Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Immunol Rev ; 311(1): 151-176, 2022 10.
Article in English | MEDLINE | ID: mdl-35909230

ABSTRACT

Inflammation is a biological process that dynamically alters the surrounding microenvironment, including participating immune cells. As a well-protected organ surrounded by specialized barriers and with immune privilege properties, the central nervous system (CNS) tightly regulates immune responses. Yet in neuroinflammatory conditions, pathogenic immunity can disrupt CNS structure and function. T cells in particular play a key role in promoting and restricting neuroinflammatory responses, while the inflamed CNS microenvironment can influence and reshape T cell function and identity. Still, the contraction of aberrant T cell responses within the CNS is not well understood. Using autoimmunity as a model, here we address the contribution of CD4 T helper (Th) cell subsets in promoting neuropathology and disease. To address the mechanisms antagonizing neuroinflammation, we focus on the control of the immune response by regulatory T cells (Tregs) and describe the counteracting processes that preserve their identity under inflammatory challenges. Finally, given the influence of the local microenvironment on immune regulation, we address how CNS-intrinsic signals reshape T cell function to mitigate abnormal immune T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes , Neuroinflammatory Diseases , Autoimmunity , Central Nervous System , Humans , T-Lymphocytes, Regulatory
2.
J Clin Immunol ; 44(4): 87, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38578402

ABSTRACT

We present a case study of a young male with a history of 22q11.2 deletion syndrome (22qDS), diagnosed with systemic capillary leak syndrome (SCLS) who presented with acute onset of diffuse anasarca and sub-comatose obtundation. We hypothesized that his co-presentation of neurological sequelae might be due to blood-brain barrier (BBB) susceptibility conferred by the 22q11.2 deletion, a phenotype that we have previously identified in 22qDS. Using pre- and post-intravenous immunoglobulins (IVIG) patient serum, we studied circulating biomarkers of inflammation and assessed the potential susceptibility of the 22qDS BBB. We employed in vitro cultures of differentiated BBB-like endothelial cells derived from a 22qDS patient and a healthy control. We found evidence of peripheral inflammation and increased serum lipopolysaccharide (LPS) alongside endothelial cells in circulation. We report that the patient's serum significantly impairs barrier function of the 22qDS BBB compared to control. Only two other cases of pediatric SCLS with neurologic symptoms have been reported, and genetic risk factors have been suggested in both instances. As the third case to be reported, our findings are consistent with the hypothesis that genetic susceptibility of the BBB conferred by genes such as claudin-5 deleted in the 22q11.2 region promoted neurologic involvement during SCLS in this patient.


Subject(s)
Capillary Leak Syndrome , DiGeorge Syndrome , Humans , Male , Child , Capillary Leak Syndrome/diagnosis , Blood-Brain Barrier , Endothelial Cells , Permeability , Inflammation
3.
Eur J Immunol ; 53(1): e2249840, 2023 01.
Article in English | MEDLINE | ID: mdl-36337041

ABSTRACT

Mice modeling the hemizygous deletion of chromosome 22q11.2 (22qMc) have been utilized to address various clinical phenotypes associated with the disease, including cardiac malformations, altered neural circuitry, and behavioral deficits. Yet, the status of the T cell compartment, an important clinical concern among 22q11.2 deletion syndrome (22qDS) patients, has not been addressed. While infancy and early childhood in 22qDS are associated with deficient T cell numbers and thymic hypoplasia, which can be severe in a small subset of patients, studies suggest normalization of the T cell counts by adulthood. We found that adult 22qMc do not exhibit thymic hypoplasia or altered thymic T cell development. Our findings that immune cell counts and inflammatory T cell activation are unaffected in 22qMc lend support to the hypothesis that human 22qDS immunodeficiencies are secondary to thymic hypoplasia, rather than intrinsic effects due to the deletion. Furthermore, the 22q11.2 deletion does not impact the differentiation capacity of T cells, nor their activity and response during inflammatory activation. Thus, 22qMc reflects the T cell compartment in adult 22qDS patients, and our findings suggest that 22qMc may serve as a novel model to address experimental and translational aspects of immunity in 22qDS.


