyelitis (EAE) is a widely used animal model of multiple sclerosis (MS), an inflammatory, demyelinating disease of the central nervous system (CNS). EAE pathogenesis involves various cell types, cytokines, chemokines, and adhesion molecules. Given the complexity of the inflammatory response in EAE, it is likely that many immune mediators still remain to be discovered. To identify novel immune mediators of EAE pathogenesis, we performed an Affymetrix gene array screen on the spinal cords of mice at the onset stage of disease. This screening identified the gene encoding lipocalin 2 (Lcn2) as being significantly upregulated. Lcn2 is a multi‐functional protein that plays a role in glial activation, matrix metalloproteinase (MMP) stabilization, and cellular iron flux. As many of these processes have been implicated in EAE, we characterized the expression and role of Lcn2 in this disease in C57BL/6 mice. We show that Lcn2 is significantly upregulated in the spinal cord throughout EAE and is expressed predominantly by monocytes and reactive astrocytes. The Lcn2 receptor, 24p3R, is also expressed on monocytes, macrophages/microglia, and astrocytes in EAE. In addition, we show that EAE severity is increased in Lcn2−/− mice as compared with wild‐type controls. Finally, we demonstrate that elevated levels of Lcn2 are detected in the plasma and cerebrospinal fluid (CSF) in MS and in immune cells in CNS lesions in MS tissue sections. These data indicate that Lcn2 is a modulator of EAE pathogenesis and suggest that it may also play a role in MS.


Brain | 2011

Central nervous system recruitment of effector memory CD8+ T lymphocytes during neuroinflammation is dependent on α4 integrin

Igal Ifergan; Hania Kebir; Jorge Ivan Alvarez; Gabriel Marceau; Monique Bernard; Lyne Bourbonnière; Josée Poirier; Pierre Duquette; Pierre J. Talbot; Nathalie Arbour; Alexandre Prat

Abstract Clonally expanded CD8+ T lymphocytes are present in multiple sclerosis lesions, as well as in the cerebrospinal fluid of patients with multiple sclerosis. In experimental autoimmune encephalomyelitis, CD8+ T lymphocytes are found in spinal cord and brainstem lesions. However, the exact phenotype of central nervous system-infiltrating CD8+ T lymphocytes and the mechanism by which these cells cross the blood–brain barrier remain largely unknown. Using cerebrospinal fluid from patients with multiple sclerosis, spinal cord from experimental autoimmune encephalomyelitis and coronavirus-induced encephalitis, we demonstrate that central nervous system-infiltrating CD8+ T lymphocytes are mostly of the effector memory phenotype (CD62L− CCR7− granzymeBhi). We further show that purified human effector memory CD8+ T lymphocytes transmigrate more readily across blood-brain barrier-endothelial cells than non-effector memory CD8+ T lymphocytes, and that blood-brain barrier endothelium promotes the selective recruitment of effector memory CD8+ T lymphocytes. Furthermore, we provide evidence for the recruitment of interferon-γ- and interleukin-17-secreting CD8+ T lymphocytes by human and mouse blood-brain barrier endothelium. Finally, we show that in vitro migration of CD8+ T lymphocytes across blood-brain barrier-endothelial cells is dependent on α4 integrin, but independent of intercellular adhesion molecule-1/leucocyte function-associated antigen-1, activated leucocyte cell adhesion molecule/CD6 and the chemokine monocyte chemotactic protein-1/CCL2. We also demonstrate that in vivo neutralization of very late antigen-4 restricts central nervous system infiltration of CD8+ T lymphocytes in active immunization and adoptive transfer experimental autoimmune encephalomyelitis, and in coronavirus-induced encephalitis. Our study thus demonstrates an active role of the blood-brain barrier in the recruitment of effector memory CD8+ T lymphocytes to the CNS compartment and defines α4 integrin as a major contributor of CD8+ T lymphocyte entry into the brain.

Collaboration


Dive into the Nathalie Arbour's collaboration.

Top Co-Authors

Avatar

Alexandre Prat

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Jack P. Antel

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Hania Kebir

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Igal Ifergan

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Romain Cayrol

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jia Newcombe

UCL Institute of Neurology

View shared research outputs
Researchain Logo
Decentralizing Knowledge