Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 1067-1078, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38676306

RÉSUMÉ

Regulatory T cells (Tregs) are essential for maintaining immune homeostasis by serving as negative regulators of adaptive immune system effector cell responses. Reduced production or function of Tregs has been implicated in several human autoimmune diseases. The cytokine interleukin 2 plays a central role in promoting Treg differentiation, survival, and function in vivo and may therefore have therapeutic benefits for autoimmune diseases. mRNA-6231 is an investigational, lipid nanoparticle-encapsulated, mRNA-based therapy that encodes a modified human interleukin 2 mutein fused to human serum albumin (HSA-IL2m). Herein, we report the development of a semi-mechanistic kinetic-pharmacodynamic model to quantify the relationship between subcutaneous dose(s) of mRNA-6231, HSA-IL2m protein expression, and Treg expansion in nonhuman primates. The nonclinical kinetic-pharmacodynamic model was extrapolated to humans using allometric scaling principles and the physiological basis of pharmacological mechanisms to predict the clinical response to therapy a priori. Model-based simulations were used to inform the dose selection and design of the first-in-human clinical study (NCT04916431). The modeling approach used to predict human responses was validated when data became available from the phase I clinical study. This validation indicates that the approach is valuable in informing clinical decision-making.


Sujet(s)
Interleukine-2 , ARN messager , Humains , Interleukine-2/pharmacocinétique , Interleukine-2/génétique , Interleukine-2/pharmacologie , Interleukine-2/administration et posologie , Animaux , ARN messager/génétique , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Nanoparticules , Modèles biologiques , Mâle , Liposomes
2.
Nat Commun ; 13(1): 3866, 2022 07 05.
Article de Anglais | MEDLINE | ID: mdl-35790728

RÉSUMÉ

Interleukin-2 (IL-2) is critical for regulatory T cell (Treg) function and homeostasis. At low doses, IL-2 can suppress immune pathologies by expanding Tregs that constitutively express the high affinity IL-2Rα subunit. However, even low dose IL-2, signaling through the IL2-Rß/γ complex, may lead to the activation of proinflammatory, non-Treg T cells, so improving specificity toward Tregs may be desirable. Here we use messenger RNAs (mRNA) to encode a half-life-extended human IL-2 mutein (HSA-IL2m) with mutations promoting reliance on IL-2Rα. Our data show that IL-2 mutein subcutaneous delivery as lipid-encapsulated mRNA nanoparticles selectively activates and expands Tregs in mice and non-human primates, and also reduces disease severity in mouse models of acute graft versus host disease and experimental autoimmune encephalomyelitis. Single cell RNA-sequencing of mouse splenic CD4+ T cells identifies multiple Treg states with distinct response dynamics following IL-2 mutein treatment. Our results thus demonstrate the potential of mRNA-encoded HSA-IL2m immunotherapy to treat autoimmune diseases.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Interleukine-2 , Animaux , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/thérapie , Interleukine-2/génétique , Sous-unité alpha du récepteur à l'interleukine-2 , Lipides , Souris , ARN messager/génétique , Lymphocytes T régulateurs
3.
Cell Rep ; 36(12): 109727, 2021 09 21.
Article de Anglais | MEDLINE | ID: mdl-34551293

RÉSUMÉ

In traumatic brain injury (TBI), a diversity of brain resident and peripherally derived myeloid cells have the potential to worsen damage and/or to assist in healing. We define the heterogeneity of microglia and macrophage phenotypes during TBI in wild-type (WT) mice and Ccr2-/- mice, which lack macrophage influx following TBI and are resistant to brain damage. We use unbiased single-cell RNA sequencing methods to uncover 25 microglia, monocyte/macrophage, and dendritic cell subsets in acute TBI and normal brains. We find alterations in transcriptional profiles of microglia subsets in Ccr2-/- TBI mice compared to WT TBI mice indicating that infiltrating monocytes/macrophages influence microglia activation to promote a type I IFN response. Preclinical pharmacological blockade of hCCR2 after injury reduces expression of IFN-responsive gene, Irf7, and improves outcomes. These data extend our understanding of myeloid cell diversity and crosstalk in brain trauma and identify therapeutic targets in myeloid subsets.


Sujet(s)
Lésions traumatiques de l'encéphale/anatomopathologie , Microglie/métabolisme , Récepteurs CCR2/génétique , Animaux , Antigènes Ly/génétique , Antigènes Ly/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Lésions traumatiques de l'encéphale/métabolisme , Chimiokine CXCL10/génétique , Chimiokine CXCL10/métabolisme , Modèles animaux de maladie humaine , Régulation négative , Humains , Facteur-7 de régulation d'interféron/génétique , Facteur-7 de régulation d'interféron/métabolisme , Interféron de type I/métabolisme , Macrophages/cytologie , Macrophages/métabolisme , Mâle , Apprentissage du labyrinthe , Souris , Souris de lignée C57BL , Souris knockout , Microglie/cytologie , Monocytes/cytologie , Monocytes/métabolisme , Récepteurs CCR2/antagonistes et inhibiteurs , Récepteurs CCR2/déficit , Récepteurs CCR2/métabolisme
4.
Biomaterials ; 272: 120786, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33839625

RÉSUMÉ

Restoring numbers and function of regulatory T cells (Tregs) is a novel therapeutic strategy for neurodegenerative disorders. Whether Treg function is boosted by adoptive cell transfer, pharmaceuticals, or immune modulators, the final result is a robust anti-inflammatory and neuronal sparing response. Herein, a newly developed lipid nanoparticle (LNP) containing mRNA encoding granulocyte-macrophage colony-stimulating factor (Gm-csf mRNA) was developed to peripherally induce Tregs and used for treatment in preclinical Parkinson's disease (PD) models. Administration of Gm-csf mRNA to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice and rats overexpressing alpha-synuclein produced dose-dependent increases in plasma GM-CSF levels and peripheral CD4+CD25+FoxP3+ Treg populations. This upregulation paralleled nigrostriatal neuroprotection, upregulated immunosuppression-associated mRNAs that led to the detection of a treatment-induced CD4+ T cell population, and decreased reactive microgliosis. The current findings strengthen prior works utilizing immune modulation by harnessing Gm-csf mRNA to augment adaptive immune function by employing a new delivery platform to treat PD and potentially other neurodegenerative disorders.


Sujet(s)
Facteur de stimulation des colonies de granulocytes et de macrophages , Maladie de Parkinson , Animaux , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Souris , Souris de lignée C57BL , Neuroprotection , Maladie de Parkinson/génétique , Maladie de Parkinson/thérapie , ARN messager/génétique , Rats
5.
Article de Anglais | MEDLINE | ID: mdl-33468560

RÉSUMÉ

OBJECTIVE: To test the hypothesis that dimethyl fumarate (DMF, Tecfidera) elicits different biological changes from DMF combined with monoethyl fumarate (MEF) (Fumaderm, a psoriasis therapy), we investigated DMF and MEF in rodents and cynomolgus monkeys. Possible translatability of findings was explored with lymphocyte counts from a retrospective cohort of patients with MS. METHODS: In rodents, we evaluated pharmacokinetic and pharmacodynamic effects induced by DMF and MEF monotherapies or in combination (DMF/MEF). Clinical implications were investigated in a retrospective, observational analysis of patients with MS treated with DMF/MEF (n = 36). RESULTS: In rodents and cynomolgus monkeys, monomethyl fumarate (MMF, the primary metabolite of DMF) exhibited higher brain penetration, whereas MEF was preferentially partitioned into the kidney. In mice, transcriptional profiling for DMF and MEF alone identified both common and distinct pharmacodynamic responses, with almost no overlap between DMF- and MEF-induced differentially expressed gene profiles in immune tissues. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated oxidative stress response pathway was exclusively regulated by DMF, whereas apoptosis pathways were activated by MEF. DMF/MEF treatment demonstrated that DMF and MEF functionally interact to modify DMF- and MEF-specific responses in unpredictable ways. In patients with MS, DMF/MEF treatment led to early and pronounced suppression of lymphocytes, predominantly CD8+ T cells. In a multivariate regression analysis, the absolute lymphocyte count (ALC) was associated with age at therapy start, baseline ALC, and DMF/MEF dosage but not with previous immunosuppressive medication and sex. Furthermore, the ALC increased in a small cohort of patients with MS (n = 6/7) after switching from DMF/MEF to DMF monotherapy. CONCLUSIONS: Fumaric acid esters exhibit different biodistribution and may elicit different biological responses; furthermore, pharmacodynamic effects of combinations differ unpredictably from monotherapy. The strong potential to induce lymphopenia in patients with MS may be a result of activation of apoptosis pathways by MEF compared with DMF.


Sujet(s)
Fumarate de diméthyle/composition chimique , Fumarate de diméthyle/pharmacologie , Fumarates/composition chimique , Fumarates/pharmacologie , Sclérose en plaques/traitement médicamenteux , Animaux , Études transversales , Fumarate de diméthyle/usage thérapeutique , Femelle , Fumarates/usage thérapeutique , Analyse de profil d'expression de gènes/méthodes , Humains , Immunosuppresseurs/composition chimique , Immunosuppresseurs/pharmacologie , Immunosuppresseurs/usage thérapeutique , Macaca fascicularis , Mâle , Souris , Souris de lignée C57BL , Sclérose en plaques/sang , Sclérose en plaques/génétique , Rats , Rat Sprague-Dawley , Études rétrospectives
6.
Mol Cell Neurosci ; 99: 103393, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31356882

RÉSUMÉ

Enhancing remyelination is a key therapeutic strategy for demyelinating diseases such as multiple sclerosis. To achieve this goal, a central challenge is being able to quantitatively and longitudinally track functional remyelination, especially with translatable biomarkers that can be performed in both preclinical models and in the clinic. We developed the methodology to stably measure multi-modal sensory evoked potentials from the skull surface over the course of months in individual mice and applied it to a genetic mouse model of oligodendrocyte ablation and demyelination. We found that auditory and somatosensory evoked potential latencies reliably increased over time during the early phase of the model and recovered spontaneously and almost completely during a later phase. Histological examination supported the interpretation that the evoked potential latency changes dynamically reflect changes in CNS myelination. Specifically, we found reduction of myelination in corresponding brain regions at the time that sensory evoked potentials were maximally impacted. Importantly, we also found that myelination levels recovered when evoked potential latencies recovered. Other changes known to associate with demyelination were also observed at the time of delayed evoked potentials, including the emergence of white matter vacuoles and increased markers for activated microglia and macrophages; these changes also fully reversed by the time that evoked potentials recovered. Our results support the hypothesis that skull-surface recorded evoked potential latencies can dynamically track CNS myelination changes. The methods developed here allow for longitudinally tracking functional myelination changes in vivo in preclinical rodent models with a quantitative biomarker that can also be applied clinically and will facilitate translational development of CNS remyelinating therapies.


Sujet(s)
Encéphalomyélite auto-immune expérimentale/physiopathologie , Potentiels évoqués auditifs , Potentiels évoqués somatosensoriels , Animaux , Électroencéphalographie/méthodes , Mâle , Souris , Souris de lignée C57BL , Gaine de myéline/métabolisme , Substance blanche/métabolisme , Substance blanche/anatomopathologie
7.
J Histochem Cytochem ; 67(3): 203-219, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30501365

RÉSUMÉ

Myelin is composed primarily of lipids and diseases affecting myelin are associated with alterations in its lipid composition. However, correlation of the spatial (in situ) distribution of lipids with the disease-associated compositional and morphological changes is not well defined. Herein we applied high resolution matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS), immunohistochemistry (IHC), and liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS) to evaluate brain lipid alterations in the dysmyelinating shiverer (Shi) mouse and cuprizone (Cz) mouse model of reversible demyelination. MALDI-IMS revealed a decrease in the spatial distribution of sulfatide (SHexCer) species, SHexCer (d42:2), and a phosphatidylcholine (PC) species, PC (36:1), in white matter regions like corpus callosum (CC) both in the Shi mouse and Cz mouse model. Changes in these lipid species were restored albeit not entirely upon spontaneous remyelination after demyelination in the Cz mouse model. Lipid distribution changes correlated with the local morphological changes as confirmed by IHC. LC-ESI-MS analyses of CC extracts confirmed the MALDI-IMS derived reductions in SHexCer and PC species. These findings highlight the role of SHexCer and PC in preserving the normal myelin architecture and our experimental approaches provide a morphological basis to define lipid abnormalities relevant to myelin diseases.


Sujet(s)
Céramides/métabolisme , Maladies démyélinisantes/métabolisme , Gaine de myéline/métabolisme , Phosphatidylcholines/métabolisme , Sulfoglycosphingolipides/métabolisme , Animaux , Corps calleux/métabolisme , Corps calleux/ultrastructure , Cuprizone/administration et posologie , Maladies démyélinisantes/induit chimiquement , Maladies démyélinisantes/génétique , Maladies démyélinisantes/anatomopathologie , Modèles animaux de maladie humaine , Immunohistochimie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Mâle , Souris , Souris transgéniques , Gaine de myéline/ultrastructure , Spectrométrie de masse ESI , Spectrométrie de masse MALDI , Substance blanche/métabolisme , Substance blanche/ultrastructure
8.
Proc Natl Acad Sci U S A ; 113(17): 4777-82, 2016 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-27078105

RÉSUMÉ

Dimethyl fumarate (DMF) (BG-12, Tecfidera) is a fumaric acid ester (FAE) that was advanced as a multiple sclerosis (MS) therapy largely for potential neuroprotection as it was recognized that FAEs are capable of activating the antioxidative transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway. However, DMF treatment in randomized controlled MS trials was associated with marked reductions in relapse rate and development of active brain MRI lesions, measures considered to reflect CNS inflammation. Here, we investigated the antiinflammatory contribution of Nrf2 in DMF treatment of the MS model, experimental autoimmune encephalomyelitis (EAE). C57BL/6 wild-type (WT) and Nrf2-deficient (Nrf2(-/-)) mice were immunized with myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 (p35-55) for EAE induction and treated with oral DMF or vehicle daily. DMF protected WT and Nrf2(-/-) mice equally well from development of clinical and histologic EAE. The beneficial effect of DMF treatment in Nrf2(-/-) and WT mice was accompanied by reduced frequencies of IFN-γ and IL-17-producing CD4(+) cells and induction of antiinflammatory M2 (type II) monocytes. DMF also modulated B-cell MHC II expression and reduced the incidence of clinical disease in a B-cell-dependent model of spontaneous CNS autoimmunity. Our observations that oral DMF treatment promoted immune modulation and provided equal clinical benefit in acute EAE in Nrf2(-/-) and WT mice, suggest that the antiinflammatory activity of DMF in treatment of MS patients may occur through alternative pathways, independent of Nrf2.


Sujet(s)
Immunité acquise/immunologie , Fumarate de diméthyle/administration et posologie , Immunité innée/immunologie , Immunomodulation/immunologie , Facteur-2 apparenté à NF-E2/immunologie , Rate/immunologie , Immunité acquise/effets des médicaments et des substances chimiques , Administration par voie orale , Animaux , Cellules cultivées , Relation dose-effet des médicaments , Femelle , Immunité innée/effets des médicaments et des substances chimiques , Facteurs immunologiques/administration et posologie , Immunomodulation/effets des médicaments et des substances chimiques , Immunosuppresseurs/administration et posologie , Mâle , Souris , Souris de lignée C57BL , Rate/cytologie , Rate/effets des médicaments et des substances chimiques
9.
Immunology ; 148(3): 276-86, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27012310

RÉSUMÉ

Interleukin-2 (IL-2) is a critical regulator of immune homeostasis through its non-redundant role in regulatory T (Treg) cell biology. There is major interest in therapeutic modulation of the IL-2 pathway to promote immune activation in the context of tumour immunotherapy or to enhance immune suppression in the context of transplantation, autoimmunity and inflammatory diseases. Antibody-mediated targeting of the high-affinity IL-2 receptor α chain (IL-2Rα or CD25) offers a direct mechanism to target IL-2 biology and is being actively explored in the clinic. In mouse models, the rat anti-mouse CD25 clone PC61 has been used extensively to investigate the biology of IL-2 and Treg cells; however, there has been controversy and conflicting data on the exact in vivo mechanistic function of PC61. Engineering antibodies to alter Fc/Fc receptor interactions can significantly alter their in vivo function. In this study, we re-engineered the heavy chain constant region of an anti-CD25 monoclonal antibody to generate variants with highly divergent Fc effector function. Using these anti-CD25 Fc variants in multiple mouse models, we investigated the in vivo impact of CD25 blockade versus depletion of CD25(+) Treg cells on immune homeostasis. We report that immune homeostasis can be maintained during CD25 blockade but aberrant T-cell activation prevails when CD25(+) Treg cells are actively depleted. These results clarify the impact of PC61 on Treg cell biology and reveal an important distinction between CD25 blockade and depletion of CD25(+) Treg cells. These findings should inform therapeutic manipulation of the IL-2 pathway by targeting the high-affinity IL-2R.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Immunoglobuline G/pharmacologie , Immunothérapie , Interleukine-2/métabolisme , Protéines de fusion recombinantes/pharmacologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Animaux , Anticorps monoclonaux/génétique , Anticorps antiviraux/immunologie , Auto-immunité/immunologie , Facteurs de transcription Forkhead/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Immunoglobuline G/génétique , Immunosuppression thérapeutique , Interleukine-2/immunologie , Déplétion lymphocytaire , Souris , Souris de lignée C57BL , Souris knockout , Ingénierie des protéines , Rats , Récepteurs du fragment Fc des IgG/génétique , Protéines de fusion recombinantes/génétique , Lymphocytes T régulateurs/immunologie
10.
Acta Neuropathol ; 130(2): 279-95, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-25920452

RÉSUMÉ

Dimethyl fumarate (DMF), recently approved as an oral immunomodulatory treatment for relapsing-remitting multiple sclerosis (MS), metabolizes to monomethyl fumarate (MMF) which crosses the blood-brain barrier and has demonstrated neuroprotective effects in experimental studies. We postulated that MMF exerts neuroprotective effects through modulation of microglia activation, a critical component of the neuroinflammatory cascade that occurs in neurodegenerative diseases such as MS. To ascertain our hypothesis and define the mechanistic pathways involved in the modulating effect of fumarates, we used real-time PCR and biochemical assays to assess changes in the molecular and functional phenotype of microglia, quantitative Western blotting to monitor activation of postulated pathway components, and ex vivo whole-cell patch clamp recording of excitatory post-synaptic currents in corticostriatal slices from mice with experimental autoimmune encephalomyelitis (EAE), a model for MS, to study synaptic transmission. We show that exposure to MMF switches the molecular and functional phenotype of activated microglia from classically activated, pro-inflammatory type to alternatively activated, neuroprotective one, through activation of the hydroxycarboxylic acid receptor 2 (HCAR2). We validate a downstream pathway mediated through the AMPK-Sirt1 axis resulting in deacetylation, and thereby inhibition, of NF-κB and, consequently, of secretion of pro-inflammatory molecules. We demonstrate through ex vivo monitoring of spontaneous glutamate-mediated excitatory post-synaptic currents of single neurons in corticostriatal slices from EAE mice that the neuroprotective effect of DMF was exerted on neurons at pre-synaptic terminals by modulating glutamate release. By exposing control slices to untreated and MMF-treated activated microglia, we confirm the modulating effect of MMF on microglia function and, thereby, its indirect neuroprotective effect at post-synaptic level. These findings, whereby DMF-induced activation of a new HCAR2-dependent pathway on microglia leads to the modulation of neuroinflammation and restores synaptic alterations occurring in EAE, represent a possible novel mechanism of action for DMF in MS.


Sujet(s)
Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Fumarates/pharmacologie , Microglie/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Récepteurs couplés aux protéines G/métabolisme , Récepteurs nicotiniques/métabolisme , Synapses/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/physiopathologie , Lignée cellulaire , Relation dose-effet des médicaments , Encéphalomyélite auto-immune expérimentale/physiopathologie , Potentiels post-synaptiques excitateurs/effets des médicaments et des substances chimiques , Femelle , Acide glutamique/métabolisme , Souris de lignée C57BL , Microglie/physiologie , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/physiologie , Sirtuine-1/métabolisme , Synapses/physiologie , Techniques de culture de tissus
13.
Bioorg Med Chem Lett ; 23(10): 3070-4, 2013 May 15.
Article de Anglais | MEDLINE | ID: mdl-23562062
14.
J Immunol ; 172(12): 7694-702, 2004 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-15187152

RÉSUMÉ

In the K/BxN mouse model of arthritis, autoantibodies against glucose-6-phosphate isomerase cause joint-specific inflammation and destruction. We have shown using micro-positron emission tomography that these glucose-6-phosphate isomerase-specific autoantibodies rapidly localize to distal joints of mice. In this study we used micro-positron emission tomography to delineate the stages involved in the development of arthritis. Localization of Abs to the joints depended upon mast cells, neutrophils, and FcRs, but not on C5. Surprisingly, anti-type II collagen Abs alone did not accumulate in the distal joints, but could be induced to do so by coinjection of irrelevant preformed immune complexes. Control Abs localized to the joint in a similar manner. Thus, immune complexes are essential initiators of arthritis by sequential activation of neutrophils and mast cells to allow Abs access to the joints, where they must bind a target Ag to initiate inflammation. Our findings support a four-stage model for the development of arthritis and identify checkpoints where the disease is reversible.


Sujet(s)
Complexe antigène-anticorps/physiologie , Arthrite/immunologie , Autoanticorps/physiologie , Animaux , Anticorps/métabolisme , Anticorps/pharmacologie , Arthrite/étiologie , Autoanticorps/métabolisme , Collagène de type II/immunologie , Complément C5/déficit , Complément C5/immunologie , Articulations/immunologie , Articulations/métabolisme , Mastocytes/immunologie , Souris , Souris transgéniques , Granulocytes neutrophiles/immunologie , Transport des protéines/physiologie , Récepteur Fc/déficit , Récepteur Fc/immunologie , Tomoscintigraphie
15.
J Exp Med ; 199(3): 323-35, 2004 Feb 02.
Article de Anglais | MEDLINE | ID: mdl-14744995

RÉSUMÉ

Incomplete deletion of KRN T cells that recognize the ubiquitously expressed self-antigen glucose-6-phosphate-isomerase (GPI) initiates an anti-GPI autoimmune cascade in K/BxN mice resulting in a humorally mediated arthritis. Transgenic (Tg) expression of a KRN T cell receptor (TCR) agonist under the major histocompatibility complex class II promoter resulted in thymic deletion with loss of anti-GPI T and B cell responses and attenuated arthritis course. However, double Tg mice succumbed to systemic autoimmunity with multiorgan inflammation and autoantibody production. Extensive thymic deletion resulted in lymphopenia and elimination of CD4+ CD25+ regulatory T cells (Tregs), but spared some CD4+ T cells expressing endogenous TCR, which oligoclonally expanded in the periphery. Disease was transferred by these T cells and prevented by cotransfer of CD4+ CD25+ Tregs. Moreover, we extended our findings to another TCR system (anti-hen egg lysozyme [HEL] TCR/HEL mice) where similarly extensive thymic deletion also resulted in disease. Thus, our studies demonstrated that central tolerance can paradoxically result in systemic autoimmunity through differential susceptibility of Tregs and autoreactive T cells to thymic deletion. Therefore, too little or too much negative selection to a self-antigen can result in systemic autoimmunity and disease.


Sujet(s)
Lymphocytes T CD4+/immunologie , Déplétion lymphocytaire , Récepteurs à l'interleukine-2/immunologie , Lymphocytes T/immunologie , Animaux , Antigènes CD/immunologie , Séquence nucléotidique , Poulets , Amorces ADN , Test ELISA , Érythrocytes/immunologie , Inflammation/immunologie , Souris , Souris transgéniques , Lysozyme/génétique , Mutagenèse , Réaction de polymérisation en chaîne , Rate/immunologie , Thymectomie
16.
Proc Natl Acad Sci U S A ; 99(22): 14368-73, 2002 Oct 29.
Article de Anglais | MEDLINE | ID: mdl-12391319

RÉSUMÉ

K/BxN mice develop an inflammatory joint disease with many features characteristic of rheumatoid arthritis. In this model, the KRN transgenic T cells and nontransgenic B cells both recognize the glycolytic enzyme glucose-6-phosphate-isomerase (GPI) as an autoantigen. Here, we followed the anti-GPI B cell response that naturally arises in K/BxN mice. The anti-GPI B cell response was robust and arose at the same time as the development of serum anti-GPI autoantibody and joint inflammation. Surprisingly, although GPI was expressed systemically, the anti-GPI B cell response was focused to the lymph nodes (LN) draining the distal joints where arthritis was evident. In lymphotoxin-beta receptor-Ig-treated mice, which lack LNs, the development of arthritis was completely inhibited up to 5-6 weeks. At later times, some arthritis did develop, but at a significantly reduced level. Thus, in this spontaneous model of autoimmunity, the LNs draining the distal joints are essential for both the inhibition and amplification of the arthritogenic B cell response. These findings imply that the immune physiology of a joint is unique, resulting in a local immune response to a systemic autoantigen.


Sujet(s)
Polyarthrite rhumatoïde/immunologie , Autoanticorps/immunologie , Autoantigènes/immunologie , Glucose 6-phosphate isomerase/immunologie , Noeuds lymphatiques/immunologie , Animaux , Lymphocytes B/immunologie , Modèles animaux de maladie humaine , Évolution de la maladie , Souris , Souris de lignée C57BL , Souris de lignée NOD
17.
Nat Immunol ; 3(4): 366-72, 2002 Apr.
Article de Anglais | MEDLINE | ID: mdl-11896393

RÉSUMÉ

In the K/BxN mouse model of rheumatoid arthritis, the transfer of autoantibodies specific for glucose-6-phosphate isomerase (GPI) into naïve mice rapidly induces joint-specific inflammation similar to that seen in human rheumatoid arthritis. The ubiquitous expression of GPI and the systemic circulation of anti-GPI immunoglobulin G (IgG) seem incongruous with the tissue specificity of this disease. By using PET (positron emission tomography), we show here that purified anti-GPI IgG localizes specifically to distal joints in the front and rear limbs within minutes of intravenous injection, reaches saturation by 20 min and remains localized for at least 24 h. In contrast, control IgG does not localize to joints or cause inflammation. The rapid kinetics of anti-GPI IgG joint localization supports a model in which autoantibodies bind directly to pre-existing extracellular GPI in normal healthy mouse joints.


Sujet(s)
Articulation talocrurale/enzymologie , Polyarthrite rhumatoïde/enzymologie , Autoantigènes/métabolisme , Glucose 6-phosphate isomerase/métabolisme , Animaux , Articulation talocrurale/imagerie diagnostique , Articulation talocrurale/immunologie , Autoantigènes/génétique , Autoantigènes/immunologie , Radio-isotopes du cuivre , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Glucose 6-phosphate isomerase/génétique , Glucose 6-phosphate isomerase/immunologie , Humains , Immunoglobuline G/immunologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris transgéniques , Radiographie , Facteurs temps , Tomoscintigraphie/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...