Subject(s)
DiGeorge Syndrome , Immunologic Deficiency Syndromes , Humans , Child, Preschool , Adult , Mice , Animals , DiGeorge Syndrome/genetics , DiGeorge Syndrome/complications , Chromosome Deletion , Thymus Gland , Immunologic Deficiency Syndromes/genetics , T-Lymphocytes
4.
Brain ; 144(6): 1670-1683, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33723591

ABSTRACT

The concerted actions of the CNS and the immune system are essential to coordinating the outcome of neuroinflammatory responses. Yet, the precise mechanisms involved in this crosstalk and their contribution to the pathophysiology of neuroinflammatory diseases largely elude us. Here, we show that the CNS-endogenous hedgehog pathway, a signal triggered as part of the host response during the inflammatory phase of multiple sclerosis and experimental autoimmune encephalomyelitis, attenuates the pathogenicity of human and mouse effector CD4 T cells by regulating their production of inflammatory cytokines. Using a murine genetic model, in which the hedgehog signalling is compromised in CD4 T cells, we show that the hedgehog pathway acts on CD4 T cells to suppress the pathogenic hallmarks of autoimmune neuroinflammation, including demyelination and axonal damage, and thus mitigates the development of experimental autoimmune encephalomyelitis. Impairment of hedgehog signalling in CD4 T cells exacerbates brain-brainstem-cerebellum inflammation and leads to the development of atypical disease. Moreover, we present evidence that hedgehog signalling regulates the pathogenic profile of CD4 T cells by limiting their production of the inflammatory cytokines granulocyte-macrophage colony-stimulating factor and interferon-γ and by antagonizing their inflammatory program at the transcriptome level. Likewise, hedgehog signalling attenuates the inflammatory phenotype of human CD4 memory T cells. From a therapeutic point of view, our study underlines the potential of harnessing the hedgehog pathway to counteract ongoing excessive CNS inflammation, as systemic administration of a hedgehog agonist after disease onset effectively halts disease progression and significantly reduces neuroinflammation and the underlying neuropathology. We thus unveil a previously unrecognized role for the hedgehog pathway in regulating pathogenic inflammation within the CNS and propose to exploit its ability to modulate this neuroimmune network as a strategy to limit the progression of ongoing neuroinflammation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Hedgehog Proteins/immunology , Inflammation/immunology , Animals , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Hedgehog Proteins/metabolism , Humans , Inflammation/metabolism , Mice , Spinal Cord/immunology , Spinal Cord/pathology
5.
Brain ; 144(5): 1351-1360, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33876226

ABSTRACT

Neuroimmune dysregulation is implicated in neuropsychiatric disorders including schizophrenia. As the blood-brain barrier is the immunological interface between the brain and the periphery, we investigated whether this vascular phenotype is intrinsically compromised in the most common genetic risk factor for schizophrenia, the 22q11.2 deletion syndrome (22qDS). Blood-brain barrier like endothelium differentiated from human 22qDS+schizophrenia-induced pluripotent stem cells exhibited impaired barrier integrity, a phenotype substantiated in a mouse model of 22qDS. The proinflammatory intercellular adhesion molecule-1 was upregulated in 22qDS+schizophrenia-induced blood-brain barrier and in 22qDS mice, indicating compromise of the blood-brain barrier immune privilege. This immune imbalance resulted in increased migration/activation of leucocytes crossing the 22qDS+schizophrenia blood-brain barrier. We also found heightened astrocyte activation in murine 22qDS, suggesting that the blood-brain barrier promotes astrocyte-mediated neuroinflammation. Finally, we substantiated these findings in post-mortem 22qDS brain tissue. Overall, the barrier-promoting and immune privilege properties of the 22qDS blood-brain barrier are compromised, and this might increase the risk for neuropsychiatric disease.


Subject(s)
22q11 Deletion Syndrome/pathology , Blood-Brain Barrier/pathology , 22q11 Deletion Syndrome/immunology , Animals , Astrocytes/metabolism , Humans , Immune Privilege/physiology , Inflammation/metabolism , Mice
6.
Nat Immunol ; 9(2): 137-45, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18157132

ABSTRACT

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.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Blood-Brain Barrier/metabolism , Brain/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/immunology , Activated-Leukocyte Cell Adhesion Molecule/analysis , Activated-Leukocyte Cell Adhesion Molecule/drug effects , Blood-Brain Barrier/chemistry , Cell Movement/drug effects , Cells, Cultured , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Proteomics
7.
Biochim Biophys Acta ; 1862(3): 472-82, 2016 03.
Article in English | MEDLINE | ID: mdl-26454208

