Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 11(19)2022 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-36231129

RESUMO

Facial motoneuron (FMN) survival is mediated by CD4+ T cells in an interleukin-10 (IL-10)-dependent manner after facial nerve axotomy (FNA), but CD4+ T cells themselves are not the source of this neuroprotective IL-10. The aims of this study were to (1) identify the temporal and cell-specific induction of IL-10 expression in the facial motor nucleus and (2) elucidate the neuroprotective capacity of this expression after axotomy. Immunohistochemistry revealed that FMN constitutively produced IL-10, whereas astrocytes were induced to make IL-10 after FNA. Il10 mRNA co-localized with microglia before and after axotomy, but microglial production of IL-10 protein was not detected. To determine whether any single source of IL-10 was critical for FMN survival, Cre/Lox mouse strains were utilized to selectively knock out IL-10 in neurons, astrocytes, and microglia. In agreement with the localization data reflecting concerted IL-10 production by multiple cell types, no single cellular source of IL-10 alone could provide neuroprotection after FNA. These findings suggest that coordinated neuronal and astrocytic IL-10 production is necessary for FMN survival and has roles in neuronal homeostasis, as well as neuroprotective trophism after axotomy.


Assuntos
Traumatismos do Nervo Facial , Núcleo do Nervo Facial , Animais , Camundongos , Axotomia , Traumatismos do Nervo Facial/genética , Traumatismos do Nervo Facial/metabolismo , Núcleo do Nervo Facial/metabolismo , Interleucina-10/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/metabolismo , Neuroproteção , RNA Mensageiro/metabolismo
2.
J Neuroinflammation ; 17(1): 121, 2020 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-32303238

RESUMO

BACKGROUND: After peripheral nerve transection, facial motoneuron (FMN) survival depends on an intact CD4+ T cell population and a central source of interleukin-10 (IL-10). However, it has not been determined previously whether CD4+ T cells participate in the central neuroprotective IL-10 cascade after facial nerve axotomy (FNA). METHODS: Immunohistochemical labeling of CD4+ T cells, pontine vasculature, and central microglia was used to determine whether CD4+ T cells cross the blood-brain barrier and enter the facial motor nucleus (FMNuc) after FNA. The importance of IL-10 signaling in CD4+ T cells was assessed by performing adoptive transfer of IL-10 receptor beta (IL-10RB)-deficient CD4+ T cells into immunodeficient mice prior to injury. Histology and qPCR were utilized to determine the impact of IL-10RB-deficient T cells on FMN survival and central gene expression after FNA. Flow cytometry was used to determine whether IL-10 signaling in T cells was necessary for their differentiation into neuroprotective subsets. RESULTS: CD4+ T cells were capable of crossing the blood-brain barrier and associating with reactive microglial nodules in the axotomized FMNuc. Full induction of central IL-10R gene expression after FNA was dependent on CD4+ T cells, regardless of their own IL-10R signaling capability. Surprisingly, CD4+ T cells lacking IL-10RB were incapable of mediating neuroprotection after axotomy and promoted increased central expression of genes associated with microglial activation, antigen presentation, T cell co-stimulation, and complement deposition. There was reduced differentiation of IL-10RB-deficient CD4+ T cells into regulatory CD4+ T cells in vitro. CONCLUSIONS: These findings support the interdependence of IL-10- and CD4+ T cell-mediated mechanisms of neuroprotection after axotomy. CD4+ T cells may potentiate central responsiveness to IL-10, while IL-10 signaling within CD4+ T cells is necessary for their ability to rescue axotomized motoneuron survival. We propose that loss of IL-10 signaling in CD4+ T cells promotes non-neuroprotective autoimmunity after FNA.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Traumatismos do Nervo Facial/metabolismo , Nervo Facial/metabolismo , Neurônios Motores/metabolismo , Receptores de Interleucina-10/biossíntese , Animais , Axotomia/métodos , Sobrevivência Celular/fisiologia , Células Cultivadas , Traumatismos do Nervo Facial/genética , Feminino , Expressão Gênica , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Interleucina-10/genética
3.
Restor Neurol Neurosci ; 36(3): 417-422, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29614705

RESUMO

BACKGROUND: When nerve transection is performed on adult rodents, a substantial population of neurons survives short-term disconnection from target, and the immune system supports this neuronal survival, however long-term survival remains unknown. Understanding the effects of permanent axotomy on cell body survival is important as target disconnection is the first pathological occurrence in fatal motoneuron diseases such as amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA). OBJECTIVE: The goal of this study was to determine if facial motoneurons (FMN) could survive permanent target disconnection up to 26 weeks post-operation (wpo) after facial nerve axotomy (FNA). In addition, the potentially additive effects of immunodeficiency and motoneuron disease on post-axotomy FMN survival were examined. METHODS: This study included three wild type (WT) mouse strains (C57BL/6J, B6SJL, and FVB/NJ) and three experimental models (RAG-2-/-: immunodeficiency; mSOD1: ALS; Smn-/-/SMN2+/+: SMA). All animals received a unilateral FNA, and FMN survival was quantified at early and extended post-operative timepoints. RESULTS: In the C57BL/6J WT group, FMN survival significantly decreased at 10 wpo (55±6%), and then remained stable out to 26 wpo (47±6%). In the RAG-2-/- and mSOD1 groups, FMN death occurred much earlier at 4 wpo, and survival plateaued at approximately 50% at 10 wpo. The SMA model and other WT strains also exhibited approximately 50% FMN survival after FNA. CONCLUSION: These results indicate that immunodeficiency and motoneuron disease accelerate axotomy-induced neuron death, but do not increase total neuron death in the context of permanent target disconnection. This consistent finding of a target disconnection-resilient motoneuron population is prevalent in other peripheral nerve injury models and in neurodegenerative disease models as well. Characterization of the distinct populations of vulnerable and resilient motoneurons may reveal new therapeutic approaches for injury and disease.


Assuntos
Doenças do Sistema Nervoso Central/patologia , Traumatismos do Nervo Facial/patologia , Nervo Facial/patologia , Neurônios Motores/patologia , Animais , Axotomia/métodos , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Camundongos Endogâmicos C57BL
4.
Int J Mol Sci ; 19(2)2018 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-29473876

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by progressive loss of lower and upper motor neurons (MN) leading to muscle weakness, paralysis and eventually death. Although a highly varied etiology results in ALS, it broadly manifests itself as sporadic and familial forms that have evident similarities in clinical symptoms and disease progression. There is a tremendous amount of knowledge on molecular mechanisms leading to loss of MNs and neuromuscular junctions (NMJ) as major determinants of disease onset, severity and progression in ALS. Specifically, two main opposing hypotheses, the dying forward and dying back phenomena, exist to account for NMJ denervation. The former hypothesis proposes that the earliest degeneration occurs at the central MNs and proceeds to the NMJ, whereas in the latter, the peripheral NMJ is the site of precipitating degeneration progressing backwards to the MN cell body. A large body of literature strongly indicates a role for the immune system in disease onset and progression via regulatory involvement at the level of both the central and peripheral nervous systems (CNS and PNS). In this review, we discuss the earliest reported immune responses with an emphasis on newly identified immune players in mutant superoxide dismutase 1 (mSOD1) transgenic mice, the gold standard mouse model for ALS.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Análise Espaço-Temporal , Animais , Humanos , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/patologia , Células de Schwann/imunologia , Células de Schwann/patologia
5.
Neural Regen Res ; 12(10): 1664-1679, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29171432

RESUMO

Amyotrophic lateral sclerosis is a motoneuron degenerative disease that is challenging to diagnose and presents with considerable variability in survival. Early identification and enhanced understanding of symptomatic patterns could aid in diagnosis and provide an avenue for monitoring disease progression. Use of the mSOD1G93A mouse model provides control of the confounding environmental factors and genetic heterogeneity seen in amyotrophic lateral sclerosis patients, while investigating underlying disease-induced changes. In the present study, we performed a longitudinal behavioral assessment paradigm and identified an early hindlimb symptom, resembling the common gait abnormality foot drop, along with an accompanying forelimb compensatory mechanism in the mSOD1G93A mouse. Following these initial changes, mSOD1 mice displayed a temporary hindlimb compensatory mechanism resembling an exaggerated steppage gait. As the disease progressed, these compensatory mechanisms were not sufficient to sustain fundamental locomotor parameters and more severe deficits appeared. We next applied these initial findings to investigate the inherent variability in B6SJL mSOD1G93A survival. We identified four behavioral variables that, when combined in a cluster analysis, identified two subpopulations with different disease progression rates: a fast progression group and a slow progression group. This behavioral assessment paradigm, with its analytical approaches, provides a method for monitoring disease progression and detecting mSOD1 subgroups with different disease severities. This affords researchers an opportunity to search for genetic modifiers or other factors that likely enhance or slow disease progression. Such factors are possible therapeutic targets with the potential to slow disease progression and provide insight into the underlying pathology and disease mechanisms.

6.
Mediators Inflamm ; 2016: 6131234, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27194826

RESUMO

An increased risk of ALS has been reported for veterans, varsity athletes, and professional football players. The mechanism underlying the increased risk in these populations has not been identified; however, it has been proposed that motor nerve injury may trigger immune responses which, in turn, can accelerate the progression of ALS. Accumulating evidence indicates that abnormal immune reactions and inflammation are involved in the pathogenesis of ALS, but the specific immune cells involved have not been clearly defined. To understand how nerve injury and immune responses may contribute to ALS development, we investigated responses of CD4(+) T cell after facial motor nerve axotomy (FNA) at a presymptomatic stage in a transgenic mouse model of ALS (B6SJL SOD1(G93A)). SOD1(G93A) mice, compared with WT mice, displayed an increase in the basal activation state of CD4(+) T cells and higher frequency of Th17 cells, which were further enhanced by FNA. In conclusion, SOD1(G93A) mice exhibit abnormal CD4(+) T cell activation with increased levels of Th17 cells prior to the onset of neurological symptoms. Motor nerve injury exacerbates Th17 cell responses and may contribute to the development of ALS, especially in those who carry genetic susceptibility to this disease.


Assuntos
Traumatismos do Nervo Facial/metabolismo , Traumatismos do Nervo Facial/patologia , Neurônios Motores/patologia , Superóxido Dismutase-1/metabolismo , Células Th17/metabolismo , Animais , Modelos Animais de Doenças , Traumatismos do Nervo Facial/imunologia , Feminino , Camundongos , Camundongos Transgênicos , Neurônios Motores/imunologia , Neurônios Motores/metabolismo , Superóxido Dismutase-1/genética , T-Linfocitopenia Idiopática CD4-Positiva/metabolismo , Células Th17/imunologia
7.
J Neuroimmune Pharmacol ; 10(4): 587-94, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26148561

RESUMO

We have established a physiologically relevant mechanism of CD4+ T cell-mediated neuroprotection involving axotomized wildtype (WT) mouse facial motoneurons (FMN) with significance in the treatment of amyotrophic lateral sclerosis (ALS), a fatal MN disease. Use of the transgenic mouse model of ALS involving expression of human mutant superoxide dismutase genes (SOD1(G93A); abbreviated here as mSOD1) has accelerated basic ALS research. Superimposition of facial nerve axotomy (FNA) on the mSOD1 mouse during pre-symptomatic stages indicates that they behave like immunodeficient mice in terms of increased FMN loss and decreased functional recovery, through a mechanism that, paradoxically, is not inherent within the MN itself, but, instead, involves a defect in peripheral immune: CNS glial cell interactions. Our goal is to utilize our WT mouse model of immune-mediated neuroprotection after FNA as a template to elucidate how a malfunctioning peripheral immune system contributes to motoneuron cell loss in the mSOD1 mouse. This review will discuss potential immune defects in ALS, as well as provide an up-to-date understanding of how the CD4+ effector T cells provide neuroprotection to motoneurons through regulation of the central microglial and astrocytic response to injury. We will discuss an IL-10 cascade within the facial nucleus that requires a functional CD4+ T cell trigger for activation. The review will discuss the role of T cells in ALS, and our recent reconstitution experiments utilizing our model of T cell-mediated neuroprotection in WT vs mSOD1 mice after FNA. Identification of defects in neural:immune interactions could provide targets for therapeutic intervention in ALS.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Linfócitos T CD4-Positivos/imunologia , Neurônios Motores/imunologia , Regeneração Nervosa/imunologia , Neuroproteção/imunologia , Animais , Humanos
8.
J Comp Neurol ; 523(18): 2752-68, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26010802

RESUMO

Disease progression rates among patients with amyotrophic lateral sclerosis (ALS) vary greatly. Although the majority of affected individuals survive 3-5 years following diagnosis, some subgroups experience a more rapidly progressing form, surviving less than 1 year, and other subgroups experience slowly progressing forms, surviving nearly 50 years. Genetic heterogeneity and environmental factors pose significant barriers in investigating patient progression rates. Similar to the case for humans, variation in survival within the mSOD1 mouse has been well documented, but different progression rates have not been investigated. The present study identifies two subgroups of B6SJL mSOD1(G93A) mice with different disease progression rates, a fast progression group (FPG) and slow progression group, as evidenced by differences in the rate of motor function decline. In addition, increased disease-associated gene expression within the FPG facial motor nucleus confirmed the presence of a more severe phenotype. We hypothesize that a more severe disease phenotype could be the result of 1) an earlier onset of axonal disconnection with a consistent degeneration rate or 2) a more severe or accelerated degenerative process. We performed a facial nerve transection axotomy in both mSOD1 subgroups prior to disease onset as a method to standardize the axonal disconnection. Instead of leading to comparable gene expression in both subgroups, this standardization did not eliminate the severe phenotype in the FPG facial nucleus, suggesting that the FPG phenotype is the result of a more severe or accelerated degenerative process. We theorize that these mSOD1 subgroups are representative of the rapid and slow disease phenotypes often experienced in ALS.


Assuntos
Esclerose Lateral Amiotrófica/diagnóstico , Esclerose Lateral Amiotrófica/genética , Mutação/genética , Superóxido Dismutase/genética , Fatores Etários , Esclerose Lateral Amiotrófica/complicações , Animais , Modelos Animais de Doenças , Progressão da Doença , Nervo Facial/metabolismo , Comportamento Alimentar/fisiologia , Microdissecção e Captura a Laser , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Motores/etiologia , Força Muscular/genética , RNA Mensageiro/metabolismo , Transtornos de Sensação/etiologia , Transfecção
9.
Brain Behav Immun ; 40: 55-60, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24911596

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease involving motoneuron (MN) axonal withdrawal and cell death. Previously, we established that facial MN (FMN) survival levels in the SOD1(G93A) transgenic mouse model of ALS are reduced and nerve regeneration is delayed, similar to immunodeficient RAG2(-/-) mice, after facial nerve axotomy. The objective of this study was to examine the functionality of SOD1(G93A) splenic microenvironment, focusing on CD4(+) T cells, with regard to defects in immune-mediated neuroprotection of injured MN. We utilized the RAG2(-/-) and SOD1(G93A) mouse models, along with the facial nerve axotomy paradigm and a variety of cellular adoptive transfers, to assess immune-mediated neuroprotection of FMN survival levels. We determined that adoptively transferred SOD1(G93A) unfractionated splenocytes into RAG2(-/-) mice were unable to support FMN survival after axotomy, but that adoptive transfer of isolated SOD1(G93A) CD4(+) T cells could. Although WT unfractionated splenocytes adoptively transferred into SOD1(G93A) mice were able to maintain FMN survival levels, WT CD4(+) T cells alone could not. Importantly, these results suggest that SOD1(G93A) CD4(+) T cells retain neuroprotective functionality when removed from a dysfunctional SOD1(G93A) peripheral splenic microenvironment. These results also indicate that the SOD1(G93A) central nervous system microenvironment is able to re-activate CD4(+) T cells for immune-mediated neuroprotection when a permissive peripheral microenvironment exists. We hypothesize that a suppressive SOD1(G93A) peripheral splenic microenvironment may compromise neuroprotective CD4(+) T cell activation and/or differentiation, which, in turn, results in impaired immune-mediated neuroprotection for MN survival after peripheral axotomy in SOD1(G93A) mice.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Linfócitos T CD4-Positivos/imunologia , Núcleo do Nervo Facial/imunologia , Neurônios Motores/imunologia , Superóxido Dismutase/genética , Transferência Adotiva , Esclerose Lateral Amiotrófica/patologia , Animais , Axotomia , Linfócitos T CD4-Positivos/transplante , Proteínas de Ligação a DNA/genética , Traumatismos do Nervo Facial , Núcleo do Nervo Facial/patologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/patologia , Superóxido Dismutase-1
10.
Semin Immunol ; 26(5): 357-68, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24486056
11.
J Comp Neurol ; 522(10): 2349-76, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24424947

RESUMO

The target disconnection theory of amyotrophic lateral sclerosis (ALS) pathogenesis suggests that disease onset is initiated by a peripheral pathological event resulting in neuromuscular junction loss and motoneuron (MN) degeneration. Presymptomatic mSOD1(G93A) mouse facial MN (FMN) are more susceptible to axotomy-induced cell death than wild-type (WT) FMN, which suggests additional CNS pathology. We have previously determined that the mSOD1 molecular response to facial nerve axotomy is phenotypically regenerative and indistinguishable from WT, whereas the surrounding microenvironment shows significant dysregulation in the mSOD1 facial nucleus. To elucidate the mechanisms underlying the enhanced mSOD1 FMN loss after axotomy, we superimposed the facial nerve axotomy model on presymptomatic mSOD1 mice and investigated gene expression for death receptor pathways after target disconnection by axotomy vs. disease progression. We determined that the TNFR1 death receptor pathway is involved in axotomy-induced FMN death in WT and is partially responsible for the mSOD1 FMN death. In contrast, an inherent mSOD1 CNS pathology resulted in a suppressed glial reaction and an upregulation in the Fas death pathway after target disconnection. We propose that the dysregulated mSOD1 glia fail to provide support the injured MN, leading to Fas-induced FMN death. Finally, we demonstrate that, during disease progression, the mSOD1 facial nucleus displays target disconnection-induced gene expression changes that mirror those induced by axotomy. This validates the use of axotomy as an investigative tool in understanding the role of peripheral target disconnection in the pathogenesis of ALS.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Morte Celular/fisiologia , Nervo Facial/fisiopatologia , Neurônios Motores/fisiologia , Degeneração Neural/fisiopatologia , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Axotomia , Progressão da Doença , Nervo Facial/patologia , Núcleo do Nervo Facial/patologia , Núcleo do Nervo Facial/fisiopatologia , Feminino , Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/fisiologia , Neurônios Motores/patologia , Degeneração Neural/patologia , Neuroglia/patologia , Neuroglia/fisiologia , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Receptor fas/metabolismo
12.
J Immunol ; 191(11): 5383-97, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24140643

RESUMO

Soluble CD23 plays a role in the positive regulation of an IgE response. Engagement of the ß2 adrenergic receptor (ß2AR) on a B cell is known to enhance the level of both soluble CD23 and IgE, although the mechanism by which this occurs is not completely understood. In this study, we report that, in comparison with a CD40 ligand/IL-4-primed murine B cell alone, ß2AR engagement on a primed B cell increased gene expression of a disintegrin and metalloproteinase (ADAM)10, which is the primary sheddase of CD23, as well as protein expression of both CD23 and ADAM10, in a protein kinase A- and p38 MAPK-dependent manner, and promoted the localization of these proteins to exosomes as early as 2 d after priming, as determined by both Western blot and flow cytometry and confirmed by electron microscopy. In comparison with isolated exosomes released from primed B cells alone, the transfer of exosomes released from ß2AR agonist-exposed primed B cells to cultures of recipient primed B cells resulted in an increase in the level of IgE produced per cell, without affecting the number of cells producing IgE, as determined by ELISPOT. These effects still occurred when a ß2AR antagonist was added along with the transfer to block residual agonist, and they failed to occur when exosomes were isolated from ß2AR-deficient B cells. These findings suggest that the mechanism responsible for mediating the ß2AR-induced increase in IgE involves a shuttling of the ß2AR-induced increase in CD23 and ADAM10 proteins to exosomes that subsequently mediate an increase in IgE.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Linfócitos B/imunologia , Exossomos/metabolismo , Imunoglobulina E/metabolismo , Proteínas de Membrana/metabolismo , Receptores de IgE/metabolismo , Proteínas ADAM/genética , Proteína ADAM10 , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Linfócitos B/efeitos dos fármacos , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Imunoglobulina E/genética , Imunoglobulina E/imunologia , Ativação Linfocitária/efeitos dos fármacos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Transporte Proteico , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/imunologia , Receptores de IgE/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
J Neurodegener Regen ; 4(1): 21-25, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24672589

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease involving progressive loss of motoneurons (MN). Axonal pathology and presynaptic deaf-ferentation precede MN degeneration during disease progression in patients and the ALS mouse model (mSOD1). Previously, we determined that a functional adaptive immune response is required for complete functional recovery following a facial nerve crush axotomy in wild-type (WT) mice. In this study, we investigated the effects of facial nerve crush axotomy on functional recovery and facial MN survival in presymptomatic mSOD1 mice, relative to WT mice. The results indicate that functional recovery and facial MN survival levels are significantly reduced in presymptomatic mSOD1, relative to WT, and similar to what has previously been observed in immunodeficient mice. It is concluded that a potential immune system defect exists in the mSOD1 mouse that negatively impacts neuronal survival and regeneration following target disconnection associated with peripheral nerve axotomy.

14.
J Immunol ; 190(2): 723-36, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23241883

RESUMO

CD86 engagement on a CD40L/IL-4-primed murine B cell activates signaling intermediates that promote NF-κB activation to increase Oct-2 and mature IgG1 mRNA and protein expression, as well as the rate of IgG1 transcription, without affecting class switch recombination. One of the most proximal signaling intermediates identified is phospholipase Cγ2, a protein reported to bind tyrosine residues, which are absent in the cytoplasmic domain of CD86. Using a proteomics-based identification approach, we show that the tyrosine-containing transmembrane adaptor proteins prohibitin (Phb)1 and Phb2 bind to CD86. The basal expression of Phb1/2 and association with CD86 was low in resting B cells, whereas the level of expression and association increased primarily after priming with CD40. The CD86-induced increase in Oct-2 and IgG1 was less when either Phb1/2 expression was reduced by short hairpin RNA or the cytoplasmic domain of CD86 was truncated or mutated at serine/threonine protein kinase C phosphorylation sites, which did not affect Phb1/2 binding to CD86. Using this approach, we also show that Phb1/2 and the CD86 cytoplasmic domain are required for the CD86-induced phosphorylation of IκBα, which we previously reported leads to NF-κB p50/p65 activation, whereas only Phb1/2 was required for the CD86-induced phosphorylation of phospholipase Cγ2 and protein kinase Cα/ß(II), which we have previously reported leads to NF-κB (p65) phosphorylation and subsequent nuclear translocation. Taken together, these findings suggest that Phb1/2 and the CD86 cytoplasmic domain cooperate to mediate CD86 signaling in a B cell through differential phosphorylation of distal signaling intermediates required to increase IgG1.


Assuntos
Linfócitos B/metabolismo , Antígeno B7-2/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transporte Ativo do Núcleo Celular , Animais , Antígeno B7-2/química , Antígenos CD40/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Feminino , Regulação da Expressão Gênica , Camundongos , NF-kappa B/metabolismo , Fosfolipase C gama/metabolismo , Proibitinas , Ligação Proteica , Proteína Quinase C/metabolismo , Proteínas Repressoras/genética
15.
Brain Behav Immun ; 26(6): 886-90, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22426430

RESUMO

BACKGROUND: The production of neurotrophic factors, such as BDNF, has generally been considered an important mechanism of immune-mediated neuroprotection. However, the ability of T cells to produce BDNF remains controversial. METHODS: In the present study, we examined mRNA and protein of BDNF using RT-PCR and western blot, respectively, in purified and reactivated CD4(+) T cells. In addition, to determine the role of BDNF derived from CD4(+) T cells, the BDNF gene was specifically deleted in T cells using the Cre-lox mouse model system. RESULTS: Our results indicate that while both mRNA expression and protein secretion of BDNF in reactivated T cells were detected at 24 h, only protein could be detected at 72 h after reactivation. The results suggest a transient up-regulation of BDNF mRNA in reactivated T cells. Furthermore, in contrast to our hypothesis that the BDNF expression is necessary for CD4(+) T cells to mediate neuroprotection, mice with CD4(+) T cells lacking BDNF expression demonstrated a similar level of facial motoneuron survival compared to their littermates that expressed BDNF, and both levels were comparable to wild-type. The results suggest that the deletion of BDNF did not impair CD4(+) T cell-mediated neuroprotection. CONCLUSION: Collectively, while CD4(+) T cells are a potential source of BDNF after nerve injury, production of BDNF is not necessary for CD4(+) T cells to mediate their neuroprotective effects.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Linfócitos T CD4-Positivos/imunologia , Traumatismos do Nervo Facial/imunologia , Animais , Axotomia , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/genética , Linfócitos T CD4-Positivos/metabolismo , Contagem de Células , Sobrevivência Celular/fisiologia , Traumatismos do Nervo Facial/metabolismo , Feminino , Citometria de Fluxo , Camundongos , Camundongos Knockout , Neurônios Motores/fisiologia , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real
16.
Metab Brain Dis ; 27(1): 23-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21975535

RESUMO

It has been well documented that both estrogen and immune cells (CD4+ T cells) mediate neuroprotection in the mouse facial nerve axotomy model. Estrogen has been shown to play an important role in regulating the immune response. However, it is unclear whether immune cell-mediated neuroprotection is dependent on estrogen signaling. In this study, using FACS staining, we confirmed that the majority of CD4+ T cells express high levels of estrogen receptor-alpha (ERα), suggesting that CD4+ T cell-mediated neuroprotection may be modulated by estrogen signaling. We previously found that immunodeficient Rag-2KO mice showed a significant increase in axotomy-induced facial motoneuron death compared to immunocompetent wild-type mice. Therefore, we investigated axotomy-induced facial motoneuron loss in immunodeficient Rag-2KO mice that received 17ß-estradiol treatment or adoptive transfer of immune cells from mice lacking functional ERα. Our results indicate that while estradiol treatment failed to rescue facial motoneurons from axotomy-induced cell death in Rag-2KO mice, immune cells lacking ERα successfully restored facial motoneuron survival in Rag-2 KO mice to a wild-type level. Collectively, we concluded that CD4+ T cell-mediated neuroprotection is independent of estrogen action through ERα.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Traumatismos do Nervo Facial/patologia , Neurônios Motores/fisiologia , Transferência Adotiva , Animais , Axotomia/métodos , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/imunologia , Sobrevivência Celular/imunologia , Sobrevivência Celular/fisiologia , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Estradiol/farmacologia , Receptor alfa de Estrogênio/imunologia , Nervo Facial/imunologia , Nervo Facial/patologia , Nervo Facial/cirurgia , Traumatismos do Nervo Facial/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/citologia , Neurônios Motores/imunologia , Neurônios Motores/metabolismo , Transdução de Sinais
17.
Brain Behav Immun ; 26(2): 195-200, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21855626

RESUMO

The role played by the beta2-adrenergic receptor (ß(2)AR) in regulating the level of T and B lymphocyte function has been studied for over half a century. During this time, we have learned that T and B lymphocytes express almost exclusively the ß(2)AR, and that the level of expression on a specific lymphocyte subset differs due to epigenetic regulation by histone and DNA methylation. We have also learned that engagement of the ß(2)AR on lymphocytes, by either norepinephrine or a selective pharmacologic ligand, regulates the level of lymphocyte activity differentially, depending on the time of receptor engagement in relation to the activation and differentiation state of the cell, the molecular signaling pathway activated, and the cytokine microenvironment. The challenge now is to determine if we understand enough about how this receptor functions on lymphocytes to predict the relevance of such regulation to overall immune homeostasis and the development/progression of human disease.


Assuntos
Linfócitos B/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Linfócitos T/fisiologia , Animais , Linfócitos B/efeitos dos fármacos , Antígeno B7-2/fisiologia , Linfócitos T CD4-Positivos/fisiologia , Humanos , Imunoglobulina G/fisiologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/fisiologia , Norepinefrina/farmacologia , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Células Th1/fisiologia , Células Th2/fisiologia
18.
J Comp Neurol ; 519(17): 3488-506, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21800301

RESUMO

Previously, we compared molecular profiles of one population of wild-type (WT) mouse facial motoneurons (FMNs) surviving with FMNs undergoing significant cell death after axotomy. Regardless of their ultimate fate, injured FMNs respond with a vigorous pro-survival/regenerative molecular response. In contrast, the neuropil surrounding the two different injured FMN populations contained distinct molecular differences that support a causative role for glial and/or immune-derived molecules in directing contrasting responses of the same cell types to the same injury. In the current investigation, we utilized the facial nerve axotomy model and a presymptomatic amyotrophic lateral sclerosis (ALS) mouse (SOD1) model to experimentally mimic the axonal die-back process observed in ALS pathogenesis without the confounding variable of disease onset. Presymptomatic SOD1 mice had a significant decrease in FMN survival compared with WT, which suggests an increased susceptibility to axotomy. Laser microdissection was used to accurately collect uninjured and axotomized facial motor nuclei of WT and presymptomatic SOD1 mice for mRNA expression pattern analyses of pro-survival/pro-regeneration genes, neuropil-specific genes, and genes involved in or responsive to the interaction of FMNs and non-neuronal cells. Axotomized presymptomatic SOD1 FMNs displayed a dynamic pro-survival/regenerative response to axotomy, similar to WT, despite increased cell death. However, significant differences were revealed when the axotomy-induced gene expression response of presymptomatic SOD1 neuropil was compared with WT. We propose that the increased susceptibility of presymptomatic SOD1 FMNs to axotomy-induced cell death and, by extrapolation, disease progression, is not intrinsic to the motoneuron, but rather involves a dysregulated response by non-neuronal cells in the surrounding neuropil.


Assuntos
Regulação da Expressão Gênica , Neurônios Motores/metabolismo , Superóxido Dismutase/biossíntese , Animais , Axotomia , Nervo Facial/metabolismo , Traumatismos do Nervo Facial/genética , Traumatismos do Nervo Facial/metabolismo , Feminino , Predisposição Genética para Doença , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Superóxido Dismutase/genética , Superóxido Dismutase-1
19.
Brain Behav Immun ; 25(5): 820-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20723599

RESUMO

We have previously shown that immunodeficient mice exhibit significant facial motoneuron (FMN) loss compared to wild-type (WT) mice after a facial nerve axotomy. Interleukin-10 (IL-10) is known as a regulatory cytokine that plays an important role in maintaining the anti-inflammatory environment within the central nervous system (CNS). IL-10 is produced by a number of different cells, including Th2 cells, and may exert an anti-apoptotic action on neurons directly. In the present study, the role of IL-10 in mediating neuroprotection following facial nerve axotomy in Rag-2- and IL-10-deficient mice was investigated. Results indicate that IL-10 is neuroprotective, but CD4+ T cells are not the requisite source of IL-10. In addition, using real-time PCR analysis of laser microdissected brainstem sections, results show that IL-10 mRNA is constitutively expressed in the facial nucleus and that a transient, significant reduction of IL-10 mRNA occurs following axotomy under immunodeficient conditions. Dual labeling immunofluorescence data show, unexpectedly, that the IL-10 receptor (IL-10R) is constitutively expressed by facial motoneurons, but is selectively induced in astrocytes within the facial nucleus after axotomy. Thus, a non-CD4+ T cell source of IL-10 is necessary for modulating both glial and neuronal events that mediate neuroprotection of injured motoneurons, but only with the cooperation of CD4+ T cells, providing an avenue of novel investigation into therapeutic approaches to prevent or reverse motoneuron diseases, such as amyotrophic lateral sclerosis (ALS).


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Sistema Nervoso Central/imunologia , Imunidade Celular/fisiologia , Interleucina-10/fisiologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Sistema Nervoso Central/fisiologia , Ensaio de Imunoadsorção Enzimática , Traumatismos do Nervo Facial/imunologia , Traumatismos do Nervo Facial/fisiopatologia , Feminino , Imunidade Celular/imunologia , Inflamação/imunologia , Inflamação/fisiopatologia , Interleucina-10/imunologia , Subunidade alfa de Receptor de Interleucina-10/imunologia , Subunidade alfa de Receptor de Interleucina-10/fisiologia , Subunidade beta de Receptor de Interleucina-10/imunologia , Subunidade beta de Receptor de Interleucina-10/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/imunologia , Neurônios Motores/fisiologia , Neurônios/imunologia , Neurônios/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Brain Behav Immun ; 25(3): 408-15, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21047549

RESUMO

We showed previously that murine naive CD4(+) T cells and T(H)1 cell clones express the beta2-adrenergic receptor (ß(2)AR), while T(H)2 cell clones do not. We report here that naive CD4(+) T cells that differentiated for 1-5 days under T(H)1 driving conditions increased ß(2)AR gene expression, while cells cultured under T(H)2 driving conditions decrease ß(2)AR gene expression. Chromatin immunoprecipitation revealed that the increase in ß(2)AR gene expression in T(H)1 cells is mediated by an increase in histone 3 (H3) and H4 acetylation, as well as an increase in histone 3 lysine 4 (H3K4) methylation. Conversely, the decrease in ß(2)AR gene expression in T(H)2 cells is mediated by a decrease in H3 and H4 acetylation and a decrease in H3K4 methylation, as well as an increase H3K9 and H3K27 methylation. The histone changes could be detected as early as 3 days of differentiating conditions. Genomic bisulfite sequencing showed that the level of methylated CpG dinucleotides within the promoter of the ß(2)AR gene was increased in T(H)2 cells as compared to naive and T(H)1 cells. Collectively, these results suggest that epigenetic mechanisms mediate maintenance and repression, respectively, of the ß(2)AR gene expression in T(H)1- and T(H)2-driven cells, providing a potential mechanism by which the level of ß(2)AR expression might be modulated pharmacologically within immune cells and other cell types in which the expression profile may change during a disease process.


Assuntos
Epigênese Genética , Receptores Adrenérgicos beta 2/metabolismo , Células Th1/metabolismo , Células Th2/metabolismo , Análise de Variância , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Metilação de DNA , Histonas/genética , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Adrenérgicos beta 2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...