Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Front Cell Neurosci ; 16: 779081, 2022.
Article in English | MEDLINE | ID: mdl-35431807

ABSTRACT

Activation of nicotinic acetylcholine receptors (nAChRs) expressed by innate immune cells can attenuate pro-inflammatory responses. Silent nAChR agonists, which down-modulate inflammation but have little or no ionotropic activity, are of outstanding clinical interest for the prevention and therapy of numerous inflammatory diseases. Here, we compare two silent nAChR agonists, phosphocholine, which is known to interact with nAChR subunits α7, α9, and α10, and pCF3-N,N-diethyl-N'-phenyl-piperazine (pCF3-diEPP), a previously identified α7 nAChR silent agonist, regarding their anti-inflammatory properties and their effects on ionotropic nAChR functions. The lipopolysaccharide (LPS)-induced release of interleukin (IL)-6 by primary murine macrophages was inhibited by pCF3-diEPP, while phosphocholine was ineffective presumably because of instability. In human whole blood cultures pCF3-diEPP inhibited the LPS-induced secretion of IL-6, TNF-α and IL-1ß. The ATP-mediated release of IL-1ß by LPS-primed human peripheral blood mononuclear leukocytes, monocytic THP-1 cells and THP-1-derived M1-like macrophages was reduced by both phosphocholine and femtomolar concentrations of pCF3-diEPP. These effects were sensitive to mecamylamine and to conopeptides RgIA4 and [V11L; V16D]ArIB, suggesting the involvement of nAChR subunits α7, α9 and/or α10. In two-electrode voltage-clamp measurements pCF3-diEPP functioned as a partial agonist and a strong desensitizer of classical human α9 and α9α10 nAChRs. Interestingly, pCF3-diEPP was more effective as an ionotropic agonist at these nAChRs than at α7 nAChR. In conclusion, phosphocholine and pCF3-diEPP are potent agonists at unconventional nAChRs expressed by monocytic and macrophage-like cells. pCF3-diEPP inhibits the LPS-induced release of pro-inflammatory cytokines, while phosphocholine is ineffective. However, both agonists signal via nAChR subunits α7, α9 and/or α10 to efficiently down-modulate the ATP-induced release of IL-1ß. Compared to phosphocholine, pCF3-diEPP is expected to have better pharmacological properties. Thus, low concentrations of pCF3-diEPP may be a therapeutic option for the treatment of inflammatory diseases including trauma-induced sterile inflammation.

2.
Brain Behav Immun ; 87: 286-300, 2020 07.
Article in English | MEDLINE | ID: mdl-31874200

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) are best known to function as ligand-gated ion channels in the nervous system. However, recent evidence suggests that nicotine modulates inflammation by desensitizing non-neuronal nAChRs, rather than by inducing channel opening. Silent agonists are molecules that selectively induce the desensitized state of nAChRs while producing little or no channel opening. A silent agonist of α7 nAChRs has recently been shown to reduce inflammation in an animal model of inflammatory pain. The objective of this study was to determine whether a silent agonist of α7 nAChRs can also effectively modulate inflammation and disease manifestation in an animal model of multiple sclerosis. We first evaluated the effects of various nAChR ligands and of an α7 nAChR-selective silent agonist, 1-ethyl-4-(3-(bromo)phenyl)piperazine (m-bromo PEP), on the modulation of mouse bone marrow-derived monocyte/macrophage (BMDM) numbers, phenotype and cytokine production. The non-competitive antagonist mecamylamine and the silent agonist m-bromo PEP reduced pro-inflammatory BMDM numbers by affecting their viability and proliferation. Both molecules also significantly reduced cytokine production by mouse BMDMs and significantly ameliorated disease in experimental autoimmune encephalomyelitis. Finally, m-bromo PEP also reduced chronic inflammatory pain in mice. Taken together, our results further support the hypothesis that nAChRs may modulate inflammation via receptor desensitization rather than channel opening. α7 nAChR-selective silent agonists may thus be a novel source of anti-inflammatory compounds that could be used for the treatment of inflammatory disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Receptors, Nicotinic , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Inflammation/drug therapy , Mice , Nicotinic Agonists/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
3.
BMC Neurol ; 17(1): 28, 2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28178923

ABSTRACT

BACKGROUND: Non-neuronal acetylcholine (ACh) restricts autoimmune responses and attenuates inflammation by cholinergic anti-inflammation pathway. To date, the implication of ACh in myasthenia gravis (MG) remained unexplored. This study aimed to investigate the possible relationship between ACh levels, anti-muscle-specific tyrosine kinase (MuSK) antibody titers, main clinical features and outcomes of MG patients. METHODS: We successfully measured ACh levels in human peripheral blood mononuclear cells (PBMCs) from 125 MG patients and 50 matched healthy controls by using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). We assessed the quantitative MG (QMG) scores for each patient and titered anti-MuSK antibody. RESULTS: We found that PBMC-derived ACh level was significantly higher in MG patients, especially in patients of class III, IV-V, compared with that in controls (0.142 ± 0.108 vs. 0.075 ± 0.014 ng/million cells, p = 0.0003) according to the Myasthenia Gravis Foundation of America clinical classification. Importantly, we also found that ACh levels were positively correlated with QMG scores (r = 0.83, p < 0.0001) and anti-MuSK Ab levels (r = 0.85, p < 0.0001). CONCLUSIONS: Our demonstration of elevated ACh levels in PBMCs of MG patients foreshadows potential new avenues for MG research and treatment.


Subject(s)
Acetylcholine/blood , Leukocytes, Mononuclear/metabolism , Myasthenia Gravis/blood , Adult , Autoantibodies/blood , Female , Humans , Male , Middle Aged , Receptor Protein-Tyrosine Kinases/blood , Receptors, Cholinergic/blood , Tandem Mass Spectrometry
4.
PLoS One ; 11(2): e0150230, 2016.
Article in English | MEDLINE | ID: mdl-26925951

ABSTRACT

It is increasingly clear that nicotinic acetylcholine receptors (nAChRs) are involved in immune regulation, and that their activation can protect against inflammatory diseases. Previous data have shown that nicotine diminishes the numbers of peripheral monocytes and macrophages, especially those of the pro-inflammatory phenotype. The goal of the present study was to determine if nicotine modulates the production of bone marrow -derived monocytes/macrophages. In this study, we first found that murine bone marrow cells express multiple nAChR subunits, and that the α7 and α9 nAChRs most predominant subtypes found in immune cells and their precursors. Using primary cultures of murine bone marrow cells, we then determined the effect of nicotine on monocyte colony-stimulating factor and interferon gamma (IFNγ)-induced monocyte production. We found that nicotine lowered the overall number of monocytes, and more specifically, inhibited the IFNγ-induced increase in pro-inflammatory monocytes by reducing cell proliferation and viability. These data suggested that nicotine diminishes the ratio of pro-inflammatory versus anti-inflammatory monocyte produced in the bone marrow. We thus confirmed this hypothesis by measuring cytokine expression, where we found that nicotine inhibited the production of the pro-inflammatory cytokines TNFα, IL-1ß and IL-12, while stimulating the secretion of IL-10, an anti-inflammatory cytokine. Finally, nicotine also reduced the number of pro-inflammatory monocytes in the bone marrow of LPS-challenged mice. Overall, our data demonstrate that both α7 and α9 nAChRs are involved in the regulation of pro-inflammatory M1 monocyte numbers.


Subject(s)
Monocytes/cytology , Monocytes/metabolism , Receptors, Nicotinic/metabolism , Animals , Cell Count , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colony-Stimulating Factors/pharmacology , Gene Expression Regulation/drug effects , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/pharmacology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Nicotine/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Nicotinic/genetics
5.
CNS Neurosci Ther ; 22(4): 298-305, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26842313

ABSTRACT

AIMS: Long noncoding RNAs (lncRNAs) play a key role in regulating immunological functions. Their impact on the chronic inflammatory disease multiple sclerosis (MS), however, remains unknown. We investigated the expression of lncRNAs in peripheral blood mononuclear cells (PBMCs) of patients with MS and attempt to explain their possible role in the process of MS. METHODS: For this study, we recruited 26 patients with MS according to the revised McDonald criteria. Then, we randomly chose 6 patients for microarray analysis. Microarray assays identified outstanding differences in lncRNA expression, which were verified through real-time PCR. LncRNA functions were annotated for target genes using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and regulatory relationships between lncRNAs and target genes were analyzed using the "cis" and "trans" model. RESULTS: There were 2353 upregulated lncRNAs, 389 downregulated lncRNAs, 1037 upregulated mRNAs, and 279 downregulated mRNAs in patients with MS compared to healthy control subjects (fold change >2.0). Real-time PCR results of six aberrant lncRNAs were consistent with the microarray data. The coexpression network comprised 864 lncRNAs and 628 mRNAs. Among differentially expressed lncRNAs, 10 lncRNAs were predicted to have 10 cis-regulated target genes, and 33 lncRNAs might regulate their trans target genes. CONCLUSIONS: We identified a subset of dysregulated lncRNAs and mRNAs. The differentially expressed lncRNAs may be important in the process of MS. However, the specific molecular mechanisms and biological functions of these lncRNAs in the pathogenesis of MS need further study.


Subject(s)
Leukocytes, Mononuclear/metabolism , Multiple Sclerosis, Relapsing-Remitting/blood , RNA, Long Noncoding/metabolism , Adult , Female , Gene Expression Profiling , Gene Ontology , Humans , Male , Microarray Analysis , Middle Aged , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Severity of Illness Index , Young Adult
6.
Sci Rep ; 6: 21819, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26898505

ABSTRACT

The aim of our present study was to determine whether message RNAs (mRNAs) and long noncoding RNAs (lncRNAs) are expressed differentially in patients with Guillain-Barré syndrome (GBS) compared with healthy controls. The mRNA and lncRNA profiles of GBS patients and healthy controls were generated by using microarray analysis. From microarray analysis, we listed 310 mRNAs and 114 lncRNAs with the mRMR software classed into two sample groups, GBS patients and healthy controls. KEGG mapping demonstrated that the top seven signal pathways may play important roles in GBS development. Several GO terms, such as cytosol, cellular macromolecular complex assembly, cell cycle, ligase activity, protein catabolic process, etc., were enriched in gene lists, suggesting a potential correlation with GBS development. Co-expression network analysis indicated that 113 lncRNAs and 303 mRNAs were included in the co-expression network. Our present study showed that these differentially expressed mRNAs and lncRNAs may play important roles in GBS development, which provides basic information for defining the mechanism(s) that promote GBS.


Subject(s)
Gene Regulatory Networks , Guillain-Barre Syndrome/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Transcriptome , Case-Control Studies , Disease Progression , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/pathology , Humans , Metabolic Networks and Pathways/genetics , Microarray Analysis , Molecular Sequence Annotation , Signal Transduction
7.
J Immunol ; 196(5): 2095-108, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26810225

ABSTRACT

Myeloid cells, including proinflammatory monocytes and neutrophils, have important roles in the pathology of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). These cells infiltrate the CNS in the early stages of disease development and contribute to the inflammatory response that is associated with symptom severity. It is thus crucial to identify and understand new mechanisms that can regulate the CNS infiltration of proinflammatory myeloid cells. Nicotinic acetylcholine receptors (nAChRs) have been increasingly studied for their immune-regulatory properties. In this study, we assessed the ability of nicotine, an nAChR ligand, to modulate proinflammatory myeloid cell numbers within the bone marrow, spleen, blood, and CNS of EAE mice. We found that nicotine significantly inhibits the infiltration of proinflammatory monocytes and neutrophils into the CNS at time points where these cells are known to play critical roles in disease pathology. In contrast, nicotine does not affect the expansion of other monocytes. We also show that nicotine exerts these effects by acting on α7 and α9 nAChR subtypes. Finally, mRNA transcript levels for CCL2 and CXCL2, chemokines involved in the chemotaxis of proinflammatory monocytes and neutrophils, respectively, are reduced in the brain of nicotine-treated EAE mice before the massive infiltration of these cells. Taken together, our data provide evidence that nAChRs can regulate proinflammatory cell infiltration into the CNS, which could be of significant value for the treatment of neuroinflammatory disorders.


Subject(s)
Chemotaxis, Leukocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Monocytes/immunology , Neutrophils/immunology , Receptors, Nicotinic/immunology , Animals , Antigens, Ly/immunology , Brain/immunology , Cell Separation , Chemotaxis, Leukocyte/drug effects , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, CCR2/immunology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/immunology
8.
Clin Rev Allergy Immunol ; 47(3): 274-88, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24057598

ABSTRACT

Guillain-Barré syndrome (GBS) is an acute and usually monophasic, neurological, demyelinating disease. Although most patients have good outcomes without sequelae after conventional plasma exchange and intravenous immunoglobulin therapy, 20% of patients continue to have severe disease and 5% die of their disease. Therefore, there is an obvious need for more acceptable and efficacious therapies. Experimental autoimmune neuritis (EAN) is the classical animal model for GBS. As there is no specific drug for GBS, several drugs targeting the humoral and cellular components of the immune response have been used to treat EAN in the endeavour to find new treatment alternatives for GBS. This review focused on some new strategies for GBS, which have been reported but have not yet been widely used, and on the main drugs which have been investigated in EAN.


Subject(s)
Guillain-Barre Syndrome/therapy , Animals , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/physiopathology , Humans
9.
Int Rev Immunol ; 32(3): 324-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23617759

ABSTRACT

IL-10 is an anti-inflammatory cytokine that plays a significant role in controlling inflammation and modulating adaptive immune responses that cause tissue damage. IL-10-producing lymphocytes contribute to the delicate balance between inflammation and immunoregulation, and are thus regarded as a kind of "regulatory cells." Dysregulation of these cells is linked with susceptibility to numerous inflammatory diseases. In this review, we summarized what is known about the regulatory effects of IL-10 produced by lymphocytes, including T cells, B cells and natural killer cells, in inflammatory diseases. We hope to augment immune responses or prevent immunopathology through making some small changes in the levels of IL-10 produced by lymphocytes, or in the cellular location where it is produced.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes, Regulatory/immunology , Immunotherapy/methods , Interleukin-10/metabolism , Killer Cells, Natural/immunology , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity , Animals , Autoimmune Diseases/therapy , Gene Expression Regulation , Humans , Immunotherapy/trends , Interleukin-10/genetics
10.
Immunol Cell Biol ; 91(3): 195-200, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23399696

ABSTRACT

Nicotine is a potent inhibitor of the immune response and is protective against experimental autoimmune encephalomyelitis (EAE). Initial studies suggested that the cholinergic system modulates inflammation via the α7-nicotinic acetylcholine receptor (nAChR) subtype. We recently have shown that effector T cells and myeloid cells constitutively express mRNAs encoding nAChR α9 and ß2 subunits and found evidence for immune system roles for non-α7-nAChRs. In the present study, we assessed the effects of nAChR α9 or ß2 subunit gene deletion on EAE onset and severity, with or without nicotine treatment. We report again that disease onset is delayed and severity is attenuated in nicotine-treated, wild-type mice, an effect that also is observed in α9 subunit knock-out (KO) mice irrespective of nicotine treatment. On the other hand, ß2 KO mice fail to recover from peak measures of disease severity regardless of nicotine treatment, despite retaining sensitivity to nicotine's attenuation of disease severity. Prior to disease onset, we found significantly less reactive oxygen species production in the central nervous system (CNS) of ß2 KO mice, elevated proportions of CNS myeloid cells but decreased ratios of CNS macrophages/microglia in α9 or ß2 KO mice, and some changes in iNOS, TNF-α and IL-1ß mRNA levels in α9 KO and/or ß2 KO mice. Our data thus suggest that ß2*- and α9*-nAChRs, in addition to α7-nAChRs, have different roles in endogenous and nicotine-dependent modulation of immune functions and could be exploited as therapeutic targets to modulate inflammation and autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophages/immunology , Microglia/immunology , Receptors, Nicotinic/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Deletion , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/pathology , Mice , Mice, Knockout , Microglia/pathology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Reactive Oxygen Species/immunology , Receptors, Nicotinic/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
Neurosci Bull ; 29(1): 83-93, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23307113

ABSTRACT

The matrix-degrading metalloproteinases (MMPs), particularly MMP-9, play important roles in the pathogenesis and development of malignant gliomas. In the present study, the oncogenic role of MMP-9 in malignant glioma cells was investigated via antisense RNA blockade in vitro and in vivo. TJ905 malignant glioma cells were transfected with pcDNA3.0 vector expressing antisense MMP-9 RNA (pcDNA-ASMMP9), which significantly decreased MMP-9 expression, and cell proliferation was assessed. For in vivo studies, U251 cells, a human malignant glioma cell line, were implanted subcutaneously into 4- to 6-week-old BALB/c nude mice. The mice bearing well-established U251 gliomas were treated with intratumoral pcDNA-AS-MMP9-Lipofectamine complex (AS-MMP-9-treated group), subcutaneous injection of endostatin (endostatin-treated group), or both (combined therapy group). Mice treated with pcDNA (empty vector)-Lipofectamine served as the control group. Four or eight weeks later, the volume and weight of tumor, MMP-9 expression, microvessel density and proliferative activity were assayed. We demonstrate that pcDNA-AS-MMP9 significantly decreased MMP-9 expression and inhibited glioma cell proliferation. Volume and weight of tumor, MMP-9 expression, microvessel density and proliferative activity in the antisense-MMP-9-treated and therapeutic alliance groups were significantly lower than those in the control group. The results suggest that MMP-9 not only promotes malignant glioma cell invasiveness, but also affects tumor cell proliferation. Blocking the expression of MMP-9 with antisense RNA substantially suppresses the malignant phenotype of glioma cells, and thus can be used as an effective therapeutic strategy for malignant gliomas.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioma/pathology , Matrix Metalloproteinase 9/metabolism , RNA, Antisense/therapeutic use , Animals , Brain Neoplasms/metabolism , Cell Proliferation , Glioma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , RNA, Antisense/metabolism , Tumor Cells, Cultured
12.
J Neurol Sci ; 312(1-2): 57-61, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21917268

ABSTRACT

Genetic analyses indicate that HLA complex genes can be involved in susceptibility to autoimmune myasthenia gravis (MG). Various HLA alleles serve as genetic elements that either predispose to or protect against MG. This study investigates the probable relationship between HLA-DQ allele polymorphisms and MG cases in northern China. The HLA-DQA1 and DQB1 alleles were determined by polymerase chain reaction/sequence-specific primers (PCR-SSP) in 84 MG patients, and the results were compared to 293 healthy controls. Our findings indicate that DQ A1*0401(P=0.008, OR: 2.5, 95%CI: 1.24-3.07) and B1*0301(P=0.000, OR: 2.29, 95%CI: 1.48-3.54) were the most frequent allele; the frequencies of DQA1*0103(P=0.000, OR:0.24, 95%CI 0.13-0.49) and DQB1*0601(P=0.001, OR:0.40, 95%CI 0.22-0.50) were significantly decreased in MG patients compared with healthy controls. Patients with thymomatous MG were positively associated with DQA1 *0401(P=0.011, OR:4.57, 95% CI 1.40-14.90) and DQB1 *0604 (P=0.001, OR:4.01, 95% CI 1.65-9.73) as compared to MG patients without thymoma. Different genetic mechanisms may exist between MG patients with thymoma and those without thymoma. The HLA-DQ associations in MG subgroups suggest that disease heterogeneity may be influenced by different genes or alleles.


Subject(s)
HLA-DQ alpha-Chains/genetics , HLA-DQ beta-Chains/genetics , Myasthenia Gravis/genetics , Thymoma/genetics , Thymus Neoplasms/genetics , Adult , Asian People/genetics , China/epidemiology , Female , Gene Frequency , Genetic Predisposition to Disease/ethnology , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Myasthenia Gravis/ethnology , Polymorphism, Genetic/genetics , Thymoma/ethnology , Thymus Neoplasms/ethnology , Young Adult
13.
Psychopharmacology (Berl) ; 218(2): 405-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21562760

ABSTRACT

RATIONALE: The synthetic progestin medroxyprogesterone acetate (MPA), widely used in hormone therapy (HT) and as the contraceptive Depo Provera, is implicated in detrimental cognitive effects in women. Recent evidence in aged ovariectomized (Ovx) rodents shows that short-term MPA treatment impairs cognition and alters the GABAergic system. OBJECTIVES: Using rats, we evaluated the long-lasting cognitive and GABAergic effects of MPA administered in young adulthood (Early-MPA), modeling contraception, and how this early exposure interacts with later MPA treatment (Late-MPA), modeling HT. METHODS: Early-MPA treatment involved weekly anti-ovulatory MPA injections (3.5 mg) from 4 to 8 months of age in ovary-intact rats. At 10 months old, rats were Ovx and weekly MPA injections were re-initiated and continued throughout testing for Late-MPA treatment. RESULTS: On the water radial-arm maze, all MPA-treated groups showed working memory impairment compared to Controls (p < 0.05); Early + Late-MPA rats were impaired on multiple dimensions of working memory (p < 0.05). On the Morris maze, Late-MPA rats showed greater overnight forgetting compared to Controls (p < 0.05). At study conclusion, MPA was detected in serum in all MPA-treated groups except Early-MPA, confirming treatment and clearance from serum in Early-MPA rats. In animals with detectable serum MPA, higher MPA levels were associated with less dorsal-hippocampal glutamic acid decarboxylase, the synthesizing enzyme for GABA (p = 0.0059). CONCLUSIONS: Findings suggest that MPA treatment leads to long-lasting cognitive impairments in the rodent, even in the absence of circulating MPA in animals given prior MPA treatment, which may relate to the GABAergic system. Further research defining the parameters of the negative impact of this widely used progestin on brain and cognition is warranted.


Subject(s)
Cognition Disorders/chemically induced , Contraceptive Agents, Female/adverse effects , Medroxyprogesterone Acetate/adverse effects , gamma-Aminobutyric Acid/metabolism , Animals , Contraceptive Agents, Female/blood , Female , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Maze Learning/drug effects , Medroxyprogesterone Acetate/blood , Memory, Short-Term/drug effects , Ovariectomy , Rats , Rats, Inbred F344 , Time Factors
14.
Exp Neurol ; 227(1): 110-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20932827

ABSTRACT

A considerable number of in vivo studies have demonstrated that the cholinergic system can dampen the peripheral immune response, and it is thought that the α7-nicotinic acetylcholine receptor (nAChR) subtype is a key mediator of this process. The goal of the present study was to determine if nicotine modulates immunological mechanisms known to be involved in the development of experimental autoimmune encephalomyelitis (EAE), a mouse model for CNS autoimmune disease, via α7-nAChRs. Here we show that nicotine exposure attenuates EAE severity and that this effect is largely abolished in nAChR α7 subunit knock-out mice. However, nicotine exposure partially retains the ability to reduce lymphocyte infiltration into the CNS, inhibit auto-reactive T cell proliferation and helper T cell cytokine production, down-regulate co-stimulatory protein expression on myeloid cells, and increase the differentiation and recruitment of regulatory T cells, even in the absence of α7-nAChRs. Diverse effects of nicotine on effector and regulatory T cells, as well as antigen-presenting cells, may be linked to differential expression patterns of nAChR subunits across these cell types. Taken together, our data show that although α7-nAChRs indeed seem to play an important role in nicotine-conferred reduction of the CNS inflammatory response and protection against EAE, other nAChR subtypes also are involved in the anti-inflammatory properties of the cholinergic system.


Subject(s)
Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Receptors, Nicotinic/metabolism , Animals , Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Flow Cytometry/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Lymph Nodes/drug effects , Lymph Nodes/pathology , Magnetic Resonance Imaging/methods , Mice , Mice, Knockout , RNA, Messenger/metabolism , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Spleen/drug effects , Spleen/pathology , Statistics, Nonparametric , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , alpha7 Nicotinic Acetylcholine Receptor
15.
Horm Behav ; 58(5): 917-28, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20849857

ABSTRACT

In women, ovarian hormone loss at menopause has been related to cognitive decline, and some studies suggest that estrogen-containing hormone therapy (HT) can mitigate these effects. Recently, the Women's Health Initiative study found that conjugated equine estrogens, the most commonly prescribed HT, do not benefit cognition. Isolated components of conjugated equine estrogens (tradename Premarin(®)) have been evaluated in vitro, with delta(8,9)-dehydroestrone (∆(8)E1) and equilin showing the strongest neuroprotective profiles. It has not been evaluated whether ∆(8)E1 or equilin impact cognition or the cholinergic system, which is affected by other estrogens and known to modulate cognition. Here, in middle-aged, ovariectomized rats, we evaluated the effects of ∆(8)E1 and equilin treatments on a cognitive battery and cholinergic nicotinic receptors (nAChR). Specifically, we used (125)I-labeled epibatidine binding to assay brain nicotinic receptor containing 4α and 2ß subunits (α4ß2-nAChR), since this nicotinic receptor subtype has been shown previously to be sensitive to other estrogens. ∆(8)E1 enhanced spatial working, recent and reference memory. ∆(8)E1 also decreased hippocampal and entorhinal cortex α4ß2-nAChR expression, which was related to spatial reference memory performance. Equilin treatment did not affect spatial memory or rat α4ß2-nAChR expression, and neither estrogen impacted (86)Rb(+) efflux, indicating lack of direct action on human α4ß2 nAChR function. Both estrogens influenced vaginal smear profiles, uterine weights, and serum luteinizing hormone levels, analogous to classic estrogens. The findings indicate that specific isolated Premarin(®) components differ in their ability to affect cognition and nAChR expression. Taken with the works of others showing ∆(8)E1-induced benefits on several dimensions of health-related concerns associated with menopause, this body of research identifies ∆(8)E1 as a new avenue to be investigated as a potential component of HT that may benefit brain health and function during aging.


Subject(s)
Cognition/drug effects , Estrogens, Conjugated (USP)/pharmacology , Receptors, Nicotinic/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Cells, Cultured , Cognition/physiology , Drug Evaluation, Preclinical , Estrogens, Conjugated (USP)/chemistry , Female , Humans , Maze Learning/drug effects , Memory/drug effects , Memory/physiology , Ovariectomy , Rats , Rats, Inbred F344 , Spatial Behavior/drug effects , Spatial Behavior/physiology , Up-Regulation/drug effects
16.
Neurobiol Learn Mem ; 93(3): 444-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20074654

ABSTRACT

In women, medroxyprogesterone acetate (MPA) is the most commonly used progestin component of hormone therapy (HT). In vitro, MPA negatively impacts markers of neuronal health and exacerbates experimentally-induced neurotoxicity. There is in vitro evidence that these factors are driven by GABAergic and neurotrophic systems. Whether these effects translate to a negative impact on brain function has not been tested in vivo, clinically or preclinically. Here we evaluate the mnemonic and neurobiological effects of MPA in the surgically menopausal rat. Aged ovariectomized (OVX) rats were given subcutaneous vehicle, natural progesterone, low-dose MPA or high-dose MPA. Multiple cognitive domains were analyzed via the water radial-arm maze (WRAM) and Morris maze (MM). Cognitive brain regions were assayed for changes in the GABAergic system by evaluating GAD protein, the synthesizing enzyme for GABA, and neurotrophins. On the WRAM, both progestin types impaired learning. Further, high-dose MPA impaired delayed memory retention on the WRAM, and exacerbated overnight forgetting on the MM. While neurotrophins were not affected by progesterone or MPA treatment, both progestin types altered GAD levels. MPA significantly and progesterone marginally decreased GAD levels in the hippocampus, and both MPA and progesterone significantly increased GAD levels in the entorhinal cortex. These findings suggest that MPA, the most commonly used progestin in HT, is detrimental to learning and two types of memory, and modulates the GABAergic system in cognitive brain regions, in aged surgically menopausal rats. These findings, combined with in vitro evidence that MPA is detrimental to neuronal health, indicates that MPA has negative effects for brain health and function.


Subject(s)
Contraceptive Agents, Female/adverse effects , Medroxyprogesterone Acetate/adverse effects , Memory Disorders/chemically induced , gamma-Aminobutyric Acid/metabolism , Animals , Female , Hippocampus/drug effects , Memory Disorders/diagnosis , Menopause , Ovariectomy , Rats , Rats, Inbred F344
17.
J Comp Neurol ; 504(6): 716-29, 2007 Oct 20.
Article in English | MEDLINE | ID: mdl-17722035

ABSTRACT

Microglia quickly react to various neurodegenerative processes by producing cytokines and eliminating cellular debris via phagocytosis. These events are also associated with an increased proliferation of microglia, which derive from resident progenitors and those present in the bone marrow. However, it is not clear whether the innate immune response by resident or newly differentiated microglia is beneficial or detrimental to the central nervous system. The aim of this study was to determine the impact of an altered immune response following acute excitotoxicity. Sodium nitroprusside (SNP) or kainic acid (KA) was administered in the brain of various groups of mice, and the extent of neurodegeneration, myelin damage, and inflammation was evaluated within a period of 2 weeks. We used synthetic glucocorticoid (GC), myeloid differentiation factor 88 (MyD88)-deficient mice to suppress nuclear factor kappaB (NF-kappaB) signaling and transgenic mice that express the thymidine kinase (TK) protein under the control of the CD11b promoter to determine the role of proliferating and infiltrating microglia in acute models of brain injury. Neurodegeneration was more extensive in GC-treated and MyD88-deficient mice, suggesting that NF-kappaB signaling and microglia activation are potent neuroprotective mechanisms in the presence of SNP. KA was also highly toxic to neurons of the amygdala in MyD88 knockout mice but not in their WT littermates. Although bone marrow-derived cells are clearly attracted to neurodegenerative areas, preventing their infiltration and differentiation did not affect the extent of SNP-related damage. These data indicate that MyD88/NF-kappaB signaling in resident non-proliferating microglia plays a critical role by restricting damage during acute excitotoxicity.


Subject(s)
Bone Marrow Transplantation/methods , Brain Injuries/pathology , Brain Injuries/prevention & control , Brain Injuries/physiopathology , Microglia/physiology , Animals , Brain Injuries/chemically induced , Disease Models, Animal , Glucocorticoids/administration & dosage , Kainic Acid , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , NF-kappa B/metabolism , Nitroprusside , Stilbamidines , Thymidine Kinase/metabolism , Time Factors
19.
Neuron ; 49(4): 489-502, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16476660

ABSTRACT

Microglia are the immune cells of the brain. Here we show a massive infiltration of highly ramified and elongated microglia within the core of amyloid plaques in transgenic mouse models of Alzheimer's disease (AD). Many of these cells originate from the bone marrow, and the beta-amyloid-40 and -42 isoforms are able to trigger this chemoattraction. These newly recruited cells also exhibit a specific immune reaction to both exogenous and endogenous beta-amyloid in the brain. Creation of a new AD transgenic mouse that expresses the thymidine kinase protein under the control of the CD11b promoter allowed us to show that blood-derived microglia and not their resident counterparts have the ability to eliminate amyloid deposits by a cell-specific phagocytic mechanism. These bone marrow-derived microglia are thus very efficient in restricting amyloid deposits. Therapeutic strategies aiming to improve their recruitment could potentially lead to a new powerful tool for the elimination of toxic senile plaques.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Bone Marrow Cells/physiology , Microglia/physiology , Plaque, Amyloid/pathology , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Bone Marrow Transplantation/methods , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Imaging, Three-Dimensional/methods , Immunohistochemistry/methods , In Situ Hybridization/methods , Indoles , Injections, Intraventricular/methods , Interleukin-1/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Membrane Cofactor Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Microglia/drug effects , Microscopy, Confocal/methods , Peptide Fragments/pharmacology , Phagocytosis/physiology , Plaque, Amyloid/metabolism , Presenilin-1 , RNA, Messenger/metabolism , Time Factors , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Whole-Body Irradiation/methods
20.
Neurol Res ; 27(7): 717-25, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16197808

ABSTRACT

It had long been thought that the central nervous system was isolated from the immune system owing to the blood-brain barrier and that this organ was unable to mount an immune reaction of its own when challenged by invading pathogens. It is now clear that the immune system has a profound impact on the central nervous system, because immune molecules found in the blood stream are able to stimulate cells within the brain. Moreover, recent studies have demonstrated that cells within the central nervous system have the capacity to produce molecules of the innate immune system and that this organ is able to generate a proper immune reaction. This topic has been extensively studied in recent years, and it is becoming clear that the innate immune system is an important modulator of the fate of neurons. Indeed, the precise role(s) of the innate immune response in neurodegenerative diseases is currently under intensive debate. In this review paper, we present evidence either supporting or opposing a role for the innate immune response in these events. The mechanisms by which pathogens interact with the brain and whether such an interaction leads to neurodegenerative disorders are also discussed.


Subject(s)
Brain Diseases/immunology , Animals , Blood-Brain Barrier/immunology , Brain Diseases/etiology , Central Nervous System/immunology , Humans , Immunity, Innate , Nerve Degeneration/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...