Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell Rep Med ; 5(5): 101546, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38703766

ABSTRACT

Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by motor neuron (MN) loss. We previously discovered that macrophage migration inhibitory factor (MIF), whose levels are extremely low in spinal MNs, inhibits mutant SOD1 misfolding and toxicity. In this study, we show that a single peripheral injection of adeno-associated virus (AAV) delivering MIF into adult SOD1G37R mice significantly improves their motor function, delays disease progression, and extends survival. Moreover, MIF treatment reduces neuroinflammation and misfolded SOD1 accumulation, rescues MNs, and corrects dysregulated pathways as observed by proteomics and transcriptomics. Furthermore, we reveal low MIF levels in human induced pluripotent stem cell-derived MNs from familial ALS patients with different genetic mutations, as well as in post mortem tissues of sporadic ALS patients. Our findings indicate that peripheral MIF administration may provide a potential therapeutic mechanism for modulating misfolded SOD1 in vivo and disease outcome in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Macrophage Migration-Inhibitory Factors , Motor Neurons , Superoxide Dismutase-1 , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Humans , Motor Neurons/metabolism , Motor Neurons/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Mice , Induced Pluripotent Stem Cells/metabolism , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/genetics , Mice, Transgenic , Dependovirus/genetics , Disease Models, Animal , Male , Mutation/genetics , Female , Protein Folding
2.
Front Immunol ; 15: 1343900, 2024.
Article in English | MEDLINE | ID: mdl-38720902

ABSTRACT

Alzheimer's disease has an increasing prevalence in the population world-wide, yet current diagnostic methods based on recommended biomarkers are only available in specialized clinics. Due to these circumstances, Alzheimer's disease is usually diagnosed late, which contrasts with the currently available treatment options that are only effective for patients at an early stage. Blood-based biomarkers could fill in the gap of easily accessible and low-cost methods for early diagnosis of the disease. In particular, immune-based blood-biomarkers might be a promising option, given the recently discovered cross-talk of immune cells of the central nervous system with those in the peripheral immune system. Here, we give a background on recent advances in research on brain-immune system cross-talk in Alzheimer's disease and review machine learning approaches, which can combine multiple biomarkers with further information (e.g. age, sex, APOE genotype) into predictive models supporting an earlier diagnosis. In addition, mechanistic modeling approaches, such as agent-based modeling open the possibility to model and analyze cell dynamics over time. This review aims to provide an overview of the current state of immune-system related blood-based biomarkers and their potential for the early diagnosis of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Biomarkers , Early Diagnosis , Alzheimer Disease/diagnosis , Alzheimer Disease/immunology , Alzheimer Disease/blood , Humans , Biomarkers/blood , Machine Learning , Animals
3.
Nat Aging ; 2(1): 60-73, 2022 01.
Article in English | MEDLINE | ID: mdl-37118355

ABSTRACT

Microglia and monocyte-derived macrophages (MDM) are key players in dealing with Alzheimer's disease. In amyloidosis mouse models, activation of microglia was found to be TREM2 dependent. Here, using Trem2-/-5xFAD mice, we assessed whether MDM act via a TREM2-dependent pathway. We adopted a treatment protocol targeting the programmed cell death ligand-1 (PD-L1) immune checkpoint, previously shown to modify Alzheimer's disease via MDM involvement. Blockade of PD-L1 in Trem2-/-5xFAD mice resulted in cognitive improvement and reduced levels of water-soluble amyloid beta1-42 with no effect on amyloid plaque burden. Single-cell RNA sequencing revealed that MDM, derived from both Trem2-/- and Trem2+/+5xFAD mouse brains, express a unique set of genes encoding scavenger receptors (for example, Mrc1, Msr1). Blockade of monocyte trafficking using anti-CCR2 antibody completely abrogated the cognitive improvement induced by anti-PD-L1 treatment in Trem2-/-5xFAD mice and similarly, but to a lesser extent, in Trem2+/+5xFAD mice. These results highlight a TREM2-independent, disease-modifying activity of MDM in an amyloidosis mouse model.


Subject(s)
Alzheimer Disease , Amyloidosis , Mice , Animals , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Macrophages/metabolism , Amyloidosis/genetics , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics
5.
Nat Commun ; 10(1): 465, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692527

ABSTRACT

Alzheimer's disease (AD) is a heterogeneous disorder with multiple etiologies. Harnessing the immune system by blocking the programmed cell death receptor (PD)-1 pathway in an amyloid beta mouse model was shown to evoke a sequence of immune responses that lead to disease modification. Here, blocking PD-L1, a PD-1 ligand, was found to have similar efficacy to that of PD-1 blocking in disease modification, in both animal models of AD and of tauopathy. Targeting PD-L1 in a tau-driven disease model resulted in increased immunomodulatory monocyte-derived macrophages within the brain parenchyma. Single cell RNA-seq revealed that the homing macrophages expressed unique scavenger molecules including macrophage scavenger receptor 1 (MSR1), which was shown here to be required for the effect of PD-L1 blockade in disease modification. Overall, our results demonstrate that immune checkpoint blockade targeting the PD-1/PD-L1 pathway leads to modification of common factors that go awry in AD and dementia, and thus can potentially provide an immunotherapy to help combat these diseases.


Subject(s)
B7-H1 Antigen/metabolism , Cognitive Dysfunction/metabolism , Macrophages/metabolism , Programmed Cell Death 1 Receptor/metabolism , Tauopathies/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Antibodies, Blocking/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Brain/immunology , Brain/metabolism , Cognitive Dysfunction/genetics , Disease Models, Animal , Humans , Macrophages/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Tauopathies/genetics
6.
Nat Commun ; 8(1): 717, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959042

ABSTRACT

During ageing, microglia acquire a phenotype that may negatively affect brain function. Here we show that ageing microglial phenotype is largely imposed by interferon type I (IFN-I) chronically present in aged brain milieu. Overexpression of IFN-ß in the CNS of adult wild-type mice, but not of mice lacking IFN-I receptor on their microglia, induces an ageing-like transcriptional microglial signature, and impairs cognitive performance. Furthermore, we demonstrate that age-related IFN-I milieu downregulates microglial myocyte-specific enhancer factor 2C (Mef2C). Immune challenge in mice lacking Mef2C in microglia results in an exaggerated microglial response and has an adverse effect on mice behaviour. Overall, our data indicate that the chronic presence of IFN-I in the brain microenvironment, which negatively affects cognitive function, is mediated via modulation of microglial activity. These findings may shed new light on other neurological conditions characterized by elevated IFN-I signalling in the brain.Microglia cells in the brain regulate immune responses, but in ageing can negatively affect brain function. Here the authors show that the chronic presence of type I interferon in aged mouse brain impedes cognitive ability by altering microglia transcriptome and limiting Mef2C, a microglia 'off' signal.


Subject(s)
Aging/immunology , Brain/immunology , Interferon-beta/immunology , MEF2 Transcription Factors/immunology , Microglia/immunology , Animals , Brain/growth & development , Brain/physiopathology , Humans , Interferon-beta/genetics , MEF2 Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL
7.
Cell ; 169(7): 1276-1290.e17, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28602351

ABSTRACT

Alzheimer's disease (AD) is a detrimental neurodegenerative disease with no effective treatments. Due to cellular heterogeneity, defining the roles of immune cell subsets in AD onset and progression has been challenging. Using transcriptional single-cell sorting, we comprehensively map all immune populations in wild-type and AD-transgenic (Tg-AD) mouse brains. We describe a novel microglia type associated with neurodegenerative diseases (DAM) and identify markers, spatial localization, and pathways associated with these cells. Immunohistochemical staining of mice and human brain slices shows DAM with intracellular/phagocytic Aß particles. Single-cell analysis of DAM in Tg-AD and triggering receptor expressed on myeloid cells 2 (Trem2)-/- Tg-AD reveals that the DAM program is activated in a two-step process. Activation is initiated in a Trem2-independent manner that involves downregulation of microglia checkpoints, followed by activation of a Trem2-dependent program. This unique microglia-type has the potential to restrict neurodegeneration, which may have important implications for future treatment of AD and other neurodegenerative diseases. VIDEO ABSTRACT.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Microglia/pathology , Phagocytes/pathology , Alzheimer Disease/genetics , Animals , Humans , Mice , Mice, Transgenic , Microglia/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Phagocytes/metabolism , Receptors, Immunologic/metabolism , Sequence Analysis, RNA , Single-Cell Analysis
8.
Cell Stem Cell ; 18(6): 689-691, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27257756

ABSTRACT

Hippocampal neurogenesis is affected throughout life by factors external to the brain, including what we eat, our gut microbiota, and the immune system. However, the mechanisms that link microbiota to neurogenesis are still puzzling. Now in Cell Reports, Möhle et al. (2016) attribute a role to Ly6C(hi) monocytes in this gut-immune-brain axis.


Subject(s)
Gastrointestinal Tract/immunology , Monocytes/immunology , Brain , Humans , Microbiota , Neurogenesis
9.
Science ; 353(6301): aad8670, 2016 Aug 19.
Article in English | MEDLINE | ID: mdl-27338705

ABSTRACT

Microglia, the resident myeloid cells of the central nervous system, play important roles in life-long brain maintenance and in pathology. Despite their importance, their regulatory dynamics during brain development have not been fully elucidated. Using genome-wide chromatin and expression profiling coupled with single-cell transcriptomic analysis throughout development, we found that microglia undergo three temporal stages of development in synchrony with the brain--early, pre-, and adult microglia--which are under distinct regulatory circuits. Knockout of the gene encoding the adult microglia transcription factor MAFB and environmental perturbations, such as those affecting the microbiome or prenatal immune activation, led to disruption of developmental genes and immune response pathways. Together, our work identifies a stepwise microglia developmental program integrating immune response pathways that may be associated with several neurodevelopmental disorders.


Subject(s)
Brain/embryology , Homeostasis/physiology , Microglia/cytology , Neurogenesis/immunology , Animals , Blood-Brain Barrier/embryology , Blood-Brain Barrier/immunology , Brain/immunology , Chromatin/metabolism , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Knockout Techniques , Histone Code , Homeostasis/genetics , Immunity/genetics , MafB Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Myeloid Cells/cytology , Neurogenesis/genetics , Single-Cell Analysis
10.
Trends Immunol ; 37(3): 181-192, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26877243

ABSTRACT

Recent findings have revealed distinct roles for type I and II interferons (IFN-I and IFN-γ) in the recruitment of immune cells to the central nervous system (CNS) and highlighted the importance of this process for brain maintenance and protection/repair. Furthermore, manipulation of IFN-I and IFN-γ pathways in pathological contexts has yielded conflicting results. We discuss these findings, focusing on two distinct conditions; relapsing remitting multiple sclerosis (RRMS) and brain aging. Using these examples, we propose that regulation of immune cell entry to the CNS is a mechanism through which interaction between IFN-I and -II can affect brain function from its anatomical borders. Deviation from homeostatic IFN-I/-II balance may contribute to distinct brain pathologies, resulting from either insufficient immune surveillance of the CNS and loss of immune-dependent protection, or overwhelming leukocyte entry and immune-mediated destruction.


Subject(s)
Aging/immunology , Brain/physiology , Interferon Type I/physiology , Interferon-gamma/physiology , Multiple Sclerosis, Relapsing-Remitting/immunology , Animals , Autoimmunity , Homeostasis , Humans , Immunologic Surveillance
11.
Nat Med ; 22(2): 135-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26779813

ABSTRACT

Systemic immune suppression may curtail the ability to mount the protective, cell-mediated immune responses that are needed for brain repair. By using mouse models of Alzheimer's disease (AD), we show that immune checkpoint blockade directed against the programmed death-1 (PD-1) pathway evokes an interferon (IFN)-γ-dependent systemic immune response, which is followed by the recruitment of monocyte-derived macrophages to the brain. When induced in mice with established pathology, this immunological response leads to clearance of cerebral amyloid-ß (Aß) plaques and improved cognitive performance. Repeated treatment sessions were required to maintain a long-lasting beneficial effect on disease pathology. These findings suggest that immune checkpoints may be targeted therapeutically in AD.


Subject(s)
Alzheimer Disease/immunology , Antibodies/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Macrophages/drug effects , Memory/drug effects , Plaque, Amyloid/immunology , Programmed Cell Death 1 Receptor/immunology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/immunology , Brain/pathology , Cognition/drug effects , Disease Models, Animal , Flow Cytometry , Humans , Immunohistochemistry , Interferon-gamma/drug effects , Interferon-gamma/immunology , Macrophages/immunology , Maze Learning/drug effects , Mice , Mice, Transgenic , Plaque, Amyloid/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sequence Analysis, RNA , Signal Transduction/drug effects
12.
Nat Commun ; 6: 7967, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26284939

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder in which chronic neuroinflammation contributes to disease escalation. Nevertheless, while immunosuppressive drugs have repeatedly failed in treating this disease, recruitment of myeloid cells to the CNS was shown to play a reparative role in animal models. Here we show, using the 5XFAD AD mouse model, that transient depletion of Foxp3(+) regulatory T cells (Tregs), or pharmacological inhibition of their activity, is followed by amyloid-ß plaque clearance, mitigation of the neuroinflammatory response and reversal of cognitive decline. We further show that transient Treg depletion affects the brain's choroid plexus, a selective gateway for immune cell trafficking to the CNS, and is associated with subsequent recruitment of immunoregulatory cells, including monocyte-derived macrophages and Tregs, to cerebral sites of plaque pathology. Our findings suggest targeting Treg-mediated systemic immunosuppression for treating AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Dentate Gyrus/pathology , Dentate Gyrus/physiology , Forkhead Transcription Factors/genetics , Gene Expression Regulation/physiology , Glatiramer Acetate/pharmacology , Immune Tolerance , Immunomodulation , Mice , Mice, Transgenic , RNA/genetics , RNA/metabolism
13.
EMBO J ; 34(13): 1816-28, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25940071

ABSTRACT

Chronic neuroinflammation is evident in brain aging and neurodegenerative disorders and is often associated with excessive nitric oxide (NO) production within the central nervous system (CNS). Under such conditions, increased NO levels are observed at the choroid plexus (CP), an epithelial layer that forms the blood-cerebrospinal fluid barrier (BCSFB) and serves as a selective gateway for leukocyte entry to the CNS in homeostasis and following injury. Here, we hypothesized that elevated cerebral NO levels interfere with CP gateway activity. We found that induction of leukocyte trafficking determinants by the CP and sequential leukocyte entry to the CSF are dependent on the CP epithelial NFκB/p65 signaling pathway, which was inhibited upon exposure to NO. Examining the CP in 5XFAD transgenic mouse model of Alzheimer's disease (AD-Tg) revealed impaired ability to mount an NFκB/p65-dependent response. Systemic administration of an NO scavenger in AD-Tg mice alleviated NFκB/p65 suppression at the CP and augmented its gateway activity. Together, our findings identify cerebral NO as a negative regulator of CP gateway activity for immune cell trafficking to the CNS.


Subject(s)
Brain/metabolism , Chemotaxis, Leukocyte/drug effects , Choroid Plexus/drug effects , Leukocytes/drug effects , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Cells, Cultured , Choroid Plexus/immunology , Choroid Plexus/metabolism , Disease Models, Animal , Female , Leukocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
J Neurosci ; 35(16): 6381-93, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904790

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating fatal motor neuron disease, for which there is currently no cure or effective treatment. In this disease, local neuroinflammation develops along the disease course and contributes to its rapid progression. In several models of CNS pathologies, circulating immune cells were shown to display an indispensable role in the resolution of the neuroinflammatory response. The recruitment of such cells to the CNS involves activation of the choroid plexus (CP) of the brain for leukocyte trafficking, through a mechanism that requires IFN-γ signaling. Here, we found that in the mutant SOD1(G93A) (mSOD1) mouse model of ALS, the CP does not support leukocyte trafficking during disease progression, due to a local reduction in IFN-γ levels. Therapeutic immunization of mSOD1 mice with a myelin-derived peptide led to CP activation, and was followed by the accumulation of immunoregulatory cells, including IL-10-producing monocyte-derived macrophages and Foxp3(+) regulatory T cells, and elevation of the neurotrophic factors IGF-1 and GDNF in the diseased spinal cord parenchyma. The immunization resulted in the attenuation of disease progression and an increased life expectancy of the mSOD1 mice. Collectively, our results demonstrate that recruitment of immunoregulatory cells to the diseased spinal cord in ALS, needed for fighting off the pathology, can be enhanced by transiently boosting peripheral immunity to myelin antigens.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Choroid Plexus/cytology , Choroid Plexus/immunology , Disease Progression , Immunization , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Movement/immunology , Disease Models, Animal , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Transgenic , Mutation , Primary Cell Culture , Spinal Cord/immunology , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , T-Lymphocytes/metabolism
15.
J Autoimmun ; 60: 40-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25911200

ABSTRACT

Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.


Subject(s)
Apoptosis/genetics , Blood-Brain Barrier/physiopathology , Neurons/pathology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factors/immunology , Animals , Apoptosis/immunology , Choroid Plexus/physiopathology , Cognition , Complement C3/immunology , Complement C4a/immunology , Complement C6/immunology , Cytokine TWEAK , Depression/genetics , Disease Models, Animal , Gliosis/genetics , Immunoglobulin G/immunology , Intercellular Adhesion Molecule-1/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Nerve Degeneration/genetics , Permeability , Signal Transduction/genetics , Signal Transduction/immunology , TWEAK Receptor , Vascular Cell Adhesion Molecule-1/metabolism , Zonula Occludens-1 Protein/biosynthesis
16.
J Autoimmun ; 54: 8-14, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25199710

ABSTRACT

Immune cell infiltration to the brain's territory was considered for decades to reflect a pathological process in which immune cells attack the central nervous system (CNS); such a process is observed in the inflammatory autoimmune disease, multiple sclerosis (MS). As neuroinflammatory processes within the CNS parenchyma are also common to other CNS pathologies, regardless of their etiology, including neurodegenerative disorders such as Alzheimer's disease (AD) and Amyotrophic lateral sclerosis (ALS), these pathologies have often been compared to MS, a disease that benefits from immunosuppressive therapy. Yet, over the last decade, it became clear that autoimmunity has a bright side, and that it plays a pivotal role in CNS repair following damage. Specifically, autoimmune T cells were found to facilitate CNS healing processes, such as in the case of sterile mechanical injuries to the brain or the spinal cord, mental stress, or biochemical insults. Even more intriguingly, autoimmune T cells were found to be involved in supporting fundamental processes of brain functional integrity, such as in the maintenance of life-long brain plasticity, including spatial learning and memory, and neurogenesis. Importantly, autoimmune T cells are part of a cellular network which, to operate efficiently and safely, requires tight regulation by other immune cell populations, such as regulatory T cells, which are indispensable for maintenance of immunological self-tolerance and homeostasis. Here, we suggest that dysregulation of the balance between peripheral immune suppression, on one hand, and protective autoimmunity, on the other, is an underlying mechanism in the emergence and progression of the neuroinflammatory response associated with chronic neurodegenerative diseases and brain aging. Mitigating chronic neuroinflammation under these conditions necessitates activation, rather than suppression, of the peripheral immune response directed against self. Accordingly, we propose that fighting off acute and chronic neurodegenerative conditions requires breaking peripheral immune tolerance to CNS self-antigens, in order to boost protective autoimmunity. Nevertheless, the optimal approach to fine tune such immune response must be individually explored for each condition.


Subject(s)
Alzheimer Disease/immunology , Amyotrophic Lateral Sclerosis/immunology , Autoantigens/immunology , Immune Tolerance , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/immunology , Brain/pathology , Chronic Disease , Humans , Multiple Sclerosis/pathology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes, Regulatory/pathology
17.
Science ; 346(6205): 89-93, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25147279

ABSTRACT

Aging-associated cognitive decline is affected by factors produced inside and outside the brain. By using multiorgan genome-wide analysis of aged mice, we found that the choroid plexus, an interface between the brain and the circulation, shows a type I interferon (IFN-I)-dependent gene expression profile that was also found in aged human brains. In aged mice, this response was induced by brain-derived signals, present in the cerebrospinal fluid. Blocking IFN-I signaling within the aged brain partially restored cognitive function and hippocampal neurogenesis and reestablished IFN-II-dependent choroid plexus activity, which is lost in aging. Our data identify a chronic aging-induced IFN-I signature, often associated with antiviral response, at the brain's choroid plexus and demonstrate its negative influence on brain function, thereby suggesting a target for ameliorating cognitive decline in aging.


Subject(s)
Aging/pathology , Brain/physiology , Choroid Plexus/metabolism , Cognition , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferon Type I/physiology , Aging/genetics , Animals , Hippocampus/cytology , Mice , Mice, Transgenic , Neurogenesis , Receptors, Interferon/genetics , Interferon gamma Receptor
18.
EMBO J ; 33(1): 7-22, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24357543

ABSTRACT

Inflammation is an integral part of the body's physiological repair mechanism, unless it remains unresolved and becomes pathological, as evident in the progressive nature of neurodegeneration. Based on studies from outside the central nervous system (CNS), it is now understood that the resolution of inflammation is an active process, which is dependent on well-orchestrated innate and adaptive immune responses. Due to the immunologically privileged status of the CNS, such resolution mechanism has been mostly ignored. Here, we discuss resolution of neuroinflammation as a process that depends on a network of immune cells operating in a tightly regulated sequence, involving the brain's choroid plexus (CP), a unique neuro-immunological interface, positioned to integrate signals it receives from the CNS parenchyma with signals coming from circulating immune cells, and to function as an on-alert gate for selective recruitment of inflammation-resolving leukocytes to the inflamed CNS parenchyma. Finally, we propose that functional dysregulation of the CP reflects a common underlying mechanism in the pathophysiology of neurodegenerative diseases, and can thus serve as a potential novel target for therapy.


Subject(s)
Brain Diseases/immunology , Brain Diseases/pathology , Choroid Plexus/immunology , Inflammation/immunology , Inflammation/pathology , Leukocytes/immunology , Neurodegenerative Diseases/physiopathology , Animals , Humans
19.
Front Immunol ; 4: 379, 2013.
Article in English | MEDLINE | ID: mdl-24312094

ABSTRACT

Reduction in T cell receptor (TCR) diversity in old age is considered as a major cause for immune complications in the elderly population. Here, we explored the consequences of aging on the TCR repertoire in mice using high-throughput sequencing (TCR-seq). We mapped the TCRß repertoire of CD4+ T cells isolated from bone marrow (BM) and spleen of young and old mice. We found that TCRß diversity is reduced in spleens of aged mice but not in their BM. Splenic CD4+ T cells were also skewed toward an effector memory phenotype in old mice, while BM cells preserved their memory phenotype with age. Analysis of Vß and Jß gene usage across samples, as well as comparison of CDR3 length distributions, showed no significant age dependent changes. However, comparison of the frequencies of amino-acid (AA) TCRß sequences between samples revealed repertoire changes that occurred at a more refined scale. The BM-derived TCRß repertoire was found to be similar among individual mice regardless of their age. In contrast, the splenic repertoire of old mice was not similar to those of young mice, but showed an increased similarity with the BM repertoire. Each old-mouse had a private set of expanded TCRß sequences. Interestingly, a fraction of these sequences was found also in the BM of the same individual, sharing the same nucleotide sequence. Together, these findings show that the composition and phenotype of the CD4+ T cell BM repertoire are relatively stable with age, while diversity of the splenic repertoire is severely reduced. This reduction is caused by idiosyncratic expansions of tens to hundreds of T cell clonotypes, which dominate the repertoire of each individual. We suggest that these private and abundant clonotypes are generated by sporadic clonal expansions, some of which correspond to pre-existing BM clonotypes. These organ- and age-specific changes of the TCRß repertoire have implications for understanding and manipulating age-associated immune decline.

20.
Brain ; 136(Pt 11): 3427-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24088808

ABSTRACT

Infiltrating T cells and monocyte-derived macrophages support central nervous system repair. Although infiltration of leucocytes to the injured central nervous system has recently been shown to be orchestrated by the brain's choroid plexus, the immunological mechanism that maintains this barrier and regulates its activity as a selective gate is poorly understood. Here, we hypothesized that CD4(+) effector memory T cells, recently shown to reside at the choroid plexus stroma, regulate leucocyte trafficking through this portal through their interactions with the choroid plexus epithelium. We found that the naïve choroid plexus is populated by T helper 1, T helper 2 and regulatory T cells, but not by encephalitogenic T cells. In vitro findings revealed that the expression of immune cell trafficking determinants by the choroid plexus epithelium is specifically induced by interferon-γ. Tumour necrosis factor-α and interferon-γ reciprocally controlled the expression of their receptors by the choroid plexus epithelium, and had a synergistic effect in inducing the epithelial expression of trafficking molecules. In vivo, interferon-γ-dependent signalling controlled trafficking through the choroid plexus; interferon-γ receptor knockout mice exhibited reduced levels of T cells and monocyte entry to the cerebrospinal fluid and impaired recovery following spinal cord injury. Moreover, reduced expression of trafficking molecules by the choroid plexus was correlated with reduced CD4(+) T cells in the choroid plexus and cerebrospinal fluid of interferon-γ receptor knockout mice. Similar effect on the expression of trafficking molecules by the choroid plexus was found in bone-marrow chimeric mice lacking interferon-γ receptor in the central nervous system, or reciprocally, lacking interferon-γ in the circulation. Collectively, our findings attribute a novel immunological plasticity to the choroid plexus epithelium, allowing it to serve, through interferon-γ signalling, as a tightly regulated entry gate into the central nervous system for circulating leucocytes immune surveillance under physiological conditions, and for repair following acute injury.


Subject(s)
Central Nervous System/immunology , Choroid Plexus/immunology , Choroid Plexus/pathology , Interferon-gamma/physiology , Animals , Cell Movement/genetics , Cell Movement/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Choroid Plexus/metabolism , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/genetics , Protein Transport/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL