Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mov Disord ; 32(10): 1482-1486, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28734065

ABSTRACT

BACKGROUND: Idiopathic REM sleep behavior disorder is a prodromal stage of Parkinson's disease and dementia with Lewy bodies. Hyposmia, reduced dopamine transporter binding, and expression of the brain metabolic PD-related pattern were each associated with increased risk of conversion to PD. The objective of this study was to study the relationship between the PD-related pattern, dopamine transporter binding, and olfaction in idiopathic REM sleep behavior disorder. METHODS: In this cross-sectional study, 21 idiopathic REM sleep behavior disorder subjects underwent 18 F-fluorodeoxyglucose PET, dopamine transporter imaging, and olfactory testing. For reference, we included 18 F-fluorodeoxyglucose PET data of 19 controls, 20 PD patients, and 22 patients with dementia with Lewy bodies. PD-related pattern expression z-scores were computed from all PET scans. RESULTS: PD-related pattern expression was higher in idiopathic REM sleep behavior disorder subjects compared with controls (P = 0.048), but lower compared with PD (P = 0.001) and dementia with Lewy bodies (P < 0.0001). PD-related pattern expression was higher in idiopathic REM sleep behavior disorder subjects with hyposmia and in subjects with an abnormal dopamine transporter scan (P < 0.05, uncorrected). CONCLUSION: PD-related pattern expression, dopamine transporter binding, and olfaction may provide complementary information for predicting phenoconversion. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Fluorodeoxyglucose F18 , Olfaction Disorders/etiology , Positron-Emission Tomography , REM Sleep Behavior Disorder , Tomography, Emission-Computed, Single-Photon , Aged , Analysis of Variance , Cross-Sectional Studies , Female , Humans , Lewy Body Disease/diagnostic imaging , Male , Middle Aged , Olfaction Disorders/diagnostic imaging , Parkinson Disease/diagnostic imaging , Psychiatric Status Rating Scales , REM Sleep Behavior Disorder/complications , REM Sleep Behavior Disorder/diagnostic imaging , REM Sleep Behavior Disorder/metabolism
2.
PLoS Biol ; 12(6): e1001874, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24893313

ABSTRACT

Mechanisms behind how the immune system signals to the brain in response to systemic inflammation are not fully understood. Transgenic mice expressing Cre recombinase specifically in the hematopoietic lineage in a Cre reporter background display recombination and marker gene expression in Purkinje neurons. Here we show that reportergene expression in neurons is caused by intercellular transfer of functional Cre recombinase messenger RNA from immune cells into neurons in the absence of cell fusion. In vitro purified secreted extracellular vesicles (EVs) from blood cells contain Cre mRNA, which induces recombination in neurons when injected into the brain. Although Cre-mediated recombination events in the brain occur very rarely in healthy animals, their number increases considerably in different injury models, particularly under inflammatory conditions, and extend beyond Purkinje neurons to other neuronal populations in cortex, hippocampus, and substantia nigra. Recombined Purkinje neurons differ in their miRNA profile from their nonrecombined counterparts, indicating physiological significance. These observations reveal the existence of a previously unrecognized mechanism to communicate RNA-based signals between the hematopoietic system and various organs, including the brain, in response to inflammation.


Subject(s)
Exosomes/metabolism , Hematopoietic System/metabolism , Inflammation/metabolism , Purkinje Cells/metabolism , RNA, Messenger/metabolism , Animals , Integrases , Mice, Transgenic , Recombination, Genetic
3.
J Neuroinflammation ; 13(1): 272, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27737697

ABSTRACT

BACKGROUND: The majority of investigations on HIV-associated neurocognitive disorders (HAND) neglect the cerebellum in spite of emerging evidence for its role in higher cognitive functions and dysfunctions in common neurodegenerative diseases. METHODS: We systematically investigated the molecular and cellular responses of the cerebellum as contributors to lentiviral infection-induced neurodegeneration, in the simian immunodeficiency virus (SIV)-infected rhesus macaque model for HIV infection and HAND. Four cohorts of animals were studied: non-infected controls, SIV-infected asymptomatic animals, and SIV-infected AIDS-diseased animals with and without brain-permeant antiretroviral treatment. The antiretroviral utilized was 6-chloro-2',3'-dideoxyguanosine (6-Cl-ddG), a CNS-permeable nucleoside reverse transcriptase inhibitor. Quantitation of granule cells and Purkinje cells, of an established biomarker of SIV infection (gp41), of microglial/monocyte/macrophage markers (IBA-1, CD68, CD163), and of the astroglial marker (GFAP) were used to reveal cell-specific cerebellar responses to lentiviral infection and antiretroviral therapy (ART). The macromolecular integrity of the blood brain barrier was tested by albumin immunohistochemistry. RESULTS: Productive CNS infection was observed in the symptomatic stage of disease, and correlated with extensive microglial/macrophage and astrocyte activation, and widespread macromolecular blood brain barrier defects. Signs of productive infection, and inflammation, were reversed upon treatment with 6-Cl-ddG, except for a residual low-grade activation of microglial cells and astrocytes. There was an extensive loss of granule cells in the SIV-infected asymptomatic cohort, which was further increased in the symptomatic stage of the disease and persisted after 6-Cl-ddG (administered after the onset of symptoms of AIDS). In the symptomatic stage, Purkinje cell density was reduced. Purkinje cell loss was likewise unaffected by 6-Cl-ddG treatment at this time. CONCLUSIONS: Our findings suggest that neurodegenerative mechanisms are triggered by SIV infection early in the disease process, i. e., preceding large-scale cerebellar productive infection and marked neuroinflammation. These affect primarily granule cells early in disease, with later involvement of Purkinje cells, indicating differential vulnerability of the two neuronal populations. The results presented here indicate a role for the cerebellum in neuro-AIDS. They also support the conclusion that, in order to attenuate the development of motor and cognitive dysfunctions in HIV-positive individuals, CNS-permeant antiretroviral therapy combined with anti-inflammatory and neuroprotective treatment is indicated even before overt signs of CNS inflammation occur.


Subject(s)
Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , Cerebellum/pathology , HIV Infections/drug therapy , HIV Infections/pathology , Neurons/drug effects , Analysis of Variance , Animals , Antigens, CD/metabolism , Calcium-Binding Proteins , Case-Control Studies , DNA-Binding Proteins/metabolism , Dideoxynucleosides/pharmacology , Dideoxynucleosides/therapeutic use , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/virology , HIV Envelope Protein gp41/metabolism , Humans , Macaca mulatta , Male , Microfilament Proteins , Microglia/drug effects , Microglia/pathology , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/virology , Neurons/metabolism
4.
J Neurochem ; 133(4): 590-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25581060

ABSTRACT

Previously, we demonstrated that systemically injected extracellular domain of neuregulin-1ß1 (Nrg1ß1), a nerve growth and differentiation factor, passes the blood-brain barrier and rescues dopaminergic neurons of substantia nigra in the 6-hydroxydopamine-mouse model of Parkinson's disease (PD). Here, we studied the effects of peripherally administered Nrg1ß1 in another toxin-based mouse model of PD. For this purpose, (i) nigrostriatal pathway injury was induced by treatment of adult wild-type mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in acute and subchronic paradigms; and (ii) Nrg1ß1 or saline (control) were administered 1 h before each MPTP injection. We found that Nrg1ß1 significantly reduced the loss of nigral dopaminergic neurons in both intoxication paradigms (7 days post-injection). However, Nrg1ß1 did not reverse MPTP-induced decrease in dopamine levels and dopaminergic fibers in the striatum. We also show that MPTP conversion to its toxic metabolite 1-methyl-4-phenylpyridinium as well as levels of dopamine transporter, mediating intracellular uptake of 1-methyl-4-phenylpyridinium, are unaffected by Nrg1ß1. Finally, neuroprotective properties of Nrg1ß1 on nigral dopaminergic neurons are specifically mediated by ErbB4 as revealed through the study of ErbB4 knockout mice. In conclusion, systemically administered Nrg1ß1 protects midbrain dopaminergic neurons against this PD-related toxic insult. Thus, Nrg1ß1 may have a benefit in the treatment of PD patients. Previously, we demonstrated that systemically administered neuregulin-1ß1 (Nrg1ß1) passes the blood-brain barrier, phosphorylates ErbB4 receptors and elevates dopamine (DA) levels in the nigrostriatal system of healthy mice. Nrg1ß1 protects nigral DAergic neurons in the 6-hydroxydopamine (6-OHDA) mouse model of Parkinson's disease (PD). Here, we show that Nrg1ß1 rescues nigral DAergic neurons also against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced cell death. ErbB4 expression is essential for the neuroprotective effect of Nrg1ß1 on midbrain DAergic neurons. Nrg1ß1 might be beneficial in PD treatment.


Subject(s)
Dopaminergic Neurons/drug effects , MPTP Poisoning/pathology , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Substantia Nigra/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Animals, Genetically Modified , Disease Models, Animal , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , MPTP Poisoning/chemically induced , Male , Mice , Mice, Inbred C57BL , Neuregulin-1/pharmacology , Neuroprotective Agents/pharmacology , Receptor, ErbB-4/deficiency , Receptor, ErbB-4/genetics , Time Factors
5.
Neurogenetics ; 15(3): 189-92, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24863655

ABSTRACT

Gain-of-function mutations of alpha-synuclein (SNCA) are known to trigger Parkinson's disease (PD) with striatal dopaminergic deficits and a reduction of spontaneous movements. The longest size variant (allele 2) of the complex microsatellite repeat Rep1 within the SNCA gene promoter is known to confer a PD risk. We now observed this Rep1 allele 2 to show significantly decreased frequency in restless legs syndrome (RLS) in a genotyping study of 258 patients versus 235 healthy controls from Germany. Given that RLS is a disease with increased spontaneous movements and with increased striatal dopamine signaling, these novel data appear plausible. The scarcity of this alpha-synuclein gain-of-function variant in RLS might suggest that a low alpha-synuclein function via the SNARE complex in presynaptic vesicle release and neurotransmission of the striatum contributes to RLS pathogenesis.


Subject(s)
Gene Frequency , Parkinson Disease/genetics , Restless Legs Syndrome/genetics , alpha-Synuclein/genetics , Adult , Aged , Aged, 80 and over , Alleles , Humans , Microsatellite Repeats , Middle Aged , Promoter Regions, Genetic
6.
Neuroimage ; 79: 191-200, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23631981

ABSTRACT

A previous clinical trial studied the effect of long-term treatment with levodopa (LD) or the dopamine agonist pramipexole (PPX) on disease progression in Parkinson disease using SPECT with the dopamine transporter (DAT)-radioligand [(123)I]ß-CIT as surrogate marker. [(123)I]ß-CIT binding declined to significantly lower levels in patients receiving LD compared to PPX. However, the interpretation of this difference as LD-induced neurotoxicity, PPX-induced neuroprotection/-regeneration, or only drug-induced regulatory changes of DAT-availability remained controversial. To address this question experimentally, we induced a subtotal lesion of the substantia nigra in mice by bilateral injection of the neurotoxin 6-hydroxydopamine. After 4 weeks, mice were treated for 20 weeks orally with LD (100mg/kg/day) or PPX (3mg/kg/day), or water (vehicle) only. The integrity of nigrostriatal projections was assessed by repeated [(123)I]FP-CIT SPECT in vivo and by immunostaining for DAT and the dopamine-synthesizing enzyme tyrosine hydroxylase (TH) after sacrifice. In sham-lesioned mice, we found that both LD and PPX treatment significantly decreased the striatal FP-CIT binding (LD: -21%; PPX: -14%) and TH-immunoreactivity (LD: -42%; PPX: -45%), but increased DAT-immunoreactivity (LD: +42%; PPX: +33%) compared to controls without dopaminergic treatment. In 6-hydroxydopamine-lesioned mice, however, neither LD nor PPX significantly influenced the stably reduced FP-CIT SPECT signal (LD: -66%; PPX: -66%; controls -66%), TH-immunoreactivity (LD: -70%; PPX: -72%; controls: -77%) and DAT-immunoreactivity (LD: -70%; PPX: -75%; controls: -75%) in the striatum or the number of TH-positive cells in the substantia nigra (LD: -88%; PPX: -88%; controls: -86%), compared to lesioned mice without dopaminergic treatment. In conclusion, chronic dopaminergic stimulation with LD or PPX induced similar adaptive presynaptic changes in healthy mice, but no discernible changes in severely lesioned mice. These findings allow to more reliably interpret the results from clinical trials using neuroimaging of DAT as surrogate parameter.


Subject(s)
Benzothiazoles/administration & dosage , Dopamine Plasma Membrane Transport Proteins/metabolism , Levodopa/administration & dosage , Neuronal Plasticity/drug effects , Parkinson Disease/metabolism , Presynaptic Terminals/metabolism , Tropanes/pharmacokinetics , Animals , Antiparkinson Agents/administration & dosage , Dopamine Agonists/administration & dosage , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Parkinson Disease/drug therapy , Pramipexole , Presynaptic Terminals/drug effects , Radiopharmaceuticals/pharmacokinetics , Tomography, Emission-Computed, Single-Photon/methods , Treatment Outcome
7.
Anal Chem ; 84(7): 3170-8, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22413784

ABSTRACT

Several complementary mass spectrometric imaging techniques allow mapping of various analytes within biological tissue sections. Laser ablation inductively coupled plasma mass spectrometry (LA-ICPMS) quantitatively detects elements and isotopes with very high sensitivity and a particularly high dynamical range. Matrix-assisted laser desorption/ionization ion mobility mass spectrometry (MALDI-IM-MS) allows a pixel-by-pixel classification and identification of biomolecules. In order to dispose of the healthy hemisphere as an internal calibrant in addition to routinely used external standards, adjacent brain sections of mice with a unilateral 6-OHDA lesion of the medial forebrain bundle were chosen as exemplary samples. We demonstrate a comprehensive way of data acquisition and analysis by coregistering mass spectrometric data on photomicrographs as common reference space and thus providing trimodal spatial information. Registering subsequent planar element maps yielded continuous 3-dimensional data sets. Furthermore, we introduce a correction of MSI data for variable slice thickness applicable to all MSI techniques. In the present case, we observed increased concentrations of iron, manganese, and copper in the lesioned substantia nigra while monounsaturated lipid levels were decreased in the identical region of interest. Our techniques provide new insights into the intricate spatial relationship of morphology and chemistry within tissue.


Subject(s)
Mass Spectrometry/methods , Microtechnology/methods , Molecular Imaging/methods , Animals , Lasers , Male , Mice , Optical Phenomena , Oxidopamine/pharmacology , Prosencephalon/drug effects , Prosencephalon/metabolism
8.
J Neurochem ; 117(6): 1066-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21517849

ABSTRACT

Neuregulin-1 (Nrg1) is genetically linked to schizophrenia, a disease caused by neurodevelopmental imbalance in dopaminergic function. The Nrg1 receptor ErbB4 is abundantly expressed on midbrain dopaminergic neurons. Nrg1 has been shown to penetrate blood-brain barrier, and peripherally administered Nrg1 activates ErbB4 and leads to a persistent hyperdopaminergic state in neonatal mice. These data prompted us to study the effect of peripheral administration of Nrg1 in the context of Parkinson's disease, a neurodegenerative disorder affecting the dopaminergic system in the adult brain. We observed that systemic injections of the extracellular domain of Nrg1ß(1) (Nrg1ß(1)-ECD) increased dopamine levels in the substantia nigra and striatum of adult mice. Nrg1ß(1)-ECD injections also significantly protected the mouse nigrostriatal dopaminergic system morphologically and functionally against 6-hydroxydopamine-induced toxicity in vivo. Moreover, Nrg1ß(1)-ECD also protected human dopaminergic neurons in vitro against 6-hydroxydopamine. In conclusion, we have identified Nrg1ß(1)-ECD as a neurotrophic factor for adult mouse and human midbrain dopaminergic neurons with peripheral administratability, warranting further investigation as therapeutic option for Parkinson's disease patients.


Subject(s)
Dopamine/metabolism , Neuregulin-1/therapeutic use , Neurons/drug effects , Parkinson Disease/drug therapy , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology
9.
Neurobiol Dis ; 42(1): 108-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21220019

ABSTRACT

Recent evidence suggests that cyclooxygenases COX1 and COX2 differentially affect brain immunity. Limited data exist about their expressional changes in neurodegenerative diseases such as neuro-AIDS. Here, we analyzed the regulation of non-neuronal COX1/2 expression in rhesus macaque brain during infection with SIV(δ670) and antiretroviral treatment. COX1 was constitutively expressed in microglia and endothelial cells and was not changed in early SIV infection. Late stage of disease was characterized by increased COX1 expression in globally activated microglia, macrophage nodules, infiltrates, and multinucleated giant cells. Endothelial COX1 expression was unaltered. In contrast, COX2 was not expressed in non-neuronal cells in the brain of uninfected and asymptomatically SIV-infected monkeys but was induced in nodule- and syncytium-forming macrophages and in endothelial cells in areas with infiltrates and SIV in monkeys with AIDS. Antiretroviral treatment of AIDS-diseased monkeys with 6-chloro-2',3'-dideoxyguanosine markedly reduced SIV burden, appearance of COX1-positive macrophage nodules, giant cells, and infiltrates, and COX2 induction in the brain. However, the number of COX1-positive diffuse microglia was still increased in antiretrovirally treated animals as compared to uninfected or asymptomatic SIV-infected monkeys. Our data imply that both COX isoforms are differentially regulated and may distinctly modulate local immune responses in the brain during lentiviral disease.


Subject(s)
Brain/enzymology , Brain/virology , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Simian Acquired Immunodeficiency Syndrome/enzymology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus , Animals , Anti-Retroviral Agents/therapeutic use , Brain/pathology , Dideoxynucleosides/therapeutic use , Disease Models, Animal , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy
10.
Neurosci Lett ; 449(3): 240-5, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-18786612

ABSTRACT

Active as well as passive immunization against beta-amlyoid (Abeta) has been proposed as a treatment to lower cerebral amyloid burden and stabilize cognitive decline in Alzheimer's disease (AD). To clarify the mechanism of action underlying passive immunization, the in vivo distribution (and sites of degradation) of peripherally administered radiolabeled human and mouse anti-Abeta antibodies were analyzed in a transgenic mouse model of AD. In APP23 mice, a model in which mutated human amyloid precursor protein is overexpressed, the biodistribution of intravenously applicated (111)indium-conjugated affinity-purified human polyclonal autoantibodies (NAbs-Abeta) was compared to that of monoclonal anti-Abeta(1-17) (6E10), anti-Abeta(17-24) antibodies (4G8) and anti-CD-20 (Rituximab), a non-Abeta targeting control. Blood clearance half-lives were 50+/-6h for Rituximab, 20-30h for NAbs-Abeta, 29+/-5h for 4G8 and 27+/-3h for 6E10. Blood activity was higher for 6E10 at 4h as compared to 4G8, Rituximab and NAbs-Abeta. At the 96h time point, Rituximab had the highest blood activity among the antibodies tested. As expected, all antibodies displayed hepatobiliary clearance. Additionally, NAbs-Abeta was excreted in the urinary tract. Liver and kidney uptake of NAbs-Abeta increased over time and was higher than in the monoclonal antibodies at 48h/96h. The brain-to-blood radioactivity ratio for NAbs-Abeta at later time points (>48h) was higher than that of 6E10, 4G8 and Rituximab. In addition, the distribution varied, with highest values found in the hippocampus. Our data indicate a cerebral accumulation of human NAbs-Abeta in the APP23 model. Further studies with human immunoglobulins and particularly with those that recognize different Abeta-epitopes are required in order to delineate in more detail the mode of action of NAbs-Abeta.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Autoantibodies/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Murine-Derived , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Humans , Indium/pharmacokinetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Rituximab , Time Factors
12.
J Neuropathol Exp Neurol ; 66(10): 901-12, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17917584

ABSTRACT

APOBEC3G restricts retrovirus replication through inducing guanosine-to-adenosine (G-to-A) hypermutations in viral DNA. Its role in brain "intrinsic immunity" has not been elucidated nor has it been convincingly demonstrated which brain cell compartments produce this defense factor in human immunodeficiency virus (HIV) infection, acquired immunodeficiency syndrome (AIDS), and antiretroviral therapy. Here, we investigated by immunohistochemistry and in situ hybridization the cell-specific regulation of APOBEC3G in rhesus macaque brains during infection with simian immunodeficiency virus (SIV) clone deltaB670, a primate model of HIV disease. We found that APOBEC3G protein and mRNA were exclusively expressed by some perivascular macrophages throughout the brain of noninfected and asymptomatic SIV-infected monkeys. Depending on virus burden, APOBEC3G was induced in microglia/macrophage-derived cells and T lymphocytes in late-stage SIV infection. Intracellularly, APOBEC3G was found in the cytoplasm and/or in the nucleus. APOBEC3G-positive cells were in close proximity to SIV gag-positive cells or were SIV-copositive. Induction of APOBEC3G was accompanied by G-to-A hypermutations in the gag and pol regions of retroviral DNA isolated from brain sections of AIDS-symptomatic monkeys. Although brain-directed treatment with antiretroviral 6-chloro-2',3'-dideoxyguanosine suppressed brain SIV burden, encephalitis and reduced cerebral APOBEC3G synthesis hypermutations were still detectable. Upregulation of APOBEC3G may restrict spread of SIV in the brain and thus limit brain damage during lentiviral infection.


Subject(s)
Cytidine Deaminase/genetics , DNA, Viral/genetics , Mutation/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Adenosine/genetics , Animals , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Viral/genetics , Guanine/metabolism , Immunohistochemistry , In Situ Hybridization , Macaca mulatta , RNA Probes , Up-Regulation/genetics
13.
Dis Model Mech ; 10(5): 619-631, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28108469

ABSTRACT

Parkinson's disease (PD) is a frequent neurodegenerative process in old age. Accumulation and aggregation of the lipid-binding SNARE complex component α-synuclein (SNCA) underlies this vulnerability and defines stages of disease progression. Determinants of SNCA levels and mechanisms of SNCA neurotoxicity have been intensely investigated. In view of the physiological roles of SNCA in blood to modulate vesicle release, we studied blood samples from a new large pedigree with SNCA gene duplication (PARK4 mutation) to identify effects of SNCA gain of function as potential disease biomarkers. Downregulation of complexin 1 (CPLX1) mRNA was correlated with genotype, but the expression of other Parkinson's disease genes was not. In global RNA-seq profiling of blood from presymptomatic PARK4 indviduals, bioinformatics detected significant upregulations for platelet activation, hemostasis, lipoproteins, endocytosis, lysosome, cytokine, Toll-like receptor signaling and extracellular pathways. In PARK4 platelets, stimulus-triggered degranulation was impaired. Strong SPP1, GZMH and PLTP mRNA upregulations were validated in PARK4. When analysing individuals with rapid eye movement sleep behavior disorder, the most specific known prodromal stage of general PD, only blood CPLX1 levels were altered. Validation experiments confirmed an inverse mutual regulation of SNCA and CPLX1 mRNA levels. In the 3'-UTR of the CPLX1 gene we identified a single nucleotide polymorphism that is significantly associated with PD risk. In summary, our data define CPLX1 as a PD risk factor and provide functional insights into the role and regulation of blood SNCA levels. The new blood biomarkers of PARK4 in this Turkish family might become useful for PD prediction.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Biomarkers/blood , Genetic Predisposition to Disease , Lewy Body Disease/blood , Nerve Tissue Proteins/genetics , Parkinson Disease/genetics , REM Sleep Behavior Disorder/blood , RNA/blood , alpha-Synuclein/deficiency , Female , Heterozygote , Humans , Lewy Body Disease/genetics , Middle Aged , Parkinson Disease/blood , REM Sleep Behavior Disorder/physiopathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , alpha-Synuclein/blood , alpha-Synuclein/genetics
14.
J Neuropathol Exp Neurol ; 65(12): 1170-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17146291

ABSTRACT

Existing data concerning the role of the delta-chemokine fractalkine (CX3CL1) and its receptor (CX3CR1) in lentivirus-induced encephalitis are limited and controversial. We explored, by quantitative in situ hybridization and immunohistochemistry, the cell-specific changes of CX3CL1 and CX3CR1 in rhesus macaque brain during simian immunodeficiency virus (SIV) infection and antiretroviral treatment. Neuronal expression of CX3CL1 was significantly reduced in cortex and striatum of AIDS-diseased monkeys as compared with uninfected and asymptomatic SIV-infected monkeys. CX3CL1 mRNA was increased in some endothelial cells and newly induced in astrocytes and macrophages focally in areas of SIV burden and inflammatory infiltrates. In most CX3CL1-positive astrocytes and macrophages, the transcription factor NF-kappaB was translocated to the nucleus. CX3CR1 was upregulated in scattered, nodule, and giant cell-forming microglia/macrophages and mononuclear infiltrates close to CX3CL1-induced cells in the brain. Treatment of AIDS monkeys with the central nervous system-permeant 6-chloro-2',3'-dideoxyguanosine fully reversed SIV burden, productive inflammation, nuclear NF-kappaB translocation as well as focal induction of CX3CL1 in astrocytes and macrophages and downregulation in neurons. In contrast, diffuse CX3CR1-positive microgliosis and GFAP-positive astrogliosis were partially reversed by 6-chloro-2',3'-dideoxyguanosine. Thus, focally induced CX3CL1 may be a target for therapeutic intervention to limit ongoing inflammatory infiltration into brain in lentivirus infection.


Subject(s)
Chemokines, CX3C/metabolism , Dideoxynucleosides/therapeutic use , Encephalitis, Viral/immunology , Membrane Proteins/metabolism , Receptors, Chemokine/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Animals , Anti-Retroviral Agents/therapeutic use , Astrocytes/immunology , Astrocytes/pathology , Astrocytes/virology , Brain/immunology , Brain/pathology , Brain/virology , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Chemokines, CX3C/genetics , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Disease Progression , Encephalitis, Viral/complications , Encephalitis, Viral/drug therapy , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelial Cells/virology , Gene Expression Regulation/drug effects , Gliosis/drug therapy , Gliosis/immunology , Gliosis/physiopathology , Immunohistochemistry , In Situ Hybridization , Macaca mulatta , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Membrane Proteins/genetics , NF-kappa B/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Chemokine/genetics , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Treatment Outcome , Viral Load
15.
Neurosci Lett ; 400(3): 187-90, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16650578

ABSTRACT

Alpha2-macroglobulin (alpha2M) as well as its receptor, the low-density lipoprotein receptor-related (LRP) and the receptor-associated protein (RAP) are involved in the clearance of cerebral A beta. Current evidence suggests that polymorphisms in the genes of alpha2M, LRP and RAP may have functional effects on the proteins. Two independent association samples of 271 AD patients and 280 representative controls were investigated whether the risk for developing AD is altered in carriers of polymorphisms in the alpha2M-gene (Va1000Ile), in the LRP-gene (Ala216Val) and in the RAP-gene (Val311Met). Genotypes were determined by standard PCR and restriction fragment length polymorphism. The results were adjusted for age, gender and apolipoprotein E-epsilon4 (APOE) polymorphism. Inheritance of alpha2M conferred a small increased risk for sporadic AD with an estimated Mantel-Haenszel odds ratio of 1.47. There was no age- or gender-dependent increase in alpha2M Val1000Ile allele frequencies in AD patients compared to controls. There was no significant difference in the allele frequencies among control and AD subjects for the LRP and RAP polymorphisms. We found no evidence of an interaction between the alpha2M and RAP or LRP with regard to conferred risk. Our data suggest that the alpha2M Val1000Ile polymorphism is weakly associated with AD. Although LRP as well as RAP seem to play an essential role in the metabolism of alpha2M and APOE, there is no increase in the genetic risk for AD in patients carrying the investigated polymorphisms.


Subject(s)
Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Risk Assessment/methods , alpha-Macroglobulins/genetics , Aged , Aged, 80 and over , Cohort Studies , Female , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Germany/epidemiology , Humans , Incidence , Male , Middle Aged , Polymorphism, Genetic , Risk Factors , Sex Distribution
16.
Neuroscience ; 331: 120-33, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27320210

ABSTRACT

l-3,4-Dihydroxyphenylalanine (l-DOPA) is the therapeutic gold standard in Parkinson's disease. However, most patients develop debilitating abnormal involuntary movements termed l-DOPA-induced dyskinesia (LID) as therapy-complicating side effects. The underlying mechanisms of LID pathogenesis are still not fully understood. Recent evidence suggests an involvement of striatal tyrosine hydroxylase (TH) protein-expressing neurons, as they are capable of endogenously producing l-DOPA and possibly dopamine. The aim of this study was to elucidate changes of TH transcription in the striatum and nucleus accumbens that occur under experimental conditions of LID. Mice with a unilateral 6-hydroxydopamine-induced lesion of the medial forebrain bundle were treated daily with l-DOPA for 15days to provoke dyskinesia. In situ hybridization analysis revealed a significant numerical decrease of TH mRNA-positive neurons in the striatum and nucleus accumbens of mice not exhibiting LID, whereas dyskinetic animals failed to show this reduction of TH transcription. Interestingly, similar changes were observed in intact non-deafferentiated striata, demonstrating an l-DOPA-responsive transcriptional TH regulation independently from nigrostriatal lesion severity. Consolidation with our previous study on TH protein level (Keber et al., 2015) impressively highlights that LID is associated with both a deficient downregulation of TH transcription and an excessive translation of TH protein in intrastriatal neurons. As TH protein levels in comparison to mRNA levels showed a stronger correlation with development and severity of LID, antidyskinetic treatment strategies should focus on translational and posttranslational modulations of TH as a promising target.


Subject(s)
Antiparkinson Agents/adverse effects , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Neurons/drug effects , Tyrosine 3-Monooxygenase/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Down-Regulation/drug effects , Dyskinesia, Drug-Induced/pathology , Enkephalins/metabolism , Male , Medial Forebrain Bundle , Mice, Inbred C57BL , Neuronal Plasticity , Neurons/metabolism , Neurons/pathology , Oxidopamine , Protein Precursors/metabolism , RNA, Messenger/metabolism
17.
J Histochem Cytochem ; 52(6): 779-88, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15150286

ABSTRACT

Monoamine storage in secretory granules is mediated by the vesicular monoamine transporters 1 and 2 (VMAT1 and VMAT2). The aim of our study was to identify monoamine-handling normal and neoplastic inflammatory cells in the skin by their expression of VMAT1 and VMAT2. Normal skin from various parts of the body, as well as 21 cases of cutaneous mastocytosis and 10 cases of cutaneous Langerhans cell histiocytosis were analyzed by immunohistochemistry, radioactive in situ hybridization, and double-fluorescence confocal microscopy. VMAT2-positive cells in the subepidermal layer were identified as mast cells by their expression of tryptase. Neoplastic mast cells in all cases of cutaneous mastocytosis retained their VMAT2 positivity. The intraepidermal VMAT2-expressing cells were identified as Langerhans cells by their CD1a positivity. VMAT2 was absent from Langerhans cell histiocytosis. VMAT2 is an excellent marker for normal and neoplastic mast cells. The expression of VMAT2 demonstrates the capacity of mast cells for monoamine storage and handling. The presence of VMAT2 in epidermal Langerhans cells revealed a previously unrecognized monoamine-handling phenotype of these cells and indicates possible involvement of amine storage and release associated with antigen presentation. Absence of VMAT2 in neoplastic Langerhans cells indicates a loss of monoamine handling capacity of these cells during tumorigenesis.


Subject(s)
Histiocytosis/metabolism , Langerhans Cells/metabolism , Mast Cells/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins , Neuropeptides , Skin Neoplasms/metabolism , Skin/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Child , Child, Preschool , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mastocytosis, Cutaneous/metabolism , Membrane Glycoproteins/genetics , Microscopy, Confocal , Middle Aged , RNA, Messenger/biosynthesis , Skin/cytology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
18.
Neurosci Lett ; 342(1-2): 132-4, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12727335

ABSTRACT

There is increasing evidence that immune mechanisms are involved in the pathogenesis of Alzheimer's disease (AD). Recently, polymorphisms of the interleukin (IL)-1 and IL-6 genes were found to be associated with late-onset AD. The immunoregulatory IL-10 downregulates synthesis of pro-inflammatory cytokines such as IL-1. Current evidence suggests that some polymorphisms in the IL-10 promoter may have functional effects on IL-10 transcription. A total of 406 German AD patients (mean age 70.2+/-10.0 years, range 50-95 years, 42% female) and 251 unrelated non-demented control subjects (mean age 66.8+/-10.6 years, range 50-93 years, 38% female) were investigated for the presence of three polymorphisms in the IL-10 promoter region (-1087A/G, -824C/T, -597C/A). No significant differences in the allelic distribution of the analyzed IL-10 polymorphisms have been found between AD patients and controls. We conclude that polymorphisms in the IL-10 promoter region do not increase the risk of developing AD.


Subject(s)
Alzheimer Disease/genetics , Interleukin-10/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Down-Regulation , Female , Genotype , Humans , Male , Middle Aged , Risk Factors
19.
Neurosci Lett ; 359(3): 195-7, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15050696

ABSTRACT

An increasing number of studies have shown that an inflammatory process is part of Parkinson's disease (PD) brain pathology. Interleukin-1 (IL-1) is a multifunctional cytokine and is considered to contribute to several inflammatory diseases. Recently, we detected an associated risk in a subgroup of PD patients with a disease onset < 50 years and a C to T transition in the IL-1alpha promoter (-889). One-hundred-seventy-six German PD patients (42.1 +/- 6.4 years; 42.4% male) and 170 unrelated age-matched control individuals (40.4 +/- 8.7 years; 57.6% male) were investigated for the presence of the IL-1alpha (-889C/T) polymorphism. No significant difference in the allelic distribution of the analyzed IL-1alpha polymorphism has been found between PD and controls. We conclude that the C/T polymorphism in the IL-1alpha promoter region at -889 does not increase the risk to develop PD.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Interleukin-1/genetics , Parkinson Disease/epidemiology , Parkinson Disease/genetics , Polymorphism, Genetic , Risk Assessment/methods , Adult , Case-Control Studies , Female , Genetic Testing , Germany/epidemiology , Humans , Male , Middle Aged , Statistics as Topic
20.
J Chem Neuroanat ; 56: 35-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24553202

ABSTRACT

Neurons partially expressing individual enzymes of dopamine (DA) biosynthesis, e.g. tyrosine hydroxylase (TH) or aromatic acid decarboxylase, are found in different areas of the central nervous system, continuously or transiently in normal and pathological conditions. This current study analyzed if TH neurons exist in target areas of ventral midbrain dopaminergic neurons and how they react to dopaminergic denervation. High power analysis of brain tissue sections revealed that TH-immunopositive neurons were present in striatum, accumbens and cortex - and several other brain areas - of healthy adult mice. DAergic denervation induced by stereotaxic injection of 6-hydroxydopamine into the medial forebrain bundle increased the number of TH expressing neurons in the striatum, accumbens and the cortex, up 40 d later. These TH neurons were not stained by specific antibodies recognizing TH phosphorylated at serine residues 19, 31 and 40, dopamine transporter and vesicular monoamine transporter type 2, but most of them expressed dopamine and cyclic AMP-regulated phosphoprotein 32kDa. Thus, mouse striatum, accumbens and cortex contain neurons which express TH with low activity, and their number increases following dopamine depletion.


Subject(s)
Brain/cytology , Neurons/cytology , Animals , Blotting, Western , Brain/metabolism , Denervation , Dopamine/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Phosphorylation , Serine/metabolism , Tyrosine 3-Monooxygenase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL