1
|
Nordengen K, Cappelletti C, Bahrami S, Frei O, Pihlstrøm L, Henriksen SP, Geut H, Rozemuller AJM, van de Berg WDJ, Andreassen OA, Toft M. Pleiotropy with sex-specific traits reveals genetic aspects of sex differences in Parkinson's disease. Brain 2024; 147:858-870. [PMID: 37671566 PMCID: PMC10907091 DOI: 10.1093/brain/awad297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
Parkinson's disease is an age-related neurodegenerative disorder with a higher incidence in males than females. The causes for this sex difference are unknown. Genome-wide association studies (GWAS) have identified 90 Parkinson's disease risk loci, but the genetic studies have not found sex-specific differences in allele frequency on autosomal chromosomes or sex chromosomes. Genetic variants, however, could exert sex-specific effects on gene function and regulation of gene expression. To identify genetic loci that might have sex-specific effects, we studied pleiotropy between Parkinson's disease and sex-specific traits. Summary statistics from GWASs were acquired from large-scale consortia for Parkinson's disease (n cases = 13 708; n controls = 95 282), age at menarche (n = 368 888 females) and age at menopause (n = 69 360 females). We applied the conditional/conjunctional false discovery rate (FDR) method to identify shared loci between Parkinson's disease and these sex-specific traits. Next, we investigated sex-specific gene expression differences in the superior frontal cortex of both neuropathologically healthy individuals and Parkinson's disease patients (n cases = 61; n controls = 23). To provide biological insights to the genetic pleiotropy, we performed sex-specific expression quantitative trait locus (eQTL) analysis and sex-specific age-related differential expression analysis for genes mapped to Parkinson's disease risk loci. Through conditional/conjunctional FDR analysis we found 11 loci shared between Parkinson's disease and the sex-specific traits age at menarche and age at menopause. Gene-set and pathway analysis of the genes mapped to these loci highlighted the importance of the immune response in determining an increased disease incidence in the male population. Moreover, we highlighted a total of nine genes whose expression or age-related expression in the human brain is influenced by genetic variants in a sex-specific manner. With these analyses we demonstrated that the lack of clear sex-specific differences in allele frequencies for Parkinson's disease loci does not exclude a genetic contribution to differences in disease incidence. Moreover, further studies are needed to elucidate the role that the candidate genes identified here could have in determining a higher incidence of Parkinson's disease in the male population.
Collapse
Affiliation(s)
- Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Chiara Cappelletti
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Faculty of Technology, Art and Design, OsloMet—Oslo Metropolitan University, 0130 Oslo, Norway
- Department of Research, Innovation and Education, Oslo University Hospital, 0424 Oslo, Norway
| | - Shahram Bahrami
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Oleksandr Frei
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Hanneke Geut
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Ole A Andreassen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
2
|
Shiraishi T, Bono K, Hiraki H, Manome Y, Oka H, Iguchi Y, Okano HJ. The impact of VPS35 D620N mutation on alternative autophagy and its reversal by estrogen in Parkinson's disease. Cell Mol Life Sci 2024; 81:103. [PMID: 38409392 PMCID: PMC10896810 DOI: 10.1007/s00018-024-05123-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/28/2024]
Abstract
VPS35 plays a key role in neurodegenerative processes in Alzheimer's disease and Parkinson's disease (PD). Many genetic studies have shown a close relationship between autophagy and PD pathophysiology, and specifically, the PD-causing D620N mutation in VPS35 has been shown to impair autophagy. However, the molecular mechanisms underlying neuronal cell death and impaired autophagy in PD are debated. Notably, increasing evidence suggests that Rab9-dependent "alternative" autophagy, which is driven by a different molecular mechanism that driving ATG5-dependent "conventional" autophagy, also contributes to neurodegenerative process. In this study, we investigated the relationship between alternative autophagy and VPS35 D620N mutant-related PD pathogenesis. We isolated iPSCs from the blood mononuclear cell population of two PD patients carrying the VPS35 D620N mutant. In addition, we used CRISPR-Cas9 to generate SH-SY5Y cells carrying the D620N variant of VPS35. We first revealed that the number of autophagic vacuoles was significantly decreased in ATG5-knockout Mouse Embryonic Fibroblast or ATG5-knockdown patient-derived dopaminergic neurons carrying the VPS35 D620N mutant compared with that of the wild type VPS35 control cells. Furthermore, estrogen, which activates alternative autophagy pathways, increased the number of autophagic vacuoles in ATG5-knockdown VPS35 D620N mutant dopaminergic neurons. Estrogen induces Rab9 phosphorylation, mediated through Ulk1 phosphorylation, ultimately regulating alternative autophagy. Moreover, estrogen reduced the apoptosis rate of VPS35 D620N neurons, and this effect of estrogen was diminished under alternative autophagy knockdown conditions. In conclusion, alternative autophagy might be important for maintaining neuronal homeostasis and may be associated with the neuroprotective effect of estrogen in PD with VPS35 D620N.
Collapse
Affiliation(s)
- Tomotaka Shiraishi
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Keiko Bono
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Hiromi Hiraki
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Yoko Manome
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan
| | - Hisayoshi Oka
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan.
| |
Collapse
|
3
|
Terrin F, Tesoriere A, Plotegher N, Dalla Valle L. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson's Disease. Cells 2023; 12:1486. [PMID: 37296608 PMCID: PMC10252697 DOI: 10.3390/cells12111486] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Sex hormones and genes on the sex chromosomes are not only key factors in the regulation of sexual differentiation and reproduction but they are also deeply involved in brain homeostasis. Their action is crucial for the development of the brain, which presents different characteristics depending on the sex of individuals. The role of these players in the brain is fundamental in the maintenance of brain function during adulthood as well, thus being important also with respect to age-related neurodegenerative diseases. In this review, we explore the role of biological sex in the development of the brain and analyze its impact on the predisposition toward and the progression of neurodegenerative diseases. In particular, we focus on Parkinson's disease, a neurodegenerative disorder that has a higher incidence in the male population. We report how sex hormones and genes encoded by the sex chromosomes could protect from the disease or alternatively predispose toward its development. We finally underline the importance of considering sex when studying brain physiology and pathology in cellular and animal models in order to better understand disease etiology and develop novel tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| | - Luisa Dalla Valle
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| |
Collapse
|
4
|
Ronchetti S, Labombarda F, Roig P, De Nicola AF, Pietranera L. Beneficial effects of the phytoestrogen genistein on hippocampal impairments of spontaneously hypertensive rats (SHR). J Neuroendocrinol 2023; 35:e13228. [PMID: 36690381 DOI: 10.1111/jne.13228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/09/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023]
Abstract
Hippocampal neuropathology is a recognized feature of the spontaneously hypertensive rat (SHR). The hippocampal alterations associate with cognitive impairment. We have shown that hippocampal abnormalities are reversed by 17β-estradiol, a steroid binding to intracellular receptors (estrogen receptor α and β subtypes) or the membrane-located G-protein coupled estradiol receptor. Genistein (GEN) is a neuroprotective phytoestrogen which binds to estrogen receptor β and G-protein coupled estradiol receptor. Here, we investigated whether GEN neuroprotection extends to SHR. For this purpose, we treated 5-month-old SHR for 2 weeks with 10 mg kg-1 daily s.c injections of GEN. We analyzed the expression of doublecortin+ neuronal progenitors, glial fibrillary acidic protein+ astrocytes and ionized calcium-binding adapter molecule 1+ microglia in the CA1 region and dentate gyrus of the hippocampus using immunocytochemistry, whereas a quantitative real-time polymerase chain reaction was used to measure the expression of pro- and anti-inflammatory factors tumor necrosis factor α, cyclooxygenase-2 and transforming growth factor β. We also evaluated hippocampal dependent memory using the novel object recognition test. The results showed a decreased number of doublecortin+ neural progenitors in the dentate gyrus of SHR that was reversed with GEN. The number of glial fibrillary acidic protein+ astrocytes in the dentate gyrus and CA1 was increased in SHR but significantly decreased by GEN treatment. Additionally, GEN shifted microglial morphology from the predominantly activated phenotype present in SHR, to the more surveillance phenotype found in normotensive rats. Furthermore, treatment with GEN decreased the mRNA of the pro-inflammatory factors tumor necrosis factor α and cyclooxygenase-2 and increased the mRNA of the anti-inflammatory factor transforming growth factor β. Discrimination index in the novel object recognition test was decreased in SHR and treatment with GEN increased this parameter. Our results indicate important neuroprotective effects of GEN at the neurochemical and behavioral level in SHR. Our data open an interesting possibility for proposing this phytoestrogen as an alternative therapy in hypertensive encephalopathy.
Collapse
Affiliation(s)
- Santiago Ronchetti
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Luciana Pietranera
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
5
|
Yuk JS, Jeong SH. Association Between Menopausal Hormone Therapy and Risk for Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1357-1367. [PMID: 37980684 PMCID: PMC10741322 DOI: 10.3233/jpd-230230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND The relationship between menopausal hormone therapy (MHT) and risk of Parkinson's disease (PD) remains controversial. OBJECTIVE This nationwide population-based cohort study investigated the association between MHT and PD development. METHODS Data from the National Health Insurance System of South Korea from 2007 to 2020 were used. The MHT group included women who underwent MHT for the first time between 2011-2014, while the non-MHT group included women who visited a healthcare provider for menopause during the same period but never received hormonal therapy. We used propensity score matching (1 : 1) to adjust for potential confounders, and Cox regression models to assess the association between MHT and PD. RESULTS We selected 303,260 female participants (n = 151,630 per MHT and non-MHT groups). The median age of the participants was 50 (48-54) years, and the follow-up period lasted 7.9 (6.9-8.9) years. Cox regression analysis revealed an increased risk of PD with MHT (hazard ratio [HR] 1.377, 95% confidence interval [CI] 1.184-1.602), particularly with tibolone (HR 1.554, 95% CI 1.297-1.861) and estrogen alone (HR 1.465, 95% CI 1.054-2.036). Tibolone and estrogen alone were linked to PD within three years; however, no association was observed after three years. In contrast, the use of combined estrogen-progesterone was linked to a higher risk of PD, which increased with the duration of MHT (HR 1.885, 95% CI 1.218-2.918 for over five years). CONCLUSIONS This study demonstrated that the MHT is closely associated with the risk of PD in a regimen- and duration-specific manner.
Collapse
Affiliation(s)
- Jin-Sung Yuk
- Department of Obstetrics and Gynecology, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Seong Ho Jeong
- Department of Neurology, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| |
Collapse
|
6
|
Chen LJ, Chen JR, Tseng GF. Modulation of striatal glutamatergic, dopaminergic and cholinergic neurotransmission pathways concomitant with motor disturbance in rats with kaolin-induced hydrocephalus. Fluids Barriers CNS 2022; 19:95. [PMID: 36437472 PMCID: PMC9701403 DOI: 10.1186/s12987-022-00393-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Hydrocephalus is characterized by abnormal accumulation of cerebrospinal fluid in the cerebral ventricles and causes motor impairments. The mechanisms underlying the motor changes remain elusive. Enlargement of ventricles compresses the striatum of the basal ganglia, a group of nuclei involved in the subcortical motor circuit. Here, we used a kaolin-injection juvenile rat model to explore the effects of acute and chronic hydrocephalus, 1 and 5 weeks post-treatment, respectively on the three major neurotransmission pathways (glutamatergic, dopaminergic and cholinergic) in the striatum. METHODS Rats were evaluated for motor impairments. Expressions of presynaptic and postsynaptic protein markers related to the glutamatergic, dopaminergic, and cholinergic connections in the striatum were evaluated. Combined intracellular dye injection and substance P immunohistochemistry were used to distinguish between direct and indirect pathway striatal medium spiny neurons (d and i-MSNs) for the analysis of their dendritic spine density changes. RESULTS Hydrocephalic rats showed compromised open-field gait behavior. However, male but not female rats displayed stereotypic movements and compromised rotarod performance. Morphologically, the increase in lateral ventricle sizes was greater in the chronic than acute hydrocephalus conditions. Biochemically, hydrocephalic rats had significantly decreased striatal levels of synaptophysin, vesicular glutamate transporter 1, and glutamatergic postsynaptic density protein 95, suggesting a reduction of corticostriatal excitation. The expression of GluR2/3 was also reduced suggesting glutamate receptor compositional changes. The densities of dendritic spines, morphological correlates of excitatory synaptic foci, on both d and i-MSNs were also reduced. Hydrocephalus altered type 1 (DR1) and 2 (DR2) dopamine receptor expressions without affecting tyrosine hydroxylase level. DR1 was decreased in acute and chronic hydrocephalus, while DR2 only started to decrease later during chronic hydrocephalus. Since dopamine excites d-MSNs through DR1 and inhibits i-MSNs via DR2, our findings suggest that hydrocephalus downregulated the direct basal ganglia neural pathway persistently and disinhibited the indirect pathway late during chronic hydrocephalus. Hydrocephalus also persistently reduced the striatal choline acetyltransferase level, suggesting a reduction of cholinergic modulation. CONCLUSIONS Hydrocephalus altered striatal glutamatergic, dopaminergic, and cholinergic neurotransmission pathways and tipped the balance between the direct and indirect basal ganglia circuits, which could have contributed to the motor impairments in hydrocephalus.
Collapse
Affiliation(s)
- Li-Jin Chen
- grid.411824.a0000 0004 0622 7222Department of Anatomy, College of Medicine, Tzu Chi University, No. 701, Section 3, Jhongyang Rd., Hualien, 97004 Taiwan
| | - Jeng-Rung Chen
- grid.260542.70000 0004 0532 3749Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Guo-Fang Tseng
- grid.411824.a0000 0004 0622 7222Department of Anatomy, College of Medicine, Tzu Chi University, No. 701, Section 3, Jhongyang Rd., Hualien, 97004 Taiwan
| |
Collapse
|
7
|
Hong S, Han K, Kim KS, Park CY. Risk of Neurodegenerative Diseases in Patients With Acromegaly: A Cohort Study. Neurology 2022; 99:e1875-e1885. [PMID: 36192177 DOI: 10.1212/wnl.0000000000201010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/09/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES A few recent studies have reported an association between insulin-like growth factor-1 (IGF-1) and neurodegenerative disease, but there was no report on any association between acromegaly and neurodegenerative disease. We investigated whether the risk of Alzheimer disease (AD), Parkinson disease, and other neurodegenerative diseases was increased among patients with acromegaly using nationwide data of Korea. METHODS We studied the association between acromegaly and Parkinson disease and dementia in 1,611 patients with acromegaly and controls (age-matched and sex-matched 8,055 participants with no diagnosis of acromegaly) from the National Health Insurance System database between 2006 and 2016 with a mean follow-up period of 7.34 years. Cox proportional hazards regression analysis was used to assess the risk of all outcomes in patients with acromegaly compared with controls with adjusting for age, sex, household income, place, type 2 diabetes, hypertension, and dyslipidemia. RESULTS The average age of the patients with acromegaly and the controls was 54.16 years (40.4% men). The incidence rate of Parkinson disease in patients with acromegaly (1.54 per 1,000 person-years) was significantly higher than that in the control group (0.55 per 1,000 person-years) (log-rank test p = 0.001). Acromegaly was associated with a higher risk of Parkinson disease (hazard ratio [HR] = 2.609, 95% CI: 1.410-2.609) than the control. In addition, acromegaly was associated with a higher risk of all-cause dementia (HR = 2.299, 95% CI: 1.362-3.881), Alzheimer disease (HR = 2.228, 95% CI: 1.191-4.168), and non-AD dementia (HR = 6.553, 95% CI: 1.754-24.482) than the control during the first 3 years after diagnosis and treatment. In subgroup analysis, diabetes was associated with higher risk of all-cause dementia (P for interaction = 0.028) in patients with acromegaly compared with controls. DISCUSSION Our study results suggest that acromegaly is associated with neurodegenerative disease. Further study is needed on the association between IGF-1/growth hormone level and neurodegenerative disease.
Collapse
Affiliation(s)
- Sangmo Hong
- From the Department of Internal Medicine (S.H.), Guri Hospital, Hanyang University, College of Medicine; Department of Statistics and Actuarial Science (K.H.), Soongsil University, Seoul; Department of Internal Medicine (K.-S.K.), CHA Bundang Medical Center, CHA University School of Medicine, Seongnam; and Department of Internal Medicine (C.-Y.P.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyungdo Han
- From the Department of Internal Medicine (S.H.), Guri Hospital, Hanyang University, College of Medicine; Department of Statistics and Actuarial Science (K.H.), Soongsil University, Seoul; Department of Internal Medicine (K.-S.K.), CHA Bundang Medical Center, CHA University School of Medicine, Seongnam; and Department of Internal Medicine (C.-Y.P.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyung-Soo Kim
- From the Department of Internal Medicine (S.H.), Guri Hospital, Hanyang University, College of Medicine; Department of Statistics and Actuarial Science (K.H.), Soongsil University, Seoul; Department of Internal Medicine (K.-S.K.), CHA Bundang Medical Center, CHA University School of Medicine, Seongnam; and Department of Internal Medicine (C.-Y.P.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol-Young Park
- From the Department of Internal Medicine (S.H.), Guri Hospital, Hanyang University, College of Medicine; Department of Statistics and Actuarial Science (K.H.), Soongsil University, Seoul; Department of Internal Medicine (K.-S.K.), CHA Bundang Medical Center, CHA University School of Medicine, Seongnam; and Department of Internal Medicine (C.-Y.P.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Pedersini R, di Mauro P, Amoroso V, Castronovo V, Zamparini M, Monteverdi S, Laini L, Schivardi G, Cosentini D, Grisanti S, Marelli S, Ferini Strambi L, Berruti A. Sleep disturbances and restless legs syndrome in postmenopausal women with early breast cancer given adjuvant aromatase inhibitor therapy. Breast 2022; 66:162-168. [PMID: 36288635 PMCID: PMC9593725 DOI: 10.1016/j.breast.2022.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
INTRODUCTION Whether adjuvant therapy with aromatase inhibitors (AIs) causes sleep disturbances or not in postmenopausal women with early breast cancer (EBC) is still a controversial issue. METHODS Between March 2014 and November 2017, validated questionnaires for assessing insomnia, anxiety, depression, quality of life (QoL) and restless legs syndrome (RLS) were administered to 160 EBC patients at baseline and after 3, 6, 12, and 24 months of AI therapy. RESULTS AI therapy significantly decreased the patients' QoL, but did not influence insomnia, anxiety or depression. However, it significantly increased the frequency and severity of RLS. Patients with RLS at baseline (19%) or who developed RLS during AI therapy (26.3%) reported statistically lower quality of sleep, higher anxiety and depression, and worse QoL compared to patients who never reported RLS (54.7%). CONCLUSION Although AI therapy does not affect sleep quality, it may increase RLS frequency. The presence of RLS could identify a group of EBC patients who may benefit from psychological support.
Collapse
Affiliation(s)
- Rebecca Pedersini
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy,SSVD Breast Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Pierluigi di Mauro
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy,Corresponding author. Oncologia Medica, ASST Spedali Civili, Piazzale Spedali Civili 1, 20123, Brescia, Italy.
| | - Vito Amoroso
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Vincenza Castronovo
- Sleep Disorders Center, Department of Clinical Neurosciences, San Raffaele Hospital and University Vita-Salute San Raffaele, Milan, Italy
| | - Manuel Zamparini
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| | | | - Lara Laini
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Greta Schivardi
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Deborah Cosentini
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Salvatore Grisanti
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Sara Marelli
- Sleep Disorders Center, Department of Clinical Neurosciences, San Raffaele Hospital and University Vita-Salute San Raffaele, Milan, Italy
| | - Luigi Ferini Strambi
- Sleep Disorders Center, Department of Clinical Neurosciences, San Raffaele Hospital and University Vita-Salute San Raffaele, Milan, Italy
| | - Alfredo Berruti
- Medical Oncology Department, ASST-Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
9
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
10
|
Qiu Y, Yu J, Tang L, Ren J, Shao M, Li S, Song Y, Cao W, Sun X. Association Between Sex Hormones and Visual Field Progression in Women With Primary Open Angle Glaucoma: A Cross-Sectional and Prospective Cohort Study. Front Aging Neurosci 2022; 13:756186. [PMID: 35002675 PMCID: PMC8741302 DOI: 10.3389/fnagi.2021.756186] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/02/2021] [Indexed: 11/15/2022] Open
Abstract
Purpose: We evaluated the level of sex hormones in female patients with primary open angle glaucoma (POAG) to determine whether they are associated with the onset and/or progression of POAG. Methods: The cross-sectional study enrolled 63 women with POAG and 56 healthy women as normal control subjects. Furthermore, 57 women with POAG were included and followed-up for at least 2 years in the cohort study. All subjects were evaluated for serum concentration of sex hormones [prolactin (PRL), luteinizing hormone (LH), testosterone (TESTO), follicle-stimulating hormone (FSH), progesterone (PROG), and estrogen (E2)] and underwent visual field (VF) examination. In the cross-sectional study, Spearman analysis, linear regression analysis, and logistic regression analysis were performed to assess risk factors for POAG in women. In the cohort study, Cox regression analyses and Kaplan–Meier survival analysis were performed to identify factors associated with VF progression in women with POAG. Results: In the cross-sectional study, the level of E2 was significantly lower in the POAG group than in the normal group (p < 0.05). Multiple logistic regression showed that the decreased level of E2 was a risk factor of POAG (OR = 0.27, 95% CI = 0.09–0.78, p < 0.05), especially in premenopausal subjects. In the cohort study, there were 29 non-progression subjects and 28 progression subjects. Patients in the progression group had significantly lower levels of E2 than those in the no progression group (p < 0.01). The decreased level of E2 at baseline was associated with POAG progression (HR = 0.08, 95% CI = 0.02–0.46, p < 0.05), especially in premenopausal subjects. Patients with POAG and with lower baseline E2 levels had significantly lower VF non-progression rates than patients with higher E2 levels (log-rank test p < 0.001), especially premenopausal subjects (log-rank test p < 0.05). Additionally, logistic regression analyses, Cox regression analyses, and Kaplan–Meier survival analysis showed that PROG, LH, FSH, and TESTO were risk factors of POAG and/or significantly associated with POAG progression. Conclusion: A decreased E2 level is a POAG risk factor and is associated with VF progression in women with POAG, especially in premenopausal subjects. Additionally, other sex hormones (PROG, LH, FSH, and TESTO) might also play a role in POAG pathogenesis.
Collapse
Affiliation(s)
- Yichao Qiu
- Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jian Yu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Li Tang
- Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jun Ren
- Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Mingxi Shao
- Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Shengjie Li
- Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai, China.,Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Yunxiao Song
- Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Wenjun Cao
- Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai, China.,Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University - Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University - Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
11
|
Chen P, Li B, Ou-Yang L. Role of estrogen receptors in health and disease. Front Endocrinol (Lausanne) 2022; 13:839005. [PMID: 36060947 PMCID: PMC9433670 DOI: 10.3389/fendo.2022.839005] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/26/2022] [Indexed: 12/14/2022] Open
Abstract
Estrogen receptors (ERs) regulate multiple complex physiological processes in humans. Abnormal ER signaling may result in various disorders, including reproductive system-related disorders (endometriosis, and breast, ovarian, and prostate cancer), bone-related abnormalities, lung cancer, cardiovascular disease, gastrointestinal disease, urogenital tract disease, neurodegenerative disorders, and cutaneous melanoma. ER alpha (ERα), ER beta (ERβ), and novel G-protein-coupled estrogen receptor 1 (GPER1) have been identified as the most prominent ERs. This review provides an overview of ERα, ERβ, and GPER1, as well as their functions in health and disease. Furthermore, the potential clinical applications and challenges are discussed.
Collapse
Affiliation(s)
| | - Bo Li
- *Correspondence: Bo Li, libo‐‐
| | | |
Collapse
|
12
|
Effect of Chronic Methylphenidate Treatment in a Female Experimental Model of Parkinsonism. Neurotox Res 2021; 39:667-676. [PMID: 33666887 DOI: 10.1007/s12640-021-00347-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/11/2021] [Accepted: 02/28/2021] [Indexed: 10/22/2022]
Abstract
Methylphenidate (MPH) is the most commonly prescribed drug for the treatment of ADHD in males and females. However, a majority of previous studies investigated the effect of MPH in only males, and little is known regarding consequences of female exposure to MPH. This is unfortunate because the few studies that have been conducted indicate that females have a greater sensitivity to MPH. Previous research in male mice has shown that chronic exposure to MPH causes dopaminergic neurons within the nigrostriatal pathway to be more sensitive to the Parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, estrogen has been shown to protect dopaminergic neurons from MPTP neurotoxicity. Therefore, in this study, we test the hypothesis that chronic MPH exposure in female mice will render dopaminergic neurons in the nigrostriatal pathway more sensitive to MPTP, and that estrogen may play a protective role. Interestingly, proestrus females exhibited greater sensitivity to MPTP, with significantly reduced dopaminergic neurons in the SN and significant increases in DA quinone production. Chronic MPH exposure contributed to GSH depletion, but surprisingly, it did not increase dopamine quinone levels or dopaminergic cell loss. There were no significant differences in anestrus animals, with the exception of a depletion in GSH seen when animals received chronic high-dose (10 mg/kg) MPH followed by MPTP. Thus, estrogen may actually sensitize neurons to MPTP in this model, and chronic MPH may contribute to GSH depletion within the striatum. This study provides insight into how chronic psychostimulant use may affect males and females differently.
Collapse
|
13
|
Kövesdi E, Szabó-Meleg E, Abrahám IM. The Role of Estradiol in Traumatic Brain Injury: Mechanism and Treatment Potential. Int J Mol Sci 2020; 22:E11. [PMID: 33374952 PMCID: PMC7792596 DOI: 10.3390/ijms22010011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023] Open
Abstract
Patients surviving traumatic brain injury (TBI) face numerous neurological and neuropsychological problems significantly affecting their quality of life. Extensive studies over the past decades have investigated pharmacological treatment options in different animal models, targeting various pathological consequences of TBI. Sex and gender are known to influence the outcome of TBI in animal models and in patients, respectively. Apart from its well-known effects on reproduction, 17β-estradiol (E2) has a neuroprotective role in brain injury. Hence, in this review, we focus on the effect of E2 in TBI in humans and animals. First, we discuss the clinical classification and pathomechanism of TBI, the research in animal models, and the neuroprotective role of E2. Based on the results of animal studies and clinical trials, we discuss possible E2 targets from early to late events in the pathomechanism of TBI, including neuroinflammation and possible disturbances of the endocrine system. Finally, the potential relevance of selective estrogenic compounds in the treatment of TBI will be discussed.
Collapse
Affiliation(s)
- Erzsébet Kövesdi
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Center for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pecs, Hungary;
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pecs, Hungary;
| | - István M. Abrahám
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Center for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pecs, Hungary;
| |
Collapse
|
14
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
15
|
Di T, Wang Y, Zhang Y, Sha S, Zeng Y, Chen L. Dopaminergic afferents from midbrain to dorsolateral bed nucleus of stria terminalis inhibit release and expression of corticotropin-releasing hormone in paraventricular nucleus. J Neurochem 2020; 154:218-234. [PMID: 32096869 DOI: 10.1111/jnc.14992] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/07/2020] [Accepted: 02/21/2020] [Indexed: 01/21/2023]
Abstract
Dopaminergic (DAergic) neurons of the midbrain ventral tegmental area (VTA) are known to regulate the hypothalamic-pituitary-adrenal (HPA) axis but have no direct projections to the paraventricular nucleus (PVN) of the hypothalamus. This study investigated whether VTA DAergic afferents modulate glutamatergic transmission-dependent GABAergic neurons in dorsolateral bed nucleus of stria terminalis (dlBNST) to affect the activity of the HPA-axis. Herein, we demonstrate that systemic administration of the neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or the VTA-injection of 1-methyl-4-phenylpyridinium ion (MPP+) in male mice (MPTP-mice and MPP+mice) caused a decline of tyrosine hydroxylase positive (TH+) cells in VTA with a reduction in TH+fibers in the dlBNST. MPTP-mice and MPP+mice displayed a clear increase in serum levels of corticosterone (CORT) and adrenocorticotropic hormone, corticotropin-releasing hormone (CRH) expression, and CRH neuron activity in PVN. The presynaptic glutamate release, glutamatergic synaptic transmission and induction of long-term potentiation in dlBNST of MPTP-mice were suppressed, and these effects were rescued by a D1-like DAergic receptor (D1R) agonist and mimicked in control dlBNST by blockade of D1R. MPTP-mice exhibited low expression of glutamic acid decarboxylase and dysfunction of the excitatory-dependent GABAergic circuit in dlBNST, and these effects were recovered by the administration of D1R agonist. Furthermore, either dlBNST-injection of D1R agonist or PVN-injection of GABAA receptor (GABAA R) agonist could correct the increased secretion and expression of CRH in MPTP-mice. The results indicate that the DAergic afferents from VTA enhance excitatory-dependent activation of GABAergic neurons in dlBNST, which suppress the activity of the HPA-axis.
Collapse
Affiliation(s)
- Tingting Di
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yajie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Sha Sha
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yanying Zeng
- Department of Gerontology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Jarras H, Bourque M, Poirier AA, Morissette M, Coulombe K, Di Paolo T, Soulet D. Neuroprotection and immunomodulation of progesterone in the gut of a mouse model of Parkinson's disease. J Neuroendocrinol 2020; 32:e12782. [PMID: 31430407 DOI: 10.1111/jne.12782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal symptoms appear in Parkinson's disease patients many years before motor symptoms, suggesting the implication of dopaminergic neurones of the gut myenteric plexus. Inflammation is also known to be increased in PD. We previously reported neuroprotection with progesterone in the brain of mice lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and hypothesised that it also has neuroprotective and immunomodulatory activities in the gut. To test this hypothesis, we investigated progesterone administered to adult male C57BL/6 mice for 10 days and treated with MPTP on day 5. In an additional experiment, progesterone was administered for 5 days following MPTP treatment. Ilea were collected on day 10 of treatment and microdissected to isolate the myenteric plexus. Dopaminergic neurones were reduced by approximately 60% and pro-inflammatory macrophages were increased by approximately 50% in MPTP mice compared to intact controls. These changes were completely prevented by progesterone administered before and after MPTP treatment and were normalised by 8 mg kg-1 progesterone administered after MPTP. In the brain of MPTP mice, brain-derived neurotrophic peptide (BDNF) and glial fibrillary acidic protein (GFAP) were associated with progesterone neuroprotection. In the myenteric plexus, increased BDNF levels compared to controls were measured in MPTP mice treated with 8 mg kg-1 progesterone started post MPTP, whereas GFAP levels remained unchanged. In conclusion, the results obtained in the present study show neuroprotective and anti-inflammatory effects of progesterone in the myenteric plexus of MPTP mice that are similar to our previous findings in the brain. Progesterone is non-feminising and could be used for both men and women in the pre-symptomatic stages of the disease.
Collapse
Affiliation(s)
- Hend Jarras
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Mélanie Bourque
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Andrée-Anne Poirier
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Katherine Coulombe
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Denis Soulet
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| |
Collapse
|
17
|
Activation of the G Protein-Coupled Estrogen Receptor (GPER) Increases Neurogenesis and Ameliorates Neuroinflammation in the Hippocampus of Male Spontaneously Hypertensive Rats. Cell Mol Neurobiol 2019; 40:711-723. [DOI: 10.1007/s10571-019-00766-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/22/2019] [Indexed: 01/20/2023]
|
18
|
A Novel Prodrug Approach for Central Nervous System-Selective Estrogen Therapy. Molecules 2019; 24:molecules24224197. [PMID: 31752337 PMCID: PMC6891678 DOI: 10.3390/molecules24224197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022] Open
Abstract
Beneficial effects of estrogens in the central nervous system (CNS) results from the synergistic combination of their well-orchestrated genomic and non-genomic actions, making them potential broad-spectrum neurotherapeutic agents. However, owing to unwanted peripheral hormonal burdens by any currently known non-invasive drug administrations, the development of estrogens as safe pharmacotherapeutic modalities cannot be realized until they are confined specifically and selectively to the site of action. We have developed small-molecule bioprecursor prodrugs carrying the para-quinol scaffold on the steroidal A-ring that are preferentially metabolized in the CNS to the corresponding estrogens. Here, we give an overview of our discovery of these prodrugs. Selected examples are shown to illustrate that, independently of the route of administrations and duration of treatments, these agents produce high concentration of estrogens only in the CNS without peripheral hormonal liability. 10β,17β-Dihydroxyestra-1,4-dien-3-one (DHED) has been the best-studied representative of this novel type of prodrugs for brain and retina health. Specific applications in preclinical animal models of centrally-regulated and estrogen-responsive human diseases, including neurodegeneration, menopausal symptoms, cognitive decline and depression, are discussed to demonstrate the translational potential of our prodrug approach for CNS-selective and gender-independent estrogen therapy with inherent therapeutic safety.
Collapse
|
19
|
Azcoitia I, Barreto GE, Garcia-Segura LM. Molecular mechanisms and cellular events involved in the neuroprotective actions of estradiol. Analysis of sex differences. Front Neuroendocrinol 2019; 55:100787. [PMID: 31513774 DOI: 10.1016/j.yfrne.2019.100787] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/27/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022]
Abstract
Estradiol, either from peripheral or central origin, activates multiple molecular neuroprotective and neuroreparative responses that, being mediated by estrogen receptors or by estrogen receptor independent mechanisms, are initiated at the membrane, the cytoplasm or the cell nucleus of neural cells. Estrogen-dependent signaling regulates a variety of cellular events, such as intracellular Ca2+ levels, mitochondrial respiratory capacity, ATP production, mitochondrial membrane potential, autophagy and apoptosis. In turn, these molecular and cellular actions of estradiol are integrated by neurons and non-neuronal cells to generate different tissue protective responses, decreasing blood-brain barrier permeability, oxidative stress, neuroinflammation and excitotoxicity and promoting synaptic plasticity, axonal growth, neurogenesis, remyelination and neuroregeneration. Recent findings indicate that the neuroprotective and neuroreparative actions of estradiol are different in males and females and further research is necessary to fully elucidate the causes for this sex difference.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - George E Barreto
- Department of Biological Sciences, School of Natural Sciences, University of Limerick, Limerick, Ireland.
| | - Luis M Garcia-Segura
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain; Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain.
| |
Collapse
|
20
|
Beneficial effect of estrogen on nigrostriatal dopaminergic neurons in drug-naïve postmenopausal Parkinson's disease. Sci Rep 2019; 9:10531. [PMID: 31324895 PMCID: PMC6642214 DOI: 10.1038/s41598-019-47026-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/08/2019] [Indexed: 12/25/2022] Open
Abstract
This study aimed to investigate the potential beneficial effects of estrogen on nigrostriatal dopaminergic neuron degeneration in postmenopausal drug-naïve Parkinson's disease (PD). Based on the ratio of lifetime estrogen exposure length to the total length of the estrogen exposure and deprivation period, postmenopausal women with drug-naïve PD were divided into low (n = 31) and high (n = 31) estrogen exposure ratio groups. We performed a comparative analysis of the striatal dopamine transporter (DAT) availability between the two groups. Additionally, we evaluated the longitudinal change in the levodopa equivalent dose per month using a linear mixed model. The motor symptoms were more severe in the low estrogen exposure ratio group than in the high estrogen exposure ratio group (P = 0.016). PD patients in the two groups had significantly lower DAT availability on all striatal sub-regions except for ventral striatum than did age- and sex-matched normal controls. When comparing the two groups, PD patients in the low estrogen exposure ratio group exhibited significantly lower DAT availability in the posterior putamen (P = 0.024) and in the ventral putamen (P = 0.036) than those in the high estrogen exposure ratio group. The estimated monthly levodopa equivalent dose changes were 10.9 in the low estrogen exposure ratio group and 7.1 in the high estrogen exposure ratio group with a significant interaction between the two groups (P = 0.001). These in vivo data provide indirect evidence showing that estrogen may elicit a beneficial effect on nigrostriatal dopamine neurons in PD.
Collapse
|
21
|
O'Neill E, Chiara Goisis R, Haverty R, Harkin A. L-alpha-aminoadipic acid restricts dopaminergic neurodegeneration and motor deficits in an inflammatory model of Parkinson's disease in male rats. J Neurosci Res 2019; 97:804-816. [PMID: 30924171 DOI: 10.1002/jnr.24420] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/21/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022]
Abstract
Neuroinflammation is a contributory factor underlying the progressive nature of dopaminergic neuronal loss within the substantia nigra (SN) of Parkinson's disease (PD) patients, albeit the role of astrocytes in this process has been relatively unexplored to date. Here, we aimed to investigate the impact of midbrain astrocytic dysfunction in the pathophysiology of intra-nigral lipopolysaccharide (LPS)-induced experimental Parkinsonism in male Wistar rats via simultaneous co-injection of the astrocytic toxin L-alpha-aminoadipic acid (L-AAA). Simultaneous intra-nigral injection of L-AAA attenuated the LPS-induced loss of tyrosine hydroxylase-positive (TH+ ) dopamine neurons in the SNpc and suppressed the affiliated degeneration of TH+ dopaminergic nerve terminals in the striatum. L-AAA also repressed LPS-induced nigrostriatal dopamine depletion and provided partial protection against ensuing motor dysfunction. L-AAA abrogated intra-nigral LPS-induced glial fibrillary acidic protein-positive (GFAP+ ) reactive astrogliosis and attenuated the LPS-mediated increases in nigral S100β expression levels in a time-dependent manner, findings which were associated with reduced ionized calcium binding adaptor molecule 1-positive (Iba1+ ) microgliosis, thus indicating a role for reactive astrocytes in sustaining microglial activation at the interface of dopaminergic neuronal loss in response to an immune stimulus. These results indicate that midbrain astrocytic dysfunction restricts the development of dopaminergic neuropathology and motor impairments in rats, highlighting reactive astrocytes as key contributors in inflammatory associated degeneration of the nigrostriatal tract.
Collapse
Affiliation(s)
- Eoin O'Neill
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Rosa Chiara Goisis
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Ruth Haverty
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| |
Collapse
|
22
|
Ruszkiewicz JA, Miranda-Vizuete A, Tinkov AA, Skalnaya MG, Skalny AV, Tsatsakis A, Aschner M. Sex-Specific Differences in Redox Homeostasis in Brain Norm and Disease. J Mol Neurosci 2019; 67:312-342. [DOI: 10.1007/s12031-018-1241-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022]
|
23
|
Song MY, Li CY, Liu XF, Xiao JY, Zhao H. Effect of 17β-oestradiol on T-type calcium channels in the lateral habenula. J Neuroendocrinol 2018; 30:e12629. [PMID: 29917292 DOI: 10.1111/jne.12629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 06/10/2018] [Accepted: 06/16/2018] [Indexed: 11/27/2022]
Abstract
T-type calcium channels (T-channels) are critical for regulating neuronal excitability. Oestrogen alters neuronal excitability by modulating the expression of T-channels. The lateral habenula (LHb), as a link between the limbic system and midbrain structures, expresses T-channels and ERs. However, little is known about the role of oestrogen with respect to modulating T-channels in the LHb. In the present study, we investigated the distribution of T-channels in 3 LHb subregions (rostral, middle and caudal) in normal female rats. Next, we analysed the influence of 17β-oestradiol (E2 ) on T-channels in the LHb in ovariectomised (OVX) rats (oil and E2 groups) using whole-cell patch clamp recording and a real-time polymerase chain reaction (PCR). In normal rats, the results obtained showed that the peak of T-type calcium current (IT ) was -474.61 ± 48.33 pA and IT density was -29.11 ± 1.93 pA/pF. The IT peak and IT density on LHb neurones gradually decreased across the rostrocaudal axis. The neuronal firing pattern varied depending on the location: burst firing was dominant (53.85%) in the rostral LHb, whereas tonic firing was dominant (79.31%) in the caudal LHb. In OVX rats, real-time PCR analysis revealed that E2 treatment decreased Cav3.3 mRNA expression in the caudal LHb. Patch clamp recording showed that E2 treatment decreased the peak IT and also reduced the low-threshold spikes (LTS) number, amplitude and width of LTS in the caudal LHb. Taken together, the results obtained in the present study suggest that E2 may inhibit T-channel activity by selectively down-regulating Cav3.3 calcium channel in the caudal LHb, leading to reduced the possibility of burst firing.
Collapse
Affiliation(s)
- Mei Ying Song
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chun Ying Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiao Feng Liu
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jin Yu Xiao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Hua Zhao
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
24
|
Krolick KN, Zhu Q, Shi H. Effects of Estrogens on Central Nervous System Neurotransmission: Implications for Sex Differences in Mental Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:105-171. [PMID: 30470289 PMCID: PMC6737530 DOI: 10.1016/bs.pmbts.2018.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nearly one of every five US individuals aged 12 years old or older lives with certain types of mental disorders. Men are more likely to use various types of substances, while women tend to be more susceptible to mood disorders, addiction, and eating disorders, all of which are risks associated with suicidal attempts. Fundamental sex differences exist in multiple aspects of the functions and activities of neurotransmitter-mediated neural circuits in the central nervous system (CNS). Dysregulation of these neural circuits leads to various types of mental disorders. The potential mechanisms of sex differences in the CNS neural circuitry regulating mood, reward, and motivation are only beginning to be understood, although they have been largely attributed to the effects of sex hormones on CNS neurotransmission pathways. Understanding this topic is important for developing prevention and treatment of mental disorders that should be tailored differently for men and women. Studies using animal models have provided important insights into pathogenesis, mechanisms, and new therapeutic approaches of human diseases, but some concerns remain to be addressed. The purpose of this chapter is to integrate human and animal studies involving the effects of the sex hormones, estrogens, on CNS neurotransmission, reward processing, and associated mental disorders. We provide an overview of existing evidence for the physiological, behavioral, cellular, and molecular actions of estrogens in the context of controlling neurotransmission in the CNS circuits regulating mood, reward, and motivation and discuss related pathology that leads to mental disorders.
Collapse
Affiliation(s)
- Kristen N Krolick
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Qi Zhu
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Haifei Shi
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States; Cellular, Molecular and Structural Biology, Miami University, Oxford, OH, United States.
| |
Collapse
|
25
|
Bonansco C, Martínez-Pinto J, Silva RA, Velásquez VB, Martorell A, Selva MV, Espinosa P, Moya PR, Cruz G, Andrés ME, Sotomayor-Zárate R. Neonatal exposure to oestradiol increases dopaminergic transmission in nucleus accumbens and morphine-induced conditioned place preference in adult female rats. J Neuroendocrinol 2018; 30:e12574. [PMID: 29377365 DOI: 10.1111/jne.12574] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 01/16/2018] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
Steroid sex hormones produce physiological effects in reproductive tissues and also in nonreproductive tissues, such as the brain, particularly in cortical, limbic and midbrain areas. Dopamine (DA) neurones involved in processes such as prolactin secretion (tuberoinfundibular system), motor circuit regulation (nigrostriatal system) and driving of motivated behaviour (mesocorticolimbic system) are specially regulated by sex hormones. Indeed, sex hormones promote neurochemical and behavioural effects induced by drugs of abuse by tuning midbrain DA neurones in adult animals. However, the long-term effects induced by neonatal exposure to sex hormones on dopaminergic neurotransmission have not been fully studied. The present study aimed to determine whether a single neonatal exposure with oestradiol valerate (EV) results in a programming of dopaminergic neurotransmission in the nucleus accumbens (NAcc) of adult female rats. To answer this question, electrophysiological, neurochemical, cellular, molecular and behavioural techniques were used. The data show that frequency but not amplitude of the spontaneous excitatory postsynaptic current is significantly increased in NAcc medium spiny neurones of EV-treated rats. In addition, DA content and release are both increased in the NAcc of EV-treated rats, caused by an increased synthesis of this neurotransmitter. These results are functionally associated with a higher percentage of EV-treated rats conditioned to morphine, a drug of abuse, compared to controls. In conclusion, neonatal programming with oestradiol increases NAcc dopaminergic neurotransmission in adulthood, which may be associated with increased reinforcing effects of drugs of abuse.
Collapse
Affiliation(s)
- C Bonansco
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - J Martínez-Pinto
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - R A Silva
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - V B Velásquez
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - A Martorell
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Escuela de Fonoaudiología, Facultad de Ciencias de la Rehabilitación, Universidad Andres Bello, Viña del Mar, Chile
| | - M V Selva
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - P Espinosa
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - P R Moya
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Núcleo Milenio Biología de Enfermedades Neuropsiquiátricas (NUMIND), Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Valparaíso, Chile
| | - G Cruz
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - M E Andrés
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - R Sotomayor-Zárate
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
26
|
Jurado-Coronel JC, Cabezas R, Ávila Rodríguez MF, Echeverria V, García-Segura LM, Barreto GE. Sex differences in Parkinson's disease: Features on clinical symptoms, treatment outcome, sexual hormones and genetics. Front Neuroendocrinol 2018; 50:18-30. [PMID: 28974386 DOI: 10.1016/j.yfrne.2017.09.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/12/2017] [Accepted: 09/29/2017] [Indexed: 01/14/2023]
Abstract
Parkinson's disease (PD) is the second most frequent age-related neurodegenerative disorder. Sex is an important factor in the development of PD, as reflected by the fact that it is more common in men than in women by an approximate ratio of 2:1. Our hypothesis is that differences in PD among men and women are highly determined by sex-dependent differences in the nigrostriatal dopaminergic system, which arise from environmental, hormonal and genetic influences. Sex hormones, specifically estrogens, influence PD pathogenesis and might play an important role in PD differences between men and women. The objective of this review was to discuss the PD physiopathology and point out sex differences in nigrostriatal degeneration, symptoms, genetics, responsiveness to treatments and biochemical and molecular mechanisms among patients suffering from this disease. Finally, we discuss the role estrogens may have on PD sex differences.
Collapse
Affiliation(s)
- Juan Camilo Jurado-Coronel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Ricardo Cabezas
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | | | - Valentina Echeverria
- Universidad San Sebastián, Fac. Cs de la Salud, Lientur 1457, Concepción, 4080871, Chile; Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL 33744, USA
| | - Luis Miguel García-Segura
- Instituto Cajal, CSIC, Madrid, Spain; CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
27
|
Cholerton B, Johnson CO, Fish B, Quinn JF, Chung KA, Peterson-Hiller AL, Rosenthal LS, Dawson TM, Albert MS, Hu SC, Mata IF, Leverenz JB, Poston KL, Montine TJ, Zabetian CP, Edwards KL. Sex differences in progression to mild cognitive impairment and dementia in Parkinson's disease. Parkinsonism Relat Disord 2018; 50:29-36. [PMID: 29478836 PMCID: PMC5943177 DOI: 10.1016/j.parkreldis.2018.02.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/17/2018] [Accepted: 02/06/2018] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Identification of factors associated with progression of cognitive symptoms in Parkinson's disease (PD) is important for treatment planning, clinical care, and design of future clinical trials. The current study sought to identify whether prediction of cognitive progression is aided by examining baseline cognitive features, and whether this differs according to stage of cognitive disease. METHODS Participants with PD in the Pacific Udall Center Clinical Consortium who had longitudinal data available and were nondemented at baseline were included in the study (n = 418). Logistic and Cox regression models were utilized to examine the relationship between cognitive, demographic, and clinical variables with risk and time to progression from no cognitive impairment to mild cognitive impairment (PD-MCI) or dementia (PDD), and from PD-MCI to PDD. RESULTS Processing speed (OR = 1.05, p = 0.009) and working memory (OR = 1.01, p = 0.03) were associated with conversion to PDD among those with PD-MCI at baseline, over and above demographic variables. Conversely, the primary predictive factor in the transition from no cognitive impairment to PD-MCI or PDD was male sex (OR = 4.47, p = 0.004), and males progressed more rapidly than females (p = 0.01). Further, among females with shorter disease duration, progression was slower than for their male counterparts, and poor baseline performance on semantic verbal fluency was associated with shorter time to cognitive impairment in females but not in males. CONCLUSIONS This study provides evidence for sex differences in the progression to cognitive impairment in PD, while specific cognitive features become more important indicators of progression with impending conversion to PDD.
Collapse
Affiliation(s)
- Brenna Cholerton
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA.
| | - Catherine O Johnson
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Brian Fish
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Joseph F Quinn
- Portland Veterans Affairs Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Kathryn A Chung
- Portland Veterans Affairs Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Amie L Peterson-Hiller
- Portland Veterans Affairs Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Liana S Rosenthal
- Neurodegeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neurodegeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shu-Ching Hu
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA; Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Ignacio F Mata
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA; Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Cyrus P Zabetian
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA; Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Karen L Edwards
- Department of Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
28
|
Miville-Godbout E, Bourque M, Morissette M, Al-Sweidi S, Smith T, Jayasinghe D, Ritchie S, Di Paolo T. Plasmalogen precursor mitigates striatal dopamine loss in MPTP mice. Brain Res 2017; 1674:70-76. [PMID: 28830769 DOI: 10.1016/j.brainres.2017.08.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022]
Abstract
Ethanolamine plasmalogens (PlsEtn) are a class of glycerophospholipids characterized by a vinyl-ether bond at the sn-1 position that play an important role in the structure and function of membranes. Previous reports have suggested a link between reduced blood and brain PlsEtn levels and Parkinson's disease (PD). We recently reported that the DHA containing plasmalogen precursor PPI-1011 protected striatal dopamine (DA) against MPTP toxicity in mice. In this paper, we further investigate the specificity requirements of the lipid side chains by testing the oleic acid-containing plasmalogen precursor PPI-1025. Male mice were treated for 10days with daily oral administration of PPI-1025 (10, 50 or 200mg/kg). On day 5 mice received MPTP and were sacrificed on Day 11. Treatment with PPI-1025 prevented MPTP-induced decrease of DA and serotonin, as well as their metabolites. In addition, PPI-1025 treatment prevented the MPTP-induced decrease of the striatal dopamine transporter (DAT) and vesicular monoamine transporter 2 (VMAT2) specific binding. Significant positive correlations were measured between striatal DA concentrations and DAT or VMAT2 specific binding, as well as with serum plasmalogen concentrations. The neuroprotective effect of PPI-1025 displayed a bell-curve dose-dependency losing effect at the highest dose tested. The similar protective response of oleic and docosahexaenoic acid (DHA)-containing plasmalogen precursors suggests that the neuroprotection observed is not only due to DHA but to the oleic substituent and the plasmalogen backbone.
Collapse
Affiliation(s)
- Edith Miville-Godbout
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, 2705 Laurier Boulevard, Quebec City, Qc G1V 4G2, Canada; Faculty of Pharmacy, Laval University, 1050, Avenue de la Médecine, Quebec City, Qc G1V 0A6, Canada
| | - Mélanie Bourque
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, 2705 Laurier Boulevard, Quebec City, Qc G1V 4G2, Canada; Faculty of Pharmacy, Laval University, 1050, Avenue de la Médecine, Quebec City, Qc G1V 0A6, Canada
| | - Marc Morissette
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, 2705 Laurier Boulevard, Quebec City, Qc G1V 4G2, Canada
| | - Sara Al-Sweidi
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, 2705 Laurier Boulevard, Quebec City, Qc G1V 4G2, Canada
| | - Tara Smith
- Med-Life Discoveries LP, 104-407 Downey Road, Saskatoon, SK S7N 4L8, Canada
| | | | - Shawn Ritchie
- Med-Life Discoveries LP, 104-407 Downey Road, Saskatoon, SK S7N 4L8, Canada
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, 2705 Laurier Boulevard, Quebec City, Qc G1V 4G2, Canada; Faculty of Pharmacy, Laval University, 1050, Avenue de la Médecine, Quebec City, Qc G1V 0A6, Canada.
| |
Collapse
|
29
|
GPR30 Activation Contributes to the Puerarin-Mediated Neuroprotection in MPP+-Induced SH-SY5Y Cell Death. J Mol Neurosci 2016; 61:227-234. [DOI: 10.1007/s12031-016-0856-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022]
|
30
|
Labandeira-Garcia JL, Rodriguez-Perez AI, Valenzuela R, Costa-Besada MA, Guerra MJ. Menopause and Parkinson's disease. Interaction between estrogens and brain renin-angiotensin system in dopaminergic degeneration. Front Neuroendocrinol 2016; 43:44-59. [PMID: 27693730 DOI: 10.1016/j.yfrne.2016.09.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023]
Abstract
The neuroprotective effects of menopausal hormonal therapy in Parkinson's disease (PD) have not yet been clarified, and it is controversial whether there is a critical period for neuroprotection. Studies in animal models and clinical and epidemiological studies indicate that estrogens induce dopaminergic neuroprotection. Recent studies suggest that inhibition of the brain renin-angiotensin system (RAS) mediates the effects of estrogens in PD models. In the substantia nigra, ovariectomy induces a decrease in levels of estrogen receptor-α (ER-α) and increases angiotensin activity, NADPH-oxidase activity and expression of neuroinflammatory markers, which are regulated by estrogen replacement therapy. There is a critical period for the neuroprotective effect of estrogen replacement therapy, and local ER-α and RAS play a major role. Astrocytes play a major role in ER-α-induced regulation of local RAS, but neurons and microglia are also involved. Interestingly, treatment with angiotensin receptor antagonists after the critical period induced neuroprotection.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| | - Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Rita Valenzuela
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria A Costa-Besada
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| |
Collapse
|
31
|
Cheng HY, Hung SH, Chu PJ. Rescue from Sexually Dimorphic Neuronal Cell Death by Estradiol and PI3 Kinase Activity. Cell Mol Neurobiol 2016; 36:767-75. [PMID: 26369912 DOI: 10.1007/s10571-015-0259-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 08/25/2015] [Indexed: 10/23/2022]
Abstract
Responses of primary hippocampal and cortical neurons derived from male and female rats to cellular stressors were studied. It is demonstrated that 17β-estradiol (E2), a potent neuroprotectant, protected the female neurons but had no effects on the male neurons from CoCl2- and glutamate-induced toxicity. Agonists of the estrogen receptor (ER) subtypes ERα and ERβ, DPN and PPT, respectively, had similar effects to E2. By contrast, effects of E2 were abolished by the ER antagonist ICI-182780, further corroborating the neuroprotective role of ERs. In male neurons, CoCl2 predominately activated the apoptosis-inducing factor (AIF)-dependent pathway and AIF translocation from the cytosol to the nucleus. In comparison, CoCl2 activated the caspase pathway and cytochrome c release in female neurons. The inhibitors of these pathways, namely DiQ for AIF and zVAD for caspase, specifically rescued CoCl2-induced cell death in male and female neurons, respectively. When zVAD and ICI-182780, and E2 were applied in combination, it was demonstrated E2 acted on the caspase pathway leading to female-specific neuroprotection. Furthermore, the PI3 kinase (PI3K) inhibitor blocked the rescue effects of DiQ and zVAD on the male and female neurons, respectively, suggesting that PI3K is a common upstream regulator for both pathways. The present study suggested that both sex-specific and nonspecific mechanisms played a role in neuronal responses to stressors and protective reagents.
Collapse
Affiliation(s)
- Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Shin-Hui Hung
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Po-Ju Chu
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
32
|
Yi H, Bao X, Tang X, Fan X, Xu H. Estrogen modulation of calretinin and BDNF expression in midbrain dopaminergic neurons of ovariectomised mice. J Chem Neuroanat 2016; 77:60-67. [PMID: 27211874 DOI: 10.1016/j.jchemneu.2016.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 05/09/2016] [Accepted: 05/18/2016] [Indexed: 11/19/2022]
Abstract
Estrogen attenuates the loss of dopamine neurons from the substantia nigra in animal models of Parkinson's disease (PD) and excitatory amino-acid induced neurotoxicity by interactions with brain-derived neurotrophic factor (BDNF), and calretinin (CR) containing dopaminergic (DA) neurons. To examine this interaction more closely, we treated the ovariectomised (OVX) mice with estrodial for 10days, and compared these mice to those OVX mice injected with the vehicle or control mice. Estrogen treatment in OVX mice had significantly more tyrosine hydroxylase (TH) positive neurons in the substantia nigra pars compacta (SNpc). Dopamine transporter (DAT) mRNA and BDNF mRNA levels in the midbrain were also significantly increased by estrogen treatment (P<0.05). OVX markedly decreased the number of TH/CR double stained cells in the SNpc (P<0.05), a trend which could be reversed by estrogen treatment. However, the number of GFAP positive cells in the substantia nigra did not show significant changes (P >0.05) after vehicle or estrodial treatment. Furthermore, we found that estrogen treatment abrogated the OVX-induced decrease in the phosphorylated AKT (p-AKT), but not p-ERK. We hypothesize that short-term treatment with estrogen confers neuroprotection to DA neurons by increasing CR in the DA neurons and BDNF in the midbrain, which possibly related to activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Hongliang Yi
- Department of Physiology, Third Military Medical University, Chongqing, 400038, PR China; Chongqing City Family Planning Institute, Chongqing, 400020, PR China
| | - Xiaohang Bao
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Xiaotong Tang
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, PR China
| | - Xiaotang Fan
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital,Third Military Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
33
|
Abstract
Globally, greater than 30 million individuals are afflicted with disorders of the nervous system accompanied by tens of thousands of new cases annually with limited, if any, treatment options. Erythropoietin (EPO) offers an exciting and novel therapeutic strategy to address both acute and chronic neurodegenerative disorders. EPO governs a number of critical protective and regenerative mechanisms that can impact apoptotic and autophagic programmed cell death pathways through protein kinase B (Akt), sirtuins, mammalian forkhead transcription factors, and wingless signaling. Translation of the cytoprotective pathways of EPO into clinically effective treatments for some neurodegenerative disorders has been promising, but additional work is necessary. In particular, development of new treatments with erythropoiesis-stimulating agents such as EPO brings several important challenges that involve detrimental vascular outcomes and tumorigenesis. Future work that can effectively and safely harness the complexity of the signaling pathways of EPO will be vital for the fruitful treatment of disorders of the nervous system.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
34
|
Kandimalla R, Reddy PH. Multiple faces of dynamin-related protein 1 and its role in Alzheimer's disease pathogenesis. Biochim Biophys Acta Mol Basis Dis 2015; 1862:814-828. [PMID: 26708942 DOI: 10.1016/j.bbadis.2015.12.018] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 01/01/2023]
Abstract
Mitochondria play a large role in neuronal function by constantly providing energy, particularly at synapses. Recent studies suggest that amyloid beta (Aβ) and phosphorylated tau interact with the mitochondrial fission protein, dynamin-related protein 1 (Drp1), causing excessive fragmentation of mitochondria and leading to abnormal mitochondrial dynamics and synaptic degeneration in Alzheimer's disease (AD) neurons. Recent research also revealed Aβ-induced and phosphorylated tau-induced changes in mitochondria, particularly affecting mitochondrial shape, size, distribution and axonal transport in AD neurons. These changes affect mitochondrial health and, in turn, could affect synaptic function and neuronal damage and ultimately leading to memory loss and cognitive impairment in patients with AD. This article highlights recent findings in the role of Drp1 in AD pathogenesis. This article also highlights Drp1 and its relationships to glycogen synthase kinase 3, cyclin-dependent kinase 5, p53, and microRNAs in AD pathogenesis.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Neurology, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Ste. E, MS 7495, Lubbock, TX 79413, United States.
| |
Collapse
|
35
|
Bessa A, Campos FL, Videira RA, Mendes-Oliveira J, Bessa-Neto D, Baltazar G. GPER: A new tool to protect dopaminergic neurons? Biochim Biophys Acta Mol Basis Dis 2015; 1852:2035-41. [DOI: 10.1016/j.bbadis.2015.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 06/24/2015] [Accepted: 07/08/2015] [Indexed: 12/11/2022]
|
36
|
Han S, Zhao B, Pan X, Song Z, Liu J, Gong Y, Wang M. Estrogen receptor variant ER-α36 is involved in estrogen neuroprotection against oxidative toxicity. Neuroscience 2015; 310:224-41. [PMID: 26383254 DOI: 10.1016/j.neuroscience.2015.09.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/05/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
Abstract
It is well known that estrogen exerts neuroprotective effect against various neuronal damages. However, the estrogen receptor (ER) that mediates estrogen neuroprotection has not been well established. In this study, we investigated the potential receptor that mediates estrogen neuroprotection and the underlying molecular mechanisms. Hydrogen peroxide (H2O2) was chosen as an agent in our study to mimic free radicals that are often involved in the pathogenesis of many degenerative diseases. We found that in human SY5Y and IMR-32 cells, the estrogen neuroprotection against H2O2 toxicity was abrogated by knockdown of a variant of estrogen receptor-α, ER-α36. We also studied the rapid estrogen signaling mediated by ER-α36 in neuroprotective effect and found the PI3K/AKT and MAPK/ERK1/2 signaling mediated by ER-α36 is involved in estrogen neuroprotection. We also found that GPER, an orphan G protein-coupled receptor, is not involved in ER-α36-mediated rapid estrogen response. Our study thus demonstrates that ER-α36-mediated rapid estrogen signaling is involved in the neuroprotection activity of estrogen against oxidative toxicity.
Collapse
Affiliation(s)
- S Han
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan 250012, Shandong, China.
| | - B Zhao
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan 250012, Shandong, China.
| | - X Pan
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China.
| | - Z Song
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan 250012, Shandong, China.
| | - J Liu
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan 250012, Shandong, China.
| | - Y Gong
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan 250012, Shandong, China.
| | - M Wang
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
37
|
Altun I, Kurutaş EB. G Protein-Coupled Estrogen Receptor Levels After Peripheral Nerve Injury in an Experimental Rat Model. World Neurosurg 2015; 84:1903-6. [PMID: 26325209 DOI: 10.1016/j.wneu.2015.08.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 08/23/2015] [Accepted: 08/24/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To assess whether G protein-coupled estrogen receptor (GPER) levels were altered during crush-induced peripheral nerve injury in an experimental rat model. METHODS Male Wistar rats (N = 80) were allocated to 1 sham and 6 study groups, and crush-type peripheral nerve injury was performed using a clamp on the sciatic nerves of study groups. In the sham group, the sciatic nerve was exposed only, and the wound was closed primarily without any surgical interventions. Peripheral nerve samples were obtained at 1 hour, 6 hours, 12 hours, 24 hours, 3 days, and 7 days. After analysis of nerve tissues by protein analysis and Western blotting, the groups were compared in terms of expression of GPER levels. RESULTS The average levels of GPER in the sham group and study groups at 1 hour, 6 hours, 12 hours, 24 hours, 3 days, and 7 days were 15.06 ng/mL ± 2.91, 3.31 ng/mL ± 0.91, 4.06 ng/mL ± 0.87, 11.94 ng/mL ± 1.15, 10.76 ng/mL ± 1.76, 9.16 ng/mL ± 2.60, and 8.49 ng/mL ± 3.55. All study groups displayed significantly lower levels of GPER compared with the sham group. CONCLUSIONS Our results demonstrate that a basal level of GPER expression occurs in peripheral nerve tissue. The lowest level was detected 1 hour after crush injury, and the highest levels of GPER were detected 12 hours and 24 hours after trauma. Further trials on larger series are required to elucidate the role of GPER in terms of protection and treatment after nerve injury.
Collapse
Affiliation(s)
- Idris Altun
- Department of Neurosurgery, Kahramanmaras Sutcu Imam University Medical Faculty, Kahramanmaras, Turkey.
| | - Ergül Belge Kurutaş
- Department of Biochemistry, Kahramanmaras Sutcu Imam University Medical Faculty, Kahramanmaras, Turkey
| |
Collapse
|
38
|
Hu Y, Deng L, Zhang J, Fang X, Mei P, Cao X, Lin J, Wei Y, Zhang X, Xu R. A Pooling Genome-Wide Association Study Combining a Pathway Analysis for Typical Sporadic Parkinson's Disease in the Han Population of Chinese Mainland. Mol Neurobiol 2015; 53:4302-18. [PMID: 26227905 DOI: 10.1007/s12035-015-9331-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 07/01/2015] [Indexed: 01/23/2023]
Abstract
Genome-wide association studies (GWAS) on sporadic Parkinson's disease (sPD) are mainly conducted in European and American populations at present, and the Han populations of Chinese mainland (HPCM) almost have not been studied yet. Here, we conducted a pooling GWAS combining a pathway analysis with 862,198 autosomal single nucleotide polymorphisms of IlluminaHumanOmniZhongHua-8 in 250 sPD and 250 controls from HPCM precluded toxicant exposure, age, and heavy coffee drinking habit interference. We revealed that among the 22 potential loci implicated, PRDM2/KIAA1026 (kgp8090149), TSG1/MANEA (kgp154172), PDE10A (kgp8130520), MDGA2 (rs9323124), ATPBD4/LOC100288892 (kgp11333367), ZFP64/TSHZ2 (kgp4156164), PAQR3/ARD1B (kgp9482779), FLJ23172/FNDC3B (kgp760898), C18orf1 (kgp348599), FLJ43860/NCRNA00051 (kgp4105983), CYP1B1/C2orf58 (kgp11353523), WNT9A/LOC728728 (rs849898), ANXA1/LOC100130911 (rs10746953), FLJ35379/LOC100132423 (kgp9550589), PLEKHN1 (kgp7172368), DMRT2/SMARCA2 (kgp10769919), ZNF396/INO80C (rs1362858), C3orf67/LOC339902 (rs6783485), LOC285194/IGSF11 (rs1879553), FGF10/MRPS30 (rs13153459), BARX1/PTPDC1 (kgp6542803), and COL5 A2 (rs11186), the peak significance was at the kgp4105983 of FLJ43860 gene in chromosome 8, the first top strongest associated locus with sPD was PRDM2 (kgp8090149) in chromosome 1, and the 24 pathways including 100 significantly associated genes were strongly associated with sPD from HPCM. The 40 genes were shared by at least two pathways. The most possible associated pathways with sPD were axon guidance, ECM-receptor interaction, neuroactive ligand-receptor interaction, tight junction, focal adhesion, gap junction, long-term depression, drug metabolism-cytochrome P450, adherens junction, endocytosis, and protein digestion and absorption. Our results indicated that these loci, pathways, and their related genes might be involved in the pathogenesis of sPD from HPCM and provided some novel evidences for further searching the genetic pathogenesis of sPD.
Collapse
Affiliation(s)
- Yakun Hu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Libing Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jie Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xin Fang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Puming Mei
- Institute of Translational Medicine, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xuebing Cao
- Department of Neurology, The Affiliated Union Hospital of Huazhong Technological University, Wuhan, 430006, Hubei, China
| | - Jiari Lin
- Institute of Translational Medicine, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi Wei
- Institute of Translational Medicine, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiong Zhang
- Department of Neurology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, Guangzhou, 510080, Guangdong, China.
| | - Renshi Xu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
39
|
Polymorphism in the Vesicular Monoamine Transporter 2 Gene Decreases the Risk of Parkinson's Disease in Han Chinese Men. PARKINSONS DISEASE 2015; 2015:903164. [PMID: 26246935 PMCID: PMC4515295 DOI: 10.1155/2015/903164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/19/2015] [Accepted: 06/24/2015] [Indexed: 02/05/2023]
Abstract
Background. Polymorphisms rs363371 and rs363324 in the vesicular monoamine transporter 2 (VMAT2) gene have been associated with risk of PD in an Italian population, and our aim is to investigate the association between the two single-nucleotide polymorphisms and PD in Han Chinese. Methods. 561 Han Chinese PD patients and 491 healthy age- and gender-matched controls were genotyped using Ligase detection reaction (LDR) method. Result. Both of patient and control groups showed similar genotype frequencies between patients and controls at both rs363371 and rs363324, as well as similar minor A allele frequencies at rs363371 (P = 0.452) and rs363324 (P = 0.413). None of the observed haplotypes showed a significant association with PD. Subgroup analysis by gender and age at onset revealed a significant association between the A allele of rs363371 and PD in Han Chinese males relative to healthy controls (OR 0.799, 95% CI 0.665 to 0.959, P = 0.016), and this association remained significant after adjusting for age (OR 0.785, 95% CI 0.652 to 0.945, P = 0.011). Conclusion. These results suggest that polymorphism of VMAT2 locus is associated with risk of PD in Han Chinese overall but that the A allele at rs363371 may protect against PD in Han Chinese males.
Collapse
|
40
|
|
41
|
Rivera HM, Christiansen KJ, Sullivan EL. The role of maternal obesity in the risk of neuropsychiatric disorders. Front Neurosci 2015; 9:194. [PMID: 26150767 PMCID: PMC4471351 DOI: 10.3389/fnins.2015.00194] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 05/16/2015] [Indexed: 12/22/2022] Open
Abstract
Recent evidence indicates that perinatal exposure to maternal obesity, metabolic disease, including diabetes and hypertension, and unhealthy maternal diet has a long-term impact on offspring behavior and physiology. During the past three decades, the prevalence of both obesity and neuropsychiatric disorders has rapidly increased. Epidemiologic studies provide evidence that maternal obesity and metabolic complications increase the risk of attention deficit hyperactivity disorder (ADHD), autism spectrum disorders, anxiety, depression, schizophrenia, eating disorders (food addiction, anorexia nervosa, and bulimia nervosa), and impairments in cognition in offspring. Animal models of maternal high-fat diet (HFD) induced obesity also document persistent changes in offspring behavior and impairments in critical neural circuitry. Animals exposed to maternal obesity and HFD consumption display hyperactivity, impairments in social behavior, increased anxiety-like and depressive-like behaviors, substance addiction, food addiction, and diminished cognition. During development, these offspring are exposed to elevated levels of nutrients (fatty acids, glucose), hormones (leptin, insulin), and inflammatory factors (C-reactive protein, interleukin, and tumor necrosis factor). Such factors appear to permanently change neuroendocrine regulation and brain development in offspring. In addition, inflammation of the offspring brain during gestation impairs the development of neural pathways critical in the regulation of behavior, such as serotoninergic, dopaminergic, and melanocortinergic systems. Dysregulation of these circuits increases the risk of mental health disorders. Given the high rates of obesity in most developed nations, it is critical that the mechanisms by which maternal obesity programs offspring behavior are thoroughly characterized. Such knowledge will be critical in the development of preventative strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Heidi M Rivera
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR, USA
| | | | - Elinor L Sullivan
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR, USA ; Department of Biology, University of Portland Portland, OR, USA
| |
Collapse
|
42
|
Rodriguez-Perez AI, Borrajo A, Valenzuela R, Lanciego JL, Labandeira-Garcia JL. Critical period for dopaminergic neuroprotection by hormonal replacement in menopausal rats. Neurobiol Aging 2014; 36:1194-208. [PMID: 25432430 DOI: 10.1016/j.neurobiolaging.2014.10.028] [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: 07/28/2014] [Revised: 10/01/2014] [Accepted: 10/24/2014] [Indexed: 10/24/2022]
Abstract
The neuroprotective effects of menopausal hormonal therapy in Parkinson's disease have not yet been clarified, and it is not known whether there is a critical period. Estrogen induced significant protection against 6-hydroxydopamine-induced dopaminergic degeneration when administered immediately or 6 weeks, but not 20 weeks after ovariectomy. In the substantia nigra, ovariectomy induced a decrease in levels of estrogen receptor-α and increased angiotensin activity, NADPH-oxidase activity, and expression of neuroinflammatory markers, which were regulated by estrogen administered immediately or 6 weeks but not 20 weeks after ovariectomy. Interestingly, treatment with angiotensin receptor antagonists after the critical period induced a significant level of neuroprotection. In cultures, treatment with 1-methyl-4-phenylpyridinium induced an increase in astrocyte-derived angiotensinogen and dopaminergic neuron death, which were inhibited by estrogen receptor α agonists. In microglial cells, estrogen receptor β agonists inhibited the angiotensin-induced increase in inflammatory markers. The results suggest that there is a critical period for the neuroprotective effect of estrogen against dopaminergic cell death, and local estrogen receptor α and renin-angiotensin system play a major role.
Collapse
Affiliation(s)
- Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Rita Valenzuela
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Jose L Lanciego
- Neurosciences Division, CIMA, University of Navarra, Pamplona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| |
Collapse
|
43
|
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
|
44
|
Duarte JM, Do KQ, Gruetter R. Longitudinal neurochemical modifications in the aging mouse brain measured in vivo by 1H magnetic resonance spectroscopy. Neurobiol Aging 2014; 35:1660-8. [DOI: 10.1016/j.neurobiolaging.2014.01.135] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 11/19/2013] [Accepted: 01/27/2014] [Indexed: 12/29/2022]
|
45
|
Liang K, Ye Y, Wang Y, Zhang J, Li C. Formononetin mediates neuroprotection against cerebral ischemia/reperfusion in rats via downregulation of the Bax/Bcl-2 ratio and upregulation PI3K/Akt signaling pathway. J Neurol Sci 2014; 344:100-4. [PMID: 24996490 DOI: 10.1016/j.jns.2014.06.033] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/09/2014] [Accepted: 06/16/2014] [Indexed: 11/28/2022]
Abstract
Isoflavone formononetin is a typical phytoestrogen isolated from Chinese medical herb red clover. It has been reported that estrogens have neuroprotective properties, and dietary intake of phytoestrogens could reduce stroke injury in cerebral ischemia/reperfusion (I/R) animal models. In the present research, we sought to investigate the molecular mechanisms underlying the neuroprotective effects of formononetin on I/R rats. Male Sprague-Dawley rats were subjected to a 2 h period of right middle cerebral artery occlusion (MCAO) followed by 24 h of reperfusion. Then neurological deficits and brain edema were evaluated. To provide insight into the functions of phosphatidylinositol 3-kinase (PI3K)/Akt and MAPK (mitogen-activated protein kinase) signaling pathway in formononetin-induced neuroprotection, the expression of ER-α, Bax, Bcl-2, p-Akt (phosphorylated protein kinase B), and p-ERK1/2 (phosphorylated extracellular signal-regulated kinases 1/2) was determined by qPCR or Western blot assay. Consequently, we found that formononetin has significantly reduced the infarcted volume and the brain water content, and improved the neurological deficit. Formononetin also exhibited an upregulation in ER-α and p-Akt, a downregulation in the ratio of Bax/Bcl-2. However, formononetin had little effect on p-ERK1/2 proteins expression. Taken together, formononetin has shown neuroprotective effects in cerebral I/R rats, and the molecular mechanisms may correlate with the downregulation of the Bax/Bcl-2 ratio and the activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Kun Liang
- Department of Emergency, Western Hospital, First Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Yu Ye
- Department of Emergency, Western Hospital, First Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Yong Wang
- Department of Physiology, Guilin Medical University, Guilin 541004, China
| | - Jianfeng Zhang
- Department of Emergency, Western Hospital, First Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Chaoqian Li
- Department of Emergency, Western Hospital, First Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| |
Collapse
|
46
|
Bondesson M, Hao R, Lin CY, Williams C, Gustafsson JÅ. Estrogen receptor signaling during vertebrate development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:142-51. [PMID: 24954179 DOI: 10.1016/j.bbagrm.2014.06.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 01/03/2023]
Abstract
Estrogen receptors are expressed and their cognate ligands produced in all vertebrates, indicative of important and conserved functions. Through evolution estrogen has been involved in controlling reproduction, affecting both the development of reproductive organs and reproductive behavior. This review broadly describes the synthesis of estrogens and the expression patterns of aromatase and the estrogen receptors, in relation to estrogen functions in the developing fetus and child. We focus on the role of estrogens for the development of reproductive tissues, as well as non-reproductive effects on the developing brain. We collate data from human, rodent, bird and fish studies and highlight common and species-specific effects of estrogen signaling on fetal development. Morphological malformations originating from perturbed estrogen signaling in estrogen receptor and aromatase knockout mice are discussed, as well as the clinical manifestations of rare estrogen receptor alpha and aromatase gene mutations in humans. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Maria Bondesson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA.
| | - Ruixin Hao
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA; DuPont Haskell Global Centers for Health and Environmental Sciences, Newark, DE, USA
| | - Chin-Yo Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA
| | - Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA; Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| |
Collapse
|
47
|
Sinclair D, Purves-Tyson TD, Allen KM, Weickert CS. Impacts of stress and sex hormones on dopamine neurotransmission in the adolescent brain. Psychopharmacology (Berl) 2014; 231:1581-99. [PMID: 24481565 PMCID: PMC3967083 DOI: 10.1007/s00213-013-3415-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
Abstract
RATIONALE Adolescence is a developmental period of complex neurobiological change and heightened vulnerability to psychiatric illness. As a result, understanding factors such as sex and stress hormones which drive brain changes in adolescence, and how these factors may influence key neurotransmitter systems implicated in psychiatric illness, is paramount. OBJECTIVES In this review, we outline the impact of sex and stress hormones at adolescence on dopamine neurotransmission, a signaling pathway which is critical to healthy brain function and has been implicated in psychiatric illness. We review normative developmental changes in dopamine, sex hormone, and stress hormone signaling during adolescence and throughout postnatal life, then highlight the interaction of sex and stress hormones and review their impacts on dopamine neurotransmission in the adolescent brain. RESULTS AND CONCLUSIONS Adolescence is a time of increased responsiveness to sex and stress hormones, during which the maturing dopaminergic neural circuitry is profoundly influenced by these factors. Testosterone, estrogen, and glucocorticoids interact with each other and have distinct, brain region-specific impacts on dopamine neurotransmission in the adolescent brain, shaping brain maturation and cognitive function in adolescence and adulthood. Some effects of stress/sex hormones on cortical and subcortical dopamine parameters bear similarities with dopaminergic abnormalities seen in schizophrenia, suggesting a possible role for sex/stress hormones at adolescence in influencing risk for psychiatric illness via modulation of dopamine neurotransmission. Stress and sex hormones may prove useful targets in future strategies for modifying risk for psychiatric illness.
Collapse
Affiliation(s)
- Duncan Sinclair
- Schizophrenia Research Institute, Sydney, Australia ,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia ,School of Psychiatry, University of New South Wales, Sydney, Australia ,Neuropsychiatric Signaling Program, Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
| | - Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney, Australia ,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia ,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Katherine M Allen
- Schizophrenia Research Institute, Sydney, Australia ,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia ,School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, Australia ,Macquarie Group Chair of Schizophrenia Research, Neuroscience Research Australia, Barker Street, Randwick, NSW 2031 Australia ,School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
48
|
17β-estradiol delays 6-OHDA-induced apoptosis by acting on Nur77 translocation from the nucleus to the cytoplasm. Neurotox Res 2013; 25:124-34. [PMID: 24277157 DOI: 10.1007/s12640-013-9442-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 11/08/2013] [Accepted: 11/13/2013] [Indexed: 12/12/2022]
Abstract
Nuclear receptors (Nurs) represent a large family of gene expression regulating proteins. Gathering evidence indicates an important role for Nurs as transcription factors in dopamine neurotransmission. Nur77, a member of the Nur superfamily, plays a role in mediating the effects of antiparkinsonian and neuroleptic drugs. Besides, Nur77 survival and apoptotic roles depend largely on its subcellular localization. Estrogens are known for their neuroprotective properties, as demonstrated in animal and clinical studies. However, their action on Nur77 translocation pertaining to neuroprotection has not been investigated yet. The aim of our study was to perform a kinetic study on the effect of neurotoxic 6-hydroxydopamine (6-OHDA) and 17β-estradiol (E2) on the subcellular localization of Nur77 with reference to the modulation of apoptosis in PC12 cells. Our results demonstrate that E2 administration alone does not affect Nur77 cytoplasmic/nuclear ratio, mRNA levels, or apoptosis in PC12 cells. The neurotoxin 6-OHDA significantly enhances cytoplasmic localization of Nur77 after merely 3 h, while precipitating apoptosis. 6-OHDA also increases Nur77 transcription, which could partly explain the rise in cytoplasmic localization of the protein. Finally, treatment with both E2 and 6-OHDA delays Nur77 accumulation in the cytoplasm and delays cell death for a few hours in our cellular paradigm. Pre-treatment with E2 does not alter the increase in levels of Nur77 mRNA produced by 6-OHDA, suggesting that a raise in nuclear translocation is likely responsible for the stabilization of the cytoplasmic/nuclear ratio until 6 h. These results suggest an intriguing cooperation between E2 and Nur77 toward cellular fate guidance.
Collapse
|
49
|
Habib P, Dreymueller D, Ludwig A, Beyer C, Dang J. Sex steroid hormone-mediated functional regulation of microglia-like BV-2 cells during hypoxia. J Steroid Biochem Mol Biol 2013; 138:195-205. [PMID: 23792783 DOI: 10.1016/j.jsbmb.2013.06.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 05/31/2013] [Accepted: 06/04/2013] [Indexed: 01/04/2023]
Abstract
17β-estradiol (E2) and progesterone (P) are neuroprotective hormones in different neurological disorders and in particular under hypoxic conditions in the brain. Both hormones dampen brain-intrinsic immune responses and regulate local glial cell function. Besides astrocytes which are functionally regulated in a manifold and complex manner, especially microglial cells are in the focus of steroid-mediated neuroprotection. In previous studies using a transient brain artery occlusion model, we demonstrated that microglial characteristics are critically modified after the administration of either E2 or P. We here studied the influence of sex steroids on the murine BV-2 microglia cell line under hypoxic conditions. Hypoxia changed the cell morphology from an amoeboid-like phenotype with processes to a rounded shape of secreting cell type. BV-2 cells expressed both estrogen receptor-β and progesterone receptors under each condition. Oxygen deprivation increased the expression of inducible nitric oxide synthetase (iNOS) and up-regulated selected cytokines and chemokines. Both hormones selectively prevented the induction of pro-inflammatory iNOS, interleukin IL-1ß, and chemokine ligand CCL5, whereas anti-inflammatory IL-10 and protective TREM 2 were up-regulated by sex steroids. Sex hormones abrogated hypoxia-dependent reduction of BV-2 phagocytic activity. We demonstrate that BV-2 microglia cells respond to hypoxia by enhanced pro-inflammatory cytokine secretion and reduced phagocytic activity. This effect is prevented by sex steroids resulting in a switch of BV-2 cells from a pro-inflammatory to a more anti-inflammatory phenotype. Anti-inflammatory effects of gonadal steroids might directly be mediated through hormone-microglia interactions in addition to known effects via astroglial regulation.
Collapse
Affiliation(s)
- Pardes Habib
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | | | | | | | | |
Collapse
|
50
|
Marin R, Casañas V, Pérez JA, Fabelo N, Fernandez CE, Diaz M. Oestrogens as modulators of neuronal signalosomes and brain lipid homeostasis related to protection against neurodegeneration. J Neuroendocrinol 2013; 25:1104-15. [PMID: 23795744 DOI: 10.1111/jne.12068] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Accepted: 06/18/2013] [Indexed: 12/19/2022]
Abstract
Oestrogens trigger several pathways at the plasma membrane that exert beneficial actions against neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Part of these actions takes place in lipid rafts, which are membrane domains with a singular protein and lipid composition. These microdomains also represent a preferential site for signalling protein complexes, or signalosomes. A plausible hypothesis is that the dynamic interaction of signalosomes with different extracellular ligands may be at the basis of neuronal maintenance against different neuropathologies. Oestrogen receptors are localised in neuronal lipid rafts, taking part of macromolecular complexes together with a voltage-dependent anion channel (VDAC), and other molecules. Oestradiol binding to its receptor at this level enhances neuroprotection against amyloid-β degeneration through the activation of different signal transduction pathways, including VDAC gating modulation. Moreover, part of the stability and functionality of signalling platforms lays on the distribution of lipid hallmarks in these microstructures, which modulate membrane physicochemical properties, thus favouring molecular interactions. Interestingly, recent findings indicate a potential role of oestrogens in the preservation of neuronal membrane physiology related to lipid homeostasis. Thus, oestrogens and docosahexaenoic acid may act synergistically to stabilise brain lipid structure by regulating neuronal lipid biosynthetic pathways, suggesting that part of the neuroprotective effects elicited by oestrogens occur through mechanisms aimed at preserving lipid homeostasis. Overall, oestrogen mechanisms of neuroprotection may occur not only by its interaction with neuronal protein targets through nongenomic and genomic mechanisms, but also through its participation in membrane architecture stabilisation via 'lipostatic' mechanisms.
Collapse
Affiliation(s)
- R Marin
- Department of Physiology, Laboratory of Cellular Neurobiology, University of La Laguna, La Laguna, Tenerife, Spain
| | | | | | | | | | | |
Collapse
|