Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cells ; 11(19)2022 10 09.
Article in English | MEDLINE | ID: mdl-36231129

ABSTRACT

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.


Subject(s)
Facial Nerve Injuries , Facial Nucleus , Animals , Mice , Axotomy , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Facial Nucleus/metabolism , Interleukin-10/metabolism , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/metabolism , Neuroprotection , RNA, Messenger/metabolism
2.
J Neuroinflammation ; 17(1): 121, 2020 Apr 17.
Article in English | MEDLINE | ID: mdl-32303238

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Facial Nerve Injuries/metabolism , Facial Nerve/metabolism , Motor Neurons/metabolism , Receptors, Interleukin-10/biosynthesis , Animals , Axotomy/methods , Cell Survival/physiology , Cells, Cultured , Facial Nerve Injuries/genetics , Female , Gene Expression , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-10/genetics
3.
Hosp Pediatr ; 9(8): 608-614, 2019 08.
Article in English | MEDLINE | ID: mdl-31308049

ABSTRACT

BACKGROUND: For infants with neonatal abstinence syndrome (NAS) in children's hospitals, treatment protocols emphasizing nonpharmacologic care have revealed improved hospital outcomes. We sought to improve NAS care within the community hospital setting through the implementation of an Eat, Sleep, Console (ESC) protocol. METHODS: Using a multidisciplinary quality improvement approach, we implemented an ESC protocol at 2 community hospitals. Primary outcomes were to decrease length of stay (LOS) by 20% and decrease scheduled morphine use to <20%. Balancing measures included transfer to a higher level of care and unplanned 30-day readmissions. Data were extracted over 2 years, from 2017 through 2018. Interventions included an emphasis on nonpharmacologic care, the initiation of 1-time morphine dosing, flexible weaning schedules for infants on morphine, and the use of ESC scoring. Data were analyzed by using statistical process control. RESULTS: A total of 304 NAS patients were admitted from January 2017 to December 2018, with 155 during the postintervention period. After implementation, mean LOS decreased from 9.0 to 6.2 days, and morphine use decreased from 57% to 23%, both with special cause variation. There were 2 unplanned readmissions in the postintervention period compared with 1 preintervention and no transfers to higher level of care in either period. CONCLUSIONS: Implementation of a nonpharmacologic care protocol within 2 community hospitals led to significant and sustained improvement in LOS and morphine exposure without compromising safety. In this study, we illustrate that evidence-based practice can be successfully implemented and sustained within community hospitals treating infants with NAS.


Subject(s)
Neonatal Abstinence Syndrome/therapy , Quality Improvement , Clinical Protocols , Combined Modality Therapy/methods , Female , Hospitals, Community , Humans , Infant, Newborn , Length of Stay/statistics & numerical data , Male , Patient Readmission/statistics & numerical data
4.
J Med Imaging Radiat Sci ; 49(4): 371-375, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30514553

ABSTRACT

INTRODUCTION: The gender landscape is changing. For professionals in health care, particularly diagnostic imaging (DI), we need better communication tools to obtain personal information from this gender diverse community. We need more specific information from patients because we are performing examinations in which radiation is involved. It is our professional duty to protect a patient's reproductive organs whenever possible, but we must know where those organs are located. In addition, we must determine if a patient could be pregnant or not. Compliance to the professional duty must also extend to transgender and nonbinary patients. Transgender patients do not express or identify the same as their sex assigned at birth; therefore, we may shield inappropriately and expose their reproductive organs unintentionally. Nonbinary patients do not identify as either male or female, and therefore, their expression does not indicate reproductive organ location. METHOD: There are currently no specific forms in DI that ask the questions we need to know to protect the public from unnecessary radiation exposure to reproductive organs. In developing the new form, we began looking at current practices in DI departments to better understand where the communication gap was and what important information would be required in the new form. RESULT: The authors have created a new intake form that accommodates all patients-regardless of age or gender. The result is the SIGE (Sex, Identity, Gender, Expression) form. DISSCUSSION: The SIGE form is inclusive and asks the necessary questions medical radiation technologists need to know in a respectful and professional manner so that we can shield gonadal tissue from ionizing radiation. In addition, the intention of the form is to help the gender diverse community to feel safe and respected in our department.