ABSTRACT

The blood-brain barrier (BBB) constitutes an elaborate structure formed by specialized capillary endothelial cells, which together with pericytes and perivascular glial cells regulates the exchanges between the central nervous system (CNS) and the periphery. Intricate interactions between the different cellular constituents of the BBB are crucial in establishing a functional BBB and maintaining the delicate homeostasis of the CNS microenvironment. In this review, we discuss the role of astrocytes and microglia in inducing and maintaining barrier properties under physiological conditions as well as their involvement during neuroinflammatory pathologies. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Subject(s)
Astrocytes/pathology , Blood-Brain Barrier/pathology , Inflammation/pathology , Leukocytes/pathology , Neuroglia/pathology , Animals , Astrocytes/immunology , Blood-Brain Barrier/immunology , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunity, Cellular , Inflammation/immunology , Leukocytes/immunology , Microglia/immunology , Microglia/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Neuroglia/immunology , Neurovascular Coupling , Pericytes/immunology , Pericytes/pathology
8.
Neurobiol Dis ; 102: 60-69, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28235673

ABSTRACT

Laquinimod is currently being tested as a therapeutic drug in multiple sclerosis. However, its exact mechanism of action is still under investigation. Tracking of fluorescently-tagged encephalitogenic T cells during experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, revealed that laquinimod significantly reduces the invasion of pathogenic effector T cells into the CNS tissue. T-cell activation, differentiation and amplification within secondary lymphoid organs after immunization with myelin antigen, their migratory capacity and re-activation within the nervous tissue were either only mildly affected or remained unchanged. Instead, laquinimod directly impacted the functionality of the CNS vasculature. The expression of tight junction proteins p120 and ZO-1 in human brain endothelial cells was up-regulated upon laquinimod treatment, resulting in a significant increase in the transendothelial electrical resistance of confluent monolayers of brain endothelial cells. Similarly, expression of the adhesion molecule activated leukocyte cell adhesion molecule (ALCAM) and inflammatory chemokines CCL2 and IP-10 was suppressed, leading to a significant reduction in the migration of memory TH1 and TH17 lymphocytes across the blood brain barrier (BBB). Our data indicate that laquinimod exerts its therapeutic effects by tightening the BBB and limiting parenchymal invasion of effector T cells, thereby reducing CNS damage.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Neuroprotective Agents/pharmacology , Quinolones/pharmacology , Adult , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/metabolism , Rats, Inbred Lew , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Young Adult
9.
J Autoimmun ; 84: 1-11, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28641926

ABSTRACT

In multiple sclerosis (MS), there is a growing interest in inhibiting the pro-inflammatory effects of granulocyte-macrophage colony-stimulating factor (GM-CSF). We sought to evaluate the therapeutic potential and underlying mechanisms of GM-CSF receptor alpha (Rα) blockade in animal models of MS. We show that GM-CSF signaling inhibition at peak of chronic experimental autoimmune encephalomyelitis (EAE) results in amelioration of disease progression. Similarly, GM-CSF Rα blockade in relapsing-remitting (RR)-EAE model prevented disease relapses and inhibited T cell responses specific for both the inducing and spread myelin peptides, while reducing activation of mDCs and inflammatory monocytes. In situ immunostaining of lesions from human secondary progressive MS (SPMS), but not primary progressive MS patients shows extensive recruitment of GM-CSF Rα+ myeloid cells. Collectively, this study reveals a pivotal role of GM-CSF in disease relapses and the benefit of GM-CSF Rα blockade as a potential novel therapeutic approach for treatment of RRMS and SPMS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Central Nervous System/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , Autoimmunity , Cell Differentiation , Cell Movement , Cells, Cultured , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Humans , Immunosuppression Therapy , Male , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Targeted Therapy , Multiple Sclerosis/therapy , Myelin Sheath/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Signal Transduction
10.
Ann Neurol ; 78(1): 39-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869475

ABSTRACT

OBJECTIVE: Although Tc17 lymphocytes are enriched in the central nervous system (CNS) of multiple sclerosis (MS) subjects and of experimental autoimmune encephalomyelitis (EAE) animals, limited information is available about their recruitment into the CNS and their role in neuroinflammation. Identification of adhesion molecules used by autoaggressive CD8(+) T lymphocytes to enter the CNS would allow further characterization of this pathogenic subset and could provide new therapeutic targets in MS. We propose that melanoma cell adhesion molecule (MCAM) is a surface marker and adhesion molecule used by pathogenic CD8(+) T lymphocytes to access the CNS. METHODS: Frequency, phenotype, and function of MCAM(+) CD8(+) T lymphocytes was characterized using a combination of ex vivo, in vitro, in situ, and in vivo approaches in humans and mice, including healthy controls, MS subjects, and EAE animals. RESULTS: Herein, we report that MCAM is expressed by human effector CD8(+) T lymphocytes and it is strikingly upregulated during MS relapses. We further demonstrate that MCAM(+) CD8(+) T lymphocytes express more interleukin 17, interferon γ, granulocyte-macrophage colony-stimulating factor, and tumor necrosis factor than MCAM(-) lymphocytes, and exhibit an enhanced killing capacity toward oligodendrocytes. MCAM blockade restricts the transmigration of CD8(+) T lymphocytes across human blood-brain barrier endothelial cells in vitro, and blocking or depleting MCAM in vivo reduces chronic neurological deficits in active, transfer, and spontaneous progressive EAE models. INTERPRETATION: Our data demonstrate that MCAM identifies encephalitogenic CD8(+) T lymphocytes, suggesting that MCAM could represent a biomarker of MS disease activity and a valid target for the treatment of neuroinflammatory conditions.


Subject(s)
Blood-Brain Barrier/metabolism , CD8-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis, Relapsing-Remitting/metabolism , Animals , Blood-Brain Barrier/immunology , CD146 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , In Vitro Techniques , Inflammation , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Mice, Transgenic , Multiple Sclerosis, Relapsing-Remitting/immunology , Oligodendroglia , Tumor Necrosis Factor-alpha/immunology
11.
J Immunol ; 193(5): 2438-54, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25049355

ABSTRACT

Disruption of the blood-brain and blood-spinal cord barriers (BBB and BSCB, respectively) and immune cell infiltration are early pathophysiological hallmarks of multiple sclerosis (MS), its animal model experimental autoimmune encephalomyelitis (EAE), and neuromyelitis optica (NMO). However, their contribution to disease initiation and development remains unclear. In this study, we induced EAE in lys-eGFP-ki mice and performed single, nonterminal intravital imaging to investigate BSCB permeability simultaneously with the kinetics of GFP(+) myeloid cell infiltration. We observed a loss in BSCB integrity within a day of disease onset, which paralleled the infiltration of GFP(+) cells into the CNS and lasted for ∼4 d. Neutrophils accounted for a significant proportion of the circulating and CNS-infiltrating myeloid cells during the preclinical phase of EAE, and their depletion delayed the onset and reduced the severity of EAE while maintaining BSCB integrity. We also show that neutrophils collected from the blood or bone marrow of EAE mice transmigrate more efficiently than do neutrophils of naive animals in a BBB cell culture model. Moreover, using intravital videomicroscopy, we demonstrate that the IL-1R type 1 governs the firm adhesion of neutrophils to the inflamed spinal cord vasculature. Finally, immunostaining of postmortem CNS material obtained from an acutely ill multiple sclerosis patient and two neuromyelitis optica patients revealed instances of infiltrated neutrophils associated with regions of BBB or BSCB leakage. Taken together, our data provide evidence that neutrophils are involved in the initial events that take place during EAE and that they are intimately linked with the status of the BBB/BSCB.


Subject(s)
Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Neutrophils/immunology , Spinal Cord/immunology , Animals , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice , Mice, Transgenic , Neuromyelitis Optica/genetics , Neuromyelitis Optica/immunology , Neuromyelitis Optica/pathology , Neutrophils/pathology , Spinal Cord/pathology
12.
Proc Natl Acad Sci U S A ; 109(24): 9505-10, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22647601

ABSTRACT

Women develop certain autoimmune diseases more often than men. It has been hypothesized that this may relate to the development of more robust T-helper (Th)1 responses in women. To test whether women exhibit a Th1 bias, we isolated naïve cluster of differentiation (CD)4(+) T cells from peripheral blood of healthy women and men and measured the proliferation and cytokine production by these cells in response to submaximal amounts of anti-CD3 and anti-CD28. We observed that CD4(+) T cells from women produced higher levels of IFNγ as well as tended to proliferate more than male CD4(+) T cells. Intriguingly, male CD4(+) T cells instead had a predilection toward IL-17A production. This sex dichotomy in Th cytokine production was found to be even more striking in the Swiss/Jackson Laboratory (SJL) mouse. Studies in mice and humans indicated that the sexual dimorphism in Th1 and Th17 cytokine production was dependent on the androgen status and the T-cell expression of peroxisome proliferator activated receptor (PPAR)α and PPARγ. Androgens increased PPARα and decreased PPARγ expression by human CD4(+) T cells. PPARα siRNA-mediated knockdown had the effect of increasing IFNγ by male CD4(+) T cells, while transfection of CD4(+) T cells with PPARγ siRNAs increased IL-17A production uniquely by female T cells. Together, our observations indicate that human T cells exhibit a sex difference in the production of IFNγ and IL-17A that may be driven by expressions of PPARα and PPARγ.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , PPAR alpha/physiology , PPAR gamma/physiology , T-Lymphocytes/metabolism , Androgens/physiology , Animals , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Sex Factors
13.
Ann Neurol ; 73(3): 341-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23463494

ABSTRACT

OBJECTIVE: 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). METHODS: Clinical and brain magnetic resonance imaging measures of disease activity were monitored serially in patients participating in the Canadian MS HSCT Study. Reconstitution kinetics of immune-cell subsets were determined by flow cytometry, whereas thymic function was assessed using T-cell receptor excision circle analyses as well as flow cytometry measurements of CD31+ recent thymic emigrants (RTEs). Functional assays were performed to track central nervous system-autoreactive antigen-specific T-cell responses, and the relative capacity to generate Th1, Th17, or Th1/17 T-cell responses. RESULTS: Complete abrogation of new clinical relapses and new focal inflammatory brain lesions throughout the 2 years of immune monitoring following treatment was associated with sustained decrease in naive T cells, in spite of restoration of both thymic function and release of RTEs during reconstitution. Re-emergence as well as in vivo expansion of autoreactive T cells to multiple myelin targets was evident in all patients studied. The reconstituted myelin-specific T cells exhibited the same Th1 and Th2 responses as preablation myelin-reactive T cells. In contrast, the post-therapy T-cell repertoire exhibited a significantly diminished capacity for Th17 responses. INTERPRETATION: Our results indicate that diminished Th17 and Th1/17 responses, rather than Th1 responses, are particularly relevant to the abrogation of new relapsing disease activity observed in this cohort of patients with aggressive MS following chemoablation and HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Lymphocyte Activation/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/surgery , Th17 Cells/immunology , Th17 Cells/pathology , Adult , Antigens, CD/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Flow Cytometry , Follow-Up Studies , Glatiramer Acetate , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Lymphocyte Activation/drug effects , Lymphocyte Count , Lymphokines/pharmacology , Male , Myelin Basic Protein/metabolism , Myelin-Oligodendrocyte Glycoprotein/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Th1 Cells/drug effects , Th1 Cells/pathology , Th17 Cells/drug effects
14.
Nat Med ; 13(10): 1173-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17828272

ABSTRACT

T(H)17 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, T(H)17 lymphocytes transmigrate efficiently across BBB-ECs, highly express granzyme B, kill human neurons and promote central nervous system inflammation through CD4+ lymphocyte recruitment.


Subject(s)
Blood-Brain Barrier/immunology , Central Nervous System/physiopathology , Inflammation/physiopathology , T-Lymphocytes, Helper-Inducer/immunology , Blood-Brain Barrier/drug effects , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Cell Membrane Permeability , Cell Movement/immunology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Granzymes/immunology , Humans , Inflammation/etiology , Interleukin-17/immunology , Interleukin-17/pharmacology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Receptors, Interleukin/immunology , T-Lymphocytes, Helper-Inducer/enzymology , Tight Junctions/metabolism , Tight Junctions/pathology
15.
Brain ; 136(Pt 6): 1760-77, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23687119

ABSTRACT

Inflammatory perivascular cuffs are comprised of leucocytes that accumulate in the perivascular space around post-capillary venules before their infiltration into the parenchyma of the central nervous system. Inflammatory perivascular cuffs are commonly found in the central nervous system of patients with multiple sclerosis and in the animal model experimental autoimmune encephalomyelitis. Leucocytes that accumulate in the perivascular space secrete matrix metalloproteinases that aid their transmigration into the neural parenchyma. We described previously that the upstream inducer of matrix metalloproteinase expression, extracellular matrix metalloproteinase inducer (CD147), was elevated in experimental autoimmune encephalomyelitis, and that its inhibition reduced leucocyte entry into the central nervous system. Here we investigated whether the expression of extracellular matrix metalloproteinase inducer varies with the temporal evolution of lesions in murine experimental autoimmune encephalomyelitis, whether it was uniformly upregulated across multiple sclerosis specimens, and whether it was a feature of inflammatory perivascular cuffs in multiple sclerosis lesions. In experimental autoimmune encephalomyelitis, elevation of extracellular matrix metalloproteinase inducer was correlated with the appearance and persistence of clinical signs of disease. In both murine and human samples, extracellular matrix metalloproteinase inducer was detected on endothelium in healthy and disease states but was dramatically increased in and around inflammatory perivascular cuffs on leucocytes, associated with matrix metalloproteinase expression, and on resident cells including microglia. Leucocyte populations that express extracellular matrix metalloproteinase inducer in multiple sclerosis lesions included CD4+ and CD8+ T lymphocytes, B lymphocytes and monocyte/macrophages. The extra-endothelial expression of extracellular matrix metalloproteinase inducer was a marker of the activity of lesions in multiple sclerosis, being present on leucocyte-containing perivascular cuffs but not in inactive lesions. By using a function-blocking antibody, we implicate extracellular matrix metalloproteinase inducer in the adhesion of leucocytes to endothelial cells and determined that its activity was more crucial on leucocytes than on endothelium in leucocyte-endothelial cell engagement in vitro. Extracellular matrix metalloproteinase inducer activity regulated the level of alpha 4 integrin on leucocytes through a mechanism associated with nuclear factor κB signalling. Blocking extracellular matrix metalloproteinase inducer attenuated the transmigration of monocytes and B lymphocytes across a model of the blood-brain barrier in culture. In summary, we describe the prominence of extracellular matrix metalloproteinase inducer in central nervous system inflammatory perivascular cuffs, emphasize its dual role in matrix metalloproteinase induction and leucocyte adhesion, and highlight the elevation of extracellular matrix metalloproteinase inducer as an orchestrator of the infiltration of leucocytes into the central nervous system parenchyma.


Subject(s)
Extracellular Matrix/enzymology , Matrix Metalloproteinases/biosynthesis , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Adult , Aged , Aged, 80 and over , Animals , Basigin/physiology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Induction/physiology , Extracellular Matrix/pathology , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Matrix Metalloproteinases/physiology , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophil Infiltration/physiology
16.
Brain ; 135(Pt 10): 2906-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22975388

ABSTRACT

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.


Subject(s)
Cell Movement/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Animals , Biomarkers/metabolism , CD146 Antigen/metabolism , CD146 Antigen/physiology , Cells, Cultured , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurogenic Inflammation/immunology , Neurogenic Inflammation/metabolism , Neurogenic Inflammation/pathology
17.
Front Pharmacol ; 14: 1098378, 2023.
Article in English | MEDLINE | ID: mdl-38094892

ABSTRACT

Introduction: Zuotai is an ancient mineral-herbal mixture containing ß-HgS in Tibetan medicine. It is used to treat nervous system diseases, similar to Chinese medicine cinnabar and Indian Ayurveda medicine Rasasindura. However, one of the key problems faced by Zuotai is that its indications are ambiguous. Our previous study found that Zuotai exhibited the activity of ameliorating depressive-like behaviors in a chronic mild stress model. However, due to the inherent limitations of animal models in simulating human disease, clear results often require more than one model for confirmation. Methods: Therefore, another depression model, chronic restraint stressed (CRS) mice, was used to validate the antidepression effect of Zuotai. Prophylactic treatment was conducted for 21 consecutive days while mice were subjected to chronic restraint stress. Results: It was observed that Zuotai and ß-HgS alleviated anhedonia, behavioral despair, stereotype behavior, and reduced exploratory and spontaneous movement in CRS mice. Zuotai and ß-HgS also reversed the increases of stress hormone corticosterone (Cort) in serum and pro-inflammatory cytokines in serum and brain, and increased the serotonin in cortex in CRS mice, with positive dose-effect relationship. The number of Ki67-positive cells in the dentate gyrus and the level of brain-derived neurotrophic factor (BDNF) in the hippocampus were slightly elevated in CRS mice treated with Zuotai; however, there was no statistically significant difference. Although Zuotai increased the total Hg concentration in main organs, the levels remained below those needed to result in observed adverse effect, at least for kidney and liver; and Zuotai showed no observed adverse effect on the brain histopathology, the cell proliferation in dentate gyrus, as well as the hippocampal and cortical organ coefficients. Conclusion: Zuotai exhibited the alleviation of depressive-like behaviors in CRS mice, accompanying with ameliorating stress hormone, peripherical and cerebral inflammation, and monoamine neurotransmitter.

18.
Nat Commun ; 14(1): 5555, 2023 09 09.
Article in English | MEDLINE | ID: mdl-37689786

ABSTRACT

Multiple Sclerosis (MS) is a chronic autoimmune inflammatory disorder of the central nervous system (CNS). Current therapies mainly target inflammatory processes during acute stages, but effective treatments for progressive MS are limited. In this context, astrocytes have gained increasing attention as they have the capacity to drive, but also suppress tissue-degeneration. Here we show that astrocytes upregulate the immunomodulatory checkpoint molecule PD-L1 during acute autoimmune CNS inflammation in response to aryl hydrocarbon receptor and interferon signaling. Using CRISPR-Cas9 genetic perturbation in combination with small-molecule and antibody-mediated inhibition of PD-L1 and PD-1 both in vivo and in vitro, we demonstrate that astrocytic PD-L1 and its interaction with microglial PD-1 is required for the attenuation of autoimmune CNS inflammation in acute and progressive stages in a mouse model of MS. Our findings suggest the glial PD-L1/PD-1 axis as a potential therapeutic target for both acute and progressive MS stages.


Subject(s)
Microglia , Multiple Sclerosis , Animals , Mice , Astrocytes , Neuroinflammatory Diseases , Programmed Cell Death 1 Receptor/genetics , B7-H1 Antigen/genetics , Inflammation
19.
Ann Neurol ; 70(5): 751-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22162058

ABSTRACT

OBJECTIVE: Blood-derived myeloid antigen-presenting cells (APCs) account for a significant proportion of the leukocytes found within lesions of multiple sclerosis (MS) and experimental allergic encephalomyelitis (EAE). These APCs along with activated microglia are thought to be pivotal in the initiation of the central nervous system (CNS)-targeted immune response in MS and EAE. However, the exact molecules that direct the migration of myeloid cells from the periphery across the blood-brain barrier (BBB) remain largely unknown. METHODS: We identified Ninjurin-1 in a proteomic screen of human BBB endothelial cells (ECs). We assessed the expression of Ninjurin-1 by BBB-ECs and immune cells, and we determined the role of Ninjurin-1 in immune cell migration to the CNS in vivo in EAE mice. RESULTS: Ninjurin-1 was found to be weakly expressed in the healthy human and mouse CNS but upregulated on BBB-ECs and on infiltrating APCs during the course of EAE and in active MS lesions. In human peripheral blood, Ninjurin-1 was predominantly expressed by monocytes, whereas it was barely detectable on T and B lymphocytes. Moreover, Ninjurin-1 neutralization specifically abrogated the adhesion and migration of human monocytes across BBB-ECs, without affecting lymphocyte recruitment. Finally, Ninjurin-1 blockade reduced clinical disease activity and histopathological indices of EAE and decreased infiltration of macrophages, dendritic cells, and APCs into the CNS. INTERPRETATION: Our study uncovers an important cell-specific role for Ninjurin-1 in the transmigration of inflammatory APCs across the BBB and further emphasizes the importance of myeloid cell recruitment during the development of neuroinflammatory lesions.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism , Nerve Growth Factors/metabolism , Animals , B-Lymphocytes/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Monocytes/metabolism , T-Lymphocytes/metabolism
20.
Brain ; 134(Pt 12): 3560-77, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22058139

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(-) granzymeB(hi)). 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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Encephalitis, Viral/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/metabolism , Multiple Sclerosis/immunology , Adult , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Coronavirus Infections/metabolism , Encephalitis, Viral/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Immunologic Memory , Mice , Middle Aged , Multiple Sclerosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL