Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
bioRxiv ; 2024 Feb 11.
Article in English | MEDLINE | ID: mdl-37398030

ABSTRACT

Paradoxically, cigarette smoking is associated with a reduced risk of Parkinson's disease (PD). This led us to hypothesize that carbon monoxide (CO) levels, which are constitutively but modestly elevated in smokers, might contribute to neuroprotection. Using rodent models of PD based on α-synuclein (αSyn) accumulation and oxidative stress, we show that low-dose CO mitigates neurodegeneration and reduces αSyn pathology. Oral CO administration activated signaling cascades mediated by heme oxygenase-1 (HO-1), which have been implicated in limiting oxidative stress, and in promoting αSyn degradation, thereby conferring neuroprotection. Consistent with a neuroprotective effect of smoking, HO-1 levels in cerebrospinal fluid were higher in human smokers compared to nonsmokers. Moreover, in PD brain samples, HO-1 levels were higher in neurons without αSyn pathology. Thus, CO in rodent PD models reduces pathology and increases oxidative stress responses, phenocopying possible protective effects of smoking evident in PD patients. These data highlight the potential for low-dose CO modulated pathways to slow symptom onset and limit pathology in PD patients.

2.
Neuroscience ; 322: 129-37, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26905951

ABSTRACT

Considerable epidemiological and laboratory data have suggested that caffeine, a nonselective adenosine receptor antagonist, may protect against the underlying neurodegeneration of parkinson's disease (PD). Although both caffeine and more specific antagonists of the A2A subtype of adenosine receptor (A2AR) have been found to confer protection in animal models of PD, the dependence of caffeine's neuroprotective effects on the A2AR is not known. To definitively determine its A2AR dependence, the effect of caffeine on 1-methyl-4-phenyl-1,2,3,6 tetra-hydropyridine (MPTP) neurotoxicity was compared in wild-type (WT) and A2AR gene global knockout (A2A KO) mice, as well as in central nervous system (CNS) cell type-specific (conditional) A2AR knockout (cKO) mice that lack the receptor either in postnatal forebrain neurons or in astrocytes. In WT and in heterozygous A2AR KO mice caffeine pretreatment (25mg/kgip) significantly attenuated MPTP-induced depletion of striatal dopamine. By contrast in homozygous A2AR global KO mice caffeine had no effect on MPTP toxicity. In forebrain neuron A2AR cKO mice, caffeine lost its locomotor stimulant effect, whereas its neuroprotective effect was mostly preserved. In astrocytic A2AR cKO mice, both caffeine's locomotor stimulant and protective properties were undiminished. Taken together, these results indicate that neuroprotection by caffeine in the MPTP model of PD relies on the A2AR, although the specific cellular localization of these receptors remains to be determined.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Caffeine/pharmacology , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , Neuroprotective Agents/pharmacology , Receptor, Adenosine A2A/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Prosencephalon/drug effects , Prosencephalon/metabolism , Receptor, Adenosine A2A/genetics
3.
Neuroscience ; 274: 242-9, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24880154

ABSTRACT

Inosine (hypoxanthine 9-beta-D-ribofuranoside), a purine nucleoside with multiple intracellular roles, also serves as an extracellular modulatory signal. On neurons, it can produce anti-inflammatory and trophic effects that confer protection against toxic influences in vivo and in vitro. The protective effects of inosine treatment might also be mediated by its metabolite urate. Urate in fact possesses potent antioxidant properties and has been reported to be protective in preclinical Parkinson's disease (PD) studies and to be an inverse risk factor for both the development and progression of PD. In this study we assessed whether inosine might protect rodent MES 23.5 dopaminergic cell line from oxidative stress in a cellular model of PD, and whether its effects could be attributed to urate. MES 23.5 cells cultured alone or in presence of enriched murine astroglial cultures MES 23.5-astrocytes co-cultures were pretreated with inosine (0.1-100 µM) for 24 h before addition of the oxidative stress inducer H2O2 (200 µM). Twenty-four hours later, cell viability was quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay or immunocytochemistry in pure and MES 23.5-astrocytes co-cultures, respectively. H2O2-toxic effect on dopaminergic cells was reduced when they were cultured with astrocytes, but not when they were cultured alone. Moreover, in MES 23.5-astrocytes co-cultures, indicators of free radical generation and oxidative damage, evaluated by nitrite (NO2(-)) release and protein carbonyl content, respectively, were attenuated. Conditioned medium experiments indicated that the protective effect of inosine relies on the release of a protective factor from inosine-stimulated astrocytes. Purine levels were measured in the cellular extract and conditioned medium using high-performance liquid chromatography (HPLC) method. Urate concentration was not significantly increased by inosine treatment however there was a significant increase in levels of other purine metabolites, such as adenosine, hypoxanthine and xanthine. In particular, in MES 23.5-astrocytes co-cultures, inosine medium content was reduced by 99% and hypoxanthine increased by 127-fold. Taken together these data raise the possibility that inosine might have a protective effect in PD that is independent of any effects mediated through its metabolite urate.


Subject(s)
Antiparkinson Agents/pharmacology , Inosine/pharmacology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Astrocytes/drug effects , Astrocytes/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Free Radicals/metabolism , Hydrogen Peroxide , Mice , Mice, Inbred C57BL , Nitrites/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Parkinsonian Disorders/physiopathology , Protein Carbonylation/drug effects , Protein Carbonylation/physiology , Purines/metabolism , Rats , Uric Acid/metabolism
4.
Neurology ; 78(15): 1138-45, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22491871

ABSTRACT

OBJECTIVE: To prospectively examine whether higher intakes of total flavonoids and their subclasses (flavanones, anthocyanins, flavan-3-ols, flavonols, flavones, and polymers) were associated with a lower risk of developing Parkinson disease (PD). METHODS: In the current analysis, we included 49,281 men in the Health Professional Follow-up Study and 80,336 women from the Nurses' Health Study. Five major sources of flavonoid-rich foods (tea, berry fruits, apples, red wine, and orange/orange juice) were also examined. Flavonoid intake was assessed using an updated food composition database and a validated food frequency questionnaire. RESULTS: We identified 805 participants (438 men and 367 women) who developed PD during 20-22 years of follow-up. In men, after adjusting for multiple confounders, participants in the highest quintile of total flavonoids had a 40%lower PD risk than those in the lowest quintile (hazard ratio [HR] = 0.60; 95% confidence interval 0.43, 0.83; p trend = 0.001). No significant relationship was observed in women (p trend = 0.62) or in pooled analyses (p trend = 0.23). In the pooled analyses for the subclasses, intakes of anthocyanins and a rich dietary source, berries, were significantly associated with a lower PD risk (HR comparing 2 extreme intake quintiles were 0.76 for anthocyanins and 0.77 for berries, respectively; p trend < 0.02 for both). CONCLUSIONS: Our findings suggest that intake of some flavonoids may reduce PD risk, particularly in men, but a protective effect of other constituents of plant foods cannot be excluded.


Subject(s)
Feeding Behavior , Flavonoids/administration & dosage , Parkinson Disease/epidemiology , Parkinson Disease/prevention & control , Adult , Aged , Anthocyanins/administration & dosage , Beverages , Citrus sinensis , Confounding Factors, Epidemiologic , Female , Flavanones/administration & dosage , Flavones/administration & dosage , Flavonols/administration & dosage , Follow-Up Studies , Fruit , Health Personnel/statistics & numerical data , Humans , Incidence , Male , Malus , Middle Aged , Odds Ratio , Prospective Studies , Risk Assessment , Surveys and Questionnaires , Tea , Wine
5.
Neuroscience ; 167(2): 475-81, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20167258

ABSTRACT

Epidemiological studies have raised the possibility of caffeine serving as a neuroprotective agent in Parkinson's disease (PD). This possibility has gained support from findings that dopaminergic neuron toxicity induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or other neurotoxins is attenuated by co-administration of caffeine in mice. Here we examined the time window of caffeine's neuroprotection as well as the effects of caffeine's metabolites (theophylline and paraxanthine) in the MPTP mouse model of PD. In the first experiment, caffeine pre-treatment (30 mg/kg ip) significantly attenuated MPTP-induced striatal dopamine depletion when it was given 10 min, 30 min, 1 h, or 2 h but not 6 h before MPTP (40 mg/kg ip) treatment. Meanwhile, caffeine post-treatment also significantly attenuated striatal dopamine loss when it was given 10 min, 30 min, 1 h or 2 h but not 4 h, 8 h or 24 h after MPTP injection. In the second experiment, both theophylline (10 or 20 mg/kg) and paraxanthine (10 or 30 mg/kg) administration (10 min before MPTP) significantly attenuated MPTP-induced dopamine depletion in mice, as did caffeine (10 mg/kg) treatment. Thus the metabolites of caffeine also provide neuroprotective effects in this mouse model of PD. The data suggest that if caffeine protects against putative toxin-induced dopaminergic neuron injury in humans, then precise temporal pairing between caffeine and toxin exposures may not be critical because the duration of neuroprotection by caffeine may be extended by protective effects of its major metabolites.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Caffeine/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/prevention & control , Theophylline/pharmacology , Animals , Caffeine/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Time Factors
6.
Neurology ; 73(16): 1286-91, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19841380

ABSTRACT

BACKGROUND: Co-occurrence of Parkinson disease (PD) and melanoma has been reported in numerous studies. If this was due to common genetic mechanisms, a positive family history of melanoma would be associated with an excessive PD risk, independent of environmental risk factors for PD. METHODS: We prospectively examined associations between a family history of melanoma and PD among 157,036 men and women free of PD at baseline (1990 for men and 1982 for women) who participated in 2 ongoing US cohorts: the Health Professional Follow-up Study and the Nurses' Health Study. Information on family history of melanoma in parents or siblings was assessed via questionnaire. Relative risks and 95% confidence intervals were estimated using Cox proportional hazards models and pooled using a fixed-effects model. RESULTS: During 14-20 years follow-up, we identified 616 incident PD cases. A family history of melanoma in a first-degree relative was associated with a higher risk of PD (multivariate relative risk = 1.85; 95% confidence interval: 1.2, 2.8; p = 0.004), after adjusting for smoking, ethnicity, caffeine intake, and other covariates. In contrast, we did not observe significant associations between a family history of colorectal, lung, prostate, or breast cancer and PD risk. Interactions between melanoma family history and age, smoking, or caffeine intake were not significant and subgroup analyses according to these factors generated similar results. CONCLUSIONS: Our findings support the notion that melanoma and Parkinson disease (PD) share common genetic components. The genetic determinants of melanoma could therefore be explored as susceptibility candidate genes for PD.


Subject(s)
Melanoma/epidemiology , Parkinson Disease/epidemiology , Family , Female , Follow-Up Studies , Genetic Predisposition to Disease , Humans , Male , Melanoma/genetics , Middle Aged , Multivariate Analysis , Parkinson Disease/genetics , Proportional Hazards Models , Prospective Studies , Risk , Risk Factors , Surveys and Questionnaires
7.
J Neurochem ; 104(1): 279-86, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18005343

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a member of neurotrophin family, enhances synaptic transmission and regulates neuronal proliferation and survival. Both BDNF and its tyrosine kinase receptors (TrkB) are highly expressed in the hippocampus, where an interaction with adenosine A(2A) receptors (A(2A)Rs) has been recently reported. In the present paper, we evaluated the role of A(2A)Rs in mediating functional effects of BDNF in hippocampus using A(2A)R knock-out (KO) mice. In hippocampal slices from WT mice, application of BDNF (10 ng/mL) increased the slope of excitatory post-synaptic field potentials (fEPSPs), an index of synaptic facilitation. This increase of fEPSP slope was abolished by the selective A(2A) antagonist ZM 241385. Similarly, genetic deletion of the A(2A)Rs abolished BDNF-induced increase of the fEPSP slope in slices from A(2A)R KO mice The reduced functional ability of BDNF in A(2A)R KO mice was correlated with the reduction in hippocampal BDNF levels. In agreement, the pharmacological blockade of A(2)Rs by systemic ZM 241385 significantly reduced BDNF levels in the hippocampus of normal mice. These results indicate that the tonic activation of A(2A)Rs is required for BDNF-induced potentiation of synaptic transmission and for sustaining a normal BDNF tone in the hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Hippocampus/physiology , Receptor, Adenosine A2A/physiology , Synaptic Transmission/drug effects , Adenosine A2 Receptor Antagonists , Animals , Dose-Response Relationship, Drug , Drug Interactions , Enzyme-Linked Immunosorbent Assay/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Mice , Mice, Knockout , Patch-Clamp Techniques/methods , Receptor, Adenosine A2A/deficiency , Triazines/pharmacology , Triazoles/pharmacology
8.
Am J Epidemiol ; 166(5): 561-7, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17584757

ABSTRACT

Oxidative stress contributes to dopaminergic neuron degeneration in Parkinson's disease. Urate, a potent antioxidant, could be neuroprotective. To determine whether higher plasma concentrations of urate predict a reduced risk of Parkinson's disease, the authors conducted a nested case-control study among participants in the Health Professionals Follow-up Study, a cohort comprising over 18,000 men who provided blood samples in 1993-1995. Eighty-four incident cases of Parkinson's disease were diagnosed through 2000, and each was randomly matched to two controls by year of birth, race, and time of blood collection. Rate ratios of Parkinson's disease according to quartile of uricemia were estimated by use of conditional logistic regression. The mean urate concentration was 5.7 mg/dl among cases and 6.1 mg/dl among controls (p = 0.01). After adjustment for age, smoking, and caffeine, the rate ratio of Parkinson's disease for the highest quartile of uricemia compared with the lowest was 0.43 (95% confidence interval: 0.18, 1.02; p(trend) = 0.017). This association was stronger in analyses excluding cases diagnosed within 4 years (median) from blood collection (rate ratio = 0.17, 95% confidence interval: 0.04, 0.69; p(trend) = 0.010). These results suggest that high plasma urate concentrations may decrease the risk of Parkinson's disease, and they raise the possibility that interventions to increase plasma urate may reduce the risk and delay the progression of Parkinson's disease.


Subject(s)
Parkinson Disease/blood , Uric Acid/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Case-Control Studies , Humans , Incidence , Logistic Models , Male , Middle Aged , Oxidative Stress , Parkinson Disease/epidemiology , Prospective Studies , Risk Factors
9.
Neurology ; 68(10): 764-8, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17339584

ABSTRACT

OBJECTIVE: To characterize further the relationship between smoking history and Parkinson disease (PD) risk by considering temporal and qualitative features of smoking exposure, including duration, average intensity, and recentness, as well as the relative importance of smoking during different periods of life. METHODS: We prospectively assessed incident PD from 1992 to 2001 among 79,977 women and 63,348 men participating in the Cancer Prevention Study II Nutrition Cohort, according to their cigarette smoking status and lifetime smoking histories. RESULTS: During follow-up, 413 participants had definite or probable PD confirmed by their treating neurologists or medical record review. Compared with never smokers, former smokers had a relative risk (RR) of 0.78 (95% CI 0.64 to 0.95) and current smokers had an RR of 0.27 (95% CI 0.13 to 0.56). On average, participants with more years smoked, more cigarettes per day, older age at quitting smoking, and fewer years since quitting smoking had lower PD risk. The relative risks and trends did not vary significantly by sex. The cumulative incidence of PD was lowest among participants who quit smoking at later ages. A 30% to 60% decreased risk of PD was apparent for smoking as early as 15 to 24 years before symptom onset, but not for smoking 25 or more years before onset. CONCLUSIONS: The lower risk of Parkinson disease among current and former smokers varied with smoking duration, intensity, and recentness. The dependence of this association on the timing of smoking during life is consistent with a biologic effect.


Subject(s)
Parkinson Disease/epidemiology , Parkinson Disease/etiology , Risk , Smoking/adverse effects , Adult , Age Factors , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Incidence , Male , Middle Aged , Odds Ratio , Prevalence , Proportional Hazards Models , Prospective Studies , Retrospective Studies , Risk Factors , Sex Factors
10.
Arthritis Rheum ; 54(8): 2632-42, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16871530

ABSTRACT

OBJECTIVE: Adenosine regulates inflammation and tissue repair, and adenosine A2A receptors promote wound healing by stimulating collagen matrix production. We therefore examined whether adenosine A2A receptors contribute to the pathogenesis of dermal fibrosis. METHODS: Collagen production by primary human dermal fibroblasts was analyzed by real-time polymerase chain reaction, 14C-proline incorporation, and Sircol assay. Intracellular signaling for dermal collagen production was investigated using inhibitors of MEK-1 and by demonstration of ERK phosphorylation. In vivo effects were studied in a bleomycin-induced dermal fibrosis model using adenosine A2A receptor-deficient wild-type littermate mice, C57BL/6 mice, and mice treated with adenosine A2A receptor antagonist. Morphometric features and levels of hydroxyproline were determined as measures of dermal fibrosis. RESULTS: Adenosine A2A receptor occupancy promoted collagen production by primary human dermal fibroblasts, which was blocked by adenosine A2A, but not A1 or A2B, receptor antagonism. Adenosine A2A receptor ligation stimulated ERK phosphorylation, and A2A receptor-mediated collagen production by dermal fibroblasts was blocked by MEK-1 inhibitors. Adenosine A2A receptor-deficient and A2A receptor antagonist-treated mice were protected from developing bleomycin-induced dermal fibrosis. CONCLUSION: These results demonstrate that adenosine A2A receptors play an active role in the pathogenesis of dermal fibrosis and suggest a novel therapeutic target in the treatment and prevention of dermal fibrosis in diseases such as scleroderma.


Subject(s)
Dermis/metabolism , Fibroblasts/metabolism , Fibrosis/metabolism , Receptor, Adenosine A2A/metabolism , Scleroderma, Diffuse/metabolism , Animals , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Dermis/drug effects , Dermis/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis/pathology , Fibrosis/prevention & control , Gene Expression , Humans , Hydroxyproline/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Scleroderma, Diffuse/chemically induced , Scleroderma, Diffuse/pathology , Scleroderma, Diffuse/prevention & control , Triazines/therapeutic use , Triazoles/therapeutic use
11.
J Neurochem ; 95(4): 1188-200, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16271052

ABSTRACT

Hippocampal metabotropic glutamate 5 receptors (mGlu5Rs) regulate both physiological and pathological responses to glutamate. Because mGlu5R activation enhances NMDA-mediated effects, and given the role played by NMDA receptors in synaptic plasticity and excitotoxicity, modulating mGlu5R may influence both the physiological and the pathological effects elicited by NMDA receptor stimulation. We evaluated whether adenosine A2A receptors (A(2A)Rs) modulated mGlu5R-dependent effects in the hippocampus, as they do in the striatum. Co-application of the A(2A)R agonist CGS 21680 with the mGlu5R agonist (RS)-2-chloro-s-hydroxyphenylglycine(CHPG) synergistically reduced field excitatory postsynaptic potentials in the CA1 area of rat hippocampal slices. Endogenous tone at A(2A)Rs seemed to be required to enable mGlu5R-mediated effects, as the ability of CHPG to potentiate NMDA effects was antagonized by the selective A(2A)R antagonist ZM 241385 in rat hippocampal slices and cultured hippocampal neurons, and abolished in the hippocampus of A(2A)R knockout mice. Evidence for the interaction between A(2A)Rs and mGlu5Rs was further strengthened by demonstrating their co-localization in hippocampal synapses. This is the first evidence showing that hippocampal A(2A)Rs and mGlu5Rs are co-located and act synergistically, and that A(2A)Rs play a permissive role in mGlu5R receptor-mediated potentiation of NMDA effects in the hippocampus.


Subject(s)
Hippocampus/cytology , N-Methylaspartate/pharmacology , Neurons/metabolism , Receptors, Adenosine A2/physiology , Receptors, Metabotropic Glutamate/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Animals , Bicuculline/pharmacology , Blotting, Western/methods , Colforsin/pharmacology , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Female , Fluorescent Antibody Technique/methods , GABA Antagonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Patch-Clamp Techniques/methods , Phenethylamines/pharmacology , Phenylacetates/pharmacology , Pregnancy , Presynaptic Terminals/metabolism , Pyridines/pharmacology , Qa-SNARE Proteins/metabolism , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
12.
Neurology ; 64(4): 664-9, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15728289

ABSTRACT

OBJECTIVE: To investigate whether greater physical activity is associated with a lower risk of Parkinson disease (PD). METHODS: The authors prospectively followed 48,574 men and 77,254 women who provided information on physical activity in 1986 or in early adulthood. During the follow-up, a total of 252 (male) and 135 (female) incident PD cases were identified. RESULTS: In men, greater baseline physical activity was associated with a lower PD risk; compared with the lowest quintile, the multivariate relative risk (RR) of PD for the highest quintile was 0.7 (95% CI 0.5 to 1.1; p value, test for trend = 0.007), and the inverse association was still present after excluding the first 10 years of follow-up (RR = 0.5; p value, test for trend = 0.02). Further, strenuous exercise in early adult life was also inversely related to PD risk in men: compared with men who regularly exercised < or =2 months/year, those with > or =10 months of strenuous exercise had a 60% lower PD risk (RR = 0.4; p value, test for trend = 0.005). In women, physical activity assessed at baseline was not related to PD risk, whereas strenuous exercise in early adulthood tended to be inversely related to PD risk later in life (highest vs lowest categories, RR = 0.5, 95% CI 0.2 to 1.4; p value, test for trend = 0.06). CONCLUSION: This study suggests either that higher levels of physical activity may lower the risk of Parkinson disease (PD) in men or that men predisposed to PD tend to avoid strenuous physical activity in their early adult years.


Subject(s)
Motor Activity , Parkinson Disease/epidemiology , Adult , Aged , Boston/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Risk , Smoking/epidemiology , Sports/statistics & numerical data , Surveys and Questionnaires
13.
Brain Res ; 1011(1): 7-13, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15140640

ABSTRACT

The non-toxic neuronal binding domain of tetanus toxin (tetanus toxin fragment C, TTC) has been used as a vector to enhance delivery of potentially therapeutic proteins to motor neurons from the periphery following an intramuscular injection. The unique binding and transport properties of this 50-kDa polypeptide suggest that it might also enhance delivery of proteins to neurons after direct injection into the CNS. Using quantitative fluorimetry, we found that labeled TTC showed vastly superior retention within brain tissue after intracerebral injection compared to a control protein (bovine serum album). Fluorescence microscopy revealed that injected TTC was not retained solely in a restricted deposit along the needle track, but was distributed through gray matter in a pattern not previously described. The distribution of injected protein within the extracellular space of the gray matter and neuropil was also seen after injection of a recombinant fusion protein comprised of TTC linked to the enzyme superoxide dismutase (TTC-SOD-1). Injections of native SOD-1 in contrast showed only minimal retention of protein along the injection track. Immunohistochemistry demonstrated that both TTC and TTC-SOD-1 were distributed in a punctate perineuronal and intraneuronal pattern similar to that seen after their retrograde transport, suggesting localization primarily in synaptic boutons. This synaptic distribution was confirmed using HRP-labeled TTC with electron microscopy along with localization within neuronal endosomes. We conclude that TTC may be a useful vector to enhance neuronal delivery of potentially therapeutic enzymes or trophic factors following direct injection into the brain.


Subject(s)
Central Nervous System/drug effects , Peptide Fragments/pharmacology , Protein Sorting Signals/drug effects , Superoxide Dismutase/metabolism , Tetanus Toxin/pharmacology , Animals , Central Nervous System/metabolism , Central Nervous System/ultrastructure , Humans , Immunohistochemistry/methods , Indicators and Reagents/pharmacology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Protein Sorting Signals/physiology , Superoxide Dismutase-1 , Time Factors , Tissue Distribution
14.
Exp Neurol ; 184(1): 285-94, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14637099

ABSTRACT

Adenosine A(2A) receptors, abundantly expressed on striatal medium spiny neurons, appear to activate signaling cascades implicated in the regulation of coexpressed ionotropic glutamatergic receptors. To evaluate the contribution of adenosinergic mechanisms to the pathogenesis of the response alterations induced by dopaminergic treatment, we studied the ability of the selective adenosine A(2A) receptor antagonist KW-6002 to prevent as well as palliate these syndromes in rodent and primate models of Parkinson's disease. In rats, KW-6002 reversed the shortened motor response produced by chronic levodopa treatment while reducing levodopa-induced hyperphosphorylation at S845 residues on AMPA receptor GluR1 subunits. In primates, KW-6002 evidenced modest antiparkinsonian activity when given alone. Once-daily coadministration of KW-6002 with apomorphine prevented the development of dyskinesias, which appeared in control animals 7-10 days after initiating apomorphine treatment. Animals initially given apomorphine plus KW-6002 for 3 weeks did not begin to manifest apomorphine-induced dyskinesias until 10-12 days after discontinuing the A(2A) antagonist. These results suggest that KW-6002 can attenuate the induction as well as the expression of motor response alterations to chronic dopaminergic stimulation in parkinsonian animals, possibly by blocking A(2A) receptor-stimulated signaling pathways. Our findings strengthen the rationale for developing A(2A) antagonists as an early treatment strategy for Parkinson's disease.


Subject(s)
Adenosine A2 Receptor Antagonists , Dopamine Agonists/toxicity , Parkinson Disease, Secondary/physiopathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Antiparkinson Agents/therapeutic use , Apomorphine/toxicity , Denervation , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/prevention & control , Levodopa/therapeutic use , Macaca fascicularis , Male , Neurons/pathology , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Phosphorylation , Purines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Sympatholytics/antagonists & inhibitors , Sympatholytics/toxicity
15.
Neurology ; 60(5): 790-5, 2003 Mar 11.
Article in English | MEDLINE | ID: mdl-12629235

ABSTRACT

BACKGROUND: Men who regularly consume caffeinated drinks have a lower risk of PD than do nondrinkers, but this relation has not been found in women. Because this sex difference could be due to hormonal effects, the authors examined prospectively the risk of PD according to use of postmenopausal hormones and caffeine intake among participants in the Nurses' Health Study. METHODS: The study population comprised 77,713 women free of PD, stroke, or cancer at baseline, who were postmenopausal at baseline or reached menopause before the end of the study. During 18 years of follow-up the authors documented 154 cases of PD. RESULTS: Overall, the risk of PD was similar in women using hormones and women who never used hormones (relative risk 1.02, 95% CI 0.69 to 1.52). Use of hormones, however, was associated with a reduced risk of PD among women with low caffeine consumption (RR 0.39, 95% CI 0.13 to 1.17), and with increased risk among women with high caffeine consumption (RR 2.44, 95% CI 0.75 to 7.86; p for interaction = 0.01). Among hormone users, women consuming six or more cups of coffee per day had a fourfold higher risk of PD (RR 3.92, 95% CI 1.49 to 10.34; p = 0.006) than did women who never drink coffee. CONCLUSION: These results suggest that caffeine reduces the risk of PD among women who do not use postmenopausal hormones, but increases risk among hormone users. Clinical trials of caffeine or estrogens in women should avoid the combined use of these agents.


Subject(s)
Coffee , Estrogen Replacement Therapy/statistics & numerical data , Parkinson Disease/epidemiology , Adult , Age of Onset , Alcohol Drinking/epidemiology , Beverages/statistics & numerical data , Caffeine/administration & dosage , Cohort Studies , Contraceptives, Oral, Hormonal/administration & dosage , Drug Interactions , Female , Humans , Middle Aged , Parity , Population Surveillance , Postmenopause , Prospective Studies , Risk Factors , Smoking/epidemiology , Surveys and Questionnaires
16.
Parkinsonism Relat Disord ; 8(1): 19-22, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11472876

ABSTRACT

The stress-activated protein kinase (SAPK) cascade serves a critical role in the apoptotic death of neuronal cells in response to a variety of cellular stresses. Recent in vitro and in vivo evidence has directly implicated this kinase in the death of dopaminergic nigral neurons in the MPTP model of Parkinson's disease (PD). To assess the involvement of c-Jun, a key transcription factor substrate of SAPK (also known as c-Jun N-terminal kinase, or JNK) in the MPTP-induced death of dopaminergic nigral neurons, we determined the ability of MPP+, the active toxin metabolite of MPTP, to induce the phosphorylated form of c-Jun in dopaminergic neurons in nigral (ventral mesencephalon) cultures. At a dose of MPP+ that specifically induces apoptotic changes in nuclear morphology in tyrosine hydroxylase-positive (dopaminergic) cells in these cultures, MPP+ induces nuclear phospho-c-Jun immunoreactivity (IR). The peak induction of phospho-c-Jun IR was observed 16h after beginning MPP+ exposure, and preceded the maximal induction of apoptotic nuclear changes by approximately 8h. These data support an important role for the SAPK/JNK pathway including its c-Jun transcriptional target in the apoptotic death of dopaminergic nigral neurons in the MPTP model of PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Dopamine Agents , Dopamine/physiology , Mesencephalon/metabolism , Neurons/metabolism , Parkinson Disease, Secondary/chemically induced , Proto-Oncogene Proteins c-jun/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Immunohistochemistry , Mesencephalon/cytology , Mesencephalon/drug effects , Neurons/drug effects , Neurons/ultrastructure , Phosphorylation , Rats , Tyrosine 3-Monooxygenase/metabolism
17.
J Neurosci ; 21(10): RC143, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11319241

ABSTRACT

Recent epidemiological studies have established an association between the common consumption of coffee or other caffeinated beverages and a reduced risk of developing Parkinson's disease (PD). To explore the possibility that caffeine helps prevent the dopaminergic deficits characteristic of PD, we investigated the effects of caffeine and the adenosine receptor subtypes through which it may act in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxin model of PD. Caffeine, at doses comparable to those of typical human exposure, attenuated MPTP-induced loss of striatal dopamine and dopamine transporter binding sites. The effects of caffeine were mimicked by several A(2A) antagonists (7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH 58261), 3,7-dimethyl-1-propargylxanthine, and (E)-1,3-diethyl-8 (KW-6002)-(3,4-dimethoxystyryl)-7-methyl-3,7-dihydro-1H-purine-2,6-dione) (KW-6002) and by genetic inactivation of the A(2A) receptor, but not by A(1) receptor blockade with 8-cyclopentyl-1,3-dipropylxanthine, suggesting that caffeine attenuates MPTP toxicity by A(2A) receptor blockade. These data establish a potential neural basis for the inverse association of caffeine with the development of PD, and they enhance the potential of A(2A) antagonists as a novel treatment for this neurodegenerative disease.


Subject(s)
Caffeine/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/drug therapy , Purinergic P1 Receptor Antagonists , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Catechols/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Immunity, Innate/genetics , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Purines/administration & dosage , Pyrimidines/administration & dosage , Receptor, Adenosine A2A , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics , Theobromine/administration & dosage , Theobromine/analogs & derivatives , Triazoles/administration & dosage , Xanthines/administration & dosage
18.
Biochem J ; 354(Pt 1): 123-30, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11171087

ABSTRACT

Agonist binding to extracellular A2A adenosine receptors (A2ARs) inhibits the activation of virtually all tested functions of T-cells and can induce apoptosis in thymocytes. The evaluation of levels of expression of these immunosuppressive receptors is expected to clarify whether the absence of spare A2ARs (no 'receptor reserve') might be one of the mechanisms of attenuation of the effects of extracellular adenosine on T-cells. A2A transcript is found in T-cells and functional receptors can be demonstrated, but the density of receptor on T-cells is too low to be detected by radioligand binding. Studies of direct radioligand binding to murine brain with the selective A2AR agonist [3H]CGS21680 (2-(4-[(2-carboxyethyl)-phenyl]ethylamino)-5'-N-ethylcarboxamidoadenosine) established that striata levels of A2AR are virtually absent from A2A knock-out mice. Mice that are heterozygous (A2AR+/-) for the A2AR express significantly decreased levels of A2AR. To test for the presence of spare receptors in T-cells we took advantage of this gene dose effect and examined whether the decrease in the number of receptors in thymocytes from A2AR+/- mice was proportionately reflected in a decrease in the functional cAMP response of T-cells to adenosine. cAMP accumulation and apoptosis induced by adenosine and by A2AR agonist are of a lower magnitude in T-cells from A2AR+/- heterozygous mice than in T-cells from A2AR+/+ littermate control mice. These results indicate that there is no A2AR reserve in murine T-cells. Strongly decreased adenosine-triggered cAMP increases were detected in thymocytes from A2AR-/- mice, suggesting that A2B adenosine receptors cannot fully compensate for the loss of A2ARs in murine T-cells. We conclude that the number of A2ARs is the limiting factor in determining the maximal cAMP response of T-lymphocytes to extracellular adenosine, thereby minimizing the immunosuppressive effects of extracellular adenosine.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Dosage , Receptors, Purinergic P1/metabolism , T-Lymphocytes/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout
19.
Proc Natl Acad Sci U S A ; 98(4): 1970-5, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11172060

ABSTRACT

The A(2A)R is largely coexpressed with D(2)Rs and enkephalin mRNA in the striatum where it modulates dopaminergic activity. Activation of the A(2A)R antagonizes D(2)R-mediated behavioral and neurochemical effects in the basal ganglia through a mechanism that may involve direct A(2A)R-D(2)R interaction. However, whether the D(2)R is required for the A(2A)R to exert its neural function is an open question. In this study, we examined the role of D(2)Rs in A(2A)R-induced behavioral and cellular responses, by using genetic knockout (KO) models (mice deficient in A(2A)Rs or D(2)Rs or both). Behavioral analysis shows that the A(2A)R agonist 2-4-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine reduced spontaneous as well as amphetamine-induced locomotion in both D(2) KO and wild-type mice. Conversely, the nonselective adenosine antagonist caffeine and the A(2A)R antagonist 8-(3-chlorostyryl)caffeine produced motor stimulation in mice lacking the D(2)R, although the stimulation was significantly attenuated. At the cellular level, A(2A)R inactivation counteracted the increase in enkephalin expression in striatopallidal neurons caused by D(2)R deficiency. Consistent with the D(2) KO phenotype, A(2A)R inactivation partially reversed both acute D(2)R antagonist (haloperidol)-induced catalepsy and chronic haloperidol-induced enkephalin mRNA expression. Together, these results demonstrate that A(2A)Rs elicit behavioral and cellular responses despite either the genetic deficiency or pharmacological blockade of D(2)Rs. Thus, A(2A)R-mediated neural functions are partially independent of D(2)Rs. Moreover, endogenous adenosine acting at striatal A(2A)Rs may be most accurately viewed as a facilitative modulator of striatal neuronal activity rather than simply as an inhibitory modulator of D(2)R neurotransmission.


Subject(s)
Adenosine/analogs & derivatives , Motor Activity/physiology , Receptors, Dopamine D2/physiology , Receptors, Purinergic P1/physiology , Adenosine/pharmacology , Amphetamines/pharmacology , Animals , Caffeine/analogs & derivatives , Caffeine/pharmacology , Catalepsy/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine Antagonists/pharmacology , Enkephalins/biosynthesis , Enkephalins/genetics , Gene Expression , Haloperidol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenethylamines/pharmacology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , RNA, Messenger , Receptor, Adenosine A2A , Receptors, Dopamine D1/biosynthesis , Receptors, Dopamine D2/biosynthesis , Receptors, Purinergic P1/biosynthesis
20.
FASEB J ; 14(13): 2065-74, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023991

ABSTRACT

Interleukin 12 (IL-12) is a crucial cytokine in the regulation of T helper 1 vs. T helper 2 immune responses. In the present study, we investigated the effect of the endogenous purine nucleoside adenosine on the production of IL-12. In mouse macrophages, adenosine suppressed IL-12 production. Although the order of potency of adenosine receptor agonists suggested the involvement of A2a receptors, data obtained with A2a receptor-deficient mice showed that the adenosine suppression of IL-12 and even TNF-alpha production is only partly mediated by A2a receptor ligation. Studies with adenosine receptor antagonists or the adenosine uptake blocker dipyridamole showed that adenosine released endogenously also decreases IL-12. Although adenosine increases IL-10 production, the inhibition of IL-12 production is independent of the increased IL-10. The mechanism of action of adenosine was not associated with alterations of the activation of the p38 and p42/p44 mitogen-activated protein kinases or the phosphorylation of the c-Jun terminal kinase. Adenosine failed to affect steady-state levels of either IL-12 p35 or p40 mRNA, but augmented IL-10 mRNA levels. In summary, adenosine inhibits IL-12 production via various adenosine receptors. These results support the notion that adenosine-based therapies might be useful in certain autoimmune and/or inflammatory diseases.


Subject(s)
Adenosine/pharmacology , Interleukin-12/biosynthesis , Macrophages/drug effects , Receptors, Purinergic P1/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Interleukin-10/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/metabolism , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Receptor, Adenosine A2A , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...