Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1243946, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37795104

RESUMEN

Introduction: The development of new autoantigen discovery techniques, like programmable phage immunoprecipitation sequencing (PhIP-Seq), has accelerated the discovery of neural-specific autoantibodies. Herein, we report the identification of a novel biomarker for paraneoplastic neurologic syndrome (PNS), Sloan-Kettering-Virus-Family-Transcriptional-Corepressor-2 (SKOR2)-IgG, utilizing PhIP-Seq. We have also performed a thorough clinical validation using normal, healthy, and disease/cancer control samples. Methods: Stored samples with unclassified staining at the junction of the Purkinje cell and the granule cell layers were analyzed by PhIP-Seq for putative autoantigen identification. The autoantigen was confirmed by recombinant antigen-expressing cell-based assay (CBA), Western blotting, and tissue immunofluorescence assay colocalization. Results: PhIP-Seq data revealed SKOR2 as the candidate autoantigen. The target antigen was confirmed by a recombinant SKOR-2-expressing, and cell lysate Western blot. Furthermore, IgG from both patient samples colocalized with a commercial SKOR2-specific IgG on cryosections of the mouse brain. Both SKOR2 IgG-positive patients had central nervous system involvement, one presenting with encephalitis and seizures (Patient 1) and the other with cognitive dysfunction, spastic ataxia, dysarthria, dysphagia, and pseudobulbar affect (Patient 2). They had a refractory progressive course and were diagnosed with adenocarcinoma (Patient 1: lung, Patient 2: gallbladder). Sera from adenocarcinoma patients without PNS (n=30) tested for SKOR2-IgG were negative. Discussion: SKOR2 IgG represents a novel biomarker for PNS associated with adenocarcinoma. Identification of additional SKOR2 IgG-positive cases will help categorize the associated neurological phenotype and the risk of underlying malignancy.


Asunto(s)
Adenocarcinoma , Síndromes Paraneoplásicos del Sistema Nervioso , Ratones , Animales , Humanos , Biomarcadores , Autoantígenos , Inmunoglobulina G
2.
Sci Rep ; 12(1): 3049, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197552

RESUMEN

Astrocytes utilize both glycolytic and mitochondrial pathways to power cellular processes that are vital to maintaining normal CNS functions. These cells also mount inflammatory and acute phase reactive programs in response to diverse stimuli. While the metabolic functions of astrocytes under homeostatic conditions are well-studied, the role of cellular bioenergetics in astrocyte reactivity is poorly understood. Teriflunomide exerts immunomodulatory effects in diseases such as multiple sclerosis by metabolically reprogramming lymphocytes and myeloid cells. We hypothesized that teriflunomide would constrain astrocytic inflammatory responses. Purified murine astrocytes were grown under serum-free conditions to prevent acquisition of a spontaneous reactive state. Stimulation with TNFα activated NFκB and increased secretion of Lcn2. TNFα stimulation increased basal respiration, maximal respiration, and ATP production in astrocytes, as assessed by oxygen consumption rate. TNFα also increased glycolytic reserve and glycolytic capacity of astrocytes but did not change the basal glycolytic rate, as assessed by measuring the extracellular acidification rate. TNFα specifically increased mitochondrial ATP production and secretion of Lcn2 required ATP generated by oxidative phosphorylation. Inhibition of dihydroorotate dehydrogenase via teriflunomide transiently increased both oxidative phosphorylation and glycolysis in quiescent astrocytes, but only the increased glycolytic ATP production was sustained over time, resulting in a bias away from mitochondrial ATP production even at doses down to 1 µM. Preconditioning with teriflunomide prevented the TNFα-induced skew toward oxidative phosphorylation, reduced mitochondrial ATP production, and reduced astrocytic inflammatory responses, suggesting that this drug may limit neuroinflammation by acting as a metabolomodulator.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Astrocitos/metabolismo , Crotonatos/farmacología , Hidroxibutiratos/farmacología , Inflamación/metabolismo , Nitrilos/farmacología , Toluidinas/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Quimiocinas/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Lipocalina 2/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Neuroinflammation ; 19(1): 22, 2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093106

RESUMEN

BACKGROUND: The pathogenic contribution of neuroinflammation to ictogenesis and epilepsy may provide a therapeutic target for reduction of seizure burden in patients that are currently underserved by traditional anti-seizure medications. The Theiler's murine encephalomyelitis virus (TMEV) model has provided important insights into the role of inflammation in ictogenesis, but questions remain regarding the relative contribution of microglia and inflammatory monocytes in this model. METHODS: Female C57BL/6 mice were inoculated by intracranial injection of 2 × 105, 5 × 104, 1.25 × 104, or 3.125 × 103 plaque-forming units (PFU) of the Daniel's strain of TMEV at 4-6 weeks of age. Infiltration of inflammatory monocytes, microglial activation, and cytokine production were measured at 24 h post-infection (hpi). Viral load, hippocampal injury, cognitive performance, and seizure burden were assessed at several timepoints. RESULTS: The intensity of inflammatory infiltration and the extent of hippocampal injury induced during TMEV encephalitis scaled with the amount of infectious virus in the initial inoculum. Cognitive performance was preserved in mice inoculated with 1.25 × 104 PFU TMEV relative to 2 × 105 PFU TMEV, but peak viral load at 72 hpi was equivalent between the inocula. CCL2 production in the brain was attenuated by 90% and TNFα and IL6 production was absent in mice inoculated with 1.25 × 104 PFU TMEV. Acute infiltration of inflammatory monocytes was attenuated by more than 80% in mice inoculated with 1.25 × 104 PFU TMEV relative to 2 × 105 PFU TMEV but microglial activation was equivalent between groups. Seizure burden was attenuated and the threshold to kainic acid-induced seizures was higher in mice inoculated with 1.25 × 104 PFU TMEV but low-level behavioral seizures persisted and the EEG exhibited reduced but detectable abnormalities. CONCLUSIONS: The size of the inflammatory monocyte response induced by TMEV scales with the amount of infectious virus in the initial inoculum, despite the development of equivalent peak infectious viral load. In contrast, the microglial response does not scale with the inoculum, as microglial hyper-ramification and increased Iba-1 expression were evident in mice inoculated with either 1.25 × 104 or 2 × 105 PFU TMEV. Inoculation conditions that drive inflammatory monocyte infiltration resulted in robust behavioral seizures and EEG abnormalities, but the low inoculum condition, associated with only microglial activation, drove a more subtle seizure and EEG phenotype.


Asunto(s)
Microglía , Theilovirus , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Monocitos/metabolismo , Convulsiones/patología
4.
Ann Neurol ; 85(4): 526-537, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30779222

RESUMEN

OBJECTIVE: We recently reported successful treatment of a child with febrile infection-related epilepsy syndrome (FIRES), a subtype of new onset refractory status epilepticus, with the recombinant interleukin-1 (IL1) receptor antagonist (IL1RA) anakinra. On this basis, we tested whether endogenous IL1RA production or function is deficient in FIRES patients. METHODS: Levels of IL1ß and IL1RA were measured in serum and cerebrospinal fluid (CSF). The inhibitory activity of endogenous IL1RA was assessed using a cell-based reporter assay. IL1RN gene variants were identified by sequencing. Expression levels for the secreted and intracellular isoforms of IL1RA were measured in patient and control cells by real-time polymerase chain reaction. RESULTS: Levels of endogenous IL1RA and IL1ß were elevated in the serum and CSF of patients with FIRES (n = 7) relative to healthy controls (n = 10). Serum from FIRES patients drove IL1R signaling activity and potentiated IL1R signaling in response to exogenous IL1ß in a cell-based reporter assay. Functional assessment of endogenous IL1RA activity in 3 FIRES patients revealed attenuated inhibition of IL1R signaling. Sequencing of IL1RN in our index patient revealed multiple variants. This was accompanied by reduced expression of intracellular but not secreted isoforms of IL1RA in the patient's peripheral blood mononuclear cells. INTERPRETATION: Our findings suggest that FIRES is associated with reduced expression of intracellular IL1RA isoforms and a functional deficiency in IL1RA inhibitory activity. These observations may provide insight into disease pathogenesis for FIRES and other inflammatory seizure disorders and may provide a valuable biomarker for therapeutic decision-making. Ann Neurol 2019;85:526-537.


Asunto(s)
Epilepsia Refractaria/metabolismo , Síndromes Epilépticos/metabolismo , Infecciones/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/sangre , Proteína Antagonista del Receptor de Interleucina 1/líquido cefalorraquídeo , Convulsiones Febriles/metabolismo , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Epilepsia Refractaria/diagnóstico , Epilepsia Refractaria/tratamiento farmacológico , Síndromes Epilépticos/diagnóstico , Síndromes Epilépticos/tratamiento farmacológico , Femenino , Células HEK293 , Humanos , Infecciones/diagnóstico , Infecciones/tratamiento farmacológico , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Masculino , Convulsiones Febriles/diagnóstico , Convulsiones Febriles/tratamiento farmacológico
5.
Ann Clin Transl Neurol ; 5(2): 172-185, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29468178

RESUMEN

Objective: Injury-associated axon-intrinsic signals are thought to underlie pathogenesis and progression in many neuroinflammatory and neurodegenerative diseases, including multiple sclerosis (MS). Retrograde interferon gamma (IFN γ) signals are known to induce expression of major histocompatibility class I (MHC I) genes in murine axons, thereby increasing the susceptibility of these axons to attack by antigen-specific CD8+ T cells. We sought to determine whether the same is true in human neurons. Methods: A novel microisolation chamber design was used to physically isolate and manipulate axons from human skin fibroblast-derived induced pluripotent stem cell (iPSC)-derived neuron-enriched neural aggregates. Fluorescent retrobeads were used to assess the fraction of neurons with projections to the distal chamber. Axons were treated with IFN γ for 72 h and expression of MHC class I and antigen presentation genes were evaluated by RT-PCR and immunofluorescence. Results: Human iPSC-derived neural stem cells maintained as 3D aggregate cultures in the cell body chamber of polymer microisolation chambers extended dense axonal projections into the fluidically isolated distal chamber. Treatment of these axons with IFN γ resulted in upregulation of MHC class I and antigen processing genes in the neuron cell bodies. IFN γ-induced MHC class I molecules were also anterogradely transported into the distal axon. Interpretation: These results provide conclusive evidence that human axons are competent to express MHC class I molecules, suggesting that inflammatory factors enriched in demyelinated lesions may render axons vulnerable to attack by autoreactive CD8+ T cells in patients with MS. Future work will be aimed at identifying pathogenic anti-axonal T cells in these patients.

6.
J Neuroinflammation ; 14(1): 238, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29202854

RESUMEN

BACKGROUND: Viral encephalitis is a dangerous compromise between the need to robustly clear pathogen from the brain and the need to protect neurons from bystander injury. Theiler's murine encephalomyelitis virus (TMEV) infection of C57Bl/6 mice is a model of viral encephalitis in which the compromise results in hippocampal damage and permanent neurological sequelae. We previously identified brain-infiltrating inflammatory monocytes as the primary driver of this hippocampal pathology, but the mechanisms involved in recruiting these cells to the brain were unclear. METHODS: Chemokine expression levels in the hippocampus were assessed by microarray, ELISA, RT-PCR, and immunofluorescence. Monocyte infiltration during acute TMEV infection was measured by flow cytometry. CCL2 levels were manipulated by immunodepletion and by specific removal from neurons in mice generated by crossing a line expressing the Cre recombinase behind the synapsin promoter to animals with floxed CCL2. RESULTS: Inoculation of the brain with TMEV induced hippocampal production of the proinflammatory chemokine CCL2 that peaked at 6 h postinfection, whereas inoculation with UV-inactivated TMEV did not elicit this response. Immunofluorescence revealed that hippocampal neurons expressed high levels of CCL2 at this timepoint. Genetic deletion of CCR2 and systemic immunodepletion of CCL2 abrogated or blunted the infiltration of inflammatory monocytes into the brain during acute infection. Specific genetic deletion of CCL2 from neurons reduced serum and hippocampal CCL2 levels and inhibited inflammatory monocyte infiltration into the brain. CONCLUSIONS: We conclude that intracranial inoculation with infectious TMEV rapidly induces the expression of CCL2 in neurons, and this cellular source is necessary for CCR2-dependent infiltration of inflammatory monocytes into the brain during the most acute stage of encephalitis. These findings highlight a unique role for neuronal production of chemokines in the initiation of leukocytic infiltration into the infected central nervous system.


Asunto(s)
Quimiocina CCL2/biosíntesis , Encefalitis Viral/mortalidad , Hipocampo/patología , Monocitos/inmunología , Neuronas/metabolismo , Animales , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/patología , Quimiotaxis de Leucocito/inmunología , Encefalitis Viral/inmunología , Encefalitis Viral/metabolismo , Encefalitis Viral/patología , Hipocampo/inmunología , Hipocampo/virología , Ratones , Ratones Endogámicos C57BL , Theilovirus
7.
Ann Neurol ; 80(6): 939-945, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27770579

RESUMEN

Febrile infection-related epilepsy syndrome (FIRES) is a devastating epileptic encephalopathy with limited treatment options and an unclear etiology. Anakinra is a recombinant version of the human interleukin-1 receptor antagonist used to treat autoinflammatory disorders. This is the first report of anakinra for treatment of a child with super-refractory status epilepticus secondary to FIRES. Anakinra was well tolerated and effective. Cerebral spinal fluid analysis revealed elevated levels of proinflammatory cytokines before treatment that normalized on anakinra, suggesting a potential pathogenic role for neuroinflammation in FIRES. Further studies are required to assess anakinra efficacy and dosing, and to further delineate disease etiology. Ann Neurol 2016;80:939-945.


Asunto(s)
Encefalitis Infecciosa/complicaciones , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Convulsiones Febriles/complicaciones , Estado Epiléptico/complicaciones , Estado Epiléptico/tratamiento farmacológico , Preescolar , Femenino , Humanos , Encefalitis Infecciosa/líquido cefalorraquídeo , Encefalitis Infecciosa/tratamiento farmacológico , Mediadores de Inflamación/líquido cefalorraquídeo , Proteínas Recombinantes/uso terapéutico , Convulsiones Febriles/líquido cefalorraquídeo , Convulsiones Febriles/tratamiento farmacológico , Estado Epiléptico/líquido cefalorraquídeo , Síndrome
8.
J Cell Sci ; 129(20): 3911-3921, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27591257

RESUMEN

Inflammatory response of blood-brain barrier (BBB) endothelial cells plays an important role in pathogenesis of many central nervous system inflammatory diseases, including multiple sclerosis; however, the molecular mechanism mediating BBB endothelial cell inflammatory response remains unclear. In this study, we first observed that knockdown of neuropilin-1 (NRP1), a co-receptor of several structurally diverse ligands, suppressed interferon-γ (IFNγ)-induced C-X-C motif chemokine 10 expression and activation of STAT1 in brain microvascular endothelial cells in a Rac1-dependent manner. Moreover, endothelial-specific NRP1-knockout mice, VECadherin-Cre-ERT2/NRP1flox/flox mice, showed attenuated disease progression during experimental autoimmune encephalomyelitis, a mouse neuroinflammatory disease model. Detailed analysis utilizing histological staining, quantitative PCR, flow cytometry and magnetic resonance imaging demonstrated that deletion of endothelial NRP1 suppressed neuron demyelination, altered lymphocyte infiltration, preserved BBB function and decreased activation of the STAT1-CXCL10 pathway. Furthermore, increased expression of NRP1 was observed in endothelial cells of acute multiple sclerosis lesions. Our data identify a new molecular mechanism of brain microvascular endothelial inflammatory response through NRP1-IFNγ crosstalk that could be a potential target for intervention of endothelial cell dysfunction in neuroinflammatory diseases.


Asunto(s)
Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Interferón gamma/farmacología , Microvasos/citología , Neuropilina-1/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Barrera Hematoencefálica/patología , Quimiocina CXCL10 , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Células Endoteliales/efectos de los fármacos , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/patología , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Factor de Transcripción STAT1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo
9.
Sci Rep ; 6: 28699, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27345730

RESUMEN

Neurologic complications associated with viral encephalitis, including seizures and cognitive impairment, are a global health issue, especially in children. We previously showed that hippocampal injury during acute picornavirus infection in mice is associated with calpain activation and is the result of neuronal death triggered by brain-infiltrating inflammatory monocytes. We therefore hypothesized that treatment with a calpain inhibitor would protect neurons from immune-mediated bystander injury. C57BL/6J mice infected with the Daniel's strain of Theiler's murine encephalomyelitis virus were treated with the FDA-approved drug ritonavir using a dosing regimen that resulted in plasma concentrations within the therapeutic range for calpain inhibition. Ritonavir treatment significantly reduced calpain activity in the hippocampus, protected hippocampal neurons from death, preserved cognitive performance, and suppressed seizure escalation, even when therapy was initiated 36 hours after disease onset. Calpain inhibition by ritonavir may be a powerful tool for preserving neurons and cognitive function and preventing neural circuit dysregulation in humans with neuroinflammatory disorders.


Asunto(s)
Calpaína/antagonistas & inhibidores , Infecciones por Cardiovirus/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/farmacología , Fármacos Neuroprotectores/farmacología , Ritonavir/farmacología , Theilovirus/metabolismo , Enfermedad Aguda , Animales , Calpaína/metabolismo , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/patología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/virología , Ratones
10.
Glia ; 62(5): 692-708, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24492996

RESUMEN

Neuromyelitis optica (NMO) is a primary astrocyte disease associated with central nervous system inflammation, demyelination, and tissue injury. Brain lesions are frequently observed in regions enriched in expression of the aquaporin-4 (AQP4) water channel, an antigenic target of the NMO IgG serologic marker. Based on observations of disease reversibility and careful characterization of NMO lesion development, we propose that the NMO IgG may induce a dynamic immunological response in astrocytes. Using primary rat astrocyte-enriched cultures and treatment with NMO patient-derived serum or purified IgG, we observed a robust pattern of gene expression changes consistent with the induction of a reactive and inflammatory phenotype in astrocytes. The reactive astrocyte factor lipocalin-2 and a broad spectrum of chemokines, cytokines, and stress response factors were induced by either NMO patient serum or purified IgG. Treatment with IgG from healthy controls had no effect. The effect is disease-specific, as serum from patients with relapsing-remitting multiple sclerosis, Sjögren's, or systemic lupus erythematosus did not induce a response in the cultures. We hypothesize that binding of the NMO IgG to AQP4 induces a cellular response that results in transcriptional and translational events within the astrocyte that are consistent with a reactive and inflammatory phenotype. Strategies aimed at reducing the inflammatory response of astrocytes may short circuit an amplification loop associated with NMO lesion development.


Asunto(s)
Astrocitos/inmunología , Inmunidad Celular/inmunología , Inmunoglobulina G/inmunología , Neuromielitis Óptica/sangre , Neuromielitis Óptica/inmunología , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunoglobulina G/farmacología , Ratas , Ratas Endogámicas Lew
11.
Sci Rep ; 2: 545, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848791

RESUMEN

Neuronal injury during acute viral infection of the brain is associated with the development of persistent cognitive deficits and seizures in humans. In C57BL/6 mice acutely infected with the Theiler's murine encephalomyelitis virus, hippocampal CA1 neurons are injured by a rapid innate immune response, resulting in profound memory deficits. In contrast, infected SJL and B6xSJL F1 hybrid mice exhibit essentially complete hippocampal and memory preservation. Analysis of brain-infiltrating leukocytes revealed that SJL mice mount a sharply attenuated inflammatory monocyte response as compared to B6 mice. Bone marrow transplantation experiments isolated the attenuation to the SJL immune system. Adoptive transfer of B6 inflammatory monocytes into acutely infected B6xSJL hosts converted these mice to a hippocampal damage phenotype and induced a cognitive deficit marked by failure to recognize a novel object. These findings show that inflammatory monocytes are the critical cellular mediator of hippocampal injury during acute picornavirus infection of the brain.


Asunto(s)
Hipocampo/inmunología , Hipocampo/virología , Monocitos/inmunología , Poliomielitis/inmunología , Poliomielitis/virología , Theilovirus/fisiología , Traslado Adoptivo , Animales , Apoptosis , Trasplante de Médula Ósea , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Inmunofenotipificación , Masculino , Ratones , Monocitos/citología , Monocitos/metabolismo , Neuronas/patología , Poliomielitis/patología , Tropismo Viral , Replicación Viral
12.
J Neuroinflammation ; 9: 50, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22405261

RESUMEN

BACKGROUND: Neuropathology caused by acute viral infection of the brain is associated with the development of persistent neurological deficits. Identification of the immune effectors responsible for injuring the brain during acute infection is necessary for the development of therapeutic strategies that reduce neuropathology but maintain immune control of the virus. METHODS: The identity of brain-infiltrating leukocytes was determined using microscopy and flow cytometry at several acute time points following intracranial infection of mice with the Theiler's murine encephalomyelitis virus. Behavioral consequences of immune cell depletion were assessed by Morris water maze. RESULTS: Inflammatory monocytes, defined as CD45hiCD11b++F4/80+Gr1+1A8-, and neutrophils, defined as CD45hiCD11b+++F4/80-Gr1+1A8+, were found in the brain at 12 h after infection. Flow cytometry of brain-infiltrating leukocytes collected from LysM: GFP reporter mice confirmed the identification of neutrophils and inflammatory monocytes in the brain. Microscopy of sections from infected LysM:GFP mice showed that infiltrating cells were concentrated in the hippocampal formation. Immunostaining confirmed that neutrophils and inflammatory monocytes were localized to the hippocampal formation at 12 h after infection. Immunodepletion of inflammatory monocytes and neutrophils but not of neutrophils only resulted in preservation of hippocampal neurons. Immunodepletion of inflammatory monocytes also preserved cognitive function as assessed by the Morris water maze. CONCLUSIONS: Neutrophils and inflammatory monocytes rapidly and robustly responded to Theiler's virus infection by infiltrating the brain. Inflammatory monocytes preceded neutrophils, but both cell types were present in the hippocampal formation at a timepoint that is consistent with a role in triggering hippocampal pathology. Depletion of inflammatory monocytes and neutrophils with the Gr1 antibody resulted in hippocampal neuroprotection and preservation of cognitive function. Specific depletion of neutrophils with the 1A8 antibody failed to preserve neurons, suggesting that inflammatory monocytes are the key effectors of brain injury during acute picornavirus infection of the brain. These effector cells may be important therapeutic targets for immunomodulatory or immunosuppressive therapies aimed at reducing or preventing central nervous system pathology associated with acute viral infection.


Asunto(s)
Infecciones por Cardiovirus/patología , Hipocampo/patología , Monocitos/patología , Infecciones por Picornaviridae/patología , Theilovirus , Enfermedad Aguda , Animales , Infecciones por Cardiovirus/inmunología , Femenino , Hipocampo/inmunología , Hipocampo/virología , Inflamación/inmunología , Inflamación/patología , Inflamación/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/virología , Infiltración Neutrófila/inmunología , Infecciones por Picornaviridae/inmunología , Theilovirus/inmunología
13.
J Vis Exp ; (52)2011 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-21694694

RESUMEN

We describe a method for preparing brain infiltrating leukocytes (BILs) from mice. We demonstrate how to infect mice with Theiler's murine encephalomyelitis virus (TMEV) via a rapid intracranial injection technique and how to purify a leukocyte-enriched population of infiltrating cells from whole brain. Briefly, mice are anesthetized with isoflurane in a closed chamber and are free-hand injected with a Hamilton syringe into the frontal cortex. Mice are then killed at various times after infection by isoflurane overdose and whole brains are extracted and homogenized in RPMI with a Tenbroeck tissue grinder. Brain homogenates are centrifuged through a continuous 30% Percoll gradient to remove the myelin and other cell debris. The cell suspension is then strained at 40 µm, washed and centrifuged on a discontinuous Ficoll-Paque Plus gradient to select and purify the leukocytes. The leukocytes are then washed and resuspended in appropriate buffers for immunophenotyping by flow cytometry. Flow cytometry reveals a population of innate immune cells at the early stages of infection in C57BL/6 mice. At 24 hours post infection, multiple subsets of immune cells are present in the BILs, with an enriched population of Gr1(+), CD11b(+) and F4/80(+)cells. Therefore, this method is useful in characterizing the immune response to acute infection in the brain.


Asunto(s)
Encéfalo/citología , Técnicas Citológicas/métodos , Leucocitos/citología , Animales , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/virología , Citometría de Flujo/métodos , Lóbulo Frontal/citología , Lóbulo Frontal/inmunología , Lóbulo Frontal/patología , Lóbulo Frontal/virología , Inmunofenotipificación/métodos , Leucocitos/inmunología , Leucocitos/patología , Ratones , Ratones Endogámicos C57BL , Theilovirus
14.
PLoS One ; 5(8): e12478, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20814579

RESUMEN

BACKGROUND: The objective of this study was to test the hypothesis that CD8+ T cells directly mediate motor disability and axon injury in the demyelinated central nervous system. We have previously observed that genetic deletion of the CD8+ T cell effector molecule perforin leads to preservation of motor function and preservation of spinal axons in chronically demyelinated mice. METHODOLOGY/PRINCIPAL FINDINGS: To determine if CD8+ T cells are necessary and sufficient to directly injure demyelinated axons, we adoptively transferred purified perforin-competent CD8+ spinal cord-infiltrating T cells into profoundly demyelinated but functionally preserved perforin-deficient host mice. Transfer of CD8+ spinal cord-infiltrating T cells rapidly and irreversibly impaired motor function, disrupted spinal cord motor conduction, and reduced the number of medium- and large-caliber spinal axons. Likewise, immunodepletion of CD8+ T cells from chronically demyelinated wildtype mice preserved motor function and limited axon loss without altering other disease parameters. CONCLUSIONS/SIGNIFICANCE: In multiple sclerosis patients, CD8+ T cells outnumber CD4+ T cells in active lesions and the number of CD8+ T cells correlates with the extent of ongoing axon injury and functional disability. Our findings suggest that CD8+ T cells may directly injure demyelinated axons and are therefore a viable therapeutic target to protect axons and motor function in patients with multiple sclerosis.


Asunto(s)
Axones/inmunología , Axones/patología , Linfocitos T CD8-positivos/inmunología , Actividad Motora/inmunología , Actividad Motora/fisiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Animales , Linfocitos T CD8-positivos/metabolismo , Enfermedades Desmielinizantes/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad/metabolismo , Leucocitos/inmunología , Masculino , Ratones , Corteza Motora/patología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Perforina/deficiencia , Perforina/metabolismo , Médula Espinal/patología
15.
J Neuropathol Exp Neurol ; 68(9): 1037-48, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19680139

RESUMEN

Axon injury is a major determinant of the loss of neurological function in patients with multiple sclerosis. It is unclear, however, whether damage to axons is an obligatory consequence of demyelination or whether it is an independent process that occurs in the permissive environment of demyelinated lesions. Previous investigations into the role of CD8 T cells and perforin in the Theiler murine encephalomyelitis virus model of multiple sclerosis have used mouse strains resistant to Theiler murine encephalomyelitis virus infection. To test the role of CD8 T cells in axon injury, we established a perforin-deficient mouse model on the H-2 major histocompatibility complex background thereby removing confounding factors related to viral biology in this Theiler murine encephalomyelitis virus-susceptible strain. This permitted direct comparison of clinical and pathological parameters between perforin-competent and perforin-deficient mice. The extent of demyelination was indistinguishable between perforin-competent and perforin-deficient H-2 mice, but chronically infected perforin-deficient mice exhibited preservation of motor function and spinal axons despite the presence of spinal cord demyelination. Thus, demyelination is necessary but insufficient for axon injury in this model; the absence of perforin protects axons without impacting demyelination. These results suggest that perforin is a key mediator of axon injury and lend additional support to the hypothesis that CD8 T cells are primarily responsible for axon damage in multiple sclerosis.


Asunto(s)
Axones/patología , Linfocitos T CD8-positivos/inmunología , Enfermedades Desmielinizantes/genética , Esclerosis Múltiple/genética , Esclerosis Múltiple/fisiopatología , Perforina/genética , Animales , Axones/inmunología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/fisiopatología , Linfocitos T CD8-positivos/patología , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/inmunología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Antígenos H-2/genética , Ratones , Ratones Noqueados , Esclerosis Múltiple/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/inmunología , Médula Espinal/patología , Médula Espinal/fisiopatología , Theilovirus/genética , Theilovirus/inmunología
16.
Am J Pathol ; 175(2): 668-84, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19608874

RESUMEN

Many viruses, including picornaviruses, have the potential to infect the central nervous system (CNS) and stimulate a neuroinflammatory immune response, especially in infants and young children. Cognitive deficits associated with CNS picornavirus infection result from injury and death of neurons that may occur due to direct viral infection or during the immune responses to virus in the brain. Previous studies have concluded that apoptosis of hippocampal neurons during picornavirus infection is a cell-autonomous event triggered by direct neuronal infection. However, these studies assessed neuron death at time points late in infection and during infections that lead to either death of the host or persistent viral infection. In contrast, many neurovirulent picornavirus infections are acute and transient, with rapid clearance of virus from the host. We provide evidence of hippocampal pathology in mice acutely infected with the Theiler's murine encephalomyelitis picornavirus. We found that CA1 pyramidal neurons exhibited several hallmarks of apoptotic death, including caspase-3 activation, DNA fragmentation, and chromatin condensation within 72 hours of infection. Critically, we also found that many of the CA1 pyramidal neurons undergoing apoptosis were not infected with virus, indicating that neuronal cell death during acute picornavirus infection of the CNS occurs in a non-cell-autonomous manner. These observations suggest that therapeutic strategies other than antiviral interventions may be useful for neuroprotection during acute CNS picornavirus infection.


Asunto(s)
Apoptosis , Hipocampo/patología , Infecciones por Picornaviridae/patología , Células Piramidales/patología , Theilovirus , Animales , Modelos Animales de Enfermedad , Hipocampo/virología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Células Piramidales/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA