Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
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
2.
Article in English | MEDLINE | ID: mdl-36241608

ABSTRACT

BACKGROUND AND OBJECTIVES: In multiple sclerosis (MS), peripheral immune cells use various cell trafficking molecules to infiltrate the CNS where they cause damage.The objective of this study was to investigate the involvement of coxsackie and adenovirus receptor-like membrane protein (CLMP) in the migration of immune cells into the CNS of patients with MS. METHODS: Expression of CLMP was measured in primary cultures of human brain endothelial cells (HBECs) and human meningeal endothelial cells (HMECs), postmortem brain samples, and peripheral blood mononuclear cells (PBMCs) from patients with MS and controls by RNA sequencing, quantitative PCR, immunohistochemistry, and flow cytometry. In vitro migration assays using HBECs and HMECs were performed to evaluate the function of CLMP. RESULTS: Using bulk RNA sequencing of primary cultures of human brain and meningeal endothelial cells (ECs), we have identified CLMP as a new potential cell trafficking molecule upregulated in inflammatory conditions. We first confirmed the upregulation of CLMP at the protein level on TNFα-activated and IFNγ-activated primary cultures of human brain and meningeal ECs. In autopsy brain specimens from patients with MS, we demonstrated an overexpression of endothelial CLMP in active MS lesions when compared with normal control brain tissue. Flow cytometry of human PBMCs demonstrated an increased frequency of CLMP+ B lymphocytes and monocytes in patients with MS, when compared with that in healthy controls. The use of a blocking antibody against CLMP reduced the migration of immune cells across the human brain and meningeal ECs in vitro. Finally, we found CLMP+ immune cell infiltrates in the perivascular area of parenchymal lesions and in the meninges of patients with MS. DISCUSSION: Collectively, our data demonstrate that CLMP is an adhesion molecule used by immune cells to access the CNS during neuroinflammatory disorders such as MS. CLMP could represent a target for a new treatment of neuroinflammatory conditions.


Subject(s)
Multiple Sclerosis , Humans , Brain/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Endothelial Cells/metabolism , Leukocytes/metabolism , Leukocytes, Mononuclear , Tumor Necrosis Factor-alpha/metabolism
4.
Nature ; 557(7707): 724-728, 2018 05.
Article in English | MEDLINE | ID: mdl-29769726

ABSTRACT

Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1-3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/microbiology , Microglia/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Central Nervous System/metabolism , Central Nervous System/microbiology , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , ErbB Receptors/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Inflammation/prevention & control , Lipopolysaccharide Receptors/metabolism , Mice , Mice, Inbred C57BL , Microglia/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Receptors, Aryl Hydrocarbon/metabolism , Symbiosis , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor alpha/metabolism , Tryptophan/deficiency , Tryptophan/metabolism , Vascular Endothelial Growth Factor B/biosynthesis , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
6.
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
7.
Cell Mol Life Sci ; 74(10): 1871-1881, 2017 05.
Article in English | MEDLINE | ID: mdl-28025672

ABSTRACT

The endothelium provides a strong barrier separating circulating blood from tissue. It also provides a significant challenge for immune cells in the bloodstream to access potential sites of infection. To mount an effective immune response, leukocytes traverse the endothelial layer in a process known as transendothelial migration. Decades of work have allowed dissection of the mechanisms through which immune cells gain access into peripheral tissues, and subsequently to inflammatory foci. However, an often under-appreciated or potentially ignored question is whether transmigrated leukocytes can leave these inflammatory sites, and perhaps even return across the endothelium and re-enter circulation. Although evidence has existed to support "reverse" transendothelial migration for a number of years, it is only recently that mechanisms associated with this process have been described. Here we review the evidence that supports both reverse transendothelial migration and reverse interstitial migration within tissues, with particular emphasis on some of the more recent studies that finally hint at potential mechanisms. Additionally, we postulate the biological significance of retrograde migration, and whether it serves as an additional mechanism to limit pathology, or provides a basis for the dissemination of systemic inflammation.


Subject(s)
Endothelial Cells/pathology , Inflammation/pathology , Monocytes/pathology , Neutrophils/pathology , T-Lymphocytes/pathology , Transendothelial and Transepithelial Migration , Animals , Endothelial Cells/immunology , Humans , Inflammation/immunology , Monocytes/immunology , Neutrophils/immunology , T-Lymphocytes/immunology
8.
J Exp Med ; 213(6): 929-49, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27139491

ABSTRACT

Growing evidence supports a role for IL-1 in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), but how it impacts neuroinflammation is poorly understood. We show that susceptibility to EAE requires activation of IL-1R1 on radiation-resistant cells via IL-1ß secreted by bone marrow-derived cells. Neutrophils and monocyte-derived macrophages (MDMs) are the main source of IL-1ß and produce this cytokine as a result of their transmigration across the inflamed blood-spinal cord barrier. IL-1R1 expression in the spinal cord is found in endothelial cells (ECs) of the pial venous plexus. Accordingly, leukocyte infiltration at EAE onset is restricted to IL-1R1(+) subpial and subarachnoid vessels. In response to IL-1ß, primary cultures of central nervous system ECs produce GM-CSF, G-CSF, IL-6, Cxcl1, and Cxcl2. Initiation of EAE or subdural injection of IL-1ß induces a similar cytokine/chemokine signature in spinal cord vessels. Furthermore, the transfer of Gr1(+) cells on the spinal cord is sufficient to induce illness in EAE-resistant IL-1ß knockout (KO) mice. Notably, transfer of Gr1(+) cells isolated from C57BL/6 mice induce massive recruitment of recipient myeloid cells compared with cells from IL-1ß KO donors, and this recruitment translates into more severe paralysis. These findings suggest that an IL-1ß-dependent paracrine loop between infiltrated neutrophils/MDMs and ECs drives neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-1beta/immunology , Macrophages/immunology , Multiple Sclerosis/immunology , Paracrine Communication/immunology , Spinal Cord/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-1beta/genetics , Macrophages/pathology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neutrophils/immunology , Neutrophils/pathology , Paracrine Communication/genetics , Spinal Cord/pathology , Transendothelial and Transepithelial Migration/genetics
9.
Curr Opin Pharmacol ; 26: 39-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26480201

ABSTRACT

The blood-brain barrier (BBB) is comprised of unique endothelial cells (ECs) that regulate the delicate central nervous system (CNS) microenvironment. During development, vasculature sprouts from the perivascular neural plexus and penetrates the CNS parenchyma. Recent studies indicate that these nascent vessels rely on radial glia (RG)-secreted factors for guidance and barrier induction. This early association also sustains astrocyte development, allowing for a tight interaction between these mature glia and ECs. The astrocyte-EC interface is crucial to BBB function and is substantially modified during pathology. Understanding the relationship between astrocytes and ECs lays the groundwork for advancing protective therapies that target neuroinflammatory disorders.


Subject(s)
Blood-Brain Barrier/physiology , Neuroglia/physiology , Animals , Homeostasis , Humans
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.
Neurobiol Dis ; 74: 14-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25448765

ABSTRACT

Early changes in the normal appearing white matter of multiple sclerosis (MS) patients precede the appearance of gadolinium-enhancing lesions. Although these findings suggest blood-brain barrier (BBB) breakdown as an important feature in MS pathogenesis, limited information is available on the BBB alterations during lesion genesis. Here, we perform a longitudinal characterization of the vascular, neuropathological and immunological changes before lesion formation in mice developing spontaneous relapsing-remitting experimental autoimmune encephalomyelitis (sRR-EAE). We found a significant upregulation of Th1 and Th17 cytokines in the periphery of sRR-EAE mice before any evident neuropathology. In the CNS, BBB and astroglial activations were the first pathological changes occurring after 45days of age and were followed by immune cell infiltration by day 50. These pathological alterations subsequently led to perivascular demyelination and disease onset. In MS, (p)reactive lesions mirrored the changes seen in early sRR-EAE by displaying considerable BBB disruption, perivascular astrogliosis, redistribution of junctional proteins and increased expression of endothelial cell adhesion molecules. Our findings suggest that BBB breach occurs before significant immune cell infiltration and demyelination. In addition, peripheral immune activation during sRR-EAE precedes CNS pathology, suggesting that outside in signaling mechanisms play a role in the development of neuroinflammatory lesions.


Subject(s)
Blood-Brain Barrier/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis, Chronic Progressive/metabolism , Multiple Sclerosis, Relapsing-Remitting/metabolism , Adult , Aged , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gliosis/metabolism , Gliosis/pathology , Humans , Longitudinal Studies , Male , Mice, Transgenic , Middle Aged , Multiple Sclerosis, Chronic Progressive/pathology , Multiple Sclerosis, Relapsing-Remitting/pathology , T-Lymphocytes/metabolism
12.
Ann Clin Transl Neurol ; 2(11): 1032-7, 2015 11.
Article in English | MEDLINE | ID: mdl-26734656

ABSTRACT

Leukocyte transmigration into the central nervous system promotes multiple sclerosis pathogenesis, yet ambiguity remains regarding the mechanisms controlling the migration of distinct immune cell subsets. Using in vitro, ex vivo and postmortem human materials, we identified a significant upregulation of junctional adhesion molecule-like expression at the blood-brain barrier, monocytes, and CD8 T cells of multiple sclerosis patients. We also detected junctional adhesion molecule-like(+) trans-migratory cups when monocytes/CD8 T cells adhered to the blood-brain barrier, however, their migratory capacity was significantly compromised when junctional adhesion molecule-like was blocked. These findings highlight a novel role for junctional adhesion molecule-like in leukocyte transmigration and its potential as a promising therapeutic target.

13.
Glia ; 61(12): 1939-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123158

ABSTRACT

The Blood Brain Barrier (BBB) is a specialized vascular structure tightly regulating central nervous system (CNS) homeostasis. Endothelial cells are the central component of the BBB and control of their barrier phenotype resides on astrocytes and pericytes. Interactions between these cells and the endothelium promote and maintain many of the physiological and metabolic characteristics that are unique to the BBB. In this review we describe recent findings related to the involvement of astroglial cells, including radial glial cells, in the induction of barrier properties during embryogenesis and adulthood. In addition, we describe changes that occur in astrocytes and endothelial cells during injury and inflammation with a particular emphasis on alterations of the BBB phenotype.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Neuroglia/metabolism , Animals , Astrocytes/metabolism , Biological Transport , Humans
14.
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
15.
Science ; 334(6063): 1727-31, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22144466

ABSTRACT

The blood-brain barrier (BBB) is composed of tightly bound endothelial cells (ECs) and perivascular astrocytes that regulate central nervous system (CNS) homeostasis. We showed that astrocytes secrete Sonic hedgehog and that BBB ECs express Hedgehog (Hh) receptors, which together promote BBB formation and integrity during embryonic development and adulthood. Using pharmacological inhibition and genetic inactivation of the Hh signaling pathway in ECs, we also demonstrated a critical role of the Hh pathway in promoting the immune quiescence of BBB ECs by decreasing the expression of proinflammatory mediators and the adhesion and migration of leukocytes, in vivo and in vitro. Overall, the Hh pathway provides a barrier-promoting effect and an endogenous anti-inflammatory balance to CNS-directed immune attacks, as occurs in multiple sclerosis.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/physiology , Brain/immunology , Endothelial Cells/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Animals , Blood-Brain Barrier/cytology , Brain/physiology , CD4-Positive T-Lymphocytes/physiology , Cell Adhesion , Cell Movement , Cells, Cultured , Chemokines/metabolism , Electric Impedance , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Hedgehog Proteins/genetics , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Permeability , Veratrum Alkaloids/pharmacology
16.
FEBS Lett ; 585(23): 3770-80, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21550344

ABSTRACT

The presence of the blood-brain barrier (BBB) restricts the movement of soluble mediators and leukocytes from the periphery to the central nervous system (CNS). Leukocyte entry into the CNS is nonetheless an early event in multiple sclerosis (MS), an inflammatory disorder of the CNS. Whether BBB dysfunction precedes immune cell infiltration or is the consequence of perivascular leukocyte accumulation remains enigmatic, but leukocyte migration modifies BBB permeability. Immune cells of MS subjects express inflammatory cytokines, reactive oxygen species (ROS) and enzymes that can facilitate their migration to the CNS by influencing BBB function, either directly or indirectly. In this review, we describe how immune cells from the peripheral blood overcome the BBB and promote CNS inflammation in MS through BBB disruption.


Subject(s)
Blood-Brain Barrier/immunology , Lymphocytes/immunology , Multiple Sclerosis/immunology , Animals , Blood-Brain Barrier/pathology , Cell Movement , Humans , Matrix Metalloproteinases/metabolism , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Reactive Oxygen Species/metabolism
17.
Biochim Biophys Acta ; 1812(2): 252-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20619340

ABSTRACT

The delicate microenvironment of the central nervous system (CNS) is protected by the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCB). These barriers function in distinct CNS compartments and their anatomical basis lay on the junctional proteins present in endothelial cells for the BBB and in the choroidal epithelium for the BCB. During neuroinflammatory conditions like multiple sclerosis (MS) and its murine model experimental autoimmune encephalomyelitis (EAE), activation or damage of the various cellular components of these barriers facilitate leukocyte infiltration leading to oligodendrocyte death, axonal damage, demyelination and lesion development. This manuscript will review in detail the features of these barriers under physiological and pathological conditions, particularly when focal immune activation promotes the loss of the BBB and BCB phenotype, the upregulation of cell adhesion molecules (CAMs) and the recruitment of immune cells.


Subject(s)
Blood-Brain Barrier/immunology , Multiple Sclerosis/immunology , Animals , Blood-Brain Barrier/pathology , Cell Adhesion Molecules/immunology , Chemokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/cerebrospinal fluid , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Integrins/immunology , Intercellular Junctions/immunology , Leukocytes/immunology , Mice , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Selectins/immunology
18.
Ann Neurol ; 66(3): 390-402, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19810097

ABSTRACT

OBJECTIVE: There is substantial evidence supporting the role of interferon (IFN)-gamma-producing T helper (T(H)) 1 and interleukin (IL)-17-expressing T(H)17 lymphocytes in multiple sclerosis (MS) and its animal model, experimental allergic encephalomyelitis (EAE). However, to date little is known about the potential cooperative interplay between these 2 cytokines. In the current study, we sought to evaluate the frequency of IFN-gamma-expressing T(H)17 lymphocytes in MS and EAE, and study their recruitment into the central nervous system (CNS). METHODS: Human T(H)17 lymphocytes were expanded in vitro from the blood of healthy controls and relapsing MS patients using IL-23. Immune cell migration to the CNS was assessed in vitro with primary cultures of human blood-brain barrier (BBB)-derived endothelial cells, and in vivo in EAE mice. RESULTS: We demonstrate that in response to IL-23, human memory lymphocytes expand into a T(H)17 phenotype, with a subpopulation of cells simultaneously expressing IFN-gamma and IL-17. We note that lymphocytes obtained from the blood of relapsing MS patients have an increased propensity to expand into IFN-gamma-producing T(H)17 cells and identify numerous T lymphocytes coexpressing IL-17 and IFN-gamma in brain tissue of MS patients. We also find lymphocytes expressing both the T(H)1- and the T(H)17-associated transcription factors ROR gamma t and T-bet, in situ and in vitro. We further provide in vitro and in vivo evidence that IFN-gamma(+) T(H)17 lymphocytes preferentially cross the human BBB and accumulate in the CNS of mice during the effector phase of EAE. INTERPRETATION: Our data underscore the involvement of IFN-gamma(+) T(H)17 lymphocytes in the pathology of MS and EAE and their preferential recruitment into the CNS during inflammatory events.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Interferon-gamma/immunology , Interleukin-17/immunology , Multiple Sclerosis/blood , T-Lymphocytes/immunology , Adult , Animals , Blood-Brain Barrier/immunology , Cell Migration Assays , Cell Migration Inhibition/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , In Vitro Techniques , Interferon-gamma/blood , Interleukin-23/immunology , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/immunology , Th1 Cells/immunology , Th2 Cells/immunology
19.
Front Biosci ; 10: 582-7, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15569599

ABSTRACT

Toll Like Receptors (TLRs) are pathogen recognition receptors (PRRs) that respond to specific pathogen associated molecular patterns (PAMPs) during microbial invasion. After TLR stimulation a series of cellular responses initiate an inflammatory response and influence specific adaptive immunity that ultimately destroy the pathogen. But the immune response is not always able to control the infection. Pathogens have developed mechanisms to overcome and evade distinct arms of vertebrate immunity. Many of these strategies have been extensively described, but with the recent discovery of TLRs additional means to manipulate the innate immune response are currently being studied. Pathogens generally inhibit TLR mediated immunity by either blocking signals that stimulate further host defense mechanisms or by down-regulating their expression. These inhibitory mechanisms have been mainly elucidated in bacterial systems, whereas in other microorganisms they remain to be identified. Here the strategies that pathogenic microbes use to subvert TLR mediated immune responses are reviewed.


Subject(s)
Gene Expression Regulation , Immune System/microbiology , Immune System/pathology , Toll-Like Receptors/metabolism , Bacterial Infections/immunology , Down-Regulation , Humans , Immunity, Innate , Inflammation , Lipopolysaccharides/metabolism , MAP Kinase Signaling System , Membrane Glycoproteins/chemistry , NF-kappa B/metabolism , Signal Transduction , Virus Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...