1
|
de Souza Lima R, Fornaguera J. Sex differences in 6-OHDA lesioned rats in a preclinical model for Parkinson's disease. Behav Brain Res 2025; 490:115642. [PMID: 40389167 DOI: 10.1016/j.bbr.2025.115642] [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: 02/06/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025]
Abstract
Although Parkinson's disease (PD) was first described in 1817, its etiology remains unclear. The primary neurotransmitter system affected in PD is the nigrostriatal dopaminergic pathway, whose dysfunction leads to the hallmark motor symptoms of the disease. These motor symptoms typically emerge only after approximately 80 % of dopaminergic neurons are lost, complicating early detection and intervention. For this reason, it is important to know more about the stages prior to the appearance of symptoms that could represent the progression of the disease. Additionally, substantial evidence suggests sex differences in PD prevalence and progression, with men more frequently affected than women. However, most preclinical studies using animal models of PD have been conducted exclusively on males, limiting our understanding of neurochemical and behavioral differences between sexes, particularly under moderate dopaminergic lesions. To address this, we used the unilateral 6-hydroxydopamine (6-OHDA) model of PD, inducing moderate lesions in the substantia nigra pars compacta (SNpc) to investigate sex-specific variations. Our study revealed distinct neurochemical and behavioral profiles in male and female animals, including sex-specific responses to amphetamine and apomorphine challenges. These pharmacological tests highlighted contrasting patterns in asymmetric and symmetric behaviors between sexes. Moreover, we considered individual variability often overlooked in animal models, which could provide critical insights into resilience or susceptibility to dopaminergic loss. These findings underscore the importance of incorporating sex as a biological variable in PD research to better understand the disease's underlying mechanisms and to improve strategies for early diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
| | - Jaime Fornaguera
- Neuroscience Research Center, University of Costa Rica, Costa Rica; Biochemistry department, School of Medicine, University of Costa Rica, Costa Rica
| |
Collapse
|
2
|
Zhou T, Wang H. The role of glutamate dehydrogenase in the ageing brain. Front Pharmacol 2025; 16:1586655. [PMID: 40356954 PMCID: PMC12066631 DOI: 10.3389/fphar.2025.1586655] [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: 03/03/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
The homeostasis of glutamate, the primary excitatory neurotransmitter in the brain and is crucial for normal brain function. The mitochondrial enzyme glutamate dehydrogenase (GDH) connects the multifunctional amino acid glutamate, which is intimately related to glutamate metabolism, to the Krebs cycle. As a result, GDH reglutes the synthesis and uptake of the chemical messenger glutamate in neuroendocrine cells, playing a crucial role in the metabolism of proteins and carbohydrates. Nonetheless, brain ageing and numerous neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease, have been linked to GDH malfunction or dysregulation. In this review, we summarize the dynamics of GDH levels in the ageing brain and provide additional details about the role of GDH in the ageing brain. Understanding the metabolic mechanisms underlying glutamate homeostasis in the aging brain and how GDH regulates glutamate-dependent metabolic processes at synapses may lead to novel therapeutic approaches for neurodegenerative and psychiatric disorders, potentially slowing the aging process and promoting brain regeneration.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Pharmaceutical and Medical equipment, Bayi Orthopedic Hospital, China RongTong Medical Healthcare Group Co. Ltd., Chengdu, China
| | - Haichuan Wang
- Department of Paediatrics, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
3
|
Limberg AS, Berg F, Köper E, Lindgraf C, Gevers C, Kumsta R, Hummel EM, Moser DA. Cell-free DNA release following psychosocial and physical stress in women and men. Transl Psychiatry 2025; 15:26. [PMID: 39863589 PMCID: PMC11763022 DOI: 10.1038/s41398-025-03242-5] [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: 04/25/2024] [Revised: 12/05/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cell-free DNA (cfDNA) is continuously shed by all cells in the body, but the regulation of this process and its physiological functions are still largely unknown. Previous research has demonstrated that both nuclear (cf-nDNA) and mitochondrial (cf-mtDNA) cfDNA levels increase in plasma in response to acute psychosocial and physical stress in males. This study further investigated these findings by testing 31 female participants (16 using oral hormonal contraception and 15 not using oral hormonal contraception), and the results were subsequently compared with those of 16 male participants. In addition, cf-nDNA and cf-mtDNA were comparatively quantified in both plasma and saliva at four time points, 2 min before and 2, 15, and 45 min after stress induction. A novel method was implemented to facilitate the straightforward collection of capillary blood by non-medical personnel for plasma analysis. While cf-mtDNA is readily detectable in body fluids due to its high copy number, the quantification of cf-nDNA is challenging due to its low abundance. To overcome this, a multiplex quantitative polymerase chain reaction (qPCR) protocol targeting L1PA2 elements, which are prevalent in the human genome, was utilized. The analysis indicated significantly elevated levels of cf-nDNA in both plasma and saliva in all participants, irrespective of gender, following psychosocial and physical stress. Conversely, neither plasma nor saliva exhibited a consistent or stress-induced release pattern for cf-mtDNA. CfDNA is a promising biomarker that is consistently released after stress in both men and women and can be detected in both plasma and saliva. However, further research is necessary to elucidate the mechanisms of cfDNA release from specific cells and to understand its biological function in the body.
Collapse
Affiliation(s)
- A S Limberg
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - F Berg
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - E Köper
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - C Lindgraf
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - C Gevers
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - R Kumsta
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
- Department of Behavioural and Cognitive Sciences, Laboratory for Stress and Gene-Environment Interplay, University of Luxemburg, Porte des Sciences, Esch-sur-Alzette, Luxembourg
| | - E M Hummel
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - D A Moser
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany.
| |
Collapse
|
4
|
Hoffmeister JD, Broadfoot CK, Schaen-Heacock NE, Lechner SA, Krasko MN, Nisbet AF, Russell J, Szot J, Glass TJ, Connor NP, Kelm-Nelson CA, Ciucci MR. Vocal and tongue exercise in early to mid-stage Parkinson disease using the Pink1-/- rat. Brain Res 2024; 1837:148958. [PMID: 38685371 PMCID: PMC11166513 DOI: 10.1016/j.brainres.2024.148958] [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: 01/24/2024] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
Vocal and swallowing deficits are common in Parkinson disease (PD). Because these impairments are resistant to dopamine replacement therapies, vocal and lingual exercise are the primary treatment, but not all individuals respond to exercise and neural mechanisms of treatment response are unclear. To explore putative mechanisms, we used the progressive Pink1-/- rat model of early to mid-stage PD and employed vocal and lingual exercises at 6- and 10-months of age in male Pink1-/- and wild type (WT) rats. We hypothesized that vocal and lingual exercise would improve vocal and tongue use dynamics and increase serotonin (5HT) immunoreactivity in related brainstem nuclei. Rats were tested at baseline and after 8 weeks of exercise or sham exercise. At early-stage PD (6 months), vocal exercise resulted in increased call complexity, but did not change intensity, while at mid-stage (10 months), vocal exercise no longer influenced vocalization complexity. Lingual exercise increased tongue force generation and reduced relative optical density of 5HT in the hypoglossal nucleus at both time points. The effects of vocal and lingual exercise at these time points are less robust than in prodromal stages observed in previous work, suggesting that early exercise interventions may yield greater benefit. Future work targeting optimization of exercise at later time points may facilitate clinical translation.
Collapse
Affiliation(s)
- J D Hoffmeister
- University of Minnesota, Dept. of Otolaryngology, 420 Delaware Street SE, Minneapolis, MN 55422, USA; University of Wisconsin-Madison, Dept. of Communication Sciences and Disorders, 1975 Willow Drive, Madison, WI 53706, USA.
| | - C K Broadfoot
- University of South Alabama, Dept. of Speech Pathology and Audiology, 5721 USA Drive N, HAHN 1119, Mobile, AL 36688, USA; University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - N E Schaen-Heacock
- University of Wisconsin-Madison, Dept. of Communication Sciences and Disorders, 1975 Willow Drive, Madison, WI 53706, USA; University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - S A Lechner
- University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - M N Krasko
- University of Wisconsin-Madison, Dept. of Communication Sciences and Disorders, 1975 Willow Drive, Madison, WI 53706, USA; University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - A F Nisbet
- University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - J Russell
- University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - J Szot
- University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - T J Glass
- University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - N P Connor
- University of Wisconsin-Madison, Dept. of Communication Sciences and Disorders, 1975 Willow Drive, Madison, WI 53706, USA; University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - C A Kelm-Nelson
- University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA.
| | - M R Ciucci
- University of Wisconsin-Madison, Dept. of Communication Sciences and Disorders, 1975 Willow Drive, Madison, WI 53706, USA; University of Wisconsin-Madison, Dept. of Surgery, Div. of Otolaryngology, 1300 University Avenue, 483 Medical Sciences Building, Madison, WI 53706, USA; University of Wisconsin-Madison, Neuroscience Training Program, 9531 WIMR II, 1111 Highland Ave., Madison, WI 53705, USA.
| |
Collapse
|
5
|
Penna DBDS, Gumiéro Costa S, Dos Santos-Rodrigues A, Pandolfo P. The association of caffeine and nandrolone decanoate modulates aversive memory and nociception in rats. Brain Res 2024; 1837:148937. [PMID: 38615923 DOI: 10.1016/j.brainres.2024.148937] [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: 01/29/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Caffeine and anabolic-androgenic steroids (AAS) are commonly used to improve muscle mass and athletic performance. Nandrolone Decanoate (ND) is one of the most abused AAS worldwide, leading to behavioral changes in both humans and rodents. Caffeine, the most widely consumed psychostimulant globally, is present in various thermogenic and gym supplements. Low and moderate doses of caffeine antagonize adenosine receptors and have been linked to improved memory and pain relief. We have previously demonstrated that consuming caffeine prevents the risk-taking behavior triggered by nandrolone. In this study, we aimed to investigate the long-term effects of ND and caffeine, either alone or in combination, on passive avoidance memory and nociception. We used the step-down and hot-plate tasks in male and female Lister Hooded rats. Our results confirmed the antinociceptive effect of caffeine and indicated that chronic administration of the ND-caffeine association promotes the evocation of aversive memory in female rats.
Collapse
Affiliation(s)
- Daniel Bussinger de Souza Penna
- Institute of Biomedical Sciences, Program of Biomedical Sciences: Physiology and Pharmacology, Federal Fluminense University, Niteroi, Brazil; Institute of Biology, Program of Neurosciences, Federal Fluminense University, Niteroi, Brazil
| | - Samara Gumiéro Costa
- Institute of Biomedical Sciences, Program of Biomedical Sciences: Physiology and Pharmacology, Federal Fluminense University, Niteroi, Brazil
| | | | - Pablo Pandolfo
- Institute of Biomedical Sciences, Program of Biomedical Sciences: Physiology and Pharmacology, Federal Fluminense University, Niteroi, Brazil; Institute of Biology, Program of Neurosciences, Federal Fluminense University, Niteroi, Brazil.
| |
Collapse
|
6
|
Wee IC, Arulsamy A, Corrigan F, Collins-Praino L. Long-Term Impact of Diffuse Traumatic Brain Injury on Neuroinflammation and Catecholaminergic Signaling: Potential Relevance for Parkinson's Disease Risk. Molecules 2024; 29:1470. [PMID: 38611750 PMCID: PMC11013319 DOI: 10.3390/molecules29071470] [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: 01/05/2024] [Revised: 02/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with an increased risk of developing Parkinson's disease (PD), though the exact mechanisms remain unclear. TBI triggers acute neuroinflammation and catecholamine dysfunction post-injury, both implicated in PD pathophysiology. The long-term impact on these pathways following TBI, however, remains uncertain. In this study, male Sprague-Dawley rats underwent sham surgery or Marmarou's impact acceleration model to induce varying TBI severities: single mild TBI (mTBI), repetitive mild TBI (rmTBI), or moderate-severe TBI (msTBI). At 12 months post-injury, astrocyte reactivity (GFAP) and microglial levels (IBA1) were assessed in the striatum (STR), substantia nigra (SN), and prefrontal cortex (PFC) using immunohistochemistry. Key enzymes and receptors involved in catecholaminergic transmission were measured via Western blot within the same regions. Minimal changes in these markers were observed, regardless of initial injury severity. Following mTBI, elevated protein levels of dopamine D1 receptors (DRD1) were noted in the PFC, while msTBI resulted in increased alpha-2A adrenoceptors (ADRA2A) in the STR and decreased dopamine beta-hydroxylase (DβH) in the SN. Neuroinflammatory changes were subtle, with a reduced number of GFAP+ cells in the SN following msTBI. However, considering the potential for neurodegenerative outcomes to manifest decades after injury, longer post-injury intervals may be necessary to observe PD-relevant alterations within these systems.
Collapse
Affiliation(s)
- Ing Chee Wee
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia;
| | - Frances Corrigan
- Head Injury Lab, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lyndsey Collins-Praino
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
7
|
Moors TE, Li S, McCaffery TD, Ho GP, Bechade PA, Pham LN, Ericsson M, Nuber S. Increased palmitoylation improves estrogen receptor alpha-dependent hippocampal synaptic deficits in a mouse model of synucleinopathy. SCIENCE ADVANCES 2023; 9:eadj1454. [PMID: 37976363 PMCID: PMC10957154 DOI: 10.1126/sciadv.adj1454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Parkinson's disease (PD) is characterized by conversion of soluble α-synuclein (αS) into intraneuronal aggregates and degeneration of neurons and neuronal processes. Indications that women with early-stage PD display milder neurodegenerative features suggest that female sex partially protects against αS pathology. We previously reported that female sex and estradiol improved αS homeostasis and PD-like phenotypes in E46K-amplified (3K) αS mice. Here, we aimed to further dissect mechanisms that drive this sex dimorphism early in disease. We observed that synaptic abnormalities were delayed in females and improved by estradiol, mediated by local estrogen receptor alpha (ERα). Aberrant ERα distribution in 3K compared to wild-type mice was paired with its decreased palmitoylation. Treatment with ML348, a de-palmitoylation inhibitor, increased ERα availability and soluble αS homeostasis, ameliorating synaptic plasticity and cognitive and motor phenotypes. Our finding that sex differences in early-disease αS-induced synaptic impairment in 3KL mice are in part mediated by palmitoylated ERα may have functional and pathogenic implications for clinical PD.
Collapse
Affiliation(s)
- Tim E. Moors
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Thomas D. McCaffery
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Gary P. H. Ho
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Pascal A. Bechade
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Luu N. Pham
- Laboratory for Drug Discovery in Neurodegeneration, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Maria Ericsson
- Electron Microscopy Laboratory, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Silke Nuber
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Al-Zaid FS, Hurley MJ, Dexter DT, Gillies GE. Neuroprotective role for RORA in Parkinson's disease revealed by analysis of post-mortem brain and a dopaminergic cell line. NPJ Parkinsons Dis 2023; 9:119. [PMID: 37500636 PMCID: PMC10374904 DOI: 10.1038/s41531-023-00563-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
Parkinson's disease (PD) is almost twice as prevalent in men, which has largely been attributed to neuroprotective effect of oestradiol in women. RORA (retinoic acid receptor-related orphan receptor alpha) regulates the transcription of central aromatase, the enzyme responsible for local oestradiol synthesis, simultaneously, RORA expression is regulated by sex hormones. Moreover, RORA protects neurones against oxidative stress, a key mechanism contributing to the loss of dopaminergic neurones in PD. Therefore, we hypothesized that there would be sex differences in RORA expression in the substantia nigra pars compacta (SNpc), which could contribute to sex differences observed in PD prevalence and pathogenesis. In a case control study, qPCR and western blot analyses were used to quantify gene and protein expression in the SNpc of post-mortem brains (n = 14 late-stage PD and 11 age and sex matched controls). The neuroprotective properties of a RORA agonist were then investigated directly using a cell culture toxin-based model of PD coupled with measures of viability, mitochondrial function and apoptosis. RORA was expressed at significantly higher levels in the SNpc from control females' brains compared to males. In PD, we found a significant increase in SNpc RORA expression in male PD compared to female PD. Treatment with a RORA agonist showed a significant neuroprotection in our cell culture model of PD and revealed significant effects on intracellular factors involved in neuronal survival and demise. This study is the first to demonstrate a sex specific pattern of RORA protein and gene expression in the SNpc of controls post-mortem human brains, and to show that this is differentially altered in male and female PD subjects, thus supporting a role for RORA in sex-specific aspects of PD. Furthermore, our in vitro PD model indicates mechanisms whereby a RORA agonist exerts its neuroprotective effect, thereby highlighting the translational potential for RORA ligands in PD.
Collapse
Affiliation(s)
- Felwah S Al-Zaid
- Department of Physiology, College of Medicine, King Saud University, Riyadh, KSA, Saudi Arabia.
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK.
| | - Michael J Hurley
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London, NW3 2PF, UK
| | - David T Dexter
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
- Parkinson's UK, 215 Vauxhall Bridge Road, London, SW1V 1EJ, UK
| | - Glenda E Gillies
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
9
|
Abstract
The lower prevalence of Parkinson disease (PD) in females is not well understood but may be partially explained by sex differences in nigrostriatal circuitry and possible neuroprotective effects of estrogen. PD motor and nonmotor symptoms differ between sexes, and women experience disparities in care including undertreatment with DBS and less access to caregiving. Our knowledge about PD in gender diverse individuals is limited. Future studies should improve our understanding of the role of hormone replacement therapy in PD, address gender-based inequities in PD care and expand our understanding of PD in SGM and marginalized communities.
Collapse
|
10
|
Koszegi Z, Cheong RY. Targeting the non-classical estrogen pathway in neurodegenerative diseases and brain injury disorders. Front Endocrinol (Lausanne) 2022; 13:999236. [PMID: 36187099 PMCID: PMC9521328 DOI: 10.3389/fendo.2022.999236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Estrogens can alter the biology of various tissues and organs, including the brain, and thus play an essential role in modulating homeostasis. Despite its traditional role in reproduction, it is now accepted that estrogen and its analogues can exert neuroprotective effects. Several studies have shown the beneficial effects of estrogen in ameliorating and delaying the progression of neurodegenerative diseases, including Alzheimer's and Parkinson's disease and various forms of brain injury disorders. While the classical effects of estrogen through intracellular receptors are more established, the impact of the non-classical pathway through receptors located at the plasma membrane as well as the rapid stimulation of intracellular signaling cascades are still under active research. Moreover, it has been suggested that the non-classical estrogen pathway plays a crucial role in neuroprotection in various brain areas. In this mini-review, we will discuss the use of compounds targeting the non-classical estrogen pathway in their potential use as treatment in neurodegenerative diseases and brain injury disorders.
Collapse
Affiliation(s)
- Zsombor Koszegi
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Y. Cheong
- Timeline Bioresearch AB, Medicon Village, Lund, Sweden
- *Correspondence: Rachel Y. Cheong,
| |
Collapse
|
11
|
Krasko MN, Hoffmeister JD, Schaen-Heacock NE, Welsch JM, Kelm-Nelson CA, Ciucci MR. Rat Models of Vocal Deficits in Parkinson's Disease. Brain Sci 2021; 11:925. [PMID: 34356159 PMCID: PMC8303338 DOI: 10.3390/brainsci11070925] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a progressive, degenerative disorder that affects 10 million people worldwide. More than 90% of individuals with PD develop hypokinetic dysarthria, a motor speech disorder that impairs vocal communication and quality of life. Despite the prevalence of vocal deficits in this population, very little is known about the pathological mechanisms underlying this aspect of disease. As such, effective treatment options are limited. Rat models have provided unique insights into the disease-specific mechanisms of vocal deficits in PD. This review summarizes recent studies investigating vocal deficits in 6-hydroxydopamine (6-OHDA), alpha-synuclein overexpression, DJ1-/-, and Pink1-/- rat models of PD. Model-specific changes to rat ultrasonic vocalization (USV), and the effects of exercise and pharmacologic interventions on USV production in these models are discussed.
Collapse
Affiliation(s)
- Maryann N. Krasko
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA; (M.N.K.); (J.D.H.); (N.E.S.-H.); (J.M.W.); (C.A.K.-N.)
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jesse D. Hoffmeister
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA; (M.N.K.); (J.D.H.); (N.E.S.-H.); (J.M.W.); (C.A.K.-N.)
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nicole E. Schaen-Heacock
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA; (M.N.K.); (J.D.H.); (N.E.S.-H.); (J.M.W.); (C.A.K.-N.)
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jacob M. Welsch
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA; (M.N.K.); (J.D.H.); (N.E.S.-H.); (J.M.W.); (C.A.K.-N.)
| | - Cynthia A. Kelm-Nelson
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA; (M.N.K.); (J.D.H.); (N.E.S.-H.); (J.M.W.); (C.A.K.-N.)
| | - Michelle R. Ciucci
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA; (M.N.K.); (J.D.H.); (N.E.S.-H.); (J.M.W.); (C.A.K.-N.)
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI 53706, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
12
|
Akintunde J, Ibrahim L, Omotosho O, Boligon A. Metabolic depletion of synaptosomal enzymes linked with neurotoxicity and ovarian dysfunction by phenolic antioxidants of Croton zambsicus leaves in rats exposed to chronic mixture of anthropogenic toxicant. Metabol Open 2021; 10:100097. [PMID: 34159306 PMCID: PMC8193606 DOI: 10.1016/j.metop.2021.100097] [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: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/01/2022] Open
Abstract
A complex mixture of organic contaminants and metals is associated with neuron-fertility disorders and studies have demonstrated that phenolic antioxidants from herbal origin, possesses a strong protective potential. This study aimed to investigate the protection of phenolic croton zambesicus (C-ZAMB) leaves against neuro-ovarian damage in rats exposed to chronic mixture of anthropogenic toxicants (EOMABRSL). The animals were divided into five groups (n = 10): Group I was given 0.5 ml of distilled water only; Group II received 0.5 ml of EOMABRSL for 98 days; Group III received 0.5 ml of EOMABRSL for 70 days and withdrew for 28 days; Group IV received 0.5 ml of EOMABRSL for 70 days +400 mg/kg phenolic C-ZAMB for 28 days; Group V received 400 mg/kg C-ZAMB only for 28 days via oral route. Both non-withdrawal and withdrawal EOMABRSL-exposed animals exhibited neuro-ovarian impairment by up-regulating neuronal 51 eco-nucleotidase (51ENT), acetylcholinesterase (AChE), butrylcholinesterase (BuChE), synaptosomal monoamine oxidase-A (MAO-A) with altered cerebral antioxidants. Similarly, exposure to EOMABRSL for 98 and 70 days caused ovarian injury by amplifying the activity of 51ENT with corresponding decline of fertility index, lactate dehydrogenase (LDH) and Δ5 17β-hydroxyl steroid dehydrogenase (Δ517β-HSD). EOMABRSL intoxication also increased the neuro-ovarian MDA content with reduced numbers of neonates. Phenolic antioxidants from C-ZAMB leaves identified by High Pressure Liquid Chromatography (HPLC) ameliorated the chronic EOMABRSL intoxication. The treatment also prevented ovarian lesions by depleting MDA content and improved antioxidant status. Thus, confirming its neuro-ovarian protection.
Collapse
Affiliation(s)
- J.K. Akintunde
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Pure and Applied Sciences, Kwara State University, Malete, P.M.B 1530, Nigeria
| | - L.B. Ibrahim
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Pure and Applied Sciences, Kwara State University, Malete, P.M.B 1530, Nigeria
| | - O.D. Omotosho
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Pure and Applied Sciences, Kwara State University, Malete, P.M.B 1530, Nigeria
| | - A.A. Boligon
- Phytochemical Research Laboratory, Department of Industrial Pharmacy, Federal University of Santa Maria, Building 26, Room 1115, Santa Maria, CEP97105-900, Brazil
| |
Collapse
|
13
|
A probabilistic atlas of the human ventral tegmental area (VTA) based on 7 Tesla MRI data. Brain Struct Funct 2021; 226:1155-1167. [PMID: 33580320 PMCID: PMC8036186 DOI: 10.1007/s00429-021-02231-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
Functional magnetic resonance imaging (fMRI) BOLD signal is commonly localized by using neuroanatomical atlases, which can also serve for region of interest analyses. Yet, the available MRI atlases have serious limitations when it comes to imaging subcortical structures: only 7% of the 455 subcortical nuclei are captured by current atlases. This highlights the general difficulty in mapping smaller nuclei deep in the brain, which can be addressed using ultra-high field 7 Tesla (T) MRI. The ventral tegmental area (VTA) is a subcortical structure that plays a pivotal role in reward processing, learning and memory. Despite the significant interest in this nucleus in cognitive neuroscience, there are currently no available, anatomically precise VTA atlases derived from 7 T MRI data that cover the full region of the VTA. Here, we first provide a protocol for multimodal VTA imaging and delineation. We then provide a data description of a probabilistic VTA atlas based on in vivo 7 T MRI data.
Collapse
|
14
|
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: 44] [Impact Index Per Article: 11.0] [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
|
15
|
Yokoi K, Hattori M, Satake Y, Tanaka Y, Sato M, Hashizume A, Hori A, Kawashima M, Hirakawa A, Watanabe H, Katsuno M. Longitudinal analysis of premotor anthropometric and serological markers of Parkinson's disease. Sci Rep 2020; 10:20524. [PMID: 33239649 PMCID: PMC7688961 DOI: 10.1038/s41598-020-77415-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 11/10/2020] [Indexed: 12/02/2022] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder in which nonmotor symptoms, such as constipation and hyposmia, precede the onset of motor symptoms by 20 years. The aim of this study was to identify biomarkers at the premotor stage of PD. We assessed the differences in longitudinal changes in anthropometric and serological indices obtained from health check-up data before and after the onset of motor symptoms between male and female PD patients and healthy subjects. We enrolled 22 male and 23 female PD patients and 60 male and 60 female healthy controls. A mixed-effects model was used to estimate the trajectory of each clinical marker over the years before and after motor symptoms onset in the PD subjects, which were then compared with the trajectories of the healthy controls. The results showed a premotor blood pressure increase in female PD patients and premotor decreases in haematocrit, total cholesterol and low-density lipoprotein cholesterol in the male patients. Our results indicated that blood pressure, haematocrit and serum cholesterol levels are potential premotor markers of PD. Additionally, the changes in anthropometric and serological indices before PD motor symptoms onset were sex specific.
Collapse
Affiliation(s)
- Katsunori Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Makoto Hattori
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yuki Satake
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yasuhiro Tanaka
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Maki Sato
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Atsushi Hashizume
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Akihiro Hori
- Kumiai Kosei Hospital, Takayama, Gifu, 5068502, Japan
| | | | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 1138510, Japan
| | - Hirohisa Watanabe
- Brain & Mind Research Centre, Nagoya University Graduate School of Medicine, Nagoya, 4668560, Japan
- Department of Neurology, Fujita Medical University, Toyoake, Aichi, 4701192, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
16
|
Conner MR, Jang D, Anderson BJ, Kritzer MF. Biological Sex and Sex Hormone Impacts on Deficits in Episodic-Like Memory in a Rat Model of Early, Pre-motor Stages of Parkinson's Disease. Front Neurol 2020; 11:942. [PMID: 33041964 PMCID: PMC7527538 DOI: 10.3389/fneur.2020.00942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/21/2020] [Indexed: 01/30/2023] Open
Abstract
Episodic memory deficits are among the earliest appearing and most commonly occurring examples of cognitive impairment in Parkinson's disease (PD). These enduring features can also predict a clinical course of rapid motor decline, significant cognitive deterioration, and the development of PD-related dementia. The lack of effective means to treat these deficits underscores the need to better understand their neurobiological bases. The prominent sex differences that characterize episodic memory in health, aging and in schizophrenia and Alzheimer's disease suggest that neuroendocrine factors may also influence episodic memory dysfunction in PD. However, while sex differences have been well-documented for many facets of PD, sex differences in, and sex hormone influences on associated episodic memory impairments have been less extensively studied and have never been examined in preclinical PD models. Accordingly, we paired bilateral neostriatal 6-hydroxydopamine (6-OHDA) lesions with behavioral testing using the What-Where-When Episodic-Like Memory (ELM) Task in adult rats to first determine whether episodic-like memory is impaired in this model. We further compared outcomes in gonadally intact female and male subjects, and in male rats that had undergone gonadectomy—with and without hormone replacement, to determine whether biological sex and/or sex hormones influenced the expression of dopamine lesioned-induced memory deficits. These studies showed that 6-OHDA lesions profoundly impaired recall for all memory domains in male and female rats. They also showed that in males, circulating gonadal hormones powerfully modulated the negative impacts of 6-OHDA lesions on What, Where, and When discriminations in domain-specific ways. Specifically, the absence of androgens was shown to fully attenuate 6-OHDA lesion-induced deficits in ELM for “Where” and to partially protect against lesion-induced deficits in ELM for “What.” In sum, these findings show that 6-OHDA lesions in rats recapitulate the vulnerability of episodic memory seen in early PD. Together with similar evidence recently obtained for spatial working memory, the present findings also showed that diminished androgen levels provide powerful, highly selective protections against the harmful effects that 6-OHDA lesions have on memory functions in male rats.
Collapse
Affiliation(s)
- Meagan R Conner
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, United States.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
| | - Doyeon Jang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
| | - Brenda J Anderson
- Department of Psychology, Stony Brook University, Stony Brook, NY, United States
| | - Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
17
|
Gamache J, Yun Y, Chiba-Falek O. Sex-dependent effect of APOE on Alzheimer's disease and other age-related neurodegenerative disorders. Dis Model Mech 2020; 13:dmm045211. [PMID: 32859588 PMCID: PMC7473656 DOI: 10.1242/dmm.045211] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The importance of apolipoprotein E (APOE) in late-onset Alzheimer's disease (LOAD) has been firmly established, but the mechanisms through which it exerts its pathogenic effects remain elusive. In addition, the sex-dependent effects of APOE on LOAD risk and endophenotypes have yet to be explained. In this Review, we revisit the different aspects of APOE involvement in neurodegeneration and neurological diseases, with particular attention to sex differences in the contribution of APOE to LOAD susceptibility. We discuss the role of APOE in a broader range of age-related neurodegenerative diseases, and summarize the biological factors linking APOE to sex hormones, drawing on supportive findings from rodent models to identify major mechanistic themes underlying the exacerbation of LOAD-associated neurodegeneration and pathology in the female brain. Additionally, we list sex-by-genotype interactions identified across neurodegenerative diseases, proposing APOE variants as a shared etiology for sex differences in the manifestation of these diseases. Finally, we present recent advancements in 'omics' technologies, which provide a new platform for more in-depth investigations of how dysregulation of this gene affects the development and progression of neurodegenerative diseases. Collectively, the evidence summarized in this Review highlights the interplay between APOE and sex as a key factor in the etiology of LOAD and other age-related neurodegenerative diseases. We emphasize the importance of careful examination of sex as a contributing factor in studying the underpinning genetics of neurodegenerative diseases in general, but particularly for LOAD.
Collapse
Affiliation(s)
- Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Young Yun
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
18
|
De Miranda BR, Fazzari M, Rocha EM, Castro S, Greenamyre JT. Sex Differences in Rotenone Sensitivity Reflect the Male-to-Female Ratio in Human Parkinson's Disease Incidence. Toxicol Sci 2020; 170:133-143. [PMID: 30907971 DOI: 10.1093/toxsci/kfz082] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is a critical need to include female subjects in disease research; however, in Parkinson's disease, where the male-to-female incidence is about 1.5-to-1, the majority of preclinical research is conducted in male animals. The mitochondrial complex I inhibitor, rotenone, is selectively toxic to dopaminergic neurons, and reproduces several neuropathological features of Parkinson's disease, including α-synuclein pathology. Rotenone has been primarily utilized in male Lewis rats; however, pilot studies in age-matched female Lewis rats revealed that our usual dose (2.8 mg/kg/day intraperitoneal [i.p.]) did not cause dopaminergic neurodegeneration. Therefore, we compared rotenone-treated males (2.8 mg/kg/day, i.p.) to females at increasing doses (2.8 mg/kg/day, 3.2 mg/kg/day, 3.6 mg/kg/day, and 1.6 mg/kg bis in die, i.p.). Female rats receiving 3.2 mg/kg, and 3.6 mg/kg rotenone displayed significant loss of dopaminergic neurons in the substantia nigra as assessed by stereology, which was accompanied by a loss of striatal dopaminergic terminals. Even at these higher doses, however, females showed less inflammation, and less accumulation of α-synuclein and transferrin, possibly as a result of preserved autophagy. Thus, the bias toward increased male incidence of human Parkinson's disease is reflected in the rotenone model. Whether such sex differences will translate into differences in responses to mechanism-driven therapeutic interventions remains to be determined.
Collapse
Affiliation(s)
- Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Marco Fazzari
- Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, 15261.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261.,Fondazione Ri.MED, Via Bandiera 11, Palermo 90133, Italy
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Sandra Castro
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| |
Collapse
|
19
|
Forns J, Verner MA, Iszatt N, Nowack N, Bach CC, Vrijheid M, Costa O, Andiarena A, Sovcikova E, Høyer BB, Wittsiepe J, Lopez-Espinosa MJ, Ibarluzea J, Hertz-Picciotto I, Toft G, Stigum H, Guxens M, Liew Z, Eggesbø M. Early Life Exposure to Perfluoroalkyl Substances (PFAS) and ADHD: A Meta-Analysis of Nine European Population-Based Studies. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:57002. [PMID: 32378965 PMCID: PMC7263458 DOI: 10.1289/ehp5444] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
INTRODUCTION To date, the evidence for an association between perfluoroalkyl substances (PFAS) exposure and attention deficit and hyperactivity disorder (ADHD) is inconclusive. OBJECTIVE We investigated the association between early life exposure to perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA), and ADHD in a collaborative study including nine European population-based studies, encompassing 4,826 mother-child pairs. METHODS Concentrations of PFOS and PFOA were measured in maternal serum/plasma during pregnancy, or in breast milk, with different timing of sample collection in each cohort. We used a validated pharmacokinetic model of pregnancy and lactation to estimate concentrations of PFOS and PFOA in children at birth and at 3, 6, 12, and 24 months of age. We classified ADHD using recommended cutoff points for each instrument used to derive symptoms scores. We used multiple imputation for missing covariates, logistic regression to model the association between PFAS exposure and ADHD in each study, and combined all adjusted study-specific effect estimates using random-effects meta-analysis. RESULTS A total of 399 children were classified as having ADHD, with a prevalence ranging from 2.3% to 7.3% in the studies. Early life exposure to PFOS or PFOA was not associated with ADHD during childhood [odds ratios (ORs) ranging from 0.96 (95% CI: 0.87, 1.06) to 1.02 (95% CI: 0.93, 1.11)]. Results from stratified models suggest potential differential effects of PFAS related to child sex and maternal education. CONCLUSION We did not identify an increased prevalence of ADHD in association with early life exposure to PFOS and PFOA. However, stratified analyses suggest that there may be an increased prevalence of ADHD in association with PFAS exposure in girls, in children from nulliparous women, and in children from low-educated mothers, all of which warrant further exploration. https://doi.org/10.1289/EHP5444.
Collapse
Affiliation(s)
- Joan Forns
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Marc-Andre Verner
- Department of Occupational and Environmental Health, School of Public Health, Université de Montréal, Montreal, Canada
- Université de Montréal Public Health Research Institute (IRSPUM), Université de Montréal, Montreal, Canada
| | - Nina Iszatt
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Nikola Nowack
- Department of Developmental Psychology, Ruhr-University Bochum, Bochum, Germany
| | - Cathrine Carlsen Bach
- Department of Pediatrics and Adolescent Medicine, Viborg Regional Hospital, Viborg, Denmark
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Martine Vrijheid
- Global Health Institute Barcelona (ISGlobal), Institute for Global Health, Barcelona, Spain
- Univeristat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Olga Costa
- Epidemiology and Environmental Health Joint Research Unit, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO)-Universitat Jaume I-Universitat de València, Valencia, Spain
| | - Ainara Andiarena
- Health Research Institute BIODONOSTIA, Donostia-San Sebastian, Basque Country, Spain
- Faculty of Psychology, University of the Basque Country (UPV/EHU), San Sebastian, Basque Country, Spain
| | - Eva Sovcikova
- Department of Environmental Medicine, Faculty of Public Health, Slovak Medical University, Bratislava, Slovak Republic
| | - Birgit Bjerre Høyer
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Jürgen Wittsiepe
- Department of Hygiene, Social and Environmental Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Maria-Jose Lopez-Espinosa
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Epidemiology and Environmental Health Joint Research Unit, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO)-Universitat Jaume I-Universitat de València, Valencia, Spain
- Department of Nursing and Chiropody, Universitat de València, Valencia, Spain
| | - Jesus Ibarluzea
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Health Research Institute BIODONOSTIA, Donostia-San Sebastian, Basque Country, Spain
- Faculty of Psychology, University of the Basque Country (UPV/EHU), San Sebastian, Basque Country, Spain
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, School of Medicine, University of California, Davis (UC Davis), Davis, California, USA
- UC Davis Medical Investigations of Neurodevelopmental Disorders Institute, UC Davis, Davis, California, USA
| | - Gunnar Toft
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Hein Stigum
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Mònica Guxens
- Global Health Institute Barcelona (ISGlobal), Institute for Global Health, Barcelona, Spain
- Univeristat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Centre, Sophia Children's Hospital, Rotterdam, Netherlands
| | - Zeyan Liew
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Merete Eggesbø
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
20
|
Lichter D, Finnegan S. Influence of Gender on Tourette Syndrome Beyond Adolescence. Eur Psychiatry 2020; 30:334-40. [DOI: 10.1016/j.eurpsy.2014.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/02/2014] [Accepted: 07/09/2014] [Indexed: 12/17/2022] Open
Abstract
AbstractAlthough boys are disproportionately affected by tics in Tourette syndrome (TS), this gender bias is attenuated in adulthood and a recent study has suggested that women may experience greater functional interference from tics than men. The authors assessed the gender distribution of adults in a tertiary University-based TS clinic population and the relative influence of gender and other variables on adult tic severity (YGTSS score) and psychosocial functioning (GAF score). We also determined retrospectively the influence of gender on change in global tic severity and overall TS impairment (YGTSS) since adolescence. Females were over-represented in relation to previously published epidemiologic surveys of both TS children and adults. Female gender was associated with a greater likelihood of tic worsening as opposed to tic improvement in adulthood; a greater likelihood of expansion as opposed to contraction of motor tic distribution; and with increased current motor tic severity and tic-related impairment. However, gender explained only a small percentage of the variance of the YGTSS global severity score and none of the variance of the GAF scale score. Psychosocial functioning was influenced most strongly by tic severity but also by a variety of comorbid neuropsychiatric disorders.
Collapse
|
21
|
Marquis JM, Lettenberger SE, Kelm-Nelson CA. Early-onset Parkinsonian behaviors in female Pink1-/- rats. Behav Brain Res 2020; 377:112175. [PMID: 31542395 PMCID: PMC6824965 DOI: 10.1016/j.bbr.2019.112175] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/01/2019] [Accepted: 08/22/2019] [Indexed: 12/26/2022]
Abstract
Parkinson disease (PD) is a progressive, neurological disease that affects millions of individuals worldwide. Although instability, rigidity, tremor, and bradykinesia are considered hallmark motor signs of the disease, these are not apparent until mid-to-late stage. In addition to limb motor impairment, individuals with PD also exhibit early-onset speech dysfunction and reduced vocal intelligibility as well as anhedonia and anxiety. Many of these clinical signs vary according to sex in humans with PD. In this study, a translational genetic rat model of early-onset PD (Pink1-/-) was used to address significant gaps in knowledge concerning sex-specific characteristics of limb sensorimotor deficits, vocal motor dysfunction, and changes in affective state. Traditional behavioral tests of limb function, ultrasonic vocalization, anxiety, and anhedonia in the Pink1-/- female rat and wildtype controls were used to test the hypothesis that behavioral performance would significantly differ between genotypes, and that these differences would increase with disease progression (age of the rat). Results demonstrate that Pink1-/- female rats do not exhibit limb sensorimotor deficits but do have significantly reduced intensity (loudness) of vocalizations, and present with anhedonia and anxiety by 8 months of age. Consistent with an early-disease model, Pink1-/- female rats do not exhibit significant decreases in nigrostriatal catecholamines/metabolites, as measured by HPLC. These results are significant in expanding knowledge of early-onset deficits in the female Pink1-/- genetic rat model of PD.
Collapse
Affiliation(s)
- Julia M Marquis
- Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA.
| | - Samantha E Lettenberger
- Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA.
| | - Cynthia A Kelm-Nelson
- Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
22
|
Mulak A. An overview of the neuroendocrine system in Parkinson's disease: what is the impact on diagnosis and treatment? Expert Rev Neurother 2019; 20:127-135. [PMID: 31829756 DOI: 10.1080/14737175.2020.1701437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: A growing body of evidence indicates that neuroendocrine interactions may occur at all levels of the brain-gut-microbiota axis, which is directly involved in the pathogenesis of Parkinson's disease (PD).Areas covered: The review presents some current and emerging concepts regarding the organization and functioning of the neuroendocrine system as well as the role of neuroendocrine disturbances in the pathophysiology and symptomatology of PD. The concept of the brain-gut-microbiota triad interactions in the neuroendocrine system and PD is proposed. In PD, dysregulation of the main neuroendocrine axes coordinated by the hypothalamus is accompanied by disruptions at the peripheral level, which involve enteroendocrine cells producing numerous neuropeptides. Moreover, the important role of the gut microbiota as a main coordinator of immune and neuroendocrine interactions is discussed. The potential diagnostic and therapeutic implications in the context of the recent developments in the fields of neuroendocrinology and neurodegeneration are also presented.Expert opinion: Unraveling complex neuroendocrine interactions in the course of PD may provide crucial diagnostic implications and novel therapeutic approaches including the application of gut neuropeptides and gut microbiota modification.
Collapse
Affiliation(s)
- Agata Mulak
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
23
|
Chen YH, Wang V, Huang EYK, Chou YC, Kuo TT, Olson L, Hoffer BJ. Delayed Dopamine Dysfunction and Motor Deficits in Female Parkinson Model Mice. Int J Mol Sci 2019; 20:ijms20246251. [PMID: 31835787 PMCID: PMC6940785 DOI: 10.3390/ijms20246251] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/22/2019] [Accepted: 12/04/2019] [Indexed: 12/21/2022] Open
Abstract
This study analyzed gender differences in the progressive dopamine (DA) deficiency phenotype in the MitoPark (MP) mouse model of Parkinson’s disease (PD) with progressive loss of DA release and reuptake in midbrain DA pathways. We found that the progressive loss of these DA presynaptic parameters begins significantly earlier in male than female MP mice. This was correlated with behavioral gender differences of both forced and spontaneous motor behavior. The degeneration of the nigrostriatal DA system in MP mice is earlier and more marked than that of the mesolimbic DA system, with male MP mice again being more strongly affected than female MP mice. After ovariectomy, DA presynaptic and behavioral changes in female mice become very similar to those of male animals. Our results suggest that estrogen, either directly or indirectly, is neuroprotective in the midbrain DA system. Our results are compatible with epidemiological data on incidence and symptom progression in PD, showing that men are more strongly affected than women at early ages.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: (Y.-H.C.); (B.J.H.)
| | - Vicki Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yu-Ching Chou
- School of Public Health, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Tung-Tai Kuo
- Graduate Institute of Computer and Communication Engineering, National Taipei University of Technology, Taipei 10608, Taiwan;
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Correspondence: (Y.-H.C.); (B.J.H.)
| |
Collapse
|
24
|
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: 78] [Impact Index Per Article: 13.0] [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
|
25
|
Female Sex and Brain-Selective Estrogen Benefit α-Synuclein Tetramerization and the PD-like Motor Syndrome in 3K Transgenic Mice. J Neurosci 2019; 39:7628-7640. [PMID: 31405930 DOI: 10.1523/jneurosci.0313-19.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
Many studies report a higher risk for Parkinson's disease (PD) and younger age of onset in men. This, and the fact that the neuropathological process underlying PD symptoms may begin before menopause, suggests that estrogen-based hormone therapy could modify this higher risk in males. However, the effects of female sex or estrogen on α-synuclein (αS) homeostasis and related PD neuropathology remain unknown. Here, we used an αS tetramer-abrogating mouse model of PD (3K) that amplifies the familial E46K PD mutation to investigate the effects of female sex and brain-selective estrogen treatment on αS tetramerization and solubility, formation of vesicle-rich αS+ aggregates, dopaminergic and cortical fiber integrity, and associated motor deficits. In male 3K mice, the motor phenotype became apparent at ∼10 weeks and increased to age 6 months, paralleled by PD-like neuropathology, whereas 3K females showed a significant delay in onset. At 6 months, this beneficial phenotypic effect in 3K females was associated with a higher αS tetramer-to-monomer ratio and less decrease in dopaminergic and cortical fiber length and quantity. Brain-selective estrogen treatment in symptomatic 3K mice significantly increased the tetramer-to-monomer ratio, turnover by autophagy of aggregate-prone monomers, and neurite complexity of surviving DAergic and cortical neurons, in parallel with benefits in motor performance. Our findings support an upstream role for αS tetramer loss in PD phenotypes and a role for estrogen in mitigating PD-like neuropathology in vivo Brain-selective estrogen therapy may be useful in delaying or reducing PD symptoms in men and postmenopausal women.SIGNIFICANCE STATEMENT The mechanisms responsible for the male-to-female preponderance in Parkinson's disease (PD) are not well understood yet important for treatment efficacy. We previously showed that abrogating native α-synuclein (αS) tetramers produces a close PD model, including dopaminergic and cortical fiber loss and a progressive motor disorder responsive to l-DOPA. Here, we analyzed sex and use 10b-17β-dihydroxyestra-1,4-dien-3-one treatment of symptomatic 3K males, and demonstrate that the beneficial effects of female sex on PD-like neuropathology can be reinstated by elevating estrogen in the male brain. The study provides evidence that 17β-estradiol restores the tetramer-to-monomer ratio by autophagy turnover of excess αS monomers, vesicle and fiber integrity in brain regions critically involved in motor behavior. These data provide the basis for understanding sex differences in αS homeostasis and the development of therapeutic approaches to treating men and postmenopausal women with PD.
Collapse
|
26
|
Nwabuobi L, Barbosa W, Sweeney M, Oyler S, Meisel T, Di Rocco A, Chodosh J, Fleisher JE. Sex-related differences in homebound advanced Parkinson's disease patients. Clin Interv Aging 2019; 14:1371-1377. [PMID: 31534322 PMCID: PMC6681424 DOI: 10.2147/cia.s203690] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/22/2019] [Indexed: 11/25/2022] Open
Abstract
Background Women with Parkinson’s disease (PD) are more likely to be older, have greater disease severity and comorbidities, and yet are less likely to receive care from a neurologist, as compared with men with PD. Within the PD population, homebound individuals are a particularly vulnerable group facing significant barriers to care, yet within this understudied population, sex-related differences have not been reported. Purpose To identify and describe differences in homebound men and women with advanced PD and related disorders, participating in an interdisciplinary home visit program. Patients and methods This was an exploratory analysis of homebound patients seen between February 2014 and July 2016 using data collected via in-person interviews and chart review. Results We enrolled 85 patients, of whom 52% were women. PD was the most common diagnosis (79%), followed by dementia with Lewy bodies (5%), and other atypical parkinsonism (16%). Men were more likely to have a PD dementia diagnosis than women (17.1% vs 2.3%, p=0.03). Women were more likely to live alone (18.1% of women had no caregiver vs 2.4% of men, p=0.05). Conclusion The role of the caregiver in facilitating safe aging-in-place is crucial. Among homebound individuals with advanced PD, women were far more likely to live alone. The absence of a spouse or care partner may be due in part to variable sex-based life expectancies. Our findings suggest that homebound women with advanced PD may face greater barriers to accessing support.
Collapse
Affiliation(s)
- Lynda Nwabuobi
- Department of Neurology, Movement Disorders Division, Columbia University Medical Center, New York, NY, USA
| | | | - Meghan Sweeney
- Department of Palliative Care, Kaiser Permanente, Lafayette, CO, USA
| | - Sarah Oyler
- Intermountain Neurosciences Institute, Intermountain Medical Center, Murray, UT, USA
| | - Talia Meisel
- State University of New York Downstate Medical Center College of Medicine, Brooklyn, NY, USA
| | - Alessandro Di Rocco
- Department of Neurology, Movement Disorders Division, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Long Island, NY, USA
| | - Joshua Chodosh
- Division of Geriatric Medicine and Palliative Care, New York University Langone Health, New York, NY, USA.,Veterans Affairs NY Harbor Healthcare System, NY, New York, USA
| | - Jori E Fleisher
- Section of Movement Disorders, Department of Neurological Sciences, Rush Medical College, Rush University, Chicago, IL, USA
| |
Collapse
|
27
|
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: 4.8] [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
|
28
|
Loiola RA, Wickstead ES, Solito E, McArthur S. Estrogen Promotes Pro-resolving Microglial Behavior and Phagocytic Cell Clearance Through the Actions of Annexin A1. Front Endocrinol (Lausanne) 2019; 10:420. [PMID: 31297095 PMCID: PMC6607409 DOI: 10.3389/fendo.2019.00420] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Local production of estrogen rapidly follows brain tissue injury, but the role this hormone plays in regulating the response to neural damage or in the modulation of mediators regulating inflammation is in many ways unclear. Using the murine BV2 microglia model as well as primary microglia from wild-type and annexin A1 (AnxA1) null mice, we have identified two related mechanisms whereby estradiol can modulate microglial behavior in a receptor specific fashion. Firstly, estradiol, via estrogen receptor β (ERβ), enhanced the phagocytic clearance of apoptotic cells, acting through increased production and release of the protein AnxA1. Secondly, stimulation of either ERβ or the G protein coupled estrogen receptor GPER promoted the adoption of an anti-inflammatory/pro-resolving phenotype, an action similarly mediated through AnxA1. Together, these data suggest the hypothesis that locally produced estrogen acts through AnxA1 to exert powerful pro-resolving actions, controlling and limiting brain inflammation and ultimately protecting this highly vulnerable organ. Given the high degree of receptor selectivity in evoking these responses, we suggest that the use of selective estrogen receptor ligands may hold therapeutic promise in the treatment of neuroinflammation, avoiding unwanted generalized effects.
Collapse
Affiliation(s)
- Rodrigo Azevedo Loiola
- John Vane Science Centre, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Laboratoire de la Barrière Hémato-Encéphalique, Faculty Jean Perrin, EA 2465, Université d'Artois, Arras, France
| | - Edward S. Wickstead
- School of Life Sciences, College of Liberal Arts & Sciences, University of Westminster, London, United Kingdom
- Barts and The London School of Medicine and Dentistry, Institute of Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Egle Solito
- John Vane Science Centre, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universitá degli Studi di Napoli Federico II, Naples, Italy
| | - Simon McArthur
- Barts and The London School of Medicine and Dentistry, Institute of Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
- *Correspondence: Simon McArthur
| |
Collapse
|
29
|
Precision medicine and drug development in Alzheimer's disease: the importance of sexual dimorphism and patient stratification. Front Neuroendocrinol 2018; 50:31-51. [PMID: 29902481 DOI: 10.1016/j.yfrne.2018.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (ND) are among the leading causes of disability and mortality. Considerable sex differences exist in the occurrence of the various manifestations leading to cognitive decline. Alzheimer's disease (AD) exhibits substantial sexual dimorphisms and disproportionately affects women. Women have a higher life expectancy compared to men and, consequently, have more lifespan to develop AD. The emerging precision medicine and pharmacology concepts - taking into account the individual genetic and biological variability relevant for disease risk, prevention, detection, diagnosis, and treatment - are expected to substantially enhance our knowledge and management of AD. Stratifying the affected individuals by sex and gender is an important basic step towards personalization of scientific research, drug development, and care. We hypothesize that sex and gender differences, extending from genetic to psychosocial domains, are highly relevant for the understanding of AD pathophysiology, and for the conceptualization of basic/translational research and for clinical therapy trial design.
Collapse
|
30
|
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: 113] [Impact Index Per Article: 16.1] [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
|
31
|
Relation among Aromatase P450 and Tumoral Growth in Human Prolactinomas. Int J Mol Sci 2017; 18:ijms18112299. [PMID: 29104246 PMCID: PMC5713269 DOI: 10.3390/ijms18112299] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 02/01/2023] Open
Abstract
The pituitary gland is part of hypothalamic-pituitary–gonadal axis, which controls development, reproduction, and aging in humans and animals. In addition, the pituitary gland is regulated mainly by hormones and neurotransmitters released from the hypothalamus and by systemic hormones secreted by target glands. Aromatase P450, the enzyme responsible for the catabolization of aromatizable androgens to estrogens, is expressed in different parts of body, including the pituitary gland. Moreover, aromatase P450 is involved in sexual dimorphism where alteration in the level of aromatase can initiate a number of diseases in both genders. On the other hand, the direct actions of estrogens, mainly estradiol, are well known for stimulating prolactin release. Numerous studies have shown that changes in the levels of estrogens, among other factors, have been implicated in the genesis and development of prolactinoma. The pituitary gland can produce estradiol locally in several types of endocrine cells, and it is possible that aromatase could be responsible for the maintenance of the population of lactotroph cells and the modulation of the action of central or peripheral regulators. Aromatase overexpression due to inappropriate gene regulation has clinical effects such as the pathogenesis of prolactinomas. The present study reports on the synthesis of pituitary aromatase, its regulation by gonadal steroids, and the physiological roles of aromatase on pituitary endocrine cells. The involvement of aromatase in the pathogenesis of pituitary tumors, mainly prolactinomas, through the auto-paracrine production of estradiol is reviewed.
Collapse
|
32
|
Preclinical models in the study of sex differences. Clin Sci (Lond) 2017; 131:449-469. [PMID: 28265036 DOI: 10.1042/cs20160847] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/13/2016] [Accepted: 01/03/2017] [Indexed: 02/06/2023]
Abstract
The biology of sex differences deals with the study of the disparities between females and males and the related biological mechanisms. Gender medicine focuses on the impact of gender and sex on human physiology, pathophysiology and clinical features of diseases that are common to women and men. The term gender refers to a complex interrelation and integration of sex-as a biological and functional determinant-and psychological and cultural behaviours (due to ethnical, social or religious background). The attention to the impact of gender differences on the pathophysiology and, therefore, on the clinical management of the most common diseases, such as cardiovascular diseases (CVD), neurodegenerative disorders, immune and autoimmune diseases as well as several tumours, is in fact often neglected. Hence, studies covering different fields of investigation and including sex differences in the pathogenesis, in diagnostic and prognostic criteria as well as in response to therapy appear mandatory. However, prerequisites for this development are preclinical studies, including in vitro and in vivo approaches. They represent the first step in the development of a drug or in the comprehension of the pathogenetic mechanisms of diseases, in turn a necessary step for the development of new or more appropriate therapeutic strategies. However, sex differences are still poorly considered and the great majority of preclinical studies do not take into account the relevance of such disparities. In this review, we describe the state of the art of these studies and provide some paradigmatic examples of key fields of investigation, such as oncology, neurology and CVD, where preclinical models should be improved.
Collapse
|
33
|
Siani F, Greco R, Levandis G, Ghezzi C, Daviddi F, Demartini C, Vegeto E, Fuzzati-Armentero MT, Blandini F. Influence of Estrogen Modulation on Glia Activation in a Murine Model of Parkinson's Disease. Front Neurosci 2017; 11:306. [PMID: 28620274 PMCID: PMC5449471 DOI: 10.3389/fnins.2017.00306] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022] Open
Abstract
Epidemiological data suggest a sexual dimorphism in Parkinson disease (PD), with women showing lower risk of developing PD. Vulnerability of the nigrostriatal pathway may be influenced by exposure to estrogenic stimulation throughout fertile life. To further address this issue, we analyzed the progression of nigrostriatal damage, microglia and astrocyte activation and microglia polarization triggered by intrastriatal injection of dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) in male, female and ovariectomized (OVX) mice, as well as in OVX mice supplemented with 17βestradiol (OVX+E). Animals were sacrificed at different time points following 6-OHDA injection and brain sections containing striatum and substantia nigra pars compacta (SNc) underwent immunohistochemistry for tyrosine hydroxylase (TH) (dopaminergic marker), immunofluorescence for IBA1 and GFAP (markers of microglia and astrocyte activation, respectively) and triple immunoflorescent to identify polarization of microglia toward the cytotoxic M1 (DAPI/IBA1/TNFα) or cytoprotective M2 (DAPI/IBA1/CD206) phenotype. SNc damage induced by 6-OHDA was significantly higher in OVX mice, as compared to all other experimental groups, at 7 and 14 days after surgery. Astrocyte activation was higher in OVX mice with respect the other experimental groups, at all time points. Microglial activation in the SNc was detected at earlier time points in male, female and OVX+E, while in OVX mice was detected at all time-points. Microglia polarization toward the M2, but not the M1, phenotype was detected in female and OVX+E mice, while the M1 phenotype was observed only in male and OVX mice. Our results support the protective effects of estrogens against nigrostriatal degeneration, suggesting that such effects may be mediated by an interaction with microglia, which tend to polarize preferentially toward an M2, cytoprotective phenotype in the presence of intense estrogenic stimulation.
Collapse
Affiliation(s)
- Francesca Siani
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological InstitutePavia, Italy
| | - Rosaria Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Center, C. Mondino National Neurological InstitutePavia, Italy
| | - Giovanna Levandis
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological InstitutePavia, Italy
| | - Cristina Ghezzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological InstitutePavia, Italy
| | - Francesca Daviddi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological InstitutePavia, Italy
| | - Chiara Demartini
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Center, C. Mondino National Neurological InstitutePavia, Italy
| | - Elisabetta Vegeto
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of MilanMilan, Italy
| | - Marie-Thérèse Fuzzati-Armentero
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological InstitutePavia, Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological InstitutePavia, Italy
| |
Collapse
|
34
|
Androgen deprivation therapy and the risk of parkinsonism in men with prostate cancer. World J Urol 2017; 35:1417-1423. [DOI: 10.1007/s00345-017-2010-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/24/2017] [Indexed: 01/03/2023] Open
|
35
|
d'Anglemont de Tassigny X. Outlook on the neuroprotective effect of estrogen. Neural Regen Res 2017; 12:1799-1800. [PMID: 29239320 PMCID: PMC5745828 DOI: 10.4103/1673-5374.219036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
36
|
Multiple Forms of Glutamate Dehydrogenase in Animals: Structural Determinants and Physiological Implications. BIOLOGY 2016; 5:biology5040053. [PMID: 27983623 PMCID: PMC5192433 DOI: 10.3390/biology5040053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/26/2016] [Accepted: 12/07/2016] [Indexed: 11/17/2022]
Abstract
Glutamate dehydrogenase (GDH) of animal cells is usually considered to be a mitochondrial enzyme. However, this enzyme has recently been reported to be also present in nucleus, endoplasmic reticulum and lysosomes. These extramitochondrial localizations are associated with moonlighting functions of GDH, which include acting as a serine protease or an ATP-dependent tubulin-binding protein. Here, we review the published data on kinetics and localization of multiple forms of animal GDH taking into account the splice variants, post-translational modifications and GDH isoenzymes, found in humans and apes. The kinetic properties of human GLUD1 and GLUD2 isoenzymes are shown to be similar to those published for GDH1 and GDH2 from bovine brain. Increased functional diversity and specific regulation of GDH isoforms due to alternative splicing and post-translational modifications are also considered. In particular, these structural differences may affect the well-known regulation of GDH by nucleotides which is related to recent identification of thiamine derivatives as novel GDH modulators. The thiamine-dependent regulation of GDH is in good agreement with the fact that the non-coenzyme forms of thiamine, i.e., thiamine triphosphate and its adenylated form are generated in response to amino acid and carbon starvation.
Collapse
|
37
|
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 PMCID: PMC11482345 DOI: 10.1007/s10571-015-0259-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
38
|
The impact of biological sex and sex hormones on cognition in a rat model of early, pre-motor Parkinson's disease. Neuroscience 2016; 345:297-314. [PMID: 27235739 DOI: 10.1016/j.neuroscience.2016.05.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/13/2016] [Accepted: 05/18/2016] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is well known for motor deficits such as bradykinesia. However, patients often experience additional deficits in working memory, behavioral selection, decision-making and other executive functions. Like other features of PD, the incidence and severity of these cognitive symptoms differ in males and females. However, preclinical models have not been used to systematically investigate the roles that sex or sex hormones may play in these complex signs. To address this, we used a Barnes maze spatial memory paradigm to compare the effects of a bilateral nigrostriatal dopamine lesion model of early PD on cognitive behaviors in adult male and female rats and in adult male rats that were gonadectomized or gonadectomized and supplemented with testosterone or estradiol. We found that dopamine lesions produced deficits in working memory and other executive operations, albeit only in male rats where circulating androgen levels were physiological. In males where androgen levels were depleted, lesions produced no additional Barnes maze deficits and attenuated those previously linked to androgen deprivation. We also found that while most measures of Barnes maze performance were unaffected by dopamine lesions in the females, lesions did induce dramatic shifts from their preferred use of thigmotactic navigation to the use of spatially guided place strategies similar to those normally preferred by males. These and other sex- and sex hormone-specific differences in the effects of nigrostriatal dopamine lesions on executive function highlight the potential of gonadal steroids as protective and/or therapeutic for the cognitive symptoms of PD. However, their complexity also indicates the need for a more thorough understanding of androgen and estrogen effects in guiding the development of hormone therapies that might effectively address these non-motor signs.
Collapse
|
39
|
Grados M, Prazak M, Saif A, Halls A. A review of animal models of obsessive-compulsive disorder: a focus on developmental, immune, endocrine and behavioral models. Expert Opin Drug Discov 2015; 11:27-43. [PMID: 26558411 DOI: 10.1517/17460441.2016.1103225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Obsessive-compulsive disorder (OCD) is a neuropsychiatric condition characterized by intrusive thoughts (obsessions) and/or repetitive behaviors (compulsions). Several models of OCD exist, many which employ behaviors such as over-grooming or hoarding as correlates for compulsive behaviors - often using a response to serotonergic agents as evidence for their validity. Recent discoveries in the genetics of OCD and the identification of aberrancies of glutamatergic, hormonal, and immune pathways in the OCD phenotype highlight a need to review existing of animal models of OCD. The focus of attention to these pathways may lead to possible new targets for drug discovery. AREAS COVERED In this review, the authors describe frameworks for animal models in OCD conceptualized as either biological (e.g., developmental, genetic, and endocrine pathways), or behavioral (e.g., repetitive grooming, and stereotypies). In addition, the authors give special attention to the emerging role of glutamate in OCD. EXPERT OPINION While many animal models for OCD demonstrate pathologic repetitive behavior phenotypes, which are relieved by serotoninergic agents, animal models based on reversal learning, perseverative responding, and neurodevelopmental mechanisms represent robust new paradigms. Glutamatergic influences in these new animal models suggest that drug discovery using neuroprotective approaches may represent a new stage for pharmacologic developments in OCD.
Collapse
Affiliation(s)
- Marco Grados
- a Department of Psychiatry , Johns Hopkins University , 1800 Orleans St. - 12th floor, Baltimore , MD 21287 , USA
| | - Michael Prazak
- b Department of Medicine , Dow University of Health Sciences , Karachi , Pakistan
| | - Aneeqa Saif
- c Department of Psychology Grand Forks , University of North Dakota , ND , USA
| | - Andrew Halls
- a Department of Psychiatry , Johns Hopkins University , 1800 Orleans St. - 12th floor, Baltimore , MD 21287 , USA
| |
Collapse
|
40
|
Kuhn C. Emergence of sex differences in the development of substance use and abuse during adolescence. Pharmacol Ther 2015; 153:55-78. [PMID: 26049025 DOI: 10.1016/j.pharmthera.2015.06.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 04/29/2015] [Indexed: 12/24/2022]
Abstract
Substance use and abuse begin during adolescence. Male and female adolescent humans initiate use at comparable rates, but males increase use faster. In adulthood, more men than women use and abuse addictive drugs. However, some women progress more rapidly from initiation of use to entry into treatment. In animal models, adolescent males and females consume addictive drugs similarly. However, reproductively mature females acquire self-administration faster, and in some models, escalate use more. Sex/gender differences exist in neurobiologic factors mediating both reinforcement (dopamine, opioids) and aversiveness (CRF, dynorphin), as well as intrinsic factors (personality, psychiatric co-morbidities) and extrinsic factors (history of abuse, environment especially peers and family) which influence the progression from initial use to abuse. Many of these important differences emerge during adolescence, and are moderated by sexual differentiation of the brain. Estradiol effects which enhance both dopaminergic and CRF-mediated processes contribute to the female vulnerability to substance use and abuse. Testosterone enhances impulsivity and sensation seeking in both males and females. Several protective factors in females also influence initiation and progression of substance use including hormonal changes of pregnancy as well as greater capacity for self-regulation and lower peak levels of impulsivity/sensation seeking. Same sex peers represent a risk factor more for males than females during adolescence, while romantic partners increase risk for women during this developmental epoch. In summary, biologic factors, psychiatric co-morbidities as well as personality and environment present sex/gender-specific risks as adolescents begin to initiate substance use.
Collapse
Affiliation(s)
- Cynthia Kuhn
- Department of Pharmacology and Cancer Biology, Box 3813, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
41
|
Acaz-Fonseca E, Duran JC, Carrero P, Garcia-Segura LM, Arevalo MA. Sex differences in glia reactivity after cortical brain injury. Glia 2015; 63:1966-1981. [DOI: 10.1002/glia.22867] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Affiliation(s)
| | - Juan C. Duran
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - Paloma Carrero
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - Luis M. Garcia-Segura
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - M. Angeles Arevalo
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| |
Collapse
|
42
|
Gillies G, Virdee K, McArthur S, Dalley J. Sex-dependent diversity in ventral tegmental dopaminergic neurons and developmental programing: A molecular, cellular and behavioral analysis. Neuroscience 2014; 282:69-85. [PMID: 24943715 PMCID: PMC4245713 DOI: 10.1016/j.neuroscience.2014.05.033] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/12/2014] [Accepted: 05/18/2014] [Indexed: 02/02/2023]
Abstract
The knowledge that diverse populations of dopaminergic neurons within the ventral tegmental area (VTA) can be distinguished in terms of their molecular, electrophysiological and functional properties, as well as their differential projections to cortical and subcortical regions has significance for key brain functions, such as the regulation of motivation, working memory and sensorimotor control. Almost without exception, this understanding has evolved from landmark studies performed in the male sex. However, converging evidence from both clinical and pre-clinical studies illustrates that the structure and functioning of the VTA dopaminergic systems are intrinsically different in males and females. This may be driven by sex differences in the hormonal environment during adulthood ('activational' effects) and development (perinatal and/or pubertal 'organizational' effects), as well as genetic factors, especially the SRY gene on the Y chromosome in males, which is expressed in a sub-population of adult midbrain dopaminergic neurons. Stress and stress hormones, especially glucocorticoids, are important factors which interact with the VTA dopaminergic systems in order to achieve behavioral adaptation and enable the individual to cope with environmental change. Here, also, there is male/female diversity not only during adulthood, but also in early life when neurobiological programing by stress or glucocorticoid exposure differentially impacts dopaminergic developmental trajectories in male and female brains. This may have enduring consequences for individual resilience or susceptibility to pathophysiological change induced by stressors in later life, with potential translational significance for sex bias commonly found in disorders involving dysfunction of the mesocorticolimbic dopaminergic systems. These findings highlight the urgent need for a better understanding of the sexual dimorphism in the VTA if we are to improve strategies for the prevention and treatment of debilitating conditions which differentially affect men and women in their prevalence and nature, including schizophrenia, attention/deficit hyperactivity disorder, autism spectrum disorders, anxiety, depression and addiction.
Collapse
Affiliation(s)
- G.E. Gillies
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK,Corresponding author. Address: Division of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK. Tel: +44-(0)-20-7594-7050.
| | - K. Virdee
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK
| | - S. McArthur
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London EC1 6BQ, UK
| | - J.W. Dalley
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychiatry, University of Cambridge, Addenbrooke’s Hospital, Hill’s Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
43
|
Zhu H, Han X, Ji D, Lv G, Xu M. Estrogen inhibits lipid peroxidation after hypoxic-ischemic brain damage in neonatal rats. Neural Regen Res 2014; 7:2424-31. [PMID: 25337092 PMCID: PMC4200716 DOI: 10.3969/j.issn.1673-5374.2012.31.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/08/2012] [Indexed: 11/18/2022] Open
Abstract
Sprague-Dawley neonatal rats within 7 days after birth were used in this study. The left common carotid artery was occluded and rats were housed in an 8% O2 environment for 2 hours to establish a hypoxic-ischemic brain damage model. 17β-estradiol (1 × 10(-5) M) was injected into the rat abdominal cavity after the model was successfully established. The left hemisphere was obtained at 12, 24, 48, 72 hours after operation. Results showed that malondialdehyde content in the left brain of neonatal rats gradually increased as modeling time prolonged, while malondialdehyde content of 17β-estrodial-treated rats significantly declined by 24 hours, reached lowest levels at 48 hours, and then peaked at 72 hours after injury. Nicotinamide-adenine dinucleotide phosphate histochemical staining showed the nitric oxide synthase-positive cells and fibers dyed blue/violet and were mainly distributed in the cortex, hippocampus and medial septal nuclei. The number of nitric oxide synthase-positive cells peaked at 48 hours and significantly decreased after 17β-estrodial treatment. Our experimental findings indicate that estrogen plays a protective role following hypoxic-ischemic brain damage by alleviating lipid peroxidation through reducing the expression of nitric oxide synthase and the content of malondialdehyde.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao Han
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, Jiangsu Key Laboratory of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Dafeng Ji
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, Jiangsu Key Laboratory of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Guangming Lv
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, Jiangsu Key Laboratory of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Meiyu Xu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
44
|
Abstract
Parkinson's disease (PD) displays a greater prevalence and earlier age at onset in men. This review addresses the concept that sex differences in PD are determined, largely, by biological sex differences in the NSDA system which, in turn, arise from hormonal, genetic and environmental influences. Current therapies for PD rely on dopamine replacement strategies to treat symptoms, and there is an urgent, unmet need for disease modifying agents. As a significant degree of neuroprotection against the early stages of clinical or experimental PD is seen, respectively, in human and rodent females compared with males, a better understanding of brain sex dimorphisms in the intact and injured NSDA system will shed light on mechanisms which have the potential to delay, or even halt, the progression of PD. Available evidence suggests that sex-specific, hormone-based therapeutic agents hold particular promise for developing treatments with optimal efficacy in men and women.
Collapse
|
45
|
Acaz-Fonseca E, Sanchez-Gonzalez R, Azcoitia I, Arevalo MA, Garcia-Segura LM. Role of astrocytes in the neuroprotective actions of 17β-estradiol and selective estrogen receptor modulators. Mol Cell Endocrinol 2014; 389:48-57. [PMID: 24444786 DOI: 10.1016/j.mce.2014.01.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 01/04/2023]
Abstract
Neuroprotective actions of 17β-estradiol (estradiol) are in part mediated by direct actions on neurons. Astrocytes, which play an essential role in the maintenance of the homeostasis of neural tissue, express estrogen receptors and are also involved in the neuroprotective actions of estradiol in the brain. Estradiol controls gliosis and regulates neuroinflammation, edema and glutamate transport acting on astrocytes. In addition, the hormone regulates the release of neurotrophic factors and other neuroprotective molecules by astrocytes. In addition, reactive astrocytes are a local source of neuroprotective estradiol for the injured brain. Since estradiol therapy is not free from peripheral risks, alternatives for the hormone have been explored. Some selective estrogen receptor modulators (SERMs), which are already in use in clinical practice for the treatment of breast cancer, osteoporosis or menopausal symptoms, exert similar actions to estradiol on astrocytes. Therefore, SERMs represent therapeutic alternatives to estradiol for the activation of astroglia-mediated neuroprotective mechanisms.
Collapse
Affiliation(s)
| | | | - Iñigo Azcoitia
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | | | | |
Collapse
|
46
|
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: 128] [Impact Index Per Article: 11.6] [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
|
47
|
Bobzean SAM, DeNobrega AK, Perrotti LI. Sex differences in the neurobiology of drug addiction. Exp Neurol 2014; 259:64-74. [PMID: 24508560 DOI: 10.1016/j.expneurol.2014.01.022] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/21/2014] [Accepted: 01/27/2014] [Indexed: 01/09/2023]
Abstract
Epidemiological data demonstrate that while women report lower rates of drug use than men, the number of current drug users and abusers who are women continues to increase. In addition women progress through the phases of addiction differently than men; women transition from casual drug use to addiction faster, are more reactive to stimuli that trigger relapse, and have higher rates of relapse then men. Sex differences in physiological and psychological responses to drugs of abuse are well documented and it is well established that estrogen effects on dopamine (DA) systems are largely responsible for these sex differences. However, the downstream mechanisms that result from interactions between estrogen and the effects of drugs of abuse on the DA system are just beginning to be explored. Here we review the basic neurocircuitry which underlies reward and addiction; highlighting the neuroadaptive changes that occur in the mesolimbic dopamine reward and anti-reward/stress pathways. We propose that sex differences in addiction are due to sex differences in the neural systems which mediate positive and negative reinforcement and that these differences are modulated by ovarian hormones. This forms a neurobehavioral basis for the search for the molecular and cellular underpinnings that uniquely guide motivational behaviors and make women more vulnerable to developing and sustaining addiction than men.
Collapse
Affiliation(s)
- Samara A M Bobzean
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Aliza K DeNobrega
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Linda I Perrotti
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
48
|
Hahn YK, Podhaizer EM, Farris SP, Miles MF, Hauser KF, Knapp PE. Effects of chronic HIV-1 Tat exposure in the CNS: heightened vulnerability of males versus females to changes in cell numbers, synaptic integrity, and behavior. Brain Struct Funct 2013; 220:605-23. [PMID: 24352707 PMCID: PMC4341022 DOI: 10.1007/s00429-013-0676-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/11/2013] [Indexed: 01/11/2023]
Abstract
HIV-associated damage to the central nervous system results in cognitive and motor deficits. Anti-retroviral therapies reduce the severity of symptoms, yet the proportion of patients affected has remained the same or increased. Although approximately half of HIV-infected patients worldwide are women, the question of whether biological sex influences outcomes of HIV infection has received little attention. We explored this question for both behavioral and cellular/morphologic endpoints, using a transgenic mouse that inducibly expresses HIV-1 Tat in the brain. After 3 months of HIV-1 Tat exposure, both sexes showed similar reduced open field ambulation. Male Tat+ mice also showed reduced forelimb grip strength and enhanced anxiety in a light–dark box assay. Tat+ males did not improve over 12 weeks of repeated rotarod testing, indicating a motor memory deficit. Male mice also had more cellular deficits in the striatum. Neither sex showed a change in volume or total neuron numbers. Both had equally reduced oligodendroglial populations and equivalent microglial increases. However, astrogliosis and microglial nitrosative stress were higher in males. Dendrites on medium spiny neurons in male Tat+ mice had fewer spines, and levels of excitatory and inhibitory pre- and post-synaptic proteins were disrupted. Our results predict sex as a determinant of HIV effects in brain. Increased behavioral deficits in males correlated with glial activation and synaptic damage, both of which are implicated in cognitive/motor impairments in patients. Tat produced by residually infected cells despite antiretroviral therapy may be an important determinant of the synaptodendritic instability and behavioral deficits accompanying chronic infection.
Collapse
Affiliation(s)
- Yun Kyung Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, PO Box 980709, Richmond, VA, 23298-0709, USA
| | | | | | | | | | | |
Collapse
|
49
|
Cereda E, Barichella M, Cassani E, Caccialanza R, Pezzoli G. Reproductive factors and clinical features of Parkinson's disease. Parkinsonism Relat Disord 2013; 19:1094-9. [DOI: 10.1016/j.parkreldis.2013.07.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/09/2013] [Accepted: 07/16/2013] [Indexed: 01/16/2023]
|
50
|
Abstract
Substantial evidence shows that the hypophyseal–pituitary–adrenal (HPA) axis and corticosteroids are involved in the process of addiction to a variety of agents, and the adrenal cortex has a key role. In general, plasma concentrations of cortisol (or corticosterone in rats or mice) increase on drug withdrawal in a manner that suggests correlation with the behavioural and symptomatic sequelae both in man and in experimental animals. Corticosteroid levels fall back to normal values in resumption of drug intake. The possible interactions between brain corticotrophin releasing hormone (CRH) and proopiomelanocortin (POMC) products and the systemic HPA, and additionally with the local CRH–POMC system in the adrenal gland itself, are complex. Nevertheless, the evidence increasingly suggests that all may be interlinked and that CRH in the brain and brain POMC products interact with the blood-borne HPA directly or indirectly. Corticosteroids themselves are known to affect mood profoundly and may themselves be addictive. Additionally, there is a heightened susceptibility for addicted subjects to relapse in conditions that are associated with change in HPA activity, such as in stress, or at different times of the day. Recent studies give compelling evidence that a significant part of the array of addictive symptoms is directly attributable to the secretory activity of the adrenal cortex and the actions of corticosteroids. Additionally, sex differences in addiction may also be attributable to adrenocortical function: in humans, males may be protected through higher secretion of DHEA (and DHEAS), and in rats, females may be more susceptible because of higher corticosterone secretion.
Collapse
|