1
|
Sex Differences in Dopaminergic Vulnerability to Environmental Toxicants - Implications for Parkinson's Disease. Curr Environ Health Rep 2022; 9:563-573. [PMID: 36201109 DOI: 10.1007/s40572-022-00380-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Sex dimorphism in Parkinson's disease (PD) is an ostensible feature of the neurological disorder, particularly as men are 1.5-2 times more likely to develop PD than women. Clinical features of the disease, such as presentation at onset, most prevalent symptoms, and response to treatment, are also affected by sex. Despite these well-known sex differences in PD risk and phenotype, the mechanisms that impart sex dimorphisms in PD remain poorly understood. RECENT FINDINGS As PD incidence is influenced by environmental factors, an intriguing pattern has recently emerged in research studies suggesting a male-specific vulnerability to dopaminergic neurodegeneration caused by neurotoxicant exposure, with relative protection in females. These new experimental data have uncovered potential mechanisms that provide clues to the source of sex differences in dopaminergic neurodegeneration and other PD pathology such as alpha-synuclein toxicity. In this review, we discuss the emerging evidence of increased male sensitivity to neurodegeneration from environmental exposures. We examine mechanisms underlying dopaminergic neurodegeneration and PD-related pathologies with evidence supporting the roles of estrogen, SRY expression, the vesicular glutamate transporter VGLUT2, and the microbiome as prospective catalysts for male vulnerability. We also highlight the importance of including sex as a biological variable, particularly when evaluating dopaminergic neurotoxicity in the context of PD.
Collapse
|
2
|
Flores-Cuadrado A, Saiz-Sanchez D, Mohedano-Moriano A, Lamas-Cenjor E, Leon-Olmo V, Martinez-Marcos A, Ubeda-Bañon I. Astrogliosis and sexually dimorphic neurodegeneration and microgliosis in the olfactory bulb in Parkinson's disease. NPJ PARKINSONS DISEASE 2021; 7:11. [PMID: 33479244 PMCID: PMC7820595 DOI: 10.1038/s41531-020-00154-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
Hyposmia is prodromal, and male sex is a risk marker for an enhanced likelihood ratio of Parkinson’s disease. The literature regarding olfactory bulb volume reduction is controversial, although the olfactory bulb has been largely reported as an early and preferential site for α-synucleinopathy. These pathological deposits have been correlated with neural loss in Nissl-stained material. However, microgliosis has rarely been studied, and astrogliosis has been virtually neglected. In the present report, α-synucleinopathy (α-synuclein), neurodegeneration (Neu-N), astrogliosis (GFAP), and microgliosis (Iba-1) were quantified, using specific markers and stereological methods. Disease, sex, age, disease duration, and post-mortem interval were considered variables for statistical analysis. No volumetric changes have been identified regarding disease or sex. α-Synucleinopathy was present throughout the OB, mainly concentrated on anterior olfactory nucleus. Neurodegeneration (reduction in Neu-N-positive cells) was statistically significant in the diseased group. Astrogliosis (increased GFAP labeling) and microgliosis (increased Iba-1 labeling) were significantly enhanced in the Parkinson’s disease group. When analyzed per sex, neurodegeneration and microgliosis differences are only present in men. These data constitute the demonstration of sex differences in neurodegeneration using specific neural markers, enhanced astrogliosis and increased microgliosis, also linked to male sex, in the human olfactory bulb in Parkinson’s disease.
Collapse
Affiliation(s)
- Alicia Flores-Cuadrado
- Neuroplasticity & Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Daniel Saiz-Sanchez
- Neuroplasticity & Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Alicia Mohedano-Moriano
- Faculty of Health Sciences, University of Castilla-La Mancha, 45600, Talavera de la Reina, Spain
| | - Elena Lamas-Cenjor
- Neuroplasticity & Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Victor Leon-Olmo
- Neuroplasticity & Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Alino Martinez-Marcos
- Neuroplasticity & Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Isabel Ubeda-Bañon
- Neuroplasticity & Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
| |
Collapse
|
3
|
Bayer J, Rusch T, Zhang L, Gläscher J, Sommer T. Dose-dependent effects of estrogen on prediction error related neural activity in the nucleus accumbens of healthy young women. Psychopharmacology (Berl) 2020; 237:745-755. [PMID: 31773208 DOI: 10.1007/s00213-019-05409-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022]
Abstract
RATIONALE Whereas the effect of the sex steroid 17-beta-estradiol (E2) on dopaminergic (DA) transmission in the nucleus accumbens (NAc) is well evidenced in female rats, studies in humans are inconsistent. Moreover, linear and inverted u-shaped dose response curves have been observed for E2's effects on hippocampal plasticity, but the shape of dose response curves for E2's effects on the NAc is much less characterized. OBJECTIVES Investigation of dose response curves for E2's effects on DA-related neural activity in the human NAc. METHODS Placebo or E2 valerate in doses of 2, 4, 6 or 12 mg was orally administered to 125 naturally cycling young women during the low-hormone menstruation phase on two consecutive days using a randomized, double-blinded design. The E2 treatment regimen induced a wide range of E2 levels, from physiological (2- and 4-mg groups; equivalent to cycle peak) to supraphysiological levels (6- and 12-mg groups; equivalent to early pregnancy). This made it possible to study different dose response functions for E2's effects on NAc activity. During E2 peak, participants performed a well-established reversal learning paradigm. We used trial-wise prediction errors (PE) estimated via a computational reinforcement learning model as a proxy for dopaminergic activity. Linear and quadratic regression analyses predicting PE-related NAc activity from salivary E2 levels were calculated. RESULTS There was a positive linear relationship between PE-associated NAc activity and salivary E2 increases. CONCLUSIONS The randomized, placebo-controlled elevation of E2 levels stimulates NAc activity in the human brain, likely mediated by dopaminergic processes.
Collapse
Affiliation(s)
- Janine Bayer
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Tessa Rusch
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Lei Zhang
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Department of Basic Psychological Research and Research Methods, University of Vienna, Liebiggasse 5, 1010, Vienna, Austria
| | - Jan Gläscher
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Tobias Sommer
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| |
Collapse
|
4
|
Dai H, Jackson CR, Davis GL, Blakely RD, McMahon DG. Is dopamine transporter-mediated dopaminergic signaling in the retina a noninvasive biomarker for attention-deficit/ hyperactivity disorder? A study in a novel dopamine transporter variant Val559 transgenic mouse model. J Neurodev Disord 2017; 9:38. [PMID: 29281965 PMCID: PMC5745861 DOI: 10.1186/s11689-017-9215-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022] Open
Abstract
Background Dopamine (DA) is a critical neuromodulator in the retina. Disruption of retinal DA synthesis and signaling significantly attenuates light-adapted, electroretinogram (ERG) responses, as well as contrast sensitivity and acuity. As these measures can be detected noninvasively, they may provide opportunities to detect disease processes linked to perturbed DA signaling. Recently, we identified a rare, functional DA transporter (DAT, SLC6A3) coding substitution, Ala559Val, in subjects with attention-deficit/hyperactivity disorder (ADHD), demonstrating that DAT Val559 imparts anomalous DA efflux (ADE) with attendant physiological, pharmacological, and behavioral phenotypes. To understand the broader impact of ADE on ADHD, noninvasive measures sensitive to DAT reversal are needed. Methods Here, we explored this question through ERG-based analysis of retinal light responses, as well as HPLC measurements of retinal DA in DAT Val559 mice. Results Male mice homozygous (HOM) for the DAT Val559 variant demonstrated increased, light-adapted ERG b-wave amplitudes compared to wild type (WT) and heterozygous (HET) mice, whereas dark-adapted responses were indistinguishable across genotypes. The elevated amplitude of the photopic light responses in HOM mice could be mimicked in WT mice by applying D1 and D4 DA receptor agonists and suppressed in HOM mice by introducing D4 antagonist, supporting elevated retinal DA signaling arising from ADE. Following the challenge with amphetamine, WT exhibited an increase in light-adapted response amplitudes, while HOM did not. Total retinal DA content was similar across genotypes. Interestingly, female DAT Val559 HOM animals revealed no significant difference in photopic ERG responses when compared with WT and HET littermates. Conclusions These data reveal that noninvasive, in vivo evaluation of retinal responses to light can reveal physiological signatures of ADE, suggesting a possible approach to the segregation of neurobehavioral disorders based on the DAT-dependent control of DA signaling.
Collapse
Affiliation(s)
- Heng Dai
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA
| | - Chad R Jackson
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.,Present address: Defense Threat Reduction Agency, 8211 Terminal Road, Lorton, VA, 22079, USA
| | - Gwynne L Davis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.
| |
Collapse
|
5
|
Hwang CJ, Choi DY, Jung YY, Lee YJ, Yun JS, Oh KW, Han SB, Oh S, Park MH, Hong JT. Inhibition of p38 pathway-dependent MPTP-induced dopaminergic neurodegeneration in estrogen receptor alpha knockout mice. Horm Behav 2016; 80:19-29. [PMID: 26836768 DOI: 10.1016/j.yhbeh.2016.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 01/24/2016] [Accepted: 01/29/2016] [Indexed: 01/20/2023]
Abstract
Approximately, 7-10 million people in the world suffer from Parkinson's disease (PD). Recently, increasing evidence has suggested the protective effect of estrogens against nigrostriatal dopaminergic damage in PD. In this study, we investigated whether estrogen affects 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced behavioral impairment in estrogen receptor alpha (ERα)-deficient mice. MPTP (15mg/kg, four times with 1.5-h interval)-induced dopaminergic neurodegeneration was evaluated in ERα wild-type (WT) and knockout (KO) mice. Larger dopamine depletion, behavioral impairments (Rotarod test, Pole test, and Gait test), activation of microglia and astrocytes, and neuroinflammation after MPTP injection were observed in ERα KO mice compared to those in WT mice. Immunostaining for tyrosine hydroxylase (TH) after MPTP injection showed fewer TH-positive neurons in ERα KO mice than WT mice. Levels of dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC, metabolite of dopamine) were also lowered in ERα KO mice after MPTP injection. Interestingly, a higher immunoreactivity for monoamine oxidase (MAO) B was found in the substantia nigra and striatum of ERα KO mice after MPTP injection. We also found an increased activation of p38 kinase (which positively regulates MAO B expression) in ERα KO mice. In vitro estrogen treatment inhibited neuroinflammation in 1-methyl-4-phenyl pyridium (MPP+)-treated cultured astrocyte cells; however, these inhibitory effects were removed by p38 inhibitor. These results indicate that ERα might be important for dopaminergic neuronal survival through inhibition of p38 pathway.
Collapse
Affiliation(s)
- Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 712-749, South Korea
| | - Yu Yeon Jung
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Young-Jung Lee
- School of Equine Science, Cheju Halla University, 38 Halladaehak-ro, Jeju-si, Jeju Special Self-Governing Province 690-708, South Korea
| | - Jae Suk Yun
- Osong Health Technology Administration Complex, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Cheongju, Chungbuk 363-700, South Korea
| | - Ki-Wan Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Seikwan Oh
- Department of Neuroscience and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-710, South Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea.
| |
Collapse
|
6
|
Azizi-Malekabadi H, Pourganji M, Zabihi H, Saeedjalali M, Hosseini M. Tamoxifen antagonizes the effects of ovarian hormones to induce anxiety and depression-like behavior in rats. ARQUIVOS DE NEURO-PSIQUIATRIA 2015; 73:132-9. [PMID: 25742583 DOI: 10.1590/0004-282x20140221] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 11/11/2014] [Indexed: 02/05/2023]
Abstract
The effects of tamoxifen (TAM) on anxiety and depression-like behavior in ovariectomized (OVX) and naïve female rats were investigated. The animals were divided into Sham-TAM, OVX-TAM, Sham and OVX groups. Tamoxifen (1 mg/kg) was administered for 4 weeks. In the forced swimming test, the immobility times in the OVX and Sham-TAM groups were higher than in the Sham group. In the open field, the numbers of central crossings in the OVX and Sham-TAM groups were lower than the number in the Sham group, and the number of peripheral crossings in the OVX group was lower than the number in the Sham group. In the elevated plus maze, the numbers of entries to the open arm among the animals in the Sham-TAM and OVX groups were lower than the number in the Sham group, while the number of entries to the open arm in the OVX-TAM group was higher than the number in the OVX group. It was shown that deletion of ovarian hormones induced anxiety and depression-like behavior. Administration of tamoxifen in naïve rats led to anxiety and depression-like behavior that was comparable with the effects of ovarian hormone deletion. It can be suggested that tamoxifen antagonizes the effects of ovarian hormones. It also seems that tamoxifen has anxiolytic effects on ovariectomized rats.
Collapse
Affiliation(s)
- Hamid Azizi-Malekabadi
- Department of Biology, Faculty of Basic Sciences, Islamic Azad University, Isfahan, Iran
| | - Masoume Pourganji
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hoda Zabihi
- Neurogenic Inflammation Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Saeedjalali
- Mashhad Technical Faculty, Technical and Vocational University, Mashhad, Iran
| | - Mahmoud Hosseini
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Deletion of ovarian hormones induces a sickness behavior in rats comparable to the effect of lipopolysaccharide. Neurol Res Int 2015; 2015:627642. [PMID: 25705518 PMCID: PMC4325213 DOI: 10.1155/2015/627642] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/26/2014] [Accepted: 01/05/2015] [Indexed: 11/17/2022] Open
Abstract
Neuroimmune factors have been proposed as the contributors to the pathogenesis of sickness behaviors. The effects of female gonadal hormones on both neuroinflammation and depression have also been well considered. In the present study, the capability of deletion of ovarian hormones to induce sickness-like behaviors in rats was compared with the effect lipopolysaccharide (LPS). The groups were including Sham, OVX, Sham-LPS, and OVX-LPS. The Sham-LPS and OVX-LPS groups were treated with LPS (250 μg/kg) two hours before conducting the behavioral tests. In the forced swimming (FST), the immobility times in both OVX and Sham-LPS groups were higher than that of Sham (P < 0.001). In open-field (OP) test, the central crossing number by OVX and Sham-LPS groups were lower than Sham (P < 0.001) while there were no significant differences between OVX-LPS and OVX groups. In elevated plus maze (EPM), the percent of entries to the open arm by both OVX and Sham-LPS groups was lower than that of Sham group (P < 0.001). The results of present study showed that deletion of ovarian hormones induced sickness behaviors in rats which were comparable to the effects of LPS. Moreover, further investigations are required in order to better understand the mechanism(s) involved.
Collapse
|
8
|
Lundin JI, Ton TG, LaCroix AZ, Longstreth W, Franklin GM, Swanson PD, Smith-Weller T, Racette BA, Checkoway H. Formulations of hormone therapy and risk of Parkinson's disease. Mov Disord 2014; 29:1631-6. [PMID: 25255692 PMCID: PMC4216612 DOI: 10.1002/mds.26037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 07/28/2014] [Accepted: 08/03/2014] [Indexed: 11/10/2022] Open
Abstract
Hormone therapy (HT) is a class of medications widely prescribed to women in the Western world. Evidence from animal models and in vitro studies suggests that estrogen may protect against nigrostriatal system injury and increase dopamine synthesis, metabolism, and transport. Existing epidemiologic research indicates a possible reduced risk of Parkinson's disease (PD) associated with HT use. The objective of this study was to evaluate PD risk associated with specific HT formulations. Neurologist-confirmed cases and age-matched controls were identified from Group Health Cooperative (GHC) of Washington State. Final analysis included 137 female cases and 227 controls. Hormone therapy use was ascertained from the GHC pharmacy database, further classified as conjugated estrogens, esterified estrogens, and progestin. Ever use of HT formulation demonstrated a suggested elevated risk with esterified estrogen use (odds ratio [OR], 3.1; 95% confidence interval [CI], 1.0-9.8), and no risk associated with conjugated estrogen use (OR, 0.6; 95% CI, 0.6-1.3). Restricting this analysis to prescriptions that included progestin further elevated the risk associated with esterified estrogen use (OR, 6.9; 95% CI, 2.1-22.9); again, no risk was associated with conjugated estrogen use (OR, 1.7; 95% CI, 0.6-5.0). The findings from this study suggest an increase in PD risk associated with esterified estrogen use combined with progestin, and no risk associated with conjugated estrogen with progestin. These findings could have important implications for choice of HT in clinical practice.
Collapse
Affiliation(s)
- Jessica I. Lundin
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Thanh G.N. Ton
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Andrea Z. LaCroix
- Department of Family and Preventive Medicine, University of California San Diego, La Jolla, CA, USA
| | - W.T. Longstreth
- Department of Neurology, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Gary M. Franklin
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Phillip D. Swanson
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Terri Smith-Weller
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Brad A. Racette
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Harvey Checkoway
- Department of Family and Preventive Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Nasuti C, Carloni M, Fedeli D, Di Stefano A, Marinelli L, Cerasa LS, Meda C, Maggi A, Gabbianelli R. Effect of 17β-estradiol on striatal dopaminergic transmission induced by permethrin in early childhood rats. CHEMOSPHERE 2014; 112:496-502. [PMID: 25048945 DOI: 10.1016/j.chemosphere.2014.05.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 05/07/2014] [Accepted: 05/13/2014] [Indexed: 06/03/2023]
Abstract
A positive effect of estrogen treatment has been observed in neurodegenerative diseases such as Parkinson's disease. Since 17β-estradiol can modulate positively dopaminergic system, here we sought to evaluate the effect of 17β-estradiol supplementation on an animal model developing dopaminergic alterations on nucleus of striatum after neonatal exposure to permethrin pesticide. The goal of the study was to verify if the co-treatment with 17β-estradiol could protect against the damage induced by pesticide exposure in early life. Permethrin treated rats showed a decrease of dopamine and Nurr1 gene expression in striatum, while a more pronounced decrease of dopamine was observed in rats co-administered with permethrin+17β-estradiol. No difference between control and permethrin treated rats was observed in both mRNA of ERα and ERβ, whereas the rats co-administered with permethrin+17β-estradiol showed a down-regulation of ERα expression. The in vitro studies showed that permethrin, at high concentration may have an antagonist effect on ERα and even more pronounced in ERβ, thus suggesting that permethrin may block the estrogen neuroprotective effects. In conclusion, in male rats, the administration of estrogen further enhanced the impairment of dopaminergic transmission due to exposure to permethrin.
Collapse
Affiliation(s)
- Cinzia Nasuti
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, MC, Italy
| | - Manuel Carloni
- School of Pharmacy, Molecular Biology Unit, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - Donatella Fedeli
- School of Pharmacy, Molecular Biology Unit, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - Antonio Di Stefano
- Dipartimento di Scienze del Farmaco, Università G. d'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Lisa Marinelli
- Dipartimento di Scienze del Farmaco, Università G. d'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Laura Serafina Cerasa
- Dipartimento di Scienze del Farmaco, Università G. d'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Clara Meda
- Center of Excellence on Neurodegenerative Diseases University of Milan, Via Balzaretti, 9 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases University of Milan, Via Balzaretti, 9 20133 Milan, Italy
| | - Rosita Gabbianelli
- School of Pharmacy, Molecular Biology Unit, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy.
| |
Collapse
|
10
|
Greene N, Lassen CF, Rugbjerg K, Ritz B. Reproductive factors and Parkinson's disease risk in Danish women. Eur J Neurol 2014; 21:1168-77, e68. [PMID: 24750445 DOI: 10.1111/ene.12450] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/17/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Parkinson's disease is more common in men than women by a ratio of about 1.5:1 and yet there is no consensus to date as to whether female reproductive factors including hormone use affect Parkinson's disease risk. Our objective was to examine the relationship between Parkinson's disease and female reproductive factors in the largest population-based Parkinson's disease case-control study to date. METHODS Seven hundred and forty-three female Parkinson's disease cases diagnosed between 1996 and 2009 were selected from the Danish National Hospital Register, diagnoses confirmed by medical record review, and the cases were matched by birth year to 765 female controls randomly selected from the Danish Civil Registration System. Covariate information was collected in computer-assisted telephone interviews covering an extensive array of topics including reproductive and lifestyle factors. RESULTS After adjusting for smoking, caffeine and alcohol use, education, age, and family Parkinson's disease history, inverse associations between Parkinson's disease and early menarche (first period at ≤11 years), oral contraceptives, high parity (≥4 children) and bilateral oophorectomy were found; adjusted odds ratios and 95% confidence limits were respectively 0.68 (0.45-1.03) for early menarche, 0.87 (0.69-1.10) for oral contraceptives, 0.79 (0.59-1.06) for high parity and 0.65 (0.45-0.94) for bilateral oophorectomy. Little support for associations between Parkinson's disease and fertile life length, age at menopause or post-menopausal hormone treatment was found. CONCLUSIONS Reproductive factors related to women's early- to mid-reproductive lives appear to be predictive of subsequent Parkinson's disease risk whereas factors occurring later in life seem less important.
Collapse
Affiliation(s)
- N Greene
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
11
|
Hormone replacement therapy and risk for neurodegenerative diseases. Int J Alzheimers Dis 2012; 2012:258454. [PMID: 22548198 PMCID: PMC3324889 DOI: 10.1155/2012/258454] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 01/17/2012] [Accepted: 01/18/2012] [Indexed: 02/02/2023] Open
Abstract
Over the past two decades, there has been a significant amount of research investigating the risks and benefits of hormone replacement therapy (HRT) with regards to neurodegenerative disease. Here, we review basic science studies, randomized clinical trials, and epidemiological studies, and discuss the putative neuroprotective effects of HRT in the context of Alzheimer's disease, Parkinson's disease, frontotemporal dementia, and HIV-associated neurocognitive disorder. Findings to date suggest a reduced risk of Alzheimer's disease and improved cognitive functioning of postmenopausal women who use 17β-estradiol. With regards to Parkinson's disease, there is consistent evidence from basic science studies for a neuroprotective effect of 17β-estradiol; however, results of clinical and epidemiological studies are inconclusive at this time, and there is a paucity of research examining the association between HRT and Parkinson's-related neurocognitive impairment. Even less understood are the effects of HRT on risk for frontotemporal dementia and HIV-associated neurocognitive disorder. Limits to the existing research are discussed, along with proposed future directions for the investigation of HRT and neurodegenerative diseases.
Collapse
|
12
|
Arad M, Weiner I. Abnormally rapid reversal learning and reduced response to antipsychotic drugs following ovariectomy in female rats. Psychoneuroendocrinology 2012; 37:200-12. [PMID: 21723667 DOI: 10.1016/j.psyneuen.2011.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 05/18/2011] [Accepted: 06/02/2011] [Indexed: 12/24/2022]
Abstract
Epidemiological and clinical life cycle studies indicate that favorable illness course and better response to antipsychotic drugs (APDs) in women with schizophrenia are positively correlated with estrogen levels. Accordingly, the estrogen hypothesis of schizophrenia proposes a neuroprotective role of estrogen in women vulnerable to schizophrenia. Previously we demonstrated in the rat that low levels of estrogen induced by ovariectomy led to disruption of latent inhibition (LI) reflecting impairment of selective attention, a core deficit of schizophrenia. LI disruption was reversed by 17β-estradiol and the atypical APD clozapine, whereas the typical APD haloperidol was ineffective unless co-administered with 17β-estradiol. Here we aimed to extend these findings by testing ovariectomized rats in another selective attention task, discrimination reversal. Ovariectomy led to a loss of selective attention as manifested in abnormally rapid reversal. The latter was normalized by high dose of 17β-estradiol (150 μg/kg) and clozapine (2.5mg/kg), but not by haloperidol (0.1mg/kg) or lower doses of 17β-estradiol (10 and 50 μg/kg). However, co-administration of haloperidol with 17β-estradiol (50 μg/kg) was effective. In sham rats low 17β-estradiol (10 μg/kg) produced rapid reversal, while high 17β-estradiol (150 μg/kg), haloperidol alone, or haloperidol-17β-estradiol combination reduced reversal speed. Clozapine did not affect reversal speed in sham rats. These results strengthen our previous results in suggesting that schizophrenia-like attentional abnormalities as well as reduced response to APDs in female rats are associated with low level of gonadal hormones. In addition, they support the possibility that estrogen may have an antipsychotic-like action in animal models.
Collapse
Affiliation(s)
- Michal Arad
- Department of Psychology, Tel-Aviv University, Tel-Aviv 69978, Israel.
| | | |
Collapse
|
13
|
Abstract
Aggregated a-synuclein is the major component of inclusions in Parkinson's disease and other synucleinopathy brains indicating that a-syn aggregation is associated with the pathogenesis of neurodegenerative disorders. Although the mechanisms underlying a-syn aggregation and toxicity are not fully elucidated, it is clear that a-syn undergoes post-translational modifications and interacts with numerous proteins and other macromolecules, metals, hormones, neurotransmitters, drugs and poisons that can all modulate its aggregation propensity. The current and most recent findings regarding the factors modulating a-syn aggregation process are discussed in detail.
Collapse
|
14
|
Simpkins JW, Perez E, Wang X, Yang S, Wen Y, Singh M. The potential for estrogens in preventing Alzheimer's disease and vascular dementia. Ther Adv Neurol Disord 2011; 2:31-49. [PMID: 19890493 DOI: 10.1177/1756285608100427] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Estrogens are the best-studied class of drugs for potential use in the prevention of Alzheimer's disease (AD). These steroids have been shown to be potent neuroprotectants both in vitro and in vivo, and to exert effects that are consistent with their potential use in prevention of AD. These include the prevention of the processing of amyloid precursor protein (APP) into beta-amyloid (Aß), the reduction in tau hyperphosphorylation, and the elimination of catastrophic attempts at neuronal mitosis. Further, epidemiological data support the efficacy of early postmenopausal use of estrogens for the delay or prevention of AD. Collectively, this evidence supports the further development of estrogen-like compounds for prevention of AD. Several approaches to enhance brain specificity of estrogen action are now underway in an attempt to reduce the side effects of chronic estrogen therapy in AD.
Collapse
Affiliation(s)
- James W Simpkins
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, Center FOR HER (Focused On Resources for her Health, Education and Research), University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | | | | | | | | |
Collapse
|
15
|
Antzoulatos E, Jakowec MW, Petzinger GM, Wood RI. MPTP Neurotoxicity and Testosterone Induce Dendritic Remodeling of Striatal Medium Spiny Neurons in the C57Bl/6 Mouse. PARKINSONS DISEASE 2011; 2011:138471. [PMID: 21765998 PMCID: PMC3134993 DOI: 10.4061/2011/138471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 03/27/2011] [Indexed: 01/20/2023]
Abstract
Nigrostriatal damage is increased in males relative to females. While estrogen is neuroprotective in females, less is known about potential protective effects of testosterone in males. We determined if castration enhances neuronal injury to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Castrates or sham-castrated mice were sacrificed 1 week following injection of MPTP (4 × 20 mg/kg) or saline (n = 11-12/group). The right striatum was immunostained for tyrosine hydroxylase (TH). The left hemisphere was stained by Golgi Cox to quantify neuronal morphology in medium spiny neurons (MSNs) of the dorsolateral striatum. MPTP reduced TH, but there was no effect of castration and no interaction. For MSN dendritic morphology, MPTP decreased the highest branch order and increased spine density on 2nd-order dendrites. Castrated males had shorter 5th-order dendrites. However, there was no interaction between gonadal status and MPTP. Thus, castration and MPTP exert nonoverlapping effects on MSN morphology with castration acting on distal dendrites and MPTP acting proximally.
Collapse
Affiliation(s)
- Eleni Antzoulatos
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
16
|
Ozsoy O, Tanriover G, Derin N, Uysal N, Demir N, Gemici B, Kencebay C, Yargicoglu P, Agar A, Aslan M. The Effect of Docosahexaenoic Acid on Visual Evoked Potentials in a Mouse Model of Parkinson’s Disease: The Role of Cyclooxygenase-2 and Nuclear Factor Kappa-B. Neurotox Res 2011; 20:250-62. [DOI: 10.1007/s12640-011-9238-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 12/26/2010] [Accepted: 01/03/2011] [Indexed: 01/04/2023]
|
17
|
Boger HA, Mannangatti P, Samuvel DJ, Saylor AJ, Bender TS, McGinty JF, Fortress AM, Zaman V, Huang P, Middaugh LD, Randall PK, Jayanthi LD, Rohrer B, Helke KL, Granholm AC, Ramamoorthy S. Effects of brain-derived neurotrophic factor on dopaminergic function and motor behavior during aging. GENES BRAIN AND BEHAVIOR 2010; 10:186-98. [PMID: 20860702 DOI: 10.1111/j.1601-183x.2010.00654.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is critical in synaptic plasticity and in the survival and function of midbrain dopamine neurons. In this study, we assessed the effects of a partial genetic deletion of BDNF on motor function and dopamine (DA) neurotransmitter measures by comparing Bdnf(+/-) with wildtype mice (WT) at different ages. Bdnf(+/-) and WT mice had similar body weights until 12 months of age; however, at 21 months, Bdnf(+/-) mice were significantly heavier than WT mice. Horizontal and vertical motor activity was reduced for Bdnf(+/-) compared to WT mice, but was not influenced by age. Performance on an accelerating rotarod declined with age for both genotypes and was exacerbated for Bdnf(+/-) mice. Body weight did not correlate with any of the three behavioral measures studied. Dopamine neurotransmitter markers indicated no genotypic difference in striatal tyrosine hydroxylase, DA transporter (DAT) or vesicular monoamine transporter 2 (VMAT2) immunoreactivity at any age. However, DA transport via DAT (starting at 12 months) and VMAT2 (starting at 3 months) as well as KCl-stimulated DA release were reduced in Bdnf(+/-) mice and declined with age suggesting an increasingly important role for BDNF in the release and uptake of DA with the aging process. These findings suggest that a BDNF expression deficit becomes more critical to dopaminergic dynamics and related behavioral activities with increasing age.
Collapse
Affiliation(s)
- H A Boger
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gillies GE, McArthur S. Independent influences of sex steroids of systemic and central origin in a rat model of Parkinson's disease: A contribution to sex-specific neuroprotection by estrogens. Horm Behav 2010; 57:23-34. [PMID: 19538962 DOI: 10.1016/j.yhbeh.2009.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 06/02/2009] [Accepted: 06/04/2009] [Indexed: 11/16/2022]
Abstract
This review considers evidence which reveals considerable complexity and sex differences in the response of the nigrostriatal dopaminergic (NSDA) system to hormonal influences. This pathway degenerates in Parkinson's disease (PD) and sex hormones contribute to sex differences in PD, where men fare worse than women. Here we discuss evidence from animal studies which allows us to hypothesize that, contrary to expectations, the acclaimed neuroprotective property of physiological concentrations of estradiol arises not by promoting NSDA neuron survival, but by targeting powerful adaptive responses in the surviving neurons, which restore striatal DA functionality until over 60% of neurons are lost. Estrogen generated locally in the NSDA region appears to promote these adaptive mechanisms in females and males to preserve striatal DA levels in the partially injured NSDA pathway. However, responses to systemic steroids differ between the sexes. In females there is general agreement that gonadal steroids and exogenous estradiol promote striatal adaptation in the partially injured NSDA pathway to protect against striatal DA loss. In contrast, the balance of evidence suggests that in males gonadal factors and exogenous estradiol have negligible or even harmful effects. Sex differences in the organization of NSDA-related circuitry may well account for these differences. Compensatory mechanisms and sexually dimorphic hard-wiring are therefore likely to represent important biological substrates for sex dimorphisms. As these processes may be targeted differentially by systemic steroids in males and females, further understanding of the underlying processes would provide valuable insights into the potential for hormone-based therapies in PD, which would need to be sex-specific. Alternatively, evidence that estrogen generated locally is protective in the injured male NSDA pathway indicates the great therapeutic potential of harnessing central steroid synthesis to ameliorate neurodegenerative disorders. A clearer understanding of the relative contributions and inter-relationships of central and systemic steroids within the NSDA system is an important goal for future studies.
Collapse
Affiliation(s)
- Glenda E Gillies
- Department of Cellular and Molecular Neuroscience, Imperial College London, Hammersmith Hospital Campus, UK.
| | | |
Collapse
|
19
|
Simpkins JW, Yi KD, Yang SH, Dykens JA. Mitochondrial mechanisms of estrogen neuroprotection. Biochim Biophys Acta Gen Subj 2009; 1800:1113-20. [PMID: 19931595 DOI: 10.1016/j.bbagen.2009.11.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 11/11/2009] [Accepted: 11/12/2009] [Indexed: 01/11/2023]
Abstract
Mitochondria have become a primary focus in our search not only for the mechanism(s) of neuronal death but also for neuroprotective drugs and therapies that can delay or prevent Alzheimer's disease and other chronic neurodegenerative conditions. This is because mitochrondria play a central role in regulating viability and death of neurons, and mitochondrial dysfunction has been shown to contribute to neuronal death seen in neurodegenerative diseases. In this article, we review the evidence for the role of mitochondria in cell death and neurodegeneration and provide evidence that estrogens have multiple effects on mitochondria that enhance or preserve mitochondrial function during pathologic circumstances such as excitotoxicity, oxidative stress, and others. As such, estrogens and novel non-hormonal analogs have come to figure prominently in our efforts to protect neurons against both acute brain injury and chronic neurodegeneration.
Collapse
Affiliation(s)
- James W Simpkins
- Department of Pharmacology & Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | | | | | | |
Collapse
|
20
|
Zheng P. Neuroactive steroid regulation of neurotransmitter release in the CNS: Action, mechanism and possible significance. Prog Neurobiol 2009; 89:134-52. [DOI: 10.1016/j.pneurobio.2009.07.001] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 03/11/2009] [Accepted: 07/02/2009] [Indexed: 12/31/2022]
|
21
|
Hirohata M, Ono K, Morinaga A, Ikeda T, Yamada M. Anti-aggregation and fibril-destabilizing effects of sex hormones on alpha-synuclein fibrils in vitro. Exp Neurol 2009; 217:434-9. [PMID: 19289119 DOI: 10.1016/j.expneurol.2009.03.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 02/19/2009] [Accepted: 03/05/2009] [Indexed: 10/21/2022]
Abstract
The alpha-synuclein aggregation in the brain is the hallmark of Lewy body diseases, including Parkinson's disease and dementia with Lewy bodies, and multiple system atrophy. Some epidemiological studies have revealed that estrogen therapy reduces the risk of Parkinson's disease in females. We examined the effects of estriol, estradiol, estrone, androstenedione, and testosterone on the formation and destabilization of alpha-synuclein fibrils at pH 7.5 and 37 degrees C in vitro, using fluorescence spectroscopy with thioflavin S and electron microscopy. These sex hormones, especially estriol, significantly exert anti-aggregation and fibril-destabilizing effects; and hence, could be valuable preventive and therapeutic agents for alpha-synucleinopathies.
Collapse
Affiliation(s)
- Mie Hirohata
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8640, Japan
| | | | | | | | | |
Collapse
|
22
|
Rodríguez-Navarro JA, Solano RM, Casarejos MJ, Gomez A, Perucho J, de Yébenes JG, Mena MA. Gender differences and estrogen effects in parkin null mice. J Neurochem 2008; 106:2143-57. [PMID: 18643794 DOI: 10.1111/j.1471-4159.2008.05569.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Estrogens are considered neurotrophic for dopamine neurons. Parkinson's disease is more frequent in males than in females, and more prevalent in females with short reproductive life. Estrogens are neuroprotective against neurotoxic agents for dopamine neurons in vivo and in vitro. Here, we have investigated the role of estrogens in wild-type (WT) and parkin null mice (PK-/-). WT mice present sexual dimorphisms in neuroprotective mechanisms (Bcl-2/Bax, chaperones, and GSH), but some of these inter-sex differences disappear in PK-/-. Tyrosine hydroxylase (TH) protein and TH+ cells decreased earlier and more severely in female than in male PK-/- mice. Neuronal cultures from midbrain of WT and PK-/- mice were treated with estradiol from 10 min to 48 h. Short-term treatments activated the mitogen-activated protein kinase pathway of WT and PK-/- neurons and the phosphatidylinositol 3'-kinase/AKT/glycogen synthase kinase-3 pathway of WT but not of PK-/- cultures. Long-term treatments with estradiol increased the number of TH+ neurons, the TH expression, and the extension of neurites, and decreased the level of apoptosis, the expression of glial fibrillary acidic protein, and the number of microglial cells in WT but not in PK-/- cultures. The levels of estrogen receptor-alpha were elevated in midbrain cultures and in the striatum of adult PK-/- male mice, suggesting that suppression of parkin changes the estrogen receptor-alpha turnover. From our data, it appears that parkin participates in the cellular estrogen response which could be of interest in the management of parkin-related Parkinson's disease patients.
Collapse
|
23
|
Postmenopausal effects of intrastriatal estrogen on catalepsy and pallidal electroencephalogram in an animal model of Parkinson's disease. Neuroscience 2008; 154:940-5. [DOI: 10.1016/j.neuroscience.2008.02.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2007] [Revised: 01/26/2008] [Accepted: 02/29/2008] [Indexed: 11/20/2022]
|
24
|
Quesada A, Lee BY, Micevych PE. PI3 kinase/Akt activation mediates estrogen and IGF-1 nigral DA neuronal neuroprotection against a unilateral rat model of Parkinson's disease. Dev Neurobiol 2008; 68:632-44. [PMID: 18278798 PMCID: PMC2667142 DOI: 10.1002/dneu.20609] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Recently, using the medial forebrain bundle (MFB) 6-hydroxydopmaine (6-OHDA) lesion rat model of Parkinson's disease (PD), we have demonstrated that blockade of central IGF-1 receptors (IGF-1R) attenuated estrogen neuroprotection of substantia nigra pars compacta (SNpc) DA neurons, but exacerbated 6-OHDA lesions in IGF-1 only treated rats (Quesada and Micevych [2004]: J Neurosci Res 75:107-116). This suggested that the IGF-1 system is a central mechanism through which estrogen acts to protect the nigrostriatal DA system. Moreover, these results also suggest that IGF-1R-induced intracellular signaling pathways are involved in the estrogen mechanism that promotes neuronal survival. In vitro, two convergent intracellular signaling pathways used by estrogen and IGF-1, the mitogen-activated protein kinase (MAPK/ERK), and phosphatidyl-inositol-3-kinase/Akt (PI3K/Akt), have been demonstrated to be neuroprotective. Continuous central infusions of MAPK/ERK and PI3K/Akt inhibitors were used to test the hypothesis that one or both of these signal transduction pathways mediates estrogen and/or IGF-1 neuroprotection of SNpc DA neurons after a unilateral administration of 6-OHDA into the MFB of rats. Motor behavior tests and tyrosine hydroxylase immunoreactivity revealed that the inhibitor of the PI3K/Akt pathway (LY294002) blocked the survival effects of both estrogen and IGF-1, while an inhibitor of the MAPK/ERK signaling (PD98059) was ineffective. Western blot analyses showed that estrogen and IGF-1 treatments increased PI3K/Akt activation in the SN; however, MAPK/ERK activation was decreased in the SN. Indeed, continuous infusions of inhibitors blocked phosphorylation of PI3K/Akt and MAPK/ERK. These findings indicate that estrogen and IGF-1-mediated SNpc DA neuronal protection is dependent on PI3K/Akt signaling, but not on the MAPK/ERK pathway.
Collapse
Affiliation(s)
- Arnulfo Quesada
- Department of Neurobiology, Brain Research Institute, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.
| | | | | |
Collapse
|
25
|
Liu LX, Chen WF, Xie JX, Wong MS. Neuroprotective effects of genistein on dopaminergic neurons in the mice model of Parkinson's disease. Neurosci Res 2007; 60:156-61. [PMID: 18054104 DOI: 10.1016/j.neures.2007.10.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 10/01/2007] [Accepted: 10/16/2007] [Indexed: 11/27/2022]
Abstract
Emerging evidence suggests beneficial effects of estrogen and estrogen-like chemicals on neurodegenerative diseases, especially Parkinson's disease (PD). Genistein, an isoflavone naturally found in soy products, displays estrogenic properties. The present study aims to investigate the neuroprotective effects of genistein on dopaminergic neurons in ovariectomized (OVX), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model mice. MPTP significantly decreased the levels of dopamine (DA) and its metabolites dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) in the striatum, which could be restored by genistein or estrogen pretreatment. MPTP-challenge with genistein or estrogen pretreatment demonstrated reduced neurotoxicity, with tyrosine hydroxylase-immunoreactive (TH-IR) neurons in the substantia nigra pars compacta (SNpc) affected to a significantly lesser extent as compared to the MPTP treated control. The reverse transcription-PCR results also confirmed that the MPTP-induced downregulation of TH, dopamine transporter (DAT) and Bcl-2 mRNA expression in the midbrain could be restored by genistein or estrogen pretreatment. These findings provide the first evidence that genistein has neuroprotective effects on dopaminergic neurons in the MPTP-induced PD mice and this effect may be attributed to enhancing Bcl-2 gene expression.
Collapse
Affiliation(s)
- Li-Xing Liu
- Department of Physiology, Medical College of Qingdao University, PR China
| | | | | | | |
Collapse
|
26
|
Liu B, Dluzen DE. OESTROGEN AND NIGROSTRIATAL DOPAMINERGIC NEURODEGENERATION: ANIMAL MODELS AND CLINICAL REPORTS OF PARKINSON'S DISEASE. Clin Exp Pharmacol Physiol 2007; 34:555-65. [PMID: 17581209 DOI: 10.1111/j.1440-1681.2007.04616.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The exact nature of oestrogen (positive, negative or no effect) in the dopaminergic neurodegenerative disorder Parkinson's disease is controversial. 2. In the present review, we summarize the data on oestrogen and nigrostriatal dopaminergic neurodegeneration in animal models and clinical reports of Parkinson's disease. 3. Most animal studies support the ability of oestrogen to function as a neuroprotectant against neurotoxins that target the nigrostriatal dopaminergic system. 4. Retrospective and prospective clinical studies generally support the findings from animal studies that oestrogen exerts a positive, or, at worst, no effect, in Parkinson's disease. 5. Oestrogen was chosen as one of the 12 neuroprotective compounds to be attractive candidates for further clinical trials (Phase II or III) in 2003.
Collapse
Affiliation(s)
- Bin Liu
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, Rootstown, Ohio 44272-0095, USA
| | | |
Collapse
|
27
|
Ragonese P, D'Amelio M, Savettieri G. Implications for estrogens in Parkinson's disease: an epidemiological approach. Ann N Y Acad Sci 2007; 1089:373-82. [PMID: 17261781 DOI: 10.1196/annals.1386.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Evidence from experimental and epidemiological studies suggests a role of sex hormones in the pathogenic process leading to neurodegenerative diseases, (i.e., Alzheimer's and Parkinson's disease). The effects of sexual steroid hormones are complex and vary with the events of women's fertile life. Estrogens are supposed to influence dopamine synthesis, metabolism, and transport; however, there is no consensus regarding the direction, locus, and mechanism of the effect of estrogens on the dopaminergic system. A neuroprotective effect of estrogens has been demonstrated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-animal models of Parkinson's disease (PD). Epidemiological studies indicate gender differences regarding the onset and the prognosis of PD. Most of the analytical studies explored the relationship between PD and exogenous estrogens. Only three studies investigated the role of endogenous estrogens in the risk of developing PD. These studies reported an increased risk of PD in conditions causing an early reduction in endogenous estrogens (early menopause, reduced fertile life length). Longer cumulative length of pregnancies has also been associated with an increased PD risk. A lack of consensus still exists on the effect of the type of menopause (surgical vs. natural) on PD risk. Finally, the effect of postmenopausal estrogen replacement therapy is still debated. Inconsistencies across studies are in part explained by the complexity of the mechanisms of action of sexual hormones and by the paucity of analytical studies.
Collapse
Affiliation(s)
- Paolo Ragonese
- Dipartimento Universitario di Neuroscienze Cliniche, Università di Palermo, Via Gaetano La Loggia 1-90129 Palermo, Italy
| | | | | |
Collapse
|
28
|
|
29
|
Lin JJ, Chen CH, Yueh KC, Chang CY, Lin SZ. A CD14 monocyte receptor polymorphism and genetic susceptibility to Parkinson's disease for females. Parkinsonism Relat Disord 2006; 12:9-13. [PMID: 16337421 DOI: 10.1016/j.parkreldis.2005.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 07/11/2005] [Accepted: 07/13/2005] [Indexed: 11/21/2022]
Abstract
Recent studies suggest that inflammation may play an important role in the pathogenesis of Parkinson's disease (PD). Because the C(-260) --> T polymorphism in the promoter of the CD14 monocyte receptor gene (pCD14) could affect the predisposition to the inflammatory response, we conducted a case-control study to investigate a possible genetic susceptibility of the pCD14 polymorphism in patients with PD. This study included 200 sporadic PD patients and 200 controls, matched by sex and case-control pairs for age at onset in the case. All observed genotype frequencies were in Hardy-Weinberg equilibrium. Results revealed that the CD14-T allele of the pCD14 polymorphism in the female PD patients existed statistically significant difference from that of the female controls (OR = 1.262, P = 0.038), but not for male. Female individuals with homozygote CD14-TT genotype were significantly increased risk of PD by 1.28 time (P = 0.027). Furthermore, a logistic regression analysis confirmed that the homozygote CD14-TT genotype was an independent risk factor for PD (OR = 1.576, P = 0.030). In conclusion, results of this study indicate the pCD14 polymorphism to be a genetic risk factor for PD in females.
Collapse
Affiliation(s)
- J J Lin
- Department of Neurology, Chushang Show-Chwan Hospital, Nantou, 557, Taiwan, ROC.
| | | | | | | | | |
Collapse
|
30
|
Gillies GE, Murray HE, Dexter D, McArthur S. Sex dimorphisms in the neuroprotective effects of estrogen in an animal model of Parkinson's disease. Pharmacol Biochem Behav 2005; 78:513-22. [PMID: 15251260 DOI: 10.1016/j.pbb.2004.04.022] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Revised: 04/19/2004] [Accepted: 04/20/2004] [Indexed: 11/27/2022]
Abstract
The incidence of certain neurological disorders, including Parkinson's disease, appears to be more prevalent in men. Studies involving estrogen treatment of ovariectomised rodents attribute this largely to the neuroprotective effects of estrogen. However, a neuroprotective role for physiological levels of circulating hormones in males and females is less clear. Using the 6-hydroxydopamine (6-OHDA) model of Parkinson's disease to lesion the nigrostriatal dopaminergic (NSDA) pathway, we have shown that in females, endogenously produced estrogen is neuroprotective, whereas in males, gonadal factors increase striatal 6-OHDA toxicity. Intriguingly, estrogen, but not dihydrotestosterone, a nonaromatizable androgen, reversed the effects of orchidectomy on lesion size, raising the novel the hypothesis that enhanced male susceptibility may be attributable to the effects of endogenous testosterone only after its aromatization to estrogen. Thus, estrogen appears to exert opposite effects in the NSDA in males and females, being neuroprotective in females, but not in males, where it may even exacerbate neurodegenerative responses, with important implications for the clinical potential of estrogen-related compounds as neuroprotective agents. Preliminary experiments support the hypothesis that sex differences in the adult NSDA may result from the organisational actions of gonadal steroids during the critical neonatal period for the masculinization of the brain. Further studies are needed to determine whether this early organisation of a sexually differentiated neural circuitry may contribute to the emergence of neurodegenerative conditions such as Parkinson's disease.
Collapse
Affiliation(s)
- Glenda E Gillies
- Department of Cellular and Molecular Neuroscience, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | | | | | | |
Collapse
|
31
|
Lin JJ, Yueh KC, Chang CY, Chen CH, Lin SZ. The homozygote AA genotype of the α1-antichymotrypsin gene may confer protection against early-onset Parkinson's disease in women. Parkinsonism Relat Disord 2004; 10:469-73. [PMID: 15542006 DOI: 10.1016/j.parkreldis.2004.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There has been increasing evidence suggesting that inflammatory response maybe involved in the pathogenesis of Parkinson's disease (PD). Alpha1-antichymotrypsin gene (ACT) has been regarded as a susceptibility factor for PD in the past, but the evidence remains controversial. This case-control study was designed to investigate the association of alpha1-antichymotrypsin gene (ACT) polymorphism between 210 Taiwanese patients with clinical definite sporadic PD and 260 controls, matched by age and sex. There were no differences of allelic frequency (A and T) and genotype polymorphism (AA, AT and TT) of the ACT in PD patients from the controls. However, there were significantly fewer early-onset PD (onset age younger than 60 years) or PD women carrying the homozygote AA genotype (ACT-AA) than in controls (p=0.046 and 0.044, respectively). Further analysis revealed that the reduced risk of ACT-AA was particularly significant among PD women with the onset age younger than 60 years (OR=0.796, 95% CI=0.749-0.847, p<0.0001). This study shows that ACT-AA may confer a modest protection against developing early-onset PD in women.
Collapse
Affiliation(s)
- J J Lin
- Department of Neurology, Chushang Show-Chwan Hospital, 75 Sec. 2 Chi-Shang Road, Chushang Jenn, Nantou 557, Taiwan.
| | | | | | | | | |
Collapse
|
32
|
Yune TY, Kim SJ, Lee SM, Lee YK, Oh YJ, Kim YC, Markelonis GJ, Oh TH. Systemic Administration of 17β-Estradiol Reduces Apoptotic Cell Death and Improves Functional Recovery following Traumatic Spinal Cord Injury in Rats. J Neurotrauma 2004; 21:293-306. [PMID: 15115604 DOI: 10.1089/089771504322972086] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent evidence indicates that estrogen exerts neuroprotective effects in both brain injury and neurodegenerative diseases. We examined the protective effect of estrogen on functional recovery after spinal cord injury (SCI) in rats. 17beta-estradiol (3, 100, or 300 microg/kg) was administered intravenously 1-2 h prior to injury (pre-treatment), and animals were then subjected to a mild, weight-drop spinal cord contusion injury. Estradiol treatment significantly improved hind limb motor function as determined by the Basso-Beattie-Bresnahan (BBB) locomotor open field behavioral rating test. Fifteen to 30 days after SCI, BBB scores were significantly higher in estradiol-treated (100 microg/kg) rats when compared to vehicle-treated rats. Morphological analysis showed that lesion sizes increased progressively in either vehicle-treated or 17beta-estradiol-treated spinal cords. However, in response to treatment with 17beta-estradiol, the lesion size was significantly reduced 18-28 days after SCI when compared to vehicle-treated controls. Terminal deoxynucleotidyl transferase-mediated UTP nickend labeling (TUNEL) staining and DNA gel electrophoresis revealed that apoptotic cell death peaked 24-48 h after injury. Also, SCI induced a marked increase in activated caspase-3 in the spinal cord, evident by 4 h after injury. However, administration of 17beta-estradiol significantly reduced the SCI-induced increase in apoptotic cell death and caspase-3 activity after SCI. Furthermore, 17beta-estradiol significantly increased expression of the anti-apoptotic genes, bcl-2 and bcl-x, after SCI while expression of the pro-apoptotic genes, bad and bax, was not affected by drug treatment. Finally, intravenous administration of 17beta-estradiol (100 microg/kg) immediately after injury (post-treatment) also significantly improved hind limb motor function 19-30 days after SCI compared to vehicle-treated controls. These data suggest that after SCI, 17 beta-estradiol treatment improved functional recovery in the injured rat, in part, by reducing apoptotic cell death.
Collapse
Affiliation(s)
- Tae Y Yune
- Biomedical Research Center, Korea Institute of Science & Technology, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Quesada A, Micevych PE. Estrogen interacts with the IGF-1 system to protect nigrostriatal dopamine and maintain motoric behavior after 6-hydroxdopamine lesions. J Neurosci Res 2004; 75:107-16. [PMID: 14689453 DOI: 10.1002/jnr.10833] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The most prominent neurochemical hallmark of Parkinson's disease (PD) is the loss of nigrostriatal dopamine (DA). Animal models of PD have concentrated on depleting DA and therapies have focused on maintaining or restoring DA. Within this context estrogen protects against 6-hydroxdopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) lesions of the nigrostriatal DA pathway. Present studies tested the hypothesis that neuroprotective estrogen actions involve activation of the insulin-like growth factor-1 (IGF-1) system. Ovariectomized rats were treated with either a single subcutaneous injection of 17beta-estradiol benzoate or centrally or peripherally IGF-1. All rats were infused unilaterally with 6-OHDA into the medial forebrain bundle (MFB) to lesion the nigrostriatal DA pathway. Tyrosine hydroxylase (TH) immunocytochemistry confirmed that rats injected with 6-OHDA had a massive loss of TH immunoreactivity in both the ipsilateral substantia nigra compacta (60% loss) and the striatum (>95% loss) compared to the contralateral side. Loss of TH immunoreactivity was correlated with loss of asymmetric forelimb movements, a behavioral assay for motor deficits. Pretreatment with estrogen or IGF-1 significantly prevented 6-OHDA-induced loss of substantia nigra compacta neurons (20% loss) and TH immunoreactivity in DA fibers in the striatum (<20% loss) and prevented the loss of asymmetric forelimb use. Blockage of IGF-1 receptors by intracerebroventricular JB-1, an IGF-1 receptor antagonist, attenuated both estrogen and IGF-1 neuroprotection of nigrostriatal DA neurons and motor behavior. These findings suggest that IGF-1 and estrogen acting through the IGF-1 system may be critical for neuroprotective effects of estrogen on nigrostriatal DA neurons in this model of PD.
Collapse
Affiliation(s)
- Arnulfo Quesada
- Department of Neurobiology, Laboratory of Neuroendocrinology, Brain Research Institute, David Geffen School of Medicine UCLA, Los Angeles, California 90095-1763, USA.
| | | |
Collapse
|
34
|
Ferraz AC, Xavier LL, Hernandes S, Sulzbach M, Viola GG, Anselmo-Franci JA, Achaval M, Da Cunha C. Failure of estrogen to protect the substantia nigra pars compacta of female rats from lesion induced by 6-hydroxydopamine. Brain Res 2003; 986:200-5. [PMID: 12965246 DOI: 10.1016/s0006-8993(03)03198-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The immunostaining for tyrosine hydroxylase (TH) in the substantia nigra pars compacta (SNpc) and in the ventral tegmental area (VTA) after intranigral infusion of 6-hydroxydopamine (6-OHDA, 6 microg/side) was analyzed in ovariectomized adult female Wistar rats. Estrogen replacement for 52 days (400-microg 17-beta-estradiol capsules) did not prevent the loss of TH-immunoreactive cells induced by 6-OHDA in the SNpc. This result indicates that the neuroprotective effect of dopaminergic mesencephalic cells is not observed with long-term estrogen replacement.
Collapse
Affiliation(s)
- Anete Curte Ferraz
- Laboratório de Fisiologia e Farmacologia do Sistema Nervoso Central, Departamento de Fisiologia e Farmacologia, Universidade Federal do Paraná, CP 19.031, 81.531-990 Curitiba, PR, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Murray HE, Pillai AV, McArthur SR, Razvi N, Datla KP, Dexter DT, Gillies GE. Dose- and sex-dependent effects of the neurotoxin 6-hydroxydopamine on the nigrostriatal dopaminergic pathway of adult rats: differential actions of estrogen in males and females. Neuroscience 2003; 116:213-22. [PMID: 12535954 DOI: 10.1016/s0306-4522(02)00578-x] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Epidemiological and clinical studies provide growing evidence for marked sex differences in the incidence of certain neurological disorders that are largely attributed to the neuroprotective effects of estrogen. Thus there is a keen interest in the clinical potential of estrogen-related compounds to act as novel therapeutic agents in conditions of neuronal injury and neurodegeneration such as Parkinson's disease. Studies employing animal models of neurodegeneration in ovariectomised female rats treated with estrogen support this hypothesis, yet experimental evidence for sex differences in the CNS response to direct neurotoxic insult is limited and, as yet, few studies have addressed the role played by endogenously produced hormones in neuroprotection. Therefore, in this study we aimed to determine (1) whether the prevailing levels of sex steroid hormones in the intact rat provide a degree of protection against neuronal assault in females compared with males and (2) whether sex differences depend solely on male/female differences in circulating estrogen levels or whether androgens could also play a role. Using the selective, centrally administered neurotoxin 6-hydroxydopamine, which induces a lesion in the nigrostriatal dopaminergic pathway similar to that seen in Parkinson's disease, we have demonstrated a sexually dimorphic (male-dominant), dose-dependent susceptibility in rats. Furthermore, following gonadectomy, dopamine depletion resulting from a submaximal dose of 6-hydroxydopamine (1 microg) was reduced in male rats, whereas in females, ovariectomy enhanced dopamine depletion. Administration of the nonaromatizable androgen dihydrotestosterone to gonadectomized animals had no significant effect on 6-hydroxydopamine toxicity in either males or females, whereas treatment of gonadectomized males and females with physiological levels of estrogen restored the extent of striatal dopamine loss to that seen in intact rats, viz, estrogen therapy reduced lesion size in females but increased it in males. Taken together, our findings strongly suggest that there are sex differences in the mechanisms whereby nigrostriatal dopaminergic neurones respond to injury. They also reveal that the reported clinically beneficial effects of estrogen in females may not be universally adopted for males. While the reasons for this gender-determined difference in response to the activational action of estrogen are unknown, we hypothesize that they may well be related to the early organizational events mediated by sex steroid hormones, which ultimately result in the sexual differentiation of the brain.
Collapse
Affiliation(s)
- H E Murray
- Department of Neuroendocrinology, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, London W12 0NN, UK.
| | | | | | | | | | | | | |
Collapse
|
36
|
Myers RE, Anderson LI, Dluzen DE. Estrogen, but not testosterone, attenuates methamphetamine-evoked dopamine output from superfused striatal tissue of female and male mice. Neuropharmacology 2003; 44:624-32. [PMID: 12668048 DOI: 10.1016/s0028-3908(03)00043-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The gonadal steroid hormone, estrogen, has the capacity to function as a neuroprotectant against methamphetamine (MA)-induced neurotoxicity of the nigrostriatal dopaminergic system within female, but not male, mice. In an attempt to understand some of the bases for this effect of estrogen, the incipient effects of MA upon evoked dopamine output from superfused striatal tissue fragments of gonadectomized female and gonadectomized as well as intact male mice were evaluated under conditions where estrogen (or testosterone) was present in the medium. The amount of dopamine evoked by MA was significantly reduced when estrogen was co-infused with MA. This attenuation was obtained with striatal tissue fragments of gonadectomized female and gonadectomized and intact male mice. In contrast to estrogen, co-infusion of testosterone failed to produce an overall statistically significant change in MA-evoked dopamine output within superfused striatal tissue fragments of gonadectomized female and male mice. In this way, the gonadal steroid hormones, estrogen and testosterone, exert differential modulatory effects upon MA-evoked dopamine output from superfused striatal tissue fragments. However, similar effects to these gonadal steroid hormones were observed between gonadectomized female and gonadectomized or intact male mice. These data reveal an absence of a sexual dimorphism in striatal responsiveness with regard to estrogen's ability to alter MA-evoked DA output. Accordingly, the sexually dimorphic capacity for estrogen to function as a neuroprotectant may involve a composite of actions upon the nigrostriatal dopaminergic system involving events/sites other than the initial stimulation of dopamine output.
Collapse
Affiliation(s)
- R E Myers
- Department of Anatomy Northeastern Ohio Universities College of Medicine (NEOUCOM), 4209 State Route 44, PO Box 95, Rootstown 44272-0095, USA
| | | | | |
Collapse
|
37
|
Dluzen DE, Park JH, Kim K. Modulation of olfactory bulb tyrosine hydroxylase and catecholamine transporter mRNA by estrogen. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 108:121-8. [PMID: 12480184 DOI: 10.1016/s0169-328x(02)00520-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Since estrogen exerts wide ranging effects within the central nervous system, it is important to investigate the sites and actions of this gonadal steroid hormone at extra-hypothalamic locations. In the present report, the effects of estrogen upon catecholaminergic function within the olfactory bulb were examined. To assess the role of estrogen at this site, ovariectomized mice received either no further hormonal treatment or were treated with estrogen, the anti-estrogen, tamoxifen, or a combination of estrogen and tamoxifen as administered in a 21-day release pellet. At 14 days post-hormonal treatment, the olfactory bulbs were assayed for mRNA levels of tyrosine hydroxylase, dopamine transporter and norepinephrine transporter using competitive-PCR. Tyrosine hydroxylase mRNA levels in either estrogen or estrogen+tamoxifen treated females were significantly decreased compared with non-hormonally treated controls. In addition, tyrosine hydroxylase mRNA levels of tamoxifen-treated mice were significantly greater than that of estrogen-treated mice. Dopamine transporter mRNA levels of tamoxifen-treated females were significantly greater than that of non-hormonally treated controls and estrogen treated mice. The combination of estrogen+tamoxifen significantly increased dopamine transporter mRNA levels compared to that of estrogen treated mice. No overall statistically significant differences in norepinephrine transporter mRNA levels were obtained among the four treatment groups. The data demonstrate that estrogen can exert significant modulatory effects upon olfactory bulb catecholaminergic function. Therefore, events which alter estrogen levels (menstrual/estrogen cycle, pregnancy/lactation, menopause, tamoxifen treatment) can modulate olfactory bulb catecholaminergic functions which may be involved with the detection and processing of olfactory stimuli.
Collapse
Affiliation(s)
- Dean E Dluzen
- Department of Anatomy, Northeastern Ohio Universities College of Medicine (NEOUCOM), 4209 State Route 44, PO Box 95, Rootstown 44272-0095, USA.
| | | | | |
Collapse
|
38
|
Gélinas S, Martinoli MG. Neuroprotective effect of estradiol and phytoestrogens on MPP+-induced cytotoxicity in neuronal PC12 cells. J Neurosci Res 2002; 70:90-6. [PMID: 12237867 DOI: 10.1002/jnr.10315] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A large body of experimental evidence supports a role for oxidative stress as a mediator of nerve cell death in Parkinson's disease. To better understand the cellular insult of oxidative stress on dopaminergic neurons, we studied the cytotoxic effect of the 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) metabolite, 1-methyl-4-phenyl pyridium (MPP(+)), on several parameters of cell distress using neuronal PC12 cells. We also measured the level of protein expression for the dopamine transporter and the estrogen receptors alpha and beta. Since estrogens have been reported to prevent neuronal degeneration caused by increased oxidative burden, we investigated the ability of 17beta-estradiol, the stereoisomer 17alpha-estradiol, and several phytoestrogens to rescue neuronal PC12 cells submitted to MPP(+)-induced cytotoxicity. Our results consistently show a protective effect of 17alpha-estradiol, 17beta-estradiol and certain phytoestrogens such as quercetin and resveratrol, in neuronal PC12 cells treated with MPP(+). In our cellular paradigm, phytoestrogens coumestrol, genistein, and kaempferol did not revert MPP(+)-induced cellular death. By Western blot, we demonstrated that administration of MPP(+) alone decrease dopamine transporter expression, while treatments with MPP(+) together with 17alpha-estradiol, 17beta-estradiol, quercetin, or resveratrol could restore dopamine transporter protein expression to control levels. Moreover, the same treatments did not modulate alpha estrogen receptor or beta estrogen receptor expression. By these studies, we aim to provide more evidence for the involvement of phytoestrogens in the process of neuroprotection and to test our hypothesis that some of these compounds may act as neuroprotective molecules and have a lesser hormonal effect than estrogens.
Collapse
Affiliation(s)
- Sylvie Gélinas
- Department of Biochemistry, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | | |
Collapse
|
39
|
Abstract
Increasing evidence suggests that estrogens may protect the nigrostriatal dopaminergic pathway affected in Parkinson's disease (PD). Animal studies show that estrogens influence the synthesis, release, and metabolism of dopamine and can modulate dopamine receptor expression and function. Some clinical studies suggest that PD symptoms may be exacerbated after menopause and delayed or alleviated with hormone replacement therapy, but others have failed to observe positive estrogenic effects. The conflicting findings suggest that several variables, including age, estrogen dose and formulation, and timing and length of dosing period, may determine whether benefits are seen and the nature of these benefits. Further investigation is therefore needed for the relationship between estrogens and the nigrostriatal dopaminergic system.
Collapse
Affiliation(s)
- Lisa M Shulman
- The Rosalyn Newman Scholar of Clinical Research in Parkinson's Disease, University of Maryland School of Medicine, 22 South Greene Street N4W46, Baltimore, MD 21201, USA.
| |
Collapse
|
40
|
Gao X, Dluzen DE. Tamoxifen abolishes estrogen's neuroprotective effect upon methamphetamine neurotoxicity of the nigrostriatal dopaminergic system. Neuroscience 2001; 103:385-94. [PMID: 11246153 DOI: 10.1016/s0306-4522(01)00014-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The effects of 17beta-estradiol and the anti-estrogen, tamoxifen, on methamphetamine-induced neurotoxicity of the nigrostriatal dopaminergic system were examined in ovariectomized CD-1 mice. In Experiment 1, striatal dopamine concentrations from estrogen treated mice were significantly greater than that from non-estrogen treated mice following methamphetamine. Dopamine concentrations from estrogen+tamoxifen+methamphetamine treated mice were decreased compared to estrogen+methamphetamine treated mice and not significantly different from those of tamoxifen+methamphetamine treated mice or mice receiving methamphetamine alone. These results suggest that estrogen is functioning as a neuroprotectant of methamphetamine-induced nigrostriatal dopaminergic neurotoxicity and that this neuroprotective effect of estrogen is abolished in the presence of tamoxifen. In Experiment 2, estrogen administration after methamphetamine treatment did not produce any significant changes in dopamine concentrations compared with methamphetamine treatment alone. The data from Experiment 2 show that estrogen cannot reverse the methamphetamine-induced neurotoxicity upon the nigrostriatal dopaminergic system. Similar results were observed for the potassium-stimulated dopamine outputs from these treatment conditions as evaluated with in vitro superfusion, although a difference between the two measures for the estrogen+methamphetamine treated group was obtained in Experiment 1. These results have important implications for estrogen-tamoxifen interactions upon the nigrostriatal dopaminergic system and the gender differences which are observed in Parkinson's disease and animal models of nigrostriatal dopaminergic neurotoxicity as well as for the proposed use of tamoxifen in pre-menopausal women at risk for breast cancer.
Collapse
Affiliation(s)
- X Gao
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272-0095, USA
| | | |
Collapse
|
41
|
Muñoz E, Pastor P, José Martí M, Valldeoriola F, Oliva R, Tolosa E. [Sporadic and familial Parkinson's disease: comparative study]. Med Clin (Barc) 2001; 116:601-4. [PMID: 11412643 DOI: 10.1016/s0025-7753(01)71919-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Several studies have shown that 13 to 33% of patients with Parkinson's disease (PD) exhibit a positive familial history. The goals of this work were to identify patients with familial PD and to analyse whether there existed distinctive features between familial and sporadic cases. PATIENTS AND METHOD 402 patients with PD from the Hospital Clínic i Universitari of Barcelona were evaluated prospectively. Clinical assessment was done using different scales in 169 patients. The disease was classified as tremorigenic, rigid or mixed according to the predominant symptoms. RESULTS The frequency of familial PD was 13%. The age at onset was not different between familial and sporadic cases but it was significantly higher in females (57.4 [13] years) than in males (54.8 [11.4] years) (p < 0.05). The tremorigenic type of PD was more common in familial cases (35.5%) (p < 0.05). In familial PD cases, the age at onset was lower in descendents (53 [13] years) than in parents (68 [7.8] years) (p = 0.001). CONCLUSIONS Genetic factors may play an important role in the development of PD and gender-associated factors may modulate the age at onset. Familial PD cases differ from sporadic cases in the higher frequency of predominantly tremorigenic forms. The lower age at onset in descendents than in parents suggests the existence of a genetic anticipation phenomenon in familial PD.
Collapse
Affiliation(s)
- E Muñoz
- Unidad de Movimientos Anormales, Servicios de Neurología, Hospital Clínic i Universitari de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Shepherd JE. Effects of estrogen on congnition mood, and degenerative brain diseases. JOURNAL OF THE AMERICAN PHARMACEUTICAL ASSOCIATION (WASHINGTON, D.C. : 1996) 2001; 41:221-8. [PMID: 11297335 DOI: 10.1016/s1086-5802(16)31233-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To review research findings on the effects of estrogen on cognition, mood, memory, and degenerative brain disease in women. DATA SOURCES English-language journal articles published primarily since 1995, retrieved from a MEDLINE search and from bibliographies of selected reviews. STUDY SELECTION Investigational studies, clinical trials, and review articles examining the effects of estrogen on the central nervous system. DATA SYNTHESIS Although scientific study of the brain is in its infancy, numerous studies indicate that estrogen is essential to optimal brain function. Estrogen has been shown to increase cerebral blood flow, act as an antiinflammatory agent, enhance activity at neuronal synapses, and exert direct neuroprotective and neurotrophic effects on brain tissue. Through these varied mechanisms, estrogen strongly influences mood and cognition, and the decline of this hormone at menopause can produce significant emotional and cognitive problems in women. CONCLUSION Pharmacists can educate women about the various mood and memory changes that can occur during perimenopause and how estrogen replacement therapy may lead to improvements in brain function. The potential use of estrogen replacement therapy to reduce the risk of Alzheimer's disease and ease the symptoms of Parkinson's disease could have a profound effect on women, their families, and society as a whole.
Collapse
|
43
|
Abstract
This review highlights recent evidence from clinical and basic science studies supporting a role for estrogen in neuroprotection. Accumulated clinical evidence suggests that estrogen exposure decreases the risk and delays the onset and progression of Alzheimer's disease and schizophrenia, and may also enhance recovery from traumatic neurological injury such as stroke. Recent basic science studies show that not only does exogenous estradiol decrease the response to various forms of insult, but the brain itself upregulates both estrogen synthesis and estrogen receptor expression at sites of injury. Thus, our view of the role of estrogen in neural function must be broadened to include not only its function in neuroendocrine regulation and reproductive behaviors, but also to include a direct protective role in response to degenerative disease or injury. Estrogen may play this protective role through several routes. Key among these are estrogen dependent alterations in cell survival, axonal sprouting, regenerative responses, enhanced synaptic transmission and enhanced neurogenesis. Some of the mechanisms underlying these effects are independent of the classically defined nuclear estrogen receptors and involve unidentified membrane receptors, direct modulation of neurotransmitter receptor function, or the known anti-oxidant activities of estrogen. Other neuroprotective effects of estrogen do depend on the classical nuclear estrogen receptor, through which estrogen alters expression of estrogen responsive genes that play a role in apoptosis, axonal regeneration, or general trophic support. Yet another possibility is that estrogen receptors in the membrane or cytoplasm alter phosphorylation cascades through direct interactions with protein kinases or that estrogen receptor signaling may converge with signaling by other trophic molecules to confer resistance to injury. Although there is clear evidence that estradiol exposure can be deleterious to some neuronal populations, the potential clinical benefits of estrogen treatment for enhancing cognitive function may outweigh the associated central and peripheral risks. Exciting and important avenues for future investigation into the protective effects of estrogen include the optimal ligand and doses that can be used clinically to confer benefit without undue risk, modulation of neurotrophin and neurotrophin receptor expression, interaction of estrogen with regulated cofactors and coactivators that couple estrogen receptors to basal transcriptional machinery, interactions of estrogen with other survival and regeneration promoting factors, potential estrogenic effects on neuronal replenishment, and modulation of phenotypic choices by neural stem cells.
Collapse
|
44
|
Dluzen DE, McDermott JL. Neuroprotective role of estrogen upon methamphetamine and related neurotoxins within the nigrostriatal dopaminergic system. Ann N Y Acad Sci 2000; 914:112-26. [PMID: 11085314 DOI: 10.1111/j.1749-6632.2000.tb05189.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this report we describe some of the data on the capacity for estrogen to function as a neuroprotectant of the nigrostriatal dopaminergic (NSDA) system. The data show that estrogen (E) can alter two different response characteristics to NSDA neurotoxins. The first being that striatal DA concentrations of ovariectomized rodents treated with E are consistently greater than non-E-treated animals in response to neurotoxins which produce degeneration of the NSDA system. The second being that E significantly reduces the amount of DA output upon initial exposure to the NSDA neurotoxin, 1-methyl-4-phenylpyridium ion (MPP+). At present, it is not known whether these two response characteristics are related. An intriguing possibility is that the E-dependent changes in initial DA output are related to the resultant neurotoxicity (attenuations in DA concentration reductions). So far our incipient findings do not seem to support this eventuality. However, additional testing on this topic is required. The present data suggest that one of the mechanisms by which E can exert these effects is through inhibition of DAT activity. This conclusion results from data which show that E produces: 1) an inhibition of [3H]DA uptake, 2) a reduction in DA clearance rates, and 3) an effect upon DA recovery that is similar to that observed to the putative DA uptake blocker, nomifensine. The capacity and significance for steroid hormones to modulate neurotransmitter transporters has been recently reviewed.
Collapse
Affiliation(s)
- D E Dluzen
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA.
| | | |
Collapse
|
45
|
Arvin M, Fedorkova L, Disshon KA, Dluzen DE, Leipheimer RE. Estrogen modulates responses of striatal dopamine neurons to MPP(+): evaluations using in vitro and in vivo techniques. Brain Res 2000; 872:160-71. [PMID: 10924688 DOI: 10.1016/s0006-8993(00)02511-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In vitro superfusion and in vivo electrochemistry were used to investigate the role of estrogen in modulating MPP(+)-induced dopamine output in the corpus striatum and nucleus accumbens of ovariectomized female rats. For in vitro superfusion experiments, dopamine and dihydroxyphenylacetic acid release were determined using HPLC with electrochemical detection from superfusion of corpus striatum fragments with Kreb's ringer phosphate buffer pulsed with MPP(+) alone or MPP(+) with estrogen. The in vivo electrochemistry experiments recorded the dopamine signal from carbon fiber microelectrodes stereotaxically passed through the corpus striatum and nucleus accumbens. Dopamine release was stimulated by pressure ejection of MPP(+) alone or in combination with estrogen through glass micropipettes fastened to the electrodes. Dopamine output from superfusion chambers which received infusion of MPP(+) with estrogen showed significantly lower output of dopamine compared with chambers which received MPP(+) alone. Outputs of dihydroxyphenylacetic acid did not increase following MPP(+) infusions. Data from the electrochemistry experiments demonstrated that estrogen significantly reduced both the amplitude and clearance rates of the MPP(+)-evoked dopamine signal in both the corpus striatum and nucleus accumbens. Results of this study demonstrate that: (1) MPP(+) evokes striatal dopamine release and this effect is significantly reduced in the presence of estrogen as determined by both in vivo electrochemistry and in vitro superfusion: (2) similar, albeit attenuated effects are observed in the nucleus accumbens as determined with in vivo electrochemistry; (3) estrogen acts to inhibit the clearance of dopamine in both the striatum and nucleus accumbens; and (4) estrogen may function as a neuroprotectant by reducing the uptake of neurotoxin into dopaminergic neurons.
Collapse
Affiliation(s)
- M Arvin
- Department of Biological Sciences, Youngstown State University, Youngstown, OH 44555, USA
| | | | | | | | | |
Collapse
|
46
|
Disshon KA, Dluzen DE. Estrogen reduces acute striatal dopamine responses in vivo to the neurotoxin MPP+ in female, but not male rats. Brain Res 2000; 868:95-104. [PMID: 10841892 DOI: 10.1016/s0006-8993(00)02329-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The effects of in vivo estrogen treatment upon MPP(+)-induced dopamine (DA) release were determined using in vivo microdialysis in female and male rats. Ovariectomized female rats were implanted or not with an estrogen pellet (0.1 mg, 17beta estradiol) and subjected to microdialysis 6 days later. After baseline DA release was determined, 5 mM MPP(+) was infused through the microdialysis probe for one 20-min interval. Perfusion resumed with normal medium for the duration of the experiment. A significant attenuation of MPP(+)-induced DA release was obtained in estrogen-treated females. One week later, striatal DA and dihydroxyphenylacetic acid (DOPAC) concentrations were determined for the lesioned and non-lesioned striata of each animal. MPP(+) infusion significantly decreased striatal DA concentrations, however, there was no effect of estrogen treatment on striatal DA depletion. This experiment was repeated using orchidectomized male rats treated with 0, 0.1, or 5 mg estradiol. In contrast to the females, no differences in MPP(+)-induced DA release were seen among these males, and there was no significant effect of the varying estrogen treatments on striatal DA or DOPAC concentrations. These results demonstrate that in vivo estrogen treatment attenuates MPP(+)-induced striatal DA release in gonadectomized female, but not male, rats.
Collapse
Affiliation(s)
- K A Disshon
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272-0095, USA
| | | |
Collapse
|
47
|
Schmidt BM, Gerdes D, Feuring M, Falkenstein E, Christ M, Wehling M. Rapid, nongenomic steroid actions: A new age? Front Neuroendocrinol 2000; 21:57-94. [PMID: 10662536 DOI: 10.1006/frne.1999.0189] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the traditional theory of steroid action, steroids bind to intracellular receptors and modulate nuclear transcription after translocation of steroid-receptor complexes into the nucleus. Due to similarities of molecular structure, specific receptors for steroids, vitamin D(3) derivatives, and thyroid hormone are considered to represent a superfamily of steroid receptors. While genomic steroid effects characterized by their delayed onset of action and their sensitivity to blockers of transcription and protein synthesis have been known for several decades, rapid actions of steroids have been more widely recognized and characterized in detail only recently. Rapid effects of steroids, thyroid hormones, and the steroid hormone metabolite of vitamin D(3), 1alpha, 25-dihydroxyvitamin D(3), on cellular signaling and function may be transmitted by specific membrane receptors. Binding sites in membranes have been characterized, exposing binding features compatible with an involvement in rapid steroid signaling. Characteristics of putative membrane receptors are completely distinct from intracellular steroid receptors, a fact which is further supported by the inability of classic steroid receptor antagonists to block nongenomic steroid actions. A putative progesterone membrane receptor has been cloned and functionally expressed with regard to progesterone binding. Development of drugs that specifically affect nongenomic action alone or even both modes of action may find applications in various, areas such as in the cardiovascular and central nervous systems and treatment of preterm labor, infertility, and electrolyte abnormalities.
Collapse
Affiliation(s)
- B M Schmidt
- Institute of Clinical Pharmacology, University of Heidelberg, Mannheim, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Disshon KA, Dluzen DE. Use of in vitro superfusion to assess the dynamics of striatal dopamine clearance: influence of estrogen. Brain Res 1999; 842:399-407. [PMID: 10526136 DOI: 10.1016/s0006-8993(99)01863-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To determine the feasibility of assessing dopamine uptake using in vitro superfusion, striatal tissue from ovariectomized female rats was infused with dopamine (1 microM), nomifensine (1 mM), or a combination of dopamine and nomifensine. Treatment with nomifensine or dopamine/nomifensine increased the recovery of dopamine in the effluent samples as compared to treatment with dopamine alone. In Experiment 2, the striatal tissue was treated with varying concentrations (0, 3, 30 or 300 nM) estradiol throughout the superfusion and subsequently given a dopamine (1 microM) challenge. The recovery of dopamine was enhanced in the presence of 3 and 30 nM estradiol. These results show that (1) in vitro superfusion can be used to dynamically evaluate dopamine recovery, and (2) estradiol, like nomifensine, increases the recovery of exogenously applied dopamine from the striata of ovariectomized female rats. Such increases in dopamine recovery with estrogen and similarities to that obtained with nomifensine suggest that estrogen may be inhibiting dopamine uptake from these striatal tissue fragments. Moreover, the doses at which estrogen can exert these effects insinuates a physiological role for this process. Our data provide a clear functional demonstration for one of the mechanisms by which estradiol can modulate striatal dopamine neurons, that of an uptake inhibitor. Such a mechanism has important implications with regard to estradiol's capacity to function as a neuroprotectant of the nigrostriatal dopaminergic system through inhibition of uptake of neurotoxins which can produce neurodegeneration of striatal dopamine neurons.
Collapse
Affiliation(s)
- K A Disshon
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, 4209 State Route 44, P.O. Box 95, Rootstown, OH 44272-0095, USA
| | | |
Collapse
|
49
|
Christ M, Haseroth K, Falkenstein E, Wehling M. Nongenomic steroid actions: fact or fantasy? VITAMINS AND HORMONES 1999; 57:325-73. [PMID: 10232053 DOI: 10.1016/s0083-6729(08)60647-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the common theory of steroid action, steroids bind to intracellular receptors and modulate nuclear transcription after translocation of steroid--receptor complexes into the nucleus. Due to homologies of molecular structure, specific receptors for steroids, vitamin D3, and thyroid hormone are considered to represent a superfamily of steroid receptors. While genomic steroid effects being characterized by their delayed onset of action and their sensitivity to blockers of transcription and protein synthesis have been known for several decades, very rapid actions of steroids have been more widely recognized and characterized in detail only recently. Rapid effects of steroids, vitamin D3, and thyroid hormones on cellular signaling and function may be transmitted by specific membrane receptors. Although no receptor of this kind has been cloned up to now, binding sites in membranes have been characterized exposing binding features compatible with an involvement in rapid steroid signaling. Characteristics of putative membrane receptors were completely different from those of intracellular steroid receptors, which was further supported by the inability of classic steroid receptor antagonists to inhibit nongenomic steroid actions. Development of drugs that specifically affect nongenomic action alone or even both modes of actions may find applications in various areas such as the cardiovascular and central nervous systems and treatment of preterm labor, infertility, and electrolyte homeostasis. To acquaint the reader with major aspects of nongenomic steroid actions, these effects on cellular function will be summarized, potentially related binding sites in membranes discussed, and the physiological or pathophysiological relevance of nonclassic actions exemplified.
Collapse
Affiliation(s)
- M Christ
- Institute of Clinical Pharmacology, Faculty for Clinical Medicine at Mannheim, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
50
|
Disshon KA, Boja JW, Dluzen DE. Inhibition of striatal dopamine transporter activity by 17beta-estradiol. Eur J Pharmacol 1998; 345:207-11. [PMID: 9600639 DOI: 10.1016/s0014-2999(98)00008-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Striatal synaptosomes from ovariectomized rats were prepared to examine the effect of 17beta-estradiol on [3H]dopamine uptake. Estradiol inhibited [3H]dopamine uptake in a dose-dependent manner, with an IC50 of 7.2 microM. Use of identical concentrations of progesterone had no effect on [3H]dopamine uptake. The effects of estradiol were exerted by decreasing the affinity of the transporter for dopamine, as revealed by a dose-dependent increase in the Km. The Km values for 0 (control), 10, and 100 microM estradiol were 108+/-11 258+/-44 and 415+/-40 nM, respectively, with each of the three concentrations tested being significantly different among each other. No statistically significant differences were obtained for the Vmax, with values for the three increasing doses being 9.2+/-0.8, 8.3+/-0.5 and 7.3+/-0.8 pmol/min per mg protein. These results demonstrate that estradiol, but not progesterone, inhibits striatal dopamine uptake by decreasing the affinity of the transporter for dopamine. Such a mechanism may serve as one of the bases for the modulatory effects of estradiol upon the nigrostriatal dopaminergic system.
Collapse
Affiliation(s)
- K A Disshon
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, Rootstown 44272-0095, USA
| | | | | |
Collapse
|