1
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
2
|
Statz M, Schleuter F, Weber H, Kober M, Plocksties F, Timmermann D, Storch A, Fauser M. Subthalamic nucleus deep brain stimulation does not alter growth factor expression in a rat model of stable dopaminergic deficiency. Neurosci Lett 2023; 814:137459. [PMID: 37625613 DOI: 10.1016/j.neulet.2023.137459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Deep brain stimulation (DBS) of the subthalamic nucleus (STN) has been a highly effective treatment option for mid-to-late-stage Parkinson's disease (PD) for decades. Besides direct effects on brain networks, neuroprotective effects of STN-DBS - potentially via alterations of growth factor expression levels - have been proposed as additional mechanisms of action. OBJECTIVE In the context of clarifying DBS mechanisms, we analyzed brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) levels in the basal ganglia, motor and parietal cortices, and dentate gyrus in an animal model of stable, severe dopaminergic deficiency. METHODS We applied one week of continuous unilateral STN-DBS in a group of stable 6-hydroxydopamine (6-OHDA) hemiparkinsonian rats (6-OHDASTIM) in comparison to a 6-OHDA control group (6-OHDASHAM) as well as healthy controls (CTRLSTIM and CTRLSHAM). BDNF and GDNF levels were determined via ELISAs. RESULTS The 6-OHDA lesion did not result in a persistent alteration in either BDNF or GDNF levels in a model of severe dopaminergic deficiency after completion of the dopaminergic degeneration. STN-DBS modestly increased BDNF levels in the entopeduncular nucleus, but even impaired BDNF and GDNF expression in cortical areas. CONCLUSIONS STN-DBS does not increase growth factor expression when applied to a model of completed, severe dopaminergic deficiency in contrast to other studies in models of modest and ongoing dopaminergic degeneration. In healthy controls, STN-DBS does not influence BDNF or GDNF expression. We consider these findings relevant for clinical purposes since DBS in PD is usually applied late in the course of the disease.
Collapse
Affiliation(s)
- Meike Statz
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Frederike Schleuter
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Hanna Weber
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Maria Kober
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Franz Plocksties
- Institute of Applied Microelectronics and Computer Engineering, University of Rostock, Albert-Einstein-Str. 26, 18119 Rostock, Germany
| | - Dirk Timmermann
- Institute of Applied Microelectronics and Computer Engineering, University of Rostock, Albert-Einstein-Str. 26, 18119 Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany; German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Mareike Fauser
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany.
| |
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: 1.0] [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
|
Pecar G, Liu S, Hooda J, Atkinson JM, Oesterreich S, Lee AV. RET signaling in breast cancer therapeutic resistance and metastasis. Breast Cancer Res 2023; 25:26. [PMID: 36918928 PMCID: PMC10015789 DOI: 10.1186/s13058-023-01622-7] [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: 05/23/2022] [Accepted: 02/16/2023] [Indexed: 03/15/2023] Open
Abstract
RET, a single-pass receptor tyrosine kinase encoded on human chromosome 10, is well known to the field of developmental biology for its role in the ontogenesis of the central and enteric nervous systems and the kidney. In adults, RET alterations have been characterized as drivers of non-small cell lung cancer and multiple neuroendocrine neoplasms. In breast cancer, RET signaling networks have been shown to influence diverse functions including tumor development, metastasis, and therapeutic resistance. While RET is known to drive the development and progression of multiple solid tumors, therapeutic agents selectively targeting RET are relatively new, though multiple multi-kinase inhibitors have shown promise as RET inhibitors in the past; further, RET has been historically neglected as a potential therapeutic co-target in endocrine-refractory breast cancers despite mounting evidence for a key pathologic role and repeated description of a bi-directional relationship with the estrogen receptor, the principal driver of most breast tumors. Additionally, the recent discovery of RET enrichment in breast cancer brain metastases suggests a role for RET inhibition specific to advanced disease. This review assesses the status of research on RET in breast cancer and evaluates the therapeutic potential of RET-selective kinase inhibitors across major breast cancer subtypes.
Collapse
Affiliation(s)
- Geoffrey Pecar
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Simeng Liu
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- School of Medicine, Tsinghua University, Beijing, China
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jagmohan Hooda
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Jennifer M Atkinson
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
5
|
Turek J, Gąsior Ł. Estrogen fluctuations during the menopausal transition are a risk factor for depressive disorders. Pharmacol Rep 2023; 75:32-43. [PMID: 36639604 PMCID: PMC9889489 DOI: 10.1007/s43440-022-00444-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023]
Abstract
Women are significantly more likely to develop depression than men. Fluctuations in the ovarian estrogen hormone levels are closely linked with women's well-being. This narrative review discusses the available knowledge on the role of estrogen in modulating brain function and the correlation between changes in estrogen levels and the development of depression. Equally discussed are the possible mechanisms underlying these effects, including the role of estrogen in modulating brain-derived neurotrophic factor activity, serotonin neurotransmission, as well as the induction of inflammatory response and changes in metabolic activity, are discussed.
Collapse
Affiliation(s)
- Justyna Turek
- Department of Neurobiology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland
| | - Łukasz Gąsior
- Department of Neurobiology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland
| |
Collapse
|
6
|
Niu P, Li L, Zhang Y, Su Z, Wang B, Liu H, Zhang S, Qiu S, Li Y. Immune regulation based on sex differences in ischemic stroke pathology. Front Immunol 2023; 14:1087815. [PMID: 36793730 PMCID: PMC9923235 DOI: 10.3389/fimmu.2023.1087815] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/02/2023] [Indexed: 01/31/2023] Open
Abstract
Ischemic stroke is one of the world's leading causes of death and disability. It has been established that gender differences in stroke outcomes prevail, and the immune response after stroke is an important factor affecting patient outcomes. However, gender disparities lead to different immune metabolic tendencies closely related to immune regulation after stroke. The present review provides a comprehensive overview of the role and mechanism of immune regulation based on sex differences in ischemic stroke pathology.
Collapse
Affiliation(s)
- Pingping Niu
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Liqin Li
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Yonggang Zhang
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Zhongzhou Su
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Binghao Wang
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - He Liu
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Shehong Zhang
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Sheng Qiu
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| | - Yuntao Li
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China.,Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuro Modulation, Huzhou, China
| |
Collapse
|
7
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
8
|
Tang T, Hu L, Liu Y, Fu X, Li J, Yan F, Cao S, Chen G. Sex-Associated Differences in Neurovascular Dysfunction During Ischemic Stroke. Front Mol Neurosci 2022; 15:860959. [PMID: 35431804 PMCID: PMC9012443 DOI: 10.3389/fnmol.2022.860959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/28/2022] [Indexed: 12/28/2022] Open
Abstract
Neurovascular units (NVUs) are basic functional units in the central nervous system and include neurons, astrocytes and vascular compartments. Ischemic stroke triggers not only neuronal damage, but also dissonance of intercellular crosstalk within the NVU. Stroke is sexually dimorphic, but the sex-associated differences involved in stroke-induced neurovascular dysfunction are studied in a limited extend. Preclinical studies have found that in rodent models of stroke, females have less neuronal loss, stronger repairing potential of astrocytes and more stable vascular conjunction; these properties are highly related to the cerebroprotective effects of female hormones. However, in humans, these research findings may be applicable only to premenopausal stroke patients. Women who have had a stroke usually have poorer outcomes compared to men, and because stoke is age-related, hormone replacement therapy for postmenopausal women may exacerbate stroke symptoms, which contradicts the findings of most preclinical studies. This stark contrast between clinical and laboratory findings suggests that understanding of neurovascular differences between the sexes is limited. Actually, apart from gonadal hormones, differences in neuroinflammation as well as genetics and epigenetics promote the sexual dimorphism of NVU functions. In this review, we summarize the confirmed sex-associated differences in NVUs during ischemic stroke and the possible contributing mechanisms. We also describe the gap between clinical and preclinical studies in terms of sexual dimorphism.
Collapse
Affiliation(s)
- Tianchi Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Libin Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Liu
- Department of Ultrasonography, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Shenglong Cao,
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Gao Chen,
| |
Collapse
|
9
|
From Menopause to Neurodegeneration-Molecular Basis and Potential Therapy. Int J Mol Sci 2021; 22:ijms22168654. [PMID: 34445359 PMCID: PMC8395405 DOI: 10.3390/ijms22168654] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
The impacts of menopause on neurodegenerative diseases, especially the changes in steroid hormones, have been well described in cell models, animal models, and humans. However, the therapeutic effects of hormone replacement therapy on postmenopausal women with neurodegenerative diseases remain controversial. The steroid hormones, steroid hormone receptors, and downstream signal pathways in the brain change with aging and contribute to disease progression. Estrogen and progesterone are two steroid hormones which decline in circulation and the brain during menopause. Insulin-like growth factor 1 (IGF-1), which plays an import role in neuroprotection, is rapidly decreased in serum after menopause. Here, we summarize the actions of estrogen, progesterone, and IGF-1 and their signaling pathways in the brain. Since the incidence of Alzheimer’s disease (AD) is higher in women than in men, the associations of steroid hormone changes and AD are emphasized. The signaling pathways and cellular mechanisms for how steroid hormones and IGF-1 provide neuroprotection are also addressed. Finally, the molecular mechanisms of potential estrogen modulation on N-methyl-d-aspartic acid receptors (NMDARs) are also addressed. We provide the viewpoint of why hormone therapy has inconclusive results based on signaling pathways considering their complex response to aging and hormone treatments. Nonetheless, while diagnosable AD may not be treatable by hormone therapy, its preceding stage of mild cognitive impairment may very well be treatable by hormone therapy.
Collapse
|
10
|
Wang G, Wang H, Zhang L, Guo F, Wu X, Liu Y. MiR-195-5p inhibits proliferation and invasion of nerve cells in Hirschsprung disease by targeting GFRA4. Mol Cell Biochem 2021; 476:2061-2073. [PMID: 33515383 DOI: 10.1007/s11010-021-04055-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/11/2021] [Indexed: 12/18/2022]
Abstract
Studies have reported that miR-195-5p plays a role in the Hirschsprung disease (HSCR). Our previous work found GDNF family receptor alpha 4 (GFRA4) is also associated with HSCR. In this study, we focused on whether miR-195-5p induces the absence of enteric neurons and enteric neural crest in HSCR by regulating GFRA4. The expression levels of GFRA4 and miR-195-5p in colon tissues were evaluated by real-time PCR (RT-PCR) assay. We overexpressed GFRA4 or miR-195-5p in SH-SY5Y cells, the cell proliferation, cell cycle, apoptosis and invasion were subsequently investigated by CCK-8 assay, EdU staining, Flow cytometry analysis and Transwell assay, respectively. We also established the xenograft model to detect the effect of miR-195-5p on tumor growth and GFRA4 and p-RET expressions. GFRA4 expression was significantly downregulated in the HSCR colon tissues when compared with that in the control tissues. Overexpression of GFRA4 significantly promoted proliferation, invasion and cell cycle arrest, and inhibited apoptosis of SH-SY5Y cells. We also proved that GFRA4 is a direct target of miR-195-5p, and miR-195-5p inhibited proliferation, invasion, cell cycle arrest and differentiation, and accelerated apoptosis in SH-SY5Y cells which can be reversed by GFRA4 overexpression. Furthermore, we demonstrated that miR-195-5p suppressed tumor growth, and observably decreased GFRA4 and p-RET expressions. Our findings suggest that miR-195-5p plays an important role in the pathogenesis of HSCR. MiR-195-5p inhibited proliferation, invasion and cell cycle arrest, and accelerated apoptosis of nerve cells by targeting GFRA4.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, China.
| | - Hefeng Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Lijuan Zhang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Feng Guo
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Xiangyu Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Yang Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, China
| |
Collapse
|
11
|
Parkinson's disease in women: Mechanisms underlying sex differences. Eur J Pharmacol 2021; 895:173862. [PMID: 33450279 DOI: 10.1016/j.ejphar.2021.173862] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/10/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
Parkinson's disease is a neurodegenerative disease which is associated with different motor, cognitive and mood-related problems. Though it has been established that Parkinson's disease is less prevalent in women in comparison to men, the differences tend to diminish with the advancing age. Different genetic, hormonal, neuroendocrinal and molecular players contribute towards the differences in the Parkinson's disease pathogenesis. Furthermore, data available with respect to the therapeutic management of Parkinson's disease in females is limited; women often tend to suffer more from the side effects of the currently available drugs. The present review highlights the sex-specific differences which play a role in the manifestation of these symptoms and side effects of the currently available therapeutic strategies. We have also discussed the current and upcoming therapeutic strategies which are in the clinical trials such as adenosine 2A (A2A) receptor antagonists, estrogen replacement therapy, α-synuclein targeting vaccines and antibodies, Botulinum toxin A, Fas-associated factor-1 (FAF-1) inhibitors, thiazolidinediones, 5-HT1A receptor agonists, dopamine D1/D5 receptor agonists, Glucagon-like peptide 1 (GLP-1) analogues and certain plant based principles for the treatment of Parkinson's disease in women.
Collapse
|
12
|
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: 5.8] [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
|
13
|
Roque C, Mendes-Oliveira J, Duarte-Chendo C, Baltazar G. The role of G protein-coupled estrogen receptor 1 on neurological disorders. Front Neuroendocrinol 2019; 55:100786. [PMID: 31513775 DOI: 10.1016/j.yfrne.2019.100786] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023]
Abstract
G protein-coupled estrogen receptor 1 (GPER) is a membrane-associated estrogen receptor (ER) associated with rapid estrogen-mediated effects. Over recent years GPER emerged has a potential therapeutic target to induce neuroprotection, avoiding the side effects elicited by the activation of classical ERs. The putative neuroprotection triggered by GPER selective activation was demonstrated in mood disorders, Alzheimer's disease or Parkinson's disease of male and female in vivo rodent models. In others, like ischemic stroke, the results are contradictory and currently there is no consensus on the role played by this receptor. However, it seems clear that sex is a biological variable that may impact the results. The major objective of this review is to provide an overview about the physiological effects of GPER in the brain and its putative contribution in neurodegenerative disorders, discussing the data about the signaling pathways involved, as well as, the diverse effects observed.
Collapse
Affiliation(s)
- C Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - J Mendes-Oliveira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - C Duarte-Chendo
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - G Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
14
|
Imamura Y, Shinozaki T, Okada-Ogawa A, Noma N, Shinoda M, Iwata K, Wada A, Abe O, Wang K, Svensson P. An updated review on pathophysiology and management of burning mouth syndrome with endocrinological, psychological and neuropathic perspectives. J Oral Rehabil 2019; 46:574-587. [PMID: 30892737 DOI: 10.1111/joor.12795] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/19/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
Burning mouth syndrome (BMS) is a chronic oro-facial pain disorder of unknown cause. It is more common in peri- and post-menopausal women, and sex hormone dysregulation is believed to be an important causative factor. Psychosocial events often trigger or exacerbate symptoms, and persons with BMS appear to be predisposed towards anxiety and depression. Atrophy of small nerve fibres in the tongue epithelium has been reported, and potential neuropathic mechanisms for BMS are now widely investigated. Historically, BMS was thought to comprise endocrinological, psychosocial and neuropathic components. Neuroprotective steroids and glial cell line-derived neurotrophic factor family ligands may have pivotal roles in the peripheral mechanisms associated with atrophy of small nerve fibres. Denervation of chorda tympani nerve fibres that innervate fungiform buds leads to alternative trigeminal innervation, which results in dysgeusia and burning pain when eating hot foods. With regard to the central mechanism of BMS, depletion of neuroprotective steroids alters the brain network-related mood and pain modulation. Peripheral mechanistic studies support the use of topical clonazepam and capsaicin for the management of BMS, and some evidence supports the use of cognitive behavioural therapy. Hormone replacement therapy may address the causes of BMS, although adverse effects prevent its use as a first-line treatment. Selective serotonin reuptake inhibitors (SSRIs) and serotonin and noradrenaline reuptake inhibitors (SNRIs) may have important benefits, and well-designed controlled studies are expected. Other treatment options to be investigated include brain stimulation and TSPO (translocator protein 18 kDa) ligands.
Collapse
Affiliation(s)
- Yoshiki Imamura
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan.,Nihon University School of Dentistry Dental Research Center, Chiyoda-ku, Tokyo, Japan
| | - Takahiro Shinozaki
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan.,Nihon University School of Dentistry Dental Research Center, Chiyoda-ku, Tokyo, Japan
| | - Akiko Okada-Ogawa
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan.,Nihon University School of Dentistry Dental Research Center, Chiyoda-ku, Tokyo, Japan
| | - Noboru Noma
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan.,Nihon University School of Dentistry Dental Research Center, Chiyoda-ku, Tokyo, Japan
| | - Masahiro Shinoda
- Nihon University School of Dentistry Dental Research Center, Chiyoda-ku, Tokyo, Japan.,Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| | - Koichi Iwata
- Nihon University School of Dentistry Dental Research Center, Chiyoda-ku, Tokyo, Japan.,Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| | - Akihiko Wada
- Department of Radiology, Faculty of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kelun Wang
- Department of Health Science and Technology, Center for Sensory-Motor Interaction, Aalborg University, Aalborg, Denmark
| | - Peter Svensson
- Department of Dentistry and Oral Health, Section for Orofacial Pain and Jaw Function, Aarhus University, Aarhus, Denmark
| |
Collapse
|
15
|
Benefits of Vitamins in the Treatment of Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9426867. [PMID: 30915197 PMCID: PMC6402202 DOI: 10.1155/2019/9426867] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/04/2019] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the elderly, which is clinically characterized by bradykinesia, resting tremor, abnormal posture balance, and hypermyotonia. Currently, the pathogenic mechanism of PD remains unclear. Numerous clinical studies as well as animal and cell experiments have found a certain relationship between the vitamin family and PD. The antioxidant properties of vitamins and their biological functions of regulating gene expression may be beneficial for the treatment of PD. Current clinical evidence indicates that proper supplementation of various vitamins can reduce the incidence of PD in the general population and improve the clinical symptoms of patients with PD; nevertheless, the safety of regular vitamin supplements still needs to be highlighted. Vitamin supplementation may be an effective adjuvant treatment for PD. In this review, we summarized the biological correlations between vitamins and PD as well as the underlying pathophysiological mechanisms. Additionally, we elaborated the therapeutic potentials of vitamins for PD.
Collapse
|
16
|
Baez-Jurado E, Rincón-Benavides MA, Hidalgo-Lanussa O, Guio-Vega G, Ashraf GM, Sahebkar A, Echeverria V, Garcia-Segura LM, Barreto GE. Molecular mechanisms involved in the protective actions of Selective Estrogen Receptor Modulators in brain cells. Front Neuroendocrinol 2019; 52:44-64. [PMID: 30223003 DOI: 10.1016/j.yfrne.2018.09.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
Synthetic selective modulators of the estrogen receptors (SERMs) have shown to protect neurons and glial cells against toxic insults. Among the most relevant beneficial effects attributed to these compounds are the regulation of inflammation, attenuation of astrogliosis and microglial activation, prevention of excitotoxicity and as a consequence the reduction of neuronal cell death. Under pathological conditions, the mechanism of action of the SERMs involves the activation of estrogen receptors (ERs) and G protein-coupled receptor for estrogens (GRP30). These receptors trigger neuroprotective responses such as increasing the expression of antioxidants and the activation of kinase-mediated survival signaling pathways. Despite the advances in the knowledge of the pathways activated by the SERMs, their mechanism of action is still not entirely clear, and there are several controversies. In this review, we focused on the molecular pathways activated by SERMs in brain cells, mainly astrocytes, as a response to treatment with raloxifene and tamoxifen.
Collapse
Affiliation(s)
- E Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - M A Rincón-Benavides
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - O Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - G Guio-Vega
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - G M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - V 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
| | - L M Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - G 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
|
17
|
Wang G, Zhang L, Wang H, Cui M, Liu W, Liu Y, Wu X. Demethylation of GFRA4 Promotes Cell Proliferation and Invasion in Hirschsprung Disease. DNA Cell Biol 2018; 37:316-324. [PMID: 29634418 DOI: 10.1089/dna.2017.3928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hirschsprung disease (HSCR) is congenital intestinal aganglionosis attributed to a failure to migrate and survive of neural crest-derived cells. Glial cell-derived neurotrophic factor alpha 4 (GFRA4) is expressed in the derivatives of the neural crest in the enteric nervous system, but whether it is related with HSCR still remains unclear. This study was designed to investigate its role and epigenetic mechanisms in HSCR in vitro. The expression of GFRA4 mRNA in HSCR tissues was determined using quantitative real-time PCR analysis. In this study, we found that GFRA4 expression was significantly reduced in HSCR tissues and cells through GFRA4 methylation by quantitative real-time PCR analysis, methylation-specific PCR, and bisulfite sequencing PCR. DNA methyltransferase inhibitor, 5-AzaC, concomitantly upregulated the protein levels of GFRA4, as well as DNA methyltransferase1 (DNMT1) and DNMT2 in SH-5YSY cells. Moreover, we found upregulated GFRA4 significantly promoted cell proliferation, cell cycle progression and invasion, but inhibited apoptosis in SH-5YSY cells, whereas GFRA4 knockdown caused the opposite effects in SH-5YSY cells by CCK-8, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and Transwell assays. In conclusion, our results support that aberrant CpG hypermethylation at least partly accounts for GFRA4 silencing in HSCR, which impairs its protective role in enteric nervous system.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Lijuan Zhang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Hefeng Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Mingyu Cui
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Yang Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Xiangyu Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| |
Collapse
|
18
|
Labandeira-Garcia JL, Costa-Besada MA, Labandeira CM, Villar-Cheda B, Rodríguez-Perez AI. Insulin-Like Growth Factor-1 and Neuroinflammation. Front Aging Neurosci 2017; 9:365. [PMID: 29163145 PMCID: PMC5675852 DOI: 10.3389/fnagi.2017.00365] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) effects on aging and neurodegeneration is still controversial. However, it is widely admitted that IGF-1 is involved in the neuroinflammatory response. In peripheral tissues, several studies showed that IGF-1 inhibited the expression of inflammatory markers, although other studies concluded that IGF-1 has proinflammatory functions. Furthermore, proinflammatory cytokines such as TNF-α impaired IGF-1 signaling. In the brain, there are controversial results on effects of IGF-1 in neuroinflammation. In addition to direct protective effects on neurons, several studies revealed anti-inflammatory effects of IGF-1 acting on astrocytes and microglia, and that IGF-1 may also inhibit blood brain barrier permeability. Altogether suggests that the aging-related decrease in IGF-1 levels may contribute to the aging-related pro-inflammatory state. IGF-1 inhibits the astrocytic response to inflammatory stimuli, and modulates microglial phenotype (IGF-1 promotes the microglial M2 and inhibits of M1 phenotype). Furthermore, IGF-1 is mitogenic for microglia. IGF-1 and estrogen interact to modulate the neuroinflammatory response and microglial and astrocytic phenotypes. Brain renin-angiotensin and IGF-1 systems also interact to modulate neuroinflammation. Induction of microglial IGF-1 by angiotensin, and possibly by other pro-inflammatory inducers, plays a major role in the repression of the M1 microglial neurotoxic phenotype and the enhancement of the transition to an M2 microglial repair/regenerative phenotype. This mechanism is impaired in aged brains. Aging-related decrease in IGF-1 may contribute to the loss of capacity of microglia to undergo M2 activation. Fine tuning of IGF-1 levels may be critical for regulating the neuroinflammatory response, and IGF-1 may be involved in inflammation in a context-dependent mode.
Collapse
Affiliation(s)
- 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), Madrid, Spain
| | - Maria A Costa-Besada
- 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), Madrid, Spain
| | - Carmen M Labandeira
- Department of Clinical Neurology, Hospital Alvaro Cunqueiro, University Hospital Complex, Vigo, Spain
| | - Begoña Villar-Cheda
- 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), Madrid, Spain
| | - Ana I Rodríguez-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), Madrid, Spain
| |
Collapse
|
19
|
Zhou CH, Zhang YH, Xue F, Xue SS, Chen YC, Gu T, Peng ZW, Wang HN. Isoflurane exposure regulates the cell viability and BDNF expression of astrocytes via upregulation of TREK‑1. Mol Med Rep 2017; 16:7305-7314. [PMID: 28944872 PMCID: PMC5865860 DOI: 10.3892/mmr.2017.7547] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/26/2017] [Indexed: 12/17/2022] Open
Abstract
Neonatal isoflurane exposure in rodents disrupts hippocampal cognitive functions, including learning and memory, and astrocytes may have an important role in this process. However, the molecular mechanisms underlying this disruption are not fully understood. The present study investigated the role of TWIK-related K+ channel (TREK-1) in isoflurane-induced cognitive impairment. Lentiviruses were used to overexpress or knockdown TREK-1 in astrocytes exposed to increasing concentrations of isoflurane or O2 for 2 h. Subsequently, the mRNA and protein expression of brain-derived neurotrophic factor (BDNF), caspase-3, Bcl-2-associated X (Bax) and TREK-1 was measured by reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. In addition, cell viability was assessed by a 2-(4-Iodophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium monosodium salt assay. The results demonstrated that, prior to manipulating TREK-1, isoflurane significantly decreased the cell viability and BDNF expression, and increased Bax, caspase-3 and TREK-1 expression was observed. However, TREK-1 overexpression in astrocytes significantly downregulated BDNF expression, and upregulated Bax and caspase-3 expression. Furthermore, lentiviral-mediated short hairpin RNA knockdown of TREK-1 effectively inhibited the isoflurane-induced changes in BDNF, Bax and caspase-3 expression. Taken together, the results of the present study indicate that isoflurane-induced cell damage in astrocytes may be associated with TREK-1-mediated inhibition of BDNF and provide a reference for the safe use of isoflurane anesthesia in infants and children.
Collapse
Affiliation(s)
- Cui-Hong Zhou
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ya-Hong Zhang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Fen Xue
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shan-Shan Xue
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yun-Chun Chen
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ting Gu
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zheng-Wu Peng
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hua-Ning Wang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
20
|
Mendes-Oliveira J, Lopes Campos F, Videira RA, Baltazar G. GPER activation is effective in protecting against inflammation-induced nigral dopaminergic loss and motor function impairment. Brain Behav Immun 2017; 64:296-307. [PMID: 28450223 DOI: 10.1016/j.bbi.2017.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/29/2022] Open
Abstract
Increasing evidence suggest that excessive inflammatory responses from overactivated microglia play a critical role in Parkinson's disease (PD), contributing to, or exacerbating, nigral dopaminergic (DA) degeneration. Recent results from our group and others demonstrated that selective activation of G protein-coupled estrogen receptor (GPER) with the agonist G1 can protect DA neurons from 1-methyl-4-phenylpyridinium (MPP+) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxins. However, it is not known whether modulation of microglial responses is one of the mechanisms by which G1 exerts its DA neuroprotective effects. We analyzed, in the N9 microglial cell line, the effect of G1 on microglial activation induced by lipopolysaccharide (LPS) exposure. The results revealed that G1 significantly decrease phagocytic activity, expression of inducible nitric oxide synthase (iNOS) and release of nitric oxide (NO) induced by LPS. To determine the relevance of this anti-inflammatory effect to the protection of nigral DA cells, the effect of G1 was analyzed in male mice injected unilaterally in the substantia nigra (SN) with LPS. Although G1 treatment did not decrease LPS-induced increase of ionized calcium binding adaptor molecule 1 (iba-1) positive cells it significantly reduced interleukin-1beta (IL-1β), cluster of differentiation 68 (CD68) and iNOS mRNA levels, and totally inhibited nigral DA cell loss and, as a consequence, protected the motor function. In summary, our findings demonstrated that the G1 agonist is able to modulate microglial responses and to protect DA neurons and motor functions against a lesion induced by an inflammatory insult. Since G1 lacks the feminizing effects associated with agonists of the classical estrogen receptors (ERs), the use of G1 to selectively activate the GPER may be a promising strategy for the development of new therapeutics for the treatment of PD and other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Julieta Mendes-Oliveira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Filipa Lopes Campos
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Rita Alexandra Videira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Graça Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
21
|
Chen WF, Wu L, Du ZR, Chen L, Xu AL, Chen XH, Teng JJ, Wong MS. Neuroprotective properties of icariin in MPTP-induced mouse model of Parkinson's disease: Involvement of PI3K/Akt and MEK/ERK signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 25:93-99. [PMID: 28190476 DOI: 10.1016/j.phymed.2016.12.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/15/2016] [Accepted: 12/28/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Epimedium sagittatum is a traditional Chinese herb normally which is used to treat the osteoporosis, cardiovascular dysfunction, and to improve neurological and sexual function in China, Korea and Japan. Icariin is the major active ingredient in Epimedium sagittatum. In the present research, we examined the neuroprotective effects of icariin on dopaminergic neurons and the possible mechanisms in a mouse model of Parkinson's disease (PD). METHODS Ovariectomized PD mice were treated with vehicle or icariin (3 days before MPTP injections) with or without the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 or mitogen-activated protein kinase kinase (MEK) inhibitor PD98059. The dopamine (DA) content in the striatum was studied by HPLC. Western blot was used to determine the protein expressions of Bcl-2, Bax and Caspase 3 in the striatum. The numbers of tyrosine hydroxylase-immunoreactive (TH-IR) neurons in the substantial nigra pars compacta (SNpc) were assessed by immunohistochemistry. The activation of Akt and ERK by icariin were detected in doparminergic MES23.5 cells. RESULTS Icariin pretreatment could ameliorate the decreased striatum DA content and the loss of TH-IR neurons in the SNpc induced by MPTP. The MPTP-induced changes of Bcl-2, Bax and caspase 3 protein expressions in the striatum could be reversed by icariin pretreatment. Blockade of PI3K/Akt or MEK/ERK signaling pathway by LY294002 or PD98059 could attenuate the increase of DA content in the striatum and TH-IR in the SNpc induced by icariin in PD mice model. Additionally, icariin treatment alone significantly induced the phosphorylation of Akt and ERK in a time dependent pattern in dopaminergic MES 23.5 cells. These effects were abolished by co-treatment with LY294002 or PD98059. CONCLUSION These data demonstrated that icariin has neuroprotective effect on dopaminergic neurons in PD mice model and the potential mechanisms might be related to PI3K/Akt and MEK/ERK pathways.
Collapse
Affiliation(s)
- Wen-Fang Chen
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao 266071, China..
| | - Lin Wu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao 266071, China.; Department of Physiology, Heze Medical College, Heze 274000, China
| | - Zhong-Rui Du
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao 266071, China
| | - Lei Chen
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao 266071, China
| | - Ai-Li Xu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao 266071, China.; Department of Physiology, Binzhou Medical University, Yantai 264003, China
| | - Xiao-Han Chen
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao 266071, China
| | - Ji-Jun Teng
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, SAR, PR China
| |
Collapse
|
22
|
Xu Q, Fan W, Ye SF, Cong YB, Qin W, Chen SY, Cai J. Cistanche tubulosa Protects Dopaminergic Neurons through Regulation of Apoptosis and Glial Cell-Derived Neurotrophic Factor: in vivo and in vitro. Front Aging Neurosci 2016; 8:295. [PMID: 28018211 PMCID: PMC5159610 DOI: 10.3389/fnagi.2016.00295] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 11/21/2016] [Indexed: 11/30/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease with the pathological hallmark of reduced nigrostriatal dopamine. In traditional Chinese medicine (TCM) clinical practice, the nanopowder of Cistanche tubulosa has therapeutic effects on PD. To identify the therapeutic mechanism, this study tested the protective effect of different doses of MPP+-induced toxicity in MES23.5 cells using the MTT assay and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice (vehicles). Immunohistochemistry was used to assess cytomorphology and tyrosine hydroxylase (TH) expression. Behavioral tests in vehicles, high performance liquid chromatography (HPLC) tests in dopamine, immunohistochemistry and western blot analysis were used to detect the expression of TH, glial cell line-derived neurotrophic factor (GDNF) and its receptors. Our results demonstrated that the C. tubulosa nanopowder improved the viability of MPP+-treated cells, increased TH expression and reduced the number of apoptotic cells. It also increased Bcl2 protein expression and suppressed Bax protein expression in MPP+-treated cells in a dose-dependent manner. In addition, C. tubulosa nanopowder improved the behavioral deficits in vehicle mice, reduced the stationary duration of swimming, enhanced the ability for spontaneous activity and increased the expression of GDNF, the GDNF family receptor alpha (GFRα1) and Ret in cells of the substantia nigra (SN). Furthermore, the protein expression of GDNF, GFRα1 and Ret increased after treatment with different doses of C. tubulosa nanopowder, with a significant difference between the high-dose and vehicle groups. The protein expression of Bcl2 and Bax were similar in the in vivo and in vitro, which suggested that C. tubulosa nanopowder has anti-apoptotic effects in neurons.
Collapse
Affiliation(s)
- Qian Xu
- Institute of Integrated Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine Fuzhou, China
| | - Wen Fan
- Department of Internal Medicine, Xiamen Hai Cang Hospital Xiamen, China
| | - Shui-Fen Ye
- Department of Geratology, Longyan First Hospital Longyan, China
| | - Yi-Bo Cong
- Institute of Integrated Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine Fuzhou, China
| | - Wei Qin
- Institute of Integrated Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine Fuzhou, China
| | - Shi-Ya Chen
- Institute of Integrated Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine Fuzhou, China
| | - Jing Cai
- Institute of Integrated Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine Fuzhou, China
| |
Collapse
|
23
|
Enterría-Morales D, López-López I, López-Barneo J, d’Anglemont de Tassigny X. Striatal GDNF Production Is Independent to Circulating Estradiol Level Despite Pan-Neuronal Activation in the Female Mouse. PLoS One 2016; 11:e0164391. [PMID: 27741271 PMCID: PMC5065215 DOI: 10.1371/journal.pone.0164391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/23/2016] [Indexed: 11/25/2022] Open
Abstract
Gender difference in Parkinson’s disease (PD) suggests that female sex steroids may promote dopaminergic neuron survival and protect them from degeneration. The glial cell line-derived neurotrophic factor (GDNF) is believed to be dopaminotrophic; thus it is considered as a potential therapeutic target in PD. Additionally, GDNF is endogenously synthetized in the caudate/putamen of humans and striatum in rodents. A neuroprotective role of estrogens on the nigrostriatal pathway via the stimulation of GDNF has been proposed. Since the GDNF-producing parvalbumin (Parv) interneurons express the estrogen receptor alpha in the mouse striatum, we sought to determine whether ectopic estrogenic compound modulates the GDNF synthesis in mice. Using an ovariectomized-estradiol (E2) replacement regimen, which reliably generates a rise of plasma estradiol, we assessed the effects of different levels of E2 on the activation of striatal neuronal populations, and GDNF production. A strong correlation was found between plasma E2 and the expression of the immediate early gene cFos in the striatum, as well as in other cortical regions. However, moderate and high E2 treatments failed to induce any striatal GDNF mRNA and protein synthesis. High E2 only stimulates cFos induction in a low percentage of striatal Parv neurons whereas the majority of cFos-positive cells are medium spiny neurons. Activation of these projecting neurons by E2 suggests a role of circulating sex steroids in the modulation of striatal neural pathways.
Collapse
Affiliation(s)
- Daniel Enterría-Morales
- Instituto de Biomedicina de Sevilla (IBIS), Departamento de Fisiología Médica y Biofísica, Hospital Universitario Virgen del Rocío/ CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ivette López-López
- Instituto de Biomedicina de Sevilla (IBIS), Departamento de Fisiología Médica y Biofísica, Hospital Universitario Virgen del Rocío/ CSIC/Universidad de Sevilla, Sevilla, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBIS), Departamento de Fisiología Médica y Biofísica, Hospital Universitario Virgen del Rocío/ CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Xavier d’Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBIS), Departamento de Fisiología Médica y Biofísica, Hospital Universitario Virgen del Rocío/ CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- * E-mail:
| |
Collapse
|
24
|
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: 4.0] [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
|
25
|
Xie H, Xiao Z, Huang J. C6 Glioma-Secreted NGF and FGF2 Regulate Neuronal APP Processing Through Up-Regulation of ADAM10 and Down-Regulation of BACE1, Respectively. J Mol Neurosci 2015; 59:334-42. [PMID: 26614345 DOI: 10.1007/s12031-015-0690-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/19/2015] [Indexed: 01/05/2023]
Abstract
Excessive accumulation of amyloid-β (Aβ) caused by cleavage of amyloid precursor protein (APP) is thought to be the primary cause of Alzheimer's disease (AD). Two key enzymes ADAM10 and BACE1 are involved in the initial cleavage of APP, resulting in the onset of two pathways, the amyloidogenic pathway and the non-amyloidogenic pathway, respectively. Altering APP metabolism towards the non-amyloidogenic pathway is thought to reduce Aβ production. It has been reported that, in vivo, exogenous neurotrophic factors make APP apt to entering the non-amyloidogenic pathway. Since astrocytes secrete a battery of neurotrophic factors, we investigated the role of astrocyte-derived factors in the dynamics of Aβ generation in neural cells. Results show that C6 glioma cell-conditioned medium (GCM), obtained from cultured astrocyte-derived C6 glioma cells, inhibit Aβ1-42 production and shift APP processing towards the non-amyloidogenic pathway in APPswe-HEK293 cells. Such effect is attributed to two key APP cleavage enzymes, ADAM10 and BACE1. Two neurotrophic factors in the GCM, nerve growth factor and fibroblast growth factor 2, are responsible for the up-regulation of ADAM10 and down-regulation of BACE1, respectively. Our findings enhance our understanding of the relationship between astrocytes and Aβ generation, indicating that stimulation of astrocytic neurotrophic factors could slow AD progression.
Collapse
Affiliation(s)
- Huiping Xie
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China
| | - Zhimin Xiao
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China.,Sanofi, Cambridge, MA, 02142, USA
| | - Jian Huang
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China.
| |
Collapse
|
26
|
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: 2.1] [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]
|
27
|
d'Anglemont de Tassigny X, Pascual A, López-Barneo J. GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway. Implications for Parkinson's disease. Front Neuroanat 2015; 9:10. [PMID: 25762899 PMCID: PMC4327623 DOI: 10.3389/fnana.2015.00010] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
The glial cell line-derived neurotrophic factor (GDNF) is a well-established trophic agent for dopaminergic (DA) neurons in vitro and in vivo. GDNF is necessary for maintenance of neuronal morphological and neurochemical phenotype and protects DA neurons from toxic damage. Numerous studies on animal models of Parkinson’s disease (PD) have reported beneficial effects of GDNF on nigrostriatal DA neuron survival. However, translation of these observations to the clinical setting has been hampered so far by side effects associated with the chronic continuous intra-striatal infusion of recombinant GDNF. In addition, double blind and placebo-controlled clinical trials have not reported any clinically relevant effect of GDNF on PD patients. In the past few years, experiments with conditional Gdnf knockout mice have suggested that GDNF is necessary for maintenance of DA neurons in adulthood. In parallel, new methodologies for exogenous GDNF delivery have been developed. Recently, it has been shown that a small population of scattered, electrically interconnected, parvalbumin positive (PV+) GABAergic interneurons is responsible for most of the GDNF produced in the rodent striatum. In addition, cholinergic striatal interneurons appear to be also involved in the modulation of striatal GDNF. In this review, we summarize current knowledge on brain GDNF delivery, homeostasis, and its effects on nigrostriatal DA neurons. Special attention is paid to the therapeutic potential of endogenous GDNF stimulation in PD.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain ; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla Seville, Spain ; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid, Spain
| |
Collapse
|
28
|
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: 2.1] [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
|
29
|
Labandeira-García JL, Garrido-Gil P, Rodriguez-Pallares J, Valenzuela R, Borrajo A, Rodríguez-Perez AI. Brain renin-angiotensin system and dopaminergic cell vulnerability. Front Neuroanat 2014; 8:67. [PMID: 25071471 PMCID: PMC4086395 DOI: 10.3389/fnana.2014.00067] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 06/24/2014] [Indexed: 01/11/2023] Open
Abstract
Although the renin-angiotensin system (RAS) was classically considered as a circulating system that regulates blood pressure, many tissues are now known to have a local RAS. Angiotensin, via type 1 receptors, is a major activator of the NADPH-oxidase complex, which mediates several key events in oxidative stress (OS) and inflammatory processes involved in the pathogenesis of major aging-related diseases. Several studies have demonstrated the presence of RAS components in the basal ganglia, and particularly in the nigrostriatal system. In the nigrostriatal system, RAS hyperactivation, via NADPH-oxidase complex activation, exacerbates OS and the microglial inflammatory response and contributes to progression of dopaminergic degeneration, which is inhibited by angiotensin receptor blockers and angiotensin converting enzyme (ACE) inhibitors. Several factors may induce an increase in RAS activity in the dopaminergic system. A decrease in dopaminergic activity induces compensatory upregulation of local RAS function in both dopaminergic neurons and glia. In addition to its role as an essential neurotransmitter, dopamine may also modulate microglial inflammatory responses and neuronal OS via RAS. Important counterregulatory interactions between angiotensin and dopamine have also been observed in several peripheral tissues. Neurotoxins and proinflammatory factors may also act on astrocytes to induce an increase in RAS activity, either independently of or before the loss of dopamine. Consistent with a major role of RAS in dopaminergic vulnerability, increased RAS activity has been observed in the nigra of animal models of aging, menopause and chronic cerebral hypoperfusion, which also showed higher dopaminergic vulnerability. Manipulation of the brain RAS may constitute an effective neuroprotective strategy against dopaminergic vulnerability and progression of Parkinson's disease.
Collapse
Affiliation(s)
- Jose L Labandeira-García
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Jannette Rodriguez-Pallares
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Rita Valenzuela
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Ana I Rodríguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| |
Collapse
|
30
|
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: 4.8] [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]
|
31
|
Fonseca CP, Gama S, Saavedra A, Baltazar G. H2O2- or l-DOPA-injured dopaminergic neurons trigger the release of soluble mediators that up-regulate striatal GDNF through different signalling pathways. Biochim Biophys Acta Mol Basis Dis 2014; 1842:927-34. [DOI: 10.1016/j.bbadis.2014.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/21/2014] [Accepted: 03/04/2014] [Indexed: 11/26/2022]
|
32
|
Rocha SM, Pires J, Esteves M, Graça B, Bernardino L. Histamine: a new immunomodulatory player in the neuron-glia crosstalk. Front Cell Neurosci 2014; 8:120. [PMID: 24817841 PMCID: PMC4012198 DOI: 10.3389/fncel.2014.00120] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/16/2014] [Indexed: 01/05/2023] Open
Abstract
Histamine is an amine acting as a major peripheral inflammatory mediator. In the brain, histamine was initially viewed as a neurotransmitter, but new evidences support its involvement in the modulation of innate immune responses. Recently, we showed that histamine modulates microglial migration and cytokine release. Its pleiotropic actions, ranging from neurotransmission to inflammation, highlight histamine as a key player in a vast array of brain physiologic activities and also in the pathogenesis of several neurodegenerative diseases. Herein, we emphasize the role of histamine as a modulator of brain immune reactions, either by acting on invading peripheral immune cells and/or on resident microglial cells. We also unveil the putative involvement of histamine in the microglial-neuronal communication. We first show that histamine modulates the release of inflammatory mediators, namely nitric oxide, by microglia cells. Consequently, the microglia secretome released upon histamine stimulation fosters dopaminergic neuronal death. These data may reveal important new pharmacological applications on the use histamine and antihistamines, particularly in the context of Parkinson’s disease.
Collapse
Affiliation(s)
- Sandra M Rocha
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior Covilhã, Portugal
| | - Joel Pires
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior Covilhã, Portugal
| | - Marta Esteves
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior Covilhã, Portugal
| | - Baltazar Graça
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior Covilhã, Portugal
| | - Liliana Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior Covilhã, Portugal
| |
Collapse
|
33
|
Smith KM, Dahodwala N. Sex differences in Parkinson's disease and other movement disorders. Exp Neurol 2014; 259:44-56. [PMID: 24681088 DOI: 10.1016/j.expneurol.2014.03.010] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 03/07/2014] [Accepted: 03/19/2014] [Indexed: 12/29/2022]
Abstract
Movement disorders including Parkinson's disease (PD), Huntington's disease (HD), chorea, tics, and Tourette's syndrome (TS) display sex differences in disease susceptibility, disease pathogenesis, and clinical presentation. PD is more common in males than in females. Epidemiologic studies suggest that exposure to endogenous and exogenous estrogen contributes to these sex differences. There is extensive evidence that estrogen prevents dopaminergic neuron depletion induced by neurotoxins in PD animal models and therefore is neuroprotective. Estrogen may also decrease the efficacy of other neuroprotective substances such as caffeine in females but not males. Sex chromosomes can exert effects independent of sex steroid hormones on the development and maintenance of the dopamine system. As a result of hormone, chromosome and other unknown effects, there are sexual dimorphisms in the basal ganglia, and at the molecular levels in dopaminergic neurons that may lead to distinct mechanisms of pathogenesis in males and females. In this review, we summarize the evidence that estrogen and selective estrogen receptor modulators are neuroprotective in PD and discuss potential mechanisms of action. We also briefly review how sex differences in basal ganglia function and dopaminergic pathways may impact HD, chorea, and tics/Tourette's syndrome. Further understanding of these sex differences may lead to novel therapeutic strategies for prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Kara M Smith
- Parkinson's Disease and Movement Disorders Center, 330 S. 9th St, 2nd Floor, Philadelphia, PA 19107, USA.
| | - Nabila Dahodwala
- Parkinson's Disease and Movement Disorders Center, 330 S. 9th St, 2nd Floor, Philadelphia, PA 19107, USA
| |
Collapse
|
34
|
Bourque M, Morissette M, Di Paolo T. Raloxifene activates G protein-coupled estrogen receptor 1/Akt signaling to protect dopamine neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice. Neurobiol Aging 2014; 35:2347-56. [PMID: 24726471 DOI: 10.1016/j.neurobiolaging.2014.03.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 12/20/2013] [Accepted: 03/14/2014] [Indexed: 11/18/2022]
Abstract
Raloxifene, used in the clinic, is reported to protect brain dopaminergic neurons in mice. Raloxifene was shown to mediate an effect through the G protein-coupled estrogen receptor 1 (GPER1). We investigated if raloxifene neuroprotective effect in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated male mice is mediated through GPER1 by using its antagonist G15. Striatal concentrations of dopamine, 3,4-dihydroxyphenylacetic acid, homovanillic acid to dopamine ratio as well as dopamine transporter and vesicular monoamine transporter 2 showed that raloxifene neuroprotection of dopaminergic neurons was blocked by G15. Protection by raloxifene was accompanied by activation of striatal Akt signaling (but not ERK1/2 signaling) and increased Bcl-2 and brain-derived neurotrophic factor levels; these effects were abolished by coadministration with G15. The effect of raloxifene was not mediated through increased levels of 17β-estradiol. MPTP mice had decreased plasma testosterone, dihydrotestosterone, and 3β-diol levels; this was prevented in raloxifene-treated MPTP mice. Our results suggest that raloxifene acted through GPER1 to mediate Akt activation, increase Bcl-2 and brain-derived neurotrophic factor levels, and protection of dopaminergic neurons and plasma androgens.
Collapse
Affiliation(s)
- Mélanie Bourque
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada; Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada; Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada.
| |
Collapse
|