Subject(s)
Diagnostic Imaging/methods , Forms as Topic , Radiology Department, Hospital/organization & administration , Transgender Persons , Canada , Diagnostic Imaging/adverse effects , Gender Identity , Genitalia/radiation effects , Humans , Patient Admission , Radiation Injuries/prevention & control , Radiation Protection
5.
Restor Neurol Neurosci ; 36(3): 417-422, 2018.
Article in English | MEDLINE | ID: mdl-29614705

ABSTRACT

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.


Subject(s)
Central Nervous System Diseases/pathology , Facial Nerve Injuries/pathology , Facial Nerve/pathology , Motor Neurons/pathology , Animals , Axotomy/methods , Cell Death/physiology , Cell Survival/physiology , Mice, Inbred C57BL
6.
Int J Mol Sci ; 19(2)2018 Feb 23.
Article in English | MEDLINE | ID: mdl-29473876

ABSTRACT

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.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Spatio-Temporal Analysis , Animals , Humans , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Neuromuscular Junction/immunology , Neuromuscular Junction/pathology , Schwann Cells/immunology , Schwann Cells/pathology
7.
Neural Regen Res ; 12(10): 1664-1679, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29171432

ABSTRACT

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.

8.
Mediators Inflamm ; 2016: 6131234, 2016.
Article in English | MEDLINE | ID: mdl-27194826

ABSTRACT

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.


Subject(s)
Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Motor Neurons/pathology , Superoxide Dismutase-1/metabolism , Th17 Cells/metabolism , Animals , Disease Models, Animal , Facial Nerve Injuries/immunology , Female , Mice , Mice, Transgenic , Motor Neurons/immunology , Motor Neurons/metabolism , Superoxide Dismutase-1/genetics , T-Lymphocytopenia, Idiopathic CD4-Positive/metabolism , Th17 Cells/immunology
9.
J Neuroimmune Pharmacol ; 10(4): 587-94, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26148561

ABSTRACT

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.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , CD4-Positive T-Lymphocytes/immunology , Motor Neurons/immunology , Nerve Regeneration/immunology , Neuroprotection/immunology , Animals , Humans
10.
J Med Chem ; 58(13): 5344-54, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26067904

ABSTRACT

A series of 6-hetaryloxy benzoxaborole compounds was designed and synthesized for a structure-activity relationship (SAR) investigation to assess the changes in antimalarial activity which result from 6-aryloxy structural variation, substituent modification on the pyrazine ring, and optimization of the side chain ester group. This SAR study discovered highly potent 6-(2-(alkoxycarbonyl)pyrazinyl-5-oxy)-1,3-dihydro-1-hydroxy-2,1-benzoxaboroles (9, 27-34) with IC50s = 0.2-22 nM against cultured Plasmodium falciparum W2 and 3D7 strains. Compound 9 also demonstrated excellent in vivo efficacy against P. berghei in infected mice (ED90 = 7.0 mg/kg).


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Malaria, Falciparum/drug therapy , Microsomes, Liver/drug effects , Plasmodium falciparum/drug effects , Pyrazines/chemistry , Pyrazines/pharmacology , Animals , Cell Survival/drug effects , Female , Humans , Jurkat Cells , Malaria, Falciparum/parasitology , Mice , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship
11.
J Comp Neurol ; 523(18): 2752-68, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26010802

ABSTRACT

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.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/genetics , Mutation/genetics , Superoxide Dismutase/genetics , Age Factors , Amyotrophic Lateral Sclerosis/complications , Animals , Disease Models, Animal , Disease Progression , Facial Nerve/metabolism , Feeding Behavior/physiology , Laser Capture Microdissection , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Disorders/etiology , Muscle Strength/genetics , RNA, Messenger/metabolism , Sensation Disorders/etiology , Transfection
12.
Glia ; 63(6): 1057-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25740080

ABSTRACT

Although the peripheral anti-inflammatory effect of norepinephrine (NE) is well documented, the mechanism by which this neurotransmitter functions as an anti-inflammatory/neuroprotective agent in the central nervous system (CNS) is unclear. This article aimed to determine the anti-inflammatory/neuroprotective effects and underlying mechanisms of NE in inflammation-based dopaminergic neurotoxicity models. In mice, NE-depleting toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected at 6 months of lipopolysaccharide (LPS)-induced neuroinflammation. It was found that NE depletion enhanced LPS-induced dopaminergic neuron loss in the substantia nigra. This piece of in vivo data prompted us to conduct a series of studies in an effort to elucidate the mechanism as to how NE affects dopamine neuron survival by using primary midbrain neuron/glia cultures. Results showed that submicromolar concentrations of NE dose-dependently protected dopaminergic neurons from LPS-induced neurotoxicity by inhibiting microglia activation and subsequent release of pro-inflammatory factors. However, NE-elicited neuroprotection was not totally abolished in cultures from ß2-adrenergic receptor (ß2-AR)-deficient mice, suggesting that novel pathways other than ß2-AR are involved. To this end, It was found that submicromolar NE dose-dependently inhibited NADPH oxidase (NOX2)-generated superoxide, which contributes to the anti-inflammatory and neuroprotective effects of NE. This novel mechanism was indeed adrenergic receptors independent since both (+) and (-) optic isomers of NE displayed the same potency. We further demonstrated that NE inhibited LPS-induced NOX2 activation by blocking the translocation of its cytosolic subunit to plasma membranes. In summary, we revealed a potential physiological role of NE in maintaining brain immune homeostasis and protecting neurons via a novel mechanism.


Subject(s)
Brain/immunology , Dopaminergic Neurons/immunology , Microglia/enzymology , NADPH Oxidases/metabolism , Norepinephrine/metabolism , Animals , Benzylamines/pharmacology , Brain/drug effects , Brain/pathology , COS Cells , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Coculture Techniques , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Homeostasis/physiology , Lipopolysaccharides/toxicity , Male , Mice, Inbred BALB C , Mice, Knockout , Microglia/drug effects , Microglia/pathology , Neurotransmitter Uptake Inhibitors/pharmacology , Rats, Inbred F344 , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism
13.
Brain Behav Immun ; 40: 55-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24911596

ABSTRACT

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.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , CD4-Positive T-Lymphocytes/immunology , Facial Nucleus/immunology , Motor Neurons/immunology , Superoxide Dismutase/genetics , Adoptive Transfer , Amyotrophic Lateral Sclerosis/pathology , Animals , Axotomy , CD4-Positive T-Lymphocytes/transplantation , DNA-Binding Proteins/genetics , Facial Nerve Injuries , Facial Nucleus/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/pathology , Superoxide Dismutase-1
15.
J Comp Neurol ; 522(10): 2349-76, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24424947

ABSTRACT

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.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Cell Death/physiology , Facial Nerve/physiopathology , Motor Neurons/physiology , Nerve Degeneration/physiopathology , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axotomy , Disease Progression , Facial Nerve/pathology , Facial Nucleus/pathology , Facial Nucleus/physiopathology , Female , Gene Expression , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Motor Neurons/pathology , Nerve Degeneration/pathology , Neuroglia/pathology , Neuroglia/physiology , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , fas Receptor/metabolism
16.
J Immunol ; 191(11): 5383-97, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24140643

ABSTRACT

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.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , B-Lymphocytes/immunology , Exosomes/metabolism , Immunoglobulin E/metabolism , Membrane Proteins/metabolism , Receptors, IgE/metabolism , ADAM Proteins/genetics , ADAM10 Protein , Adrenergic beta-2 Receptor Agonists/pharmacology , Amyloid Precursor Protein Secretases/genetics , Animals , B-Lymphocytes/drug effects , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Lymphocyte Activation/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Transport , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/immunology , Receptors, IgE/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Immunol ; 190(2): 723-36, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23241883

ABSTRACT

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.


Subject(s)
B-Lymphocytes/metabolism , B7-2 Antigen/metabolism , Protein Interaction Domains and Motifs , Repressor Proteins/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Animals , B7-2 Antigen/chemistry , CD40 Antigens/metabolism , Cell Line , Cell Nucleus/metabolism , Female , Gene Expression Regulation , Mice , NF-kappa B/metabolism , Phospholipase C gamma/metabolism , Prohibitins , Protein Binding , Protein Kinase C/metabolism , Repressor Proteins/genetics
18.
J Neurodegener Regen ; 4(1): 21-25, 2013.
Article in English | MEDLINE | ID: mdl-24672589

ABSTRACT

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.

19.
FEBS Lett ; 586(19): 3410-4, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22841723

ABSTRACT

We have used boron-based molecules to create novel, competitive, reversible inhibitors of phosphodiesterase 4 (PDE4). The co-crystal structure reveals a binding configuration which is unique compared to classical catechol PDE4 inhibitors, with boron binding to the activated water in the bimetal center. These phenoxybenzoxaboroles can be optimized to generate submicromolar potency enzyme inhibitors, which inhibit TNF-α, IL-2, IFN-γ, IL-5 and IL-10 activities in vitro and show safety and efficacy for topical treatment of human psoriasis. They provide a valuable new route for creating novel potent anti-PDE4 inhibitors.


Subject(s)
Boron Compounds/chemistry , Boron Compounds/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Binding, Competitive , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Catalytic Domain , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cytokines/biosynthesis , Humans , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Metals/chemistry , Models, Molecular , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
J Pharm Biomed Anal ; 70: 344-53, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22687459

ABSTRACT

There is an increasing interest in in vivo metabolite identification in early drug discovery in order to (i) give a more complete picture of metabolic profile in investigational animal models, (ii) propose phase I and phase II metabolites using the same pharmacokinetic/toxicokinetic study samples, (iii) expose metabolically labile groups where chemical modifications could improve stability, and (iv) enable early safety assessment of metabolites. In the early discovery stage of our anti-inflammatory program, one novel benzoxaborole, AN6414, exhibiting both PDE4 enzyme and TNFα inhibition activities, became our primary candidate for further investigation. The traditional metabolite identifications usually require high dosed samples with long data scans and analysis. In this study, we conducted quick and more selective core-structure related precursor scans followed by daughter ion scans and identified a total of 10 major phase I and phase II metabolites using rat plasma samples from a toxicokinetic study at an oral dosing of 30 mg/kg. Plasma samples were treated with solid phase extraction (SPE) prior to LC/MS/MS. An AB SCIEX API 4000 QTRAP mass spectrometer coupled with a Shimadzu LC system was used for LC/MS/MS analysis. We found the major metabolites of AN6414 to be oxidative deboronation, protodeboronation, oxidation products and their sulfate-conjugated species. This analysis drove analoging efforts which improved the pharmacokinetic profile, namely, lowering clearance and increasing exposure relative to AN6414. Toxicity predictions by the software program DEREK suggest the identified potential metabolites to be safe.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Boron Compounds/pharmacokinetics , Chromatography, Liquid , Drug Discovery/methods , Phosphodiesterase 4 Inhibitors/pharmacology , Pyridines/pharmacokinetics , Tandem Mass Spectrometry , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/urine , Area Under Curve , Biotransformation , Boron Compounds/administration & dosage , Boron Compounds/blood , Boron Compounds/toxicity , Female , Male , Metabolic Clearance Rate , Models, Biological , Oxidation-Reduction , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/blood , Phosphodiesterase 4 Inhibitors/toxicity , Pyridines/administration & dosage , Pyridines/blood , Pyridines/toxicity , Rats , Rats, Sprague-Dawley , Risk Assessment , Software , Solid Phase Extraction , Sulfates/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...