1
|
Bentall L, Parr‐Brownlie L. Sexual Dimorphism in Levodopa-Induced Dyskinesia Following Parkinson's Disease: Uncharted Territory. Eur J Neurosci 2025; 61:e70144. [PMID: 40360439 PMCID: PMC12075048 DOI: 10.1111/ejn.70144] [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: 11/23/2024] [Revised: 04/12/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Sexual dimorphism is well-documented in Parkinson's disease (PD); however, when it comes to levodopa-induced dyskinesia (LID), epidemiological and clinical findings are scarce. This is an oversight because recent studies show significant correlations between LID risk and female sex. Estrogen strongly impacts neuronal function, affecting cognitive tasks such as movement, object recognition, and reward. In movement pathways, estrogen increases dopamine synthesis, transmission, and regulation, resulting in neuroprotection for PD in women. However, following menopause, PD prevalence, symptom severity, and LID risk increase for women. Consequently, early to mid-life estrogen state is neuroprotective, but later in life becomes a risk factor for PD and LID. This review explores estrogen's action in the brain, specifically within the dopamine system. Sexual dimorphism is described for the prevalence and onset of PD and LID. We examine the cellular basis of estrogen's role in sexual dimorphism and integrate these ideas to hypothesize why the risk for LID is higher for women, than men, with PD. Lastly, this review proposes that women with PD need their symptoms to be considered and managed differently to males. Treatment of women with PD should be based on their menopausal stage, as estrogen may be masking, exacerbating, or complicating symptoms. Importantly, we present these concepts to stimulate discussion among clinical and bench scientists so that key experiments can be conducted to examine the mechanisms underlying LID, so they can be prevented to improve the quality of life for women and men living with PD in the future.
Collapse
|
2
|
Hernandez G, Zhao J, Niu Z, MacGowan D, Capolicchio T, Song A, Gul S, Moiz A, Herrera I, Day JJ, Flores C. Amphetamine in Adolescence Induces a Sex-Specific Mesolimbic Dopamine Phenotype in the Adult Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640363. [PMID: 40060609 PMCID: PMC11888448 DOI: 10.1101/2025.02.26.640363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Drugs of abuse in adolescence impact brain maturation and increase psychiatric risk, with differences in sensitivity between males and females. Amphetamine in adolescent male, but not female mice, causes dopamine axons intended to innervate the nucleus accumbens and to grow ectopically to the prefrontal cortex (PFC). This is mediated by drug-induced downregulation of the Netrin-1 receptor DCC. How off-target dopamine axons function in the adult PFC remains to be determined. Here we report that males and females show place preference for amphetamine in adolescence. However, only in males, amphetamine increases PFC dopamine transporter expression in adulthood: leading to aberrant baseline dopamine transients, faster dopamine release, and exaggerated responses to acute methylphenidate. Upregulation of DCC in adolescence, using CRISPRa, prevents all these changes. Mesolimbic dopamine axons rerouted to the PFC in adolescence retain anatomical and functional phenotypes of their intended target, rendering males enduringly vulnerable to the harmful effects of drugs of abuse.
Collapse
Affiliation(s)
- G Hernandez
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - J Zhao
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Z Niu
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - D MacGowan
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
| | - T Capolicchio
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
| | - A Song
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - S Gul
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - A Moiz
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - I Herrera
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - J J Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C Flores
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Department of Psychiatry McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, QC, Canada
| |
Collapse
|
3
|
Chen Y, Hu J, Zhang Y, Peng L, Li X, Li C, Wu X, Wang C. Epilepsy therapy beyond neurons: unveiling astrocytes as cellular targets. Neural Regen Res 2025; 21:01300535-990000000-00659. [PMID: 39819836 PMCID: PMC12094549 DOI: 10.4103/nrr.nrr-d-24-01035] [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: 10/15/2024] [Revised: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
ABSTRACT Epilepsy is a leading cause of disability and mortality worldwide. However, despite the availability of more than 20 antiseizure medications, more than one-third of patients continue to experience seizures. Given the urgent need to explore new treatment strategies for epilepsy, recent research has highlighted the potential of targeting gliosis, metabolic disturbances, and neural circuit abnormalities as therapeutic strategies. Astrocytes, the largest group of nonneuronal cells in the central nervous system, play several crucial roles in maintaining ionic and energy metabolic homeostasis in neurons, regulating neurotransmitter levels, and modulating synaptic plasticity. This article briefly reviews the critical role of astrocytes in maintaining balance within the central nervous system. Building on previous research, we discuss how astrocyte dysfunction contributes to the onset and progression of epilepsy through four key aspects: the imbalance between excitatory and inhibitory neuronal signaling, dysregulation of metabolic homeostasis in the neuronal microenvironment, neuroinflammation, and the formation of abnormal neural circuits. We summarize relevant basic research conducted over the past 5 years that has focused on modulating astrocytes as a therapeutic approach for epilepsy. We categorize the therapeutic targets proposed by these studies into four areas: restoration of the excitation-inhibition balance, reestablishment of metabolic homeostasis, modulation of immune and inflammatory responses, and reconstruction of abnormal neural circuits. These targets correspond to the pathophysiological mechanisms by which astrocytes contribute to epilepsy. Additionally, we need to consider the potential challenges and limitations of translating these identified therapeutic targets into clinical treatments. These limitations arise from interspecies differences between humans and animal models, as well as the complex comorbidities associated with epilepsy in humans. We also highlight valuable future research directions worth exploring in the treatment of epilepsy and the regulation of astrocytes, such as gene therapy and imaging strategies. The findings presented in this review may help open new therapeutic avenues for patients with drug-resistant epilepsy and for those suffering from other central nervous system disorders associated with astrocytic dysfunction.
Collapse
Affiliation(s)
- Yuncan Chen
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayi Hu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lulu Peng
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cong Li
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xunyi Wu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cong Wang
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Shanghai, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
4
|
Nelson KH, Freels DL, Carter JS, Wood SK, Denton AR, Hopkins JL, Goldsmith ST, Lewandowski SI, Scofield MD, Reichel CM. Perirhinal cortex to the nucleus accumbens circuit in novelty salience following methamphetamine self-administration. ADDICTION NEUROSCIENCE 2024; 13:100181. [PMID: 39664755 PMCID: PMC11633022 DOI: 10.1016/j.addicn.2024.100181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Methamphetamine (meth) use disorder is part of an overarching use disorder that encompasses continued drug seeking and an increased risk of returning to drug use following periods of abstaining. Chronic meth use results in drug-induced cortical plasticity in the perirhinal cortex (PRC) that mediates responses to novelty. PRH projection targets are numerous and include the nucleus accumbens core (NAc). Whereas the PRH-prefrontal cortex is involved in object recognition; we propose that the PRH-NAc is involved in novelty salience. Rats underwent short-access (ShA, 1 hr) or long-access (LgA, 6 hr) meth self-administration (SA). We then used a dual viral strategy to inhibit or activate PRH-NAc during a novel cue test in which rats were presented with meth-associated and novel levers. Response patterns on these levers differ depending on the meth access protocol: ShA meth SA results in equal responding on both novel- and meth-associated levers, whereas LgA meth results in perseverative responding on the meth-associated lever. Inactivation of the PRH-NAc increased responding on the meth lever relative to the novel lever, resulting in a LgA behavioral phenotype. In contrast, activation in LgA rats was without a behavioral effect. We also report that male LgA sucrose SA animals perseverated on the novel lever rather than the meth-associated lever, which contrast their meth SA counter parts and female specific patterns of behavior. These data open a new line of interest in the role of the PRH-NAc circuit in novelty salience through identification of the behavioral relevance of this circuit.
Collapse
Affiliation(s)
- Katharine H. Nelson
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Dylan L. Freels
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Jordan S. Carter
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Samuel K. Wood
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Adam R. Denton
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Jordan L. Hopkins
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Sarah T. Goldsmith
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Stacia I. Lewandowski
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Michael D. Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Carmela M. Reichel
- Department of Neuroscience, Medical University of South Carolina, Basic Science Building 416, MSC 510, 173 Ashley Avenue, Charleston, SC 29425, USA
| |
Collapse
|
5
|
Armenta-Resendiz M, Carter JS, Hunter Z, Taniguchi M, Reichel CM, Lavin A. Sex differences in behavior, cognitive, and physiological recovery following methamphetamine administration. Psychopharmacology (Berl) 2024; 241:2331-2345. [PMID: 38953940 PMCID: PMC11513735 DOI: 10.1007/s00213-024-06638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
Intact executive functions are required for proper performance of cognitive tasks and relies on balance of excitatory and inhibitory (E/I) transmission in the medial prefrontal cortex (mPFC). Hypofrontality is a state of decreased activity in the mPFC and is seen in several neuropsychiatric conditions, including substance use disorders. People who chronically use methamphetamine (meth) develop hypofrontality and concurrent changes in cognitive processing across several domains. Despite the fact that there are sex difference in substance use disorders, few studies have considered sex as a biological variable regarding meth-mediated hypoactivity in mPFC and concurrent cognitive deficits. Hypofrontality along with changes in cognition are emulated in rodent models following repeated meth administration. Here, we used a meth sensitization regimen to study sex differences in a Temporal Order Memory (TOM) task following short (7 days) or prolonged (28 days) periods of abstinence. GABAergic transmission, GABAA receptor (GABAAR) and GABA Transporter (GAT) mRNA expression in the mPFC were evaluated with patch-clamp recordings and RT-qPCR, respectively. Both sexes sensitized to the locomotor activating effects of meth, with the effect persisting in females. After short abstinence, males and females had impaired TOM and increased GABAergic transmission. Female rats recovered from these changes after prolonged abstinence, whereas male rats showed enduring changes. In general, meth appears to elicit an overall decrease in GABAAR expression after short abstinence; whereas GABA transporters are decreased in meth female rats after prolonged abstinence. These results show sex differences in the long-term effects of repeated meth exposure and suggest that females have neuroprotective mechanisms that alleviate some of the meth-mediated cognitive deficits.
Collapse
Affiliation(s)
| | - Jordan S Carter
- Department of Neuroscience, MUSC, 173 Ashley Ave 403BSB, Charleston, SC, 29425, USA
| | - Zachariah Hunter
- Department of Neuroscience, MUSC, 173 Ashley Ave 403BSB, Charleston, SC, 29425, USA
| | - Makoto Taniguchi
- Department of Neuroscience, MUSC, 173 Ashley Ave 403BSB, Charleston, SC, 29425, USA
| | - Carmela M Reichel
- Department of Neuroscience, MUSC, 173 Ashley Ave 403BSB, Charleston, SC, 29425, USA
| | - Antonieta Lavin
- Department of Neuroscience, MUSC, 173 Ashley Ave 403BSB, Charleston, SC, 29425, USA.
| |
Collapse
|
6
|
Simonetti A, Ferrara OM, Kotzalidis GD, Bernardi E, Restaino A, Moccia L, Camardese G, Mazza M, Scambia G, Sani G. Suicide in Gynecological and Breast Cancer: A Systematic Review. Psychooncology 2024; 33:e70007. [PMID: 39439077 DOI: 10.1002/pon.70007] [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: 02/27/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Depression and suicide rates are high among cancer sufferers. Women with breast and gynecological cancer show high levels of distress, depressive symptoms, cognitive impairment, and anxiety. Understanding suicide rates and risk factors in this population would represent a viable tool in planning tailored, prevention strategies. The objective of this study was to estimate suicide rate and identify the determinants of suicide risk in women with breast and other gynecologic cancer. METHODS A systematic research was performed in PubMed and PsycINFO from anytime to September 26, 2023. The following search strategy was used: (Gynaecol* OR Gynecolog*) AND (cancer OR tumor OR tumor OR neoplas* OR malignan*) AND suicid*. In this review, we adhered to PRISMA statement. RESULTS Nine papers met inclusion criteria. Women with breast or gynecological cancers showed higher suicide rates compared to the general population. Ovarian cancer was associated with higher suicide risk and suicidal ideation compared to other gynecological cancers. The extent of surgical demolition was positively associated with both. Psychological factors, such as self-perceived burden and alexithymia, might also influence suicidal thinking. CONCLUSIONS Women with breast and gynecological cancer are at high risk of suicide. Intervention aimed to reduce burden related to psychological factors might help reducing such risk.
Collapse
Affiliation(s)
- Alessio Simonetti
- Department of Neuroscience, Sensory Organs and Chest, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Ottavia Marianna Ferrara
- Department of Neuroscience, Sensory Organs and Chest, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Georgios D Kotzalidis
- Department of Neuroscience, Sensory Organs and Chest, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Sensory Organs and Chest, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Evelina Bernardi
- Department of Neuroscience, Sensory Organs and Chest, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Restaino
- Department of Neuroscience, Sensory Organs and Chest, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorenzo Moccia
- Department of Neuroscience, Sensory Organs and Chest, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Camardese
- Department of Neuroscience, Sensory Organs and Chest, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marianna Mazza
- Department of Neuroscience, Sensory Organs and Chest, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Gynecology Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Gynecology Oncology Unit, Department of Woman and Child Health and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gabriele Sani
- Department of Neuroscience, Sensory Organs and Chest, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Sensory Organs and Chest, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Han L, Gu JQ, Mao JH, Liu XQ, Jiao Z. Insights into the population pharmacokinetics and pharmacodynamics of quetiapine: a systematic review. Expert Rev Clin Pharmacol 2024; 17:57-72. [PMID: 38108086 DOI: 10.1080/17512433.2023.2295428] [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: 09/06/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Quetiapine exhibits notable pharmacokinetic and pharmacodynamic (PK/PD) variability, the origins of which are poorly understood. This systematic review summarizes published population PK/PD studies and identifies significant covariates accounting for this variability to inform precision dosing. METHODS We systematically searched the PubMed, Web of Science, and Embase databases and compared study characteristics, model parameters, and covariate effects. Visual predictive distributions were used to compare different models. Forest plots and Monte Carlo simulations were used to assess the influence of covariates. RESULTS Six population PK and three population PK/PD studies were included. The median apparent clearance in adults was 87.7 L/h. Strong and moderate cytochrome P450 3A4 inducers increased the apparent clearance approximately fourfold, while strong cytochrome P450 3A4 inhibitors reduced it by 93%. The half-maximum effect concentrations were 82.8 ng/mL for the Brief Psychiatric Rating Scale and 583 ng/mL for dopamine D2 receptor occupancy. Both treatment duration and quetiapine exposure were associated with weight gain. CONCLUSIONS Concurrent administration of potent or moderate CYP3A4 inducers and inhibitors need to be avoided in quetiapine-treated patients. When co-medication is required, it is recommended to adjust the dosage based on therapeutic drug monitoring. Additional research is warranted to delineate the dose-exposure-response relationships of quetiapine and active metabolite norquetiapine in pediatrics, geriatrics, hepatically-impaired patients, and women using contraceptives or are pregnant or menopausal. PROSPERO REGISTRATION CRD42023446654.
Collapse
Affiliation(s)
- Lu Han
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Qin Gu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jue-Hui Mao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiao-Qin Liu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Diaz JC, Dunaway K, Zuniga C, Sheil E, Sadeghian K, Auger AP, Baldo BA. Delayed estrogen actions diminish food consumption without changing food approach, motor activity, or hypothalamic activation elicited by corticostriatal µ-opioid signaling. Neuropsychopharmacology 2023; 48:1952-1962. [PMID: 37640922 PMCID: PMC10584984 DOI: 10.1038/s41386-023-01711-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/01/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
Mu-opioid receptor (μ-OR) signaling in forebrain sites including nucleus accumbens (Acb) and ventromedial prefrontal cortex (vmPFC) modulates reward-driven feeding and may play a role in the pathophysiology of disordered eating. In preclinical models, intra-Acb or intra-vmPFC μ-OR stimulation causes overeating and vigorous responding for food rewards. These effects have been studied mainly in male animals, despite demonstrated sex differences and estrogen modulation of central reward systems. Hence, the present study investigated sex differences and estrogen modulation of intra-Acb and intra-vmPFC μ-OR-driven feeding behaviors. First, the dose-related effects of intra-Acb and intra-vmPFC infusions of the μ-OR-selective agonist, DAMGO, were compared among intact female, ovariectomized (OVX) female, and intact male rats. The DAMGO feeding dose-effect function was flattened in intact females relative to the robust, dose-dependent effects observed in OVX females and intact males. Thus, in intact females, intra-Acb DAMGO failed to elevate food intake relative to vehicle, while intra-vmPFC DAMGO elevated food intake, but to a smaller degree compared to males and OVX females. Next, to explore the possible role of estrogen in mediating the diminished DAMGO response observed in intact females, OVX rats were given intra-Acb or intra-vmPFC infusions of DAMGO either immediately after a subcutaneous injection of 17-beta-estradiol 3-benzoate (EB; 5 μg/0.1 mL) or 24 h after EB injection. Intra-Acb DAMGO effects were not changed at the immediate post-EB time point. At the delayed post-EB timepoint, significant lordosis was noted and the duration of intra-Acb DAMGO-driven feeding bouts was significantly reduced, with no change in the number of bouts initiated, locomotor hyperactivity, or Fos immunoreactivity in hypothalamic feeding and arousal systems. Similarly, EB failed to alter the motor-activational effects of intra-vmPFC DAMGO while reducing feeding. These findings indicate that delayed, presumably genomically mediated estrogen actions modulate the μ-OR-generated motivational state by reducing consummatory activity while sparing goal-approach and general arousal/activity. The results additionally suggest that EB regulation of consummatory activity occurs outside of forebrain-μ-OR control of hypothalamic systems.
Collapse
Affiliation(s)
- Julio C Diaz
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Kate Dunaway
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
- College of Letters and Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Carla Zuniga
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth Sheil
- College of Letters and Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Ken Sadeghian
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Anthony P Auger
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian A Baldo
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
9
|
Dutra-Tavares AC, Souza TP, Silva JO, Semeão KA, Mello FF, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Neonatal phencyclidine as a model of sex-biased schizophrenia symptomatology in adolescent mice. Psychopharmacology (Berl) 2023; 240:2111-2129. [PMID: 37530885 DOI: 10.1007/s00213-023-06434-3] [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: 04/22/2023] [Accepted: 07/22/2023] [Indexed: 08/03/2023]
Abstract
Sex-biased differences in schizophrenia are evident in several features of the disease, including symptomatology and response to pharmacological treatments. As a neurodevelopmental disorder, these differences might originate early in life and emerge later during adolescence. Considering that the disruption of the glutamatergic system during development is known to contribute to schizophrenia, we hypothesized that the neonatal phencyclidine model could induce sex-dependent behavioral and neurochemical changes associated with this disorder during adolescence. C57BL/6 mice received either saline or phencyclidine (5, 10, or 20 mg/kg) on postnatal days (PN) 7, 9, and 11. Behavioral assessment occurred in late adolescence (PN48-50), when mice were submitted to the open field, social interaction, and prepulse inhibition tests. Either olanzapine or saline was administered before each test. The NMDAR obligatory GluN1 subunit and the postsynaptic density protein 95 (PSD-95) were evaluated in the frontal cortex and hippocampus at early (PN30) and late (PN50) adolescence. Neonatal phencyclidine evoked dose-dependent deficits in all analyzed behaviors and males were more susceptible. Males also had reduced GluN1 expression in the frontal cortex at PN30. There were late-emergent effects at PN50. Cortical GluN1 was increased in both sexes, while phencyclidine increased cortical and decreased hippocampal PSD-95 in females. Olanzapine failed to mitigate most phencyclidine-evoked alterations. In some instances, this antipsychotic aggravated the deficits or potentiated subthreshold effects. These results lend support to the use of neonatal phencyclidine as a sex-biased neurodevelopmental preclinical model of schizophrenia. Olanzapine null effects and deleterious outcomes suggest that its use during adolescence should be further evaluated.
Collapse
Affiliation(s)
- Ana Carolina Dutra-Tavares
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Thainá P Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Juliana O Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Keila A Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Felipe F Mello
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Claudio C Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro (UERJ), RJ, São Gonçalo, Brazil
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil.
| |
Collapse
|
10
|
Seib DR, Tobiansky DJ, Meitzen J, Floresco SB, Soma KK. Neurosteroids and the mesocorticolimbic system. Neurosci Biobehav Rev 2023; 153:105356. [PMID: 37567491 PMCID: PMC11915106 DOI: 10.1016/j.neubiorev.2023.105356] [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: 12/29/2022] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
The mesocorticolimbic system coordinates executive functions, such as working memory and behavioral flexibility. This circuit includes dopaminergic projections from the ventral tegmental area to the nucleus accumbens and medial prefrontal cortex. In this review, we summarize evidence that cells in multiple nodes of the mesocorticolimbic system produce neurosteroids (steroids synthesized in the nervous system) and express steroid receptors. Here, we focus on neuroandrogens (androgens synthesized in the nervous system), neuroestrogens (estrogens synthesized in the nervous system), and androgen and estrogen receptors. We also summarize how (neuro)androgens and (neuro)estrogens affect dopamine signaling in the mesocorticolimbic system and regulate executive functions. Taken together, the data suggest that steroids produced in the gonads and locally in the brain modulate higher-order cognition and executive functions.
Collapse
Affiliation(s)
- Désirée R Seib
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| | - Daniel J Tobiansky
- Department of Biology and Neuroscience Program, St. Mary's College of Maryland, St. Mary's City, MD, USA
| | - John Meitzen
- Department of Biological Sciences and Center for Human Health and the Environment, NC State University, Raleigh, NC, USA
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K Soma
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
11
|
Hersey M, Bartole MK, Jones CS, Newman AH, Tanda G. Are There Prevalent Sex Differences in Psychostimulant Use Disorder? A Focus on the Potential Therapeutic Efficacy of Atypical Dopamine Uptake Inhibitors. Molecules 2023; 28:5270. [PMID: 37446929 PMCID: PMC10343811 DOI: 10.3390/molecules28135270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Psychostimulant use disorders (PSUD) affect a growing number of men and women and exert sizable public health and economic burdens on our global society. Notably, there are some sex differences in the onset of dependence, relapse rates, and treatment success with PSUD observed in preclinical and clinical studies. The subtle sex differences observed in the behavioral aspects of PSUD may be associated with differences in the neurochemistry of the dopaminergic system between sexes. Preclinically, psychostimulants have been shown to increase synaptic dopamine (DA) levels and may downregulate the dopamine transporter (DAT). This effect is greatest in females during the high estradiol phase of the estrous cycle. Interestingly, women have been shown to be more likely to begin drug use at younger ages and report higher levels of desire to use cocaine than males. Even though there is currently no FDA-approved medication, modafinil, a DAT inhibitor approved for use in the treatment of narcolepsy and sleep disorders, has shown promise in the treatment of PSUD among specific populations of affected individuals. In this review, we highlight the therapeutic potential of modafinil and other atypical DAT inhibitors focusing on the lack of sex differences in the actions of these agents.
Collapse
Affiliation(s)
| | | | | | | | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA; (M.H.); (M.K.B.); (C.S.J.); (A.H.N.)
| |
Collapse
|
12
|
Nadalin S, Zatković L, Peitl V, Karlović D, Vidrih B, Puljić A, Pavlić SD, Buretić-Tomljanović A. Association between PLA2 gene polymorphisms and treatment response to antipsychotic medications: A study of antipsychotic-naïve first-episode psychosis patients and nonadherent chronic psychosis patients. Prostaglandins Leukot Essent Fatty Acids 2023; 194:102578. [PMID: 37290257 DOI: 10.1016/j.plefa.2023.102578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023]
Abstract
Here we investigated whether antipsychotic treatment was influenced by three polymorphisms: rs10798059 (BanI) in the phospholipase A2 (PLA2)G4A gene, rs4375 in PLA2G6, and rs1549637 in PLA2G4C. A total of 186 antipsychotic-naïve first-episode psychosis patients or nonadherent chronic psychosis individuals (99 males and 87 females) were genotyped by polymerase chain reaction analysis/restriction fragment length polymorphism. At baseline, and after 8 weeks of treatment with various antipsychotic medications, we assessed patients' Positive and Negative Syndrome Scale (PANSS) scores, PANSS factors, and metabolic syndrome-related parameters (fasting plasma lipid and glucose levels, and body mass index). We found that PLA2G4A polymorphism influenced changes in PANSS psychopathology, and PLA2G6 polymorphism influenced changes in PANSS psychopathology and metabolic parameters. PLA2G4C polymorphism did not show any impact on PANSS psychopathology or metabolic parameters. The polymorphisms' effect sizes were estimated as moderate to strong, with contributions ranging from around 6.2-15.7%. Furthermore, the polymorphisms' effects manifested in a gender-specific manner.
Collapse
Affiliation(s)
- Sergej Nadalin
- Department of Psychiatry, General Hospital "Dr. Josip Benčević", Slavonski Brod, Croatia; School of Medicine, Catholic University of Croatia, Zagreb, Croatia.
| | - Lena Zatković
- Hospital pharmacy, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | - Vjekoslav Peitl
- School of Medicine, Catholic University of Croatia, Zagreb, Croatia; Department of Psychiatry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Dalibor Karlović
- School of Medicine, Catholic University of Croatia, Zagreb, Croatia; Department of Psychiatry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Branka Vidrih
- School of Medicine, Catholic University of Croatia, Zagreb, Croatia; Department of Psychiatry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Antonia Puljić
- School of Medicine, Catholic University of Croatia, Zagreb, Croatia; Department of Psychiatry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Sanja Dević Pavlić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Alena Buretić-Tomljanović
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
13
|
Mishra P, Davies DA, Albensi BC. The Interaction Between NF-κB and Estrogen in Alzheimer's Disease. Mol Neurobiol 2023; 60:1515-1526. [PMID: 36512265 DOI: 10.1007/s12035-022-03152-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
Post-menopausal women are at a higher risk of developing Alzheimer's disease (AD) than males. The higher rates of AD in women are associated with the sharp decline in the estrogen levels after menopause. Estrogen has been shown to downregulate inflammatory cytokines in the central nervous system (CNS), which has a neuroprotective role against neurodegenerative diseases including AD. Sustained neuroinflammation is associated with neurodegeneration and contributes to AD. Nuclear factor kappa-B (NF-κB) is a transcription factor involved with the modulation of inflammation and interacts with estrogen to influence the progression of AD. Application of 17β-estradiol (E2) has been shown to inhibit NF-κB, thereby reducing transcription of NF-κB target genes. Despite accumulating evidence showing that estrogens have beneficial effects in pre-clinical AD studies, there are mixed results with hormone replacement therapy in clinical trials. Furthering our understanding of how NF-κB interacts with estrogen and alters the progression of neurodegenerative disorders including AD, should be beneficial and result in the development of novel therapeutics.
Collapse
Affiliation(s)
- Pranav Mishra
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada.,Department of Pharmacology & Therapeutics, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Don A Davies
- Department of Biology, York University, Toronto, ON, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada. .,Department of Pharmacology & Therapeutics, College of Medicine, University of Manitoba, Winnipeg, MB, Canada. .,Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.
| |
Collapse
|
14
|
Kövesdi E, Udvarácz I, Kecskés A, Szőcs S, Farkas S, Faludi P, Jánosi TZ, Ábrahám IM, Kovács G. 17β-estradiol does not have a direct effect on the function of striatal cholinergic interneurons in adult mice in vitro. Front Endocrinol (Lausanne) 2023; 13:993552. [PMID: 36686456 PMCID: PMC9848397 DOI: 10.3389/fendo.2022.993552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
The striatum is an essential component of the basal ganglia that is involved in motor control, action selection and motor learning. The pathophysiological changes of the striatum are present in several neurological and psychiatric disorder including Parkinson's and Huntington's diseases. The striatal cholinergic neurons are the main regulators of striatal microcircuitry. It has been demonstrated that estrogen exerts various effects on neuronal functions in dopaminergic and medium spiny neurons (MSN), however little is known about how the activity of cholinergic interneurons are influenced by estrogens. In this study we examined the acute effect of 17β-estradiol on the function of striatal cholinergic neurons in adult mice in vitro. We also tested the effect of estrus cycle and sex on the spontaneous activity of cholinergic interneurons in the striatum. Our RNAscope experiments showed that ERα, ERβ, and GPER1 receptor mRNAs are expressed in some striatal cholinergic neurons at a very low level. In cell-attached patch clamp experiments, we found that a high dose of 17β-estradiol (100 nM) affected the spontaneous firing rate of these neurons only in old males. Our findings did not demonstrate any acute effect of a low concentration of 17β-estradiol (100 pM) or show any association of estrus cycle or sex with the activity of striatal cholinergic neurons. Although estrogen did not induce changes in the intrinsic properties of neurons, indirect effects via modulation of the synaptic inputs of striatal cholinergic interneurons cannot be excluded.
Collapse
Affiliation(s)
- Erzsébet Kövesdi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Ildikó Udvarácz
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Angéla Kecskés
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Szilárd Szőcs
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Szidónia Farkas
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Péter Faludi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Tibor Z. Jánosi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - István M. Ábrahám
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Gergely Kovács
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| |
Collapse
|
15
|
Shahini N, Salimi Z, Kiani D, Raftari A, Ziaee M. Relationship of serum estradiol and progesterone with symptoms and sex difference in schizophrenia: A cross-sectional study in Iran. Front Psychiatry 2023; 14:1075780. [PMID: 36970277 PMCID: PMC10030605 DOI: 10.3389/fpsyt.2023.1075780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/25/2023] [Indexed: 03/29/2023] Open
Abstract
Background Schizophrenia is a devastating disease characterized by frequent relapses, cognitive decline, and emotional and functional disability, with unknown causes. The phenomenology and clinical course of schizophrenic disorders are different between the two genders, which is thought to be related mainly to the effects of steroid sex hormones on the nervous system. Regarding inconsistencies in the studies, we aimed to compare the levels of estradiol and progesterone between schizophrenia patients and healthy individuals. Methods This cross-sectional study was conducted on 66 patients referred to the specialized clinical psychiatric ward of a teaching hospital in the north of Iran, for 5 months in 2021. Thirty-three schizophrenia patients confirmed by a psychiatrist based on DSM5 criteria were included in the case group, and 33 individuals without a psychiatric disease were included in the control group. We completed a demographic information checklist for each patient, along with the Simpson-Angus extrapyramidal side effect scale (SAS) for drug side effects and the positive and negative syndrome scale (PANSS) for the severity of the disease symptoms. Then, a 3-ml blood sample was taken from each participant to determine the serum levels of estradiol and progesterone. The data were analyzed by SPSS16 software. Results Thirty-four (51.5%) and 32 (48.5%) participants in this study were male and female, respectively. The mean serum level of estradiol was 22.33 ± 13.65 pm/dl in schizophrenia patients and 29.36 ± 21.32 pm/dl in the control group, showing no significant difference between the two groups (P = 0.4). However, the mean serum level of progesterone was significantly lower in schizophrenia patients (0.37 ± 1.39 pm/dl) than in control subjects (3.15 ± 5.73 pm/dl) (P < 0.001). The PANSS and SAS scores were not significantly correlated with the level of sex hormones (P > 0.05). Serum estradiol and progesterone levels based on sex significantly differed between the two groups (except for female estradiol). Conclusion Considering the hormonal differences between schizophrenia patients and control subjects, determining hormonal levels in these patients and using complementary hormonal therapies with estradiol or similar compounds can be beneficial as the starting point of schizophrenia treatment, where therapeutic responses can draw the future developmental framework.
Collapse
Affiliation(s)
- Najmeh Shahini
- Golestan Research Center of Psychiatry (GRCP), Golestan University of Medical Sciences, Gorgan, Iran
- Clinical Research Development Unit (CRDU), 5 Azar Hospital, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zanireh Salimi
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dorsa Kiani
- Golestan Research Center of Psychiatry (GRCP), Golestan University of Medical Sciences, Gorgan, Iran
- Dorsa Kiani,
| | - Ahmad Raftari
- Golestan Research Center of Psychiatry (GRCP), Golestan University of Medical Sciences, Gorgan, Iran
| | - Maliheh Ziaee
- Department of Community Medicine, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
- *Correspondence: Maliheh Ziaee,
| |
Collapse
|
16
|
Mirtazapine attenuates the cocaine-induced locomotor sensitization in male and female C57BL/6J and BALBA/cJ mouse. Pharmacol Biochem Behav 2023; 222:173507. [PMID: 36481182 DOI: 10.1016/j.pbb.2022.173507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Clinical studies have described the efficacy of various therapeutic approaches. Results are inconsistent and clinical application is limited. Clinical trials have suggested that individual variability in the response to pharmacological therapies and sex affects the efficacy of some antidepressant drugs. Mouse strain-dependent variability influenced the response to antidepressant drugs. Some mouse strains respond faster and better to antidepressants than other mouse strains. We recently reported a series of preclinical studies that showed that dosing of mirtazapine, a noradrenergic and serotonergic antidepressant, in male and female Wistar rats decreased cocaine-induced locomotor activity and attenuated the induction and expression of cocaine-induced locomotor sensitization. Therefore, the aim of this study was to evaluate the mirtazapine effects, on cocaine-induced locomotor activity and cocaine-induced locomotor sensitization in male and female mice of the C57BL/6J and BALB/cJ strains, which differ in sensitivity to the cocaine motor effects and response to antidepressant drugs. METHODS Male and female BALB/cJ and C57BL/6J inbred mice (20-25 g) were daily dosed with 10 mg/kg of cocaine during the induction and expression of locomotor sensitization. During drug withdrawal, cocaine was withdrawn, and the groups received daily mirtazapine (30 mg/kg, i.p.) or saline. Mirtazapine was administered 30 min before cocaine. After each administration, locomotor activity for each animal was recorded for 30 min in transparent Plexiglass activity chambers. RESULTS Cocaine-induced locomotor activity were greater in C57BL/6J strain mice than BALB/cJ strain mice during the induction and expression phase of locomotor sensitization. The female mice of both strains showed a higher cocaine locomotor response than males and mirtazapine significantly decreased cocaine-induced locomotor activity, as well as the induction and expression of locomotor sensitization, regardless of mouse strain or sex. CONCLUSION The results suggest mirtazapine may be considered an effective therapeutic option to treat cocaine use disorder in men and women with very diverse genetic backgrounds.
Collapse
|
17
|
Hilz EN, Lee HJ. Estradiol and progesterone in female reward-learning, addiction, and therapeutic interventions. Front Neuroendocrinol 2023; 68:101043. [PMID: 36356909 DOI: 10.1016/j.yfrne.2022.101043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/24/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022]
Abstract
Sex steroid hormones like estradiol (E2) and progesterone (P4) guide the sexual organization and activation of the developing brain and control female reproductive behavior throughout the lifecycle; importantly, these hormones modulate functional activity of not just the endocrine system, but most of the nervous system including the brain reward system. The effects of E2 and P4 can be seen in the processing of and memory for rewarding stimuli and in the development of compulsive reward-seeking behaviors like those seen in substance use disorders. Women are at increased risk of developing substance use disorders; however, the origins of this sex difference are not well understood and therapeutic interventions targeting ovarian hormones have produced conflicting results. This article reviews the contribution of the E2 and P4 in females to functional modulation of the brain reward system, their possible roles in origins of addiction vulnerability, and the development and treatment of compulsive reward-seeking behaviors.
Collapse
Affiliation(s)
- Emily N Hilz
- The University of Texas at Austin, Department of Pharmacology, USA.
| | - Hongjoo J Lee
- The University of Texas at Austin, Department of Psychology, USA; The University of Texas at Austin, Institute for Neuroscience, USA
| |
Collapse
|
18
|
Silvani A, Ghorayeb I, Manconi M, Li Y, Clemens S. Putative Animal Models of Restless Legs Syndrome: A Systematic Review and Evaluation of Their Face and Construct Validity. Neurotherapeutics 2023; 20:154-178. [PMID: 36536233 PMCID: PMC10119375 DOI: 10.1007/s13311-022-01334-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Restless legs syndrome (RLS) is a sensorimotor disorder that severely affects sleep. It is characterized by an urge to move the legs, which is often accompanied by periodic limb movements during sleep. RLS has a high prevalence in the population and is usually a life-long condition. While its origins remain unclear, RLS is initially highly responsive to treatment with dopaminergic agonists that target D2-like receptors, in particular D2 and D3, but the long-term response is often unsatisfactory. Over the years, several putative animal models for RLS have been developed, mainly based on the epidemiological and neurochemical link with iron deficiency, treatment efficacy of D2-like dopaminergic agonists, or genome-wide association studies that identified risk factors in the patient population. Here, we present the first systematic review of putative animal models of RLS, provide information about their face and construct validity, and report their role in deciphering the underlying pathophysiological mechanisms that may cause or contribute to RLS. We propose that identifying the causal links between genetic risk factors, altered organ functions, and changes to molecular pathways in neural circuitry will eventually lead to more effective new treatment options that bypass the side effects of the currently used therapeutics in RLS, especially for long-term therapy.
Collapse
Affiliation(s)
- Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Ravenna Campus, Ravenna, Italy
| | - Imad Ghorayeb
- Département de Neurophysiologie Clinique, Pôle Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, Université de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, CNRS, Bordeaux, France
| | - Mauro Manconi
- Sleep Medicine Unit, Neurocenter of Southern Switzerland, EOC, Ospedale Civico, Lugano, Switzerland
- Department of Neurology, University Hospital, Inselspital, Bern, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Yuqing Li
- Department of Neurology, College of Medicine, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
19
|
Appetitive Motivation and Associated Neurobiology Change Differentially across the Life Course of Mouse Offspring Exposed to Peri- and Postnatal High Fat Feeding. Nutrients 2022; 14:nu14235161. [PMID: 36501191 PMCID: PMC9735866 DOI: 10.3390/nu14235161] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/07/2022] Open
Abstract
Alterations in neural pathways that regulate appetitive motivation may contribute to increased obesity risk in offspring born to mothers fed a high fat (HF) diet. However, current findings on the impact of maternal obesity on motivation in offspring are inconclusive, and there is no information about the long-lasting effects in aged animals. This study examined the longitudinal effect of perinatal and chronic postnatal HF intake on appetitive motivation in young and aged offspring. Female C57Bl/6 were fed either a control (C) or HF diet before mating through to lactation. At weaning, offspring were maintained on the C or HF diet, generating the following four diet groups: C/C, C/HF, HF/C, and HF/HF based on the pre/post weaning diet. At 6 months, motivation was higher in HF/C females, but lower in male and female C/HF and HF/HF mice. By 12 months, this difference was lost, as C-fed animals became less motivated, while motivation increased in HF-fed mice. The mRNA levels of dopamine receptor 1 and 2 increased with age, while cannabinoid receptor 1 and μ-opioid receptor expression remained stable or decreased in mesolimbic and mesocortical dopaminergic pathways. Results from this study suggest that perinatal and chronic postnatal HF feeding produced opposite effects on appetitive motivation in young adult offspring mice, which was also reflected in the shift in motivation over time. These results have significant implications for patterns of hedonic eating across the life course and the relative risk of obesity at different time points.
Collapse
|
20
|
Stuursma A, Lanjouw L, Idema DL, de Bock GH, Mourits MJE. Surgical Menopause and Bilateral Oophorectomy: Effect of Estrogen-Progesterone and Testosterone Replacement Therapy on Psychological Well-being and Sexual Functioning; A Systematic Literature Review. J Sex Med 2022; 19:1778-1789. [PMID: 36175351 DOI: 10.1016/j.jsxm.2022.08.191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Besides experiencing vasomotor symptoms, after surgical menopause and bilateral salpingo-oophorectomy (BSO), women experience moderate to severe psychological and sexual symptoms. AIMS To systematically review and meta-analyze the effect of systemic hormone replacement therapy (sHRT) on psychological well-being and sexual functioning in women after surgical menopause and BSO. METHODS Medline/Pubmed, EMBASE and PsychInfo were systematically searched until November 2021. Randomized controlled trials investigating the effect of sHRT on psychological well-being and/or sexual functioning in surgically menopausal women and women after BSO were eligible for inclusion. Two independent authors performed study selection, risk of bias assessment and data extraction. Standardized mean differences (SMDs) were calculated. OUTCOMES Primary outcomes for psychological well-being were defined as overall psychological well-being, depression, and anxiety. Primary outcomes for sexual functioning were defined as overall sexual functioning, sexual desire, and sexual satisfaction. All outcomes were assessed on short (≤12 weeks) or medium term (13-26 weeks). RESULTS Twelve studies were included. Estradiol had a beneficial effect on depressed mood on short term 3-6 years after surgery or 2 years (median) after surgery with high heterogeneity (SMD: -1.37, 95%CI: -2.38 to -0.37, P = .007, I2 79%). Testosterone had a beneficial effect on overall sexual functioning on short to medium term 4.6 years (mean) after surgery (SMD 0.38, 95%CI 0.11-0.65, I2 0%) and on sexual desire on medium term at least 3-12 months after surgery (SMD 0.38, 95%CI 0.19-0.56, I2 54%). For most studies, risk of bias was uncertain. CLINICAL IMPLICATIONS Estradiol may beneficially affect psychological symptoms after surgical menopause or BSO and testosterone might improve sexual desire and overall sexual functioning. STRENGTHS AND LIMITATIONS This review only included patient-reported outcomes, thereby reflected perceived and not simply objective symptoms in surgically menopausal women and women after BSO. The small number of studies highly varied in nature and bias could not be excluded, therefore our results should be interpreted with great caution. CONCLUSION Independent randomized controlled clinical trials investigating the effects of estrogen-progesterone and testosterone on psychological and sexual symptoms after surgical menopause are needed. PROSPERO REGISTRATION NUMBER CRD42019136698. Stuursma A, Lanjouw L, Idema DL, et al. Surgical Menopause and Bilateral Oophorectomy: Effect of Estrogen-Progesterone and Testosterone Replacement Therapy on Psychological Well-being and Sexual Functioning: A Systematic Literature Review. J Sex Med 2022;19:1778-1789.
Collapse
Affiliation(s)
- Annechien Stuursma
- Department of Obstetrics & Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Lieke Lanjouw
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Demy L Idema
- Julius Center, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marian J E Mourits
- Department of Obstetrics & Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Effects of Early Life Exposure to Sex Hormones on Neurochemical and Behavioral Responses to Psychostimulants in Adulthood: Implications in Drug Addiction. Int J Mol Sci 2022; 23:ijms23126575. [PMID: 35743018 PMCID: PMC9223714 DOI: 10.3390/ijms23126575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 12/03/2022] Open
Abstract
Early life exposure to sex hormones affects several brain areas involved in regulating locomotor and motivation behaviors. Our group has shown that neonatal exposure to testosterone propionate (TP) or estradiol valerate (EV) affected the brain dopamine (DA) system in adulthood. Here, we studied the long-lasting effects of neonatal exposure to sex hormones on behavioral and neurochemical responses to amphetamine (AMPH) and methylphenidate (MPD). Our results show that AMPH-induced locomotor activity was higher in female than male control rats. The conditioned place preference (CPP) to AMPH was only observed in EV male rats. In EV female rats, AMPH did not increase locomotor activity, but MPD-induced CPP was observed in control, EV and TP female rats. Using in vivo brain microdialysis, we observed that AMPH-induced extracellular DA levels were lower in nucleus accumbens (NAcc) of EV and TP female rats than control rats. In addition, MPD did not increase NAcc extracellular DA levels in EV rats. Using in vivo fast-scan cyclic voltammetry in striatum, MPD-induced DA reuptake was higher in EV than control rats. In summary, our results show that early life exposure to sex hormones modulates mesolimbic and nigrostriatal DA neurons producing opposite neurochemical effects induced by psychostimulant drugs in NAcc or striatum.
Collapse
|
22
|
Analysis of female enrollment in clinical trials for alcohol and substance use disorders: Is it time for sex-informed pharmacotherapy? Contemp Clin Trials 2022; 118:106784. [PMID: 35618230 DOI: 10.1016/j.cct.2022.106784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Women represent an increasing number of individuals with alcohol and substance use disorders (ASUDs), and sex-differences might affect results of interventional clinical trials (CTs). We aim at assessing the proportion of women and the reporting of sex-stratified and female-specific data in CTs for ASUDs. METHODS We extracted data from ClinicalTrials.gov on Phase 1-3 CTs of investigational drugs for ASUDs conducted from 2000 to 2021 and identified articles related to these trials. We determined the average proportions of women enrolled per trial overall, over time, and by disease area and trial phase. Next, we calculated the proportion of articles reporting sex-stratified and female-specific data. RESULTS In the 234 CTs identified, the overall proportion of women was 33.4% [95% CI: 32.7%-33.9%]), with an increasing temporal trend. Women's participation was higher in CTs of investigational drugs for tobacco (43.5% [95% CI: 42.4% -44.5%]) and alcohol use disorder (35.9% [95% CI: 34.54%-37.21%]), and closely mirrored their representation in the disease populations (46% and 37%). Conversely, women were underrepresented in clinical trials of drugs for stimulants and cocaine use disorders (25.8% [95% CI: 24.6%-27.1%]) and opioid use disorders (25.9% [95% CI:24.2%-27.7%]). Nine publications reported sex-stratified data in the method and/or result section, whereas none documented female-specific data. CONCLUSIONS Enrollment of women in ASUDs CTs has increased over time but remains low in several disease areas. This, together with the low rates of reporting of sex-stratified data, calls for an adequate inclusion of sex in the design and analysis of CTs for ASUDs.
Collapse
|
23
|
Abstract
Most psychiatric illnesses, such as schizophrenia, show profound sex differences in incidence, clinical presentation, course, and outcome. Fortunately, more recently the literature on sex differences and (to a lesser extent) effects of sex steroid hormones is expanding, and in this review we have focused on such studies in psychosis, both from a clinical/epidemiological and preclinical/animal model perspective. We begin by briefly describing the clinical evidence for sex differences in schizophrenia epidemiology, symptomatology, and pathophysiology. We then detail sex differences and sex hormone effects in behavioral animal models of psychosis, specifically psychotropic drug-induced locomotor hyperactivity and disruption of prepulse inhibition. We expand on the preclinical data to include developmental and genetic models of psychosis, such as the maternal immune activation model and neuregulin transgenic animals, respectively. Finally, we suggest several recommendations for future studies, in order to facilitate a better understanding of sex differences in the development of psychosis.
Collapse
|
24
|
Shigemoto Y, Matsuda H, Kimura Y, Chiba E, Ohnishi M, Nakaya M, Maikusa N, Ogawa M, Mukai Y, Takahashi Y, Sako K, Toyama H, Inui Y, Taki Y, Nagayama H, Ono K, Kono A, Sekiguchi K, Hirano S, Sato N. Voxel-based analysis of age and gender effects on striatal [ 123I] FP-CIT binding in healthy Japanese adults. Ann Nucl Med 2022; 36:460-467. [PMID: 35174441 DOI: 10.1007/s12149-022-01725-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/01/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Although previous studies have investigated age and gender effects on striatal subregional dopamine transporter (DaT) binding, these studies were mostly based on a conventional regions of interest-based analysis. Here, we investigated age and gender effects on striatal DaT binding at the voxel level, using a multicenter database of [(123)I] N-omega-fluoropropyl-2beta-carbomethoxy-3beta-{4-iodophenyl}nortropane ([(123)I] FP-CIT)-single photon emission computed tomography (SPECT) scans in 256 healthy Japanese adults. METHODS We used the Southampton method to calculate the specific binding ratios (SBRs) of each subject's striatum and then converted the [123I] FP-CIT SPECT images to quantitative SBRs images. To investigate the effects of age and gender effects on striatal DaT binding, we performed a voxel-based analysis using statistical parametric mapping. Gender differences were also compared between young to middle-aged subjects and elderly subjects (age threshold: 60 years). RESULTS When all subjects were explored as a group, DaT binding throughout the striatum decreased with advancing age. Among all subjects, the females showed higher DaT binding in the bilateral caudate compared to the males. In the young to middle-aged subjects, the females showed higher DaT binding throughout the striatum (with a slight caudate predominance) versus the males. In the elderly, there were no gender differences in striatal DaT binding. CONCLUSION Our findings of striatal subregional age- and gender-related differences may provide useful information to construct a more detailed DaT database in healthy Japanese subjects.
Collapse
Affiliation(s)
- Yoko Shigemoto
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan.,Drug Discovery and Cyclotron Research Center, Southern TOHOKU Research Institute for Neuroscience, Koriyama, 963-8052, Japan
| | - Hiroshi Matsuda
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan. .,Drug Discovery and Cyclotron Research Center, Southern TOHOKU Research Institute for Neuroscience, Koriyama, 963-8052, Japan. .,Southern TOHOKU Research Institute for Neuroscience, Shin-Otemachi Building 6F (621), 2-2-1, Otemachi, Chiyoda-ku, Tokyo, 199-0004, Japan.
| | - Yukio Kimura
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| | - Emiko Chiba
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| | - Masahiro Ohnishi
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| | - Moto Nakaya
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| | - Norihide Maikusa
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| | - Masayo Ogawa
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| | - Yohei Mukai
- Department of Neurology, National Center Hospital of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuji Takahashi
- Department of Neurology, National Center Hospital of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Kazuya Sako
- Department of Neurology, Nakamura Memorial Hospital, Sapporo, Japan
| | - Hiroshi Toyama
- Department of Radiology, Fujita Health University, Toyoake, Japan
| | - Yoshitaka Inui
- Department of Radiology, Fujita Health University, Toyoake, Japan
| | - Yasuyuki Taki
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hiroshi Nagayama
- Department of Neurology, Graduate School of Medicine, Nippon Medical School Bunkyo-Ku, Tokyo, Japan
| | - Kenjiro Ono
- Division of Neurology, Department of Internal Medicine, Showa University School of Medicine, Tokyo, Japan.,Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Atsushi Kono
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Sekiguchi
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shigeki Hirano
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Noriko Sato
- Department of Radiology, National Center Hospital of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan
| |
Collapse
|
25
|
Sheng JA, Tan SML, Hale TM, Handa RJ. Androgens and Their Role in Regulating Sex Differences in the Hypothalamic/Pituitary/Adrenal Axis Stress Response and Stress-Related Behaviors. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2022; 2:261-274. [PMID: 35024695 PMCID: PMC8744007 DOI: 10.1089/andro.2021.0021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Androgens play a pivotal role during development. These gonadal hormones and their receptors exert organizational actions that shape brain morphology in regions controlling the stress regulatory systems in a male-specific manner. Specifically, androgens drive sex differences in the hypothalamic/pituitary/adrenal (HPA) axis and corresponding hypothalamic neuropeptides. While studies have examined the role of estradiol and its receptors in sex differences in the HPA axis and associated behaviors, the role of androgens remains far less studied. Androgens are generally thought to modulate the HPA axis through the activation of androgen receptors (ARs). They can also impact the HPA axis through reduction to estrogenic metabolites that can bind estrogen receptors in the brain and periphery. Such regulation of the HPA axis stress response by androgens can often result in sex-biased risk factors for stress-related disorders, such as anxiety and depression. This review focuses on the biosynthesis pathways and molecular actions of androgens and their nuclear receptors. The impact of androgens on hypothalamic neuropeptide systems (corticotropin-releasing hormone, arginine vasopressin, oxytocin, dopamine, and serotonin) that control the stress response and stress-related disorders is discussed. Finally, this review discusses potential therapeutics involving androgens (androgen replacement therapies, selective AR modulator therapies) and ongoing clinical trials.
Collapse
Affiliation(s)
- Julietta A Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah M L Tan
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Taben M Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Arizona, USA
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
26
|
McNealy KR, Houser SD, Barrett ST, Bevins RA. Investigating sex differences and the effect of drug exposure order in the sensory reward-enhancing effects of nicotine and d-amphetamine alone and in combination. Neuropharmacology 2022; 202:108845. [PMID: 34678376 PMCID: PMC8627442 DOI: 10.1016/j.neuropharm.2021.108845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Nicotine enhances the rewarding effects of other environmental stimuli; this reward-enhancement encourages and maintains nicotine consumption. Nicotine use precedes other psychostimulant use, but receiving a stimulant prescription also predicts future smoking. Previously, no study has investigated effects of drug exposure order in reward-enhancement, nor with nicotine and d-amphetamine. Thus, we aimed to investigate how drug exposure order impacted the reward-enhancing effects of nicotine and d-amphetamine, alone and in combination. We used 20 male and 20 female Sprague-Dawley rats. Enhancement was investigated within-subjects by examining responding maintained by a visual stimulus reinforcer following a pre-session injection of either d-amphetamine (Sal, 0.1, 0.3, or 0.6 mg/kg) or nicotine (Sal, 0.03, 0.06, 0.1, 0.3 mg/kg). Twenty rats (10 M, 10 F) completed enhancement testing with nicotine before d-amphetamine. The other 20 rats (10 M, 10 F) completed testing with d-amphetamine before nicotine. Following these phases, rats were then given two pre-session injections: one of d-amphetamine (Sal, 0.1, 0.3, or 0.6 mg/kg) and another of nicotine (Sal, 0.03, 0.06, 0.1, or 0.3 mg/kg). Experiencing amphetamine before nicotine increased reward-enhancing effects of nicotine. Females exhibited greater effects of d-amphetamine on reward-enhancement, with no effect of exposure order. During the interaction phase, receiving nicotine before amphetamine enhanced the interaction between nicotine and d-amphetamine for females whereas amphetamine before nicotine heightened this interaction for males. From this, prior and current amphetamine use, in addition to sex, should be considered when treating nicotine dependency and when examining factors driving poly-substance use involving nicotine and d-amphetamine. Keywords: Adderall, ADHD, Dexedrine, operant, smoking, polysubstance use.
Collapse
Affiliation(s)
- Kathleen R McNealy
- Department of Psychology University of Nebraska-Lincoln, 238 Burnett Hall, Lincoln, NE, 68588-0308, USA
| | - Sydney D Houser
- Department of Psychology University of Nebraska-Lincoln, 238 Burnett Hall, Lincoln, NE, 68588-0308, USA
| | - Scott T Barrett
- Department of Psychology University of Nebraska-Lincoln, 238 Burnett Hall, Lincoln, NE, 68588-0308, USA
| | - Rick A Bevins
- Department of Psychology University of Nebraska-Lincoln, 238 Burnett Hall, Lincoln, NE, 68588-0308, USA.
| |
Collapse
|
27
|
Fisher VL, Ortiz LS, Powers AR. A computational lens on menopause-associated psychosis. Front Psychiatry 2022; 13:906796. [PMID: 35990063 PMCID: PMC9381820 DOI: 10.3389/fpsyt.2022.906796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
Abstract
Psychotic episodes are debilitating disease states that can cause extreme distress and impair functioning. There are sex differences that drive the onset of these episodes. One difference is that, in addition to a risk period in adolescence and early adulthood, women approaching the menopause transition experience a second period of risk for new-onset psychosis. One leading hypothesis explaining this menopause-associated psychosis (MAP) is that estrogen decline in menopause removes a protective factor against processes that contribute to psychotic symptoms. However, the neural mechanisms connecting estrogen decline to these symptoms are still not well understood. Using the tools of computational psychiatry, links have been proposed between symptom presentation and potential algorithmic and biological correlates. These models connect changes in signaling with symptom formation by evaluating changes in information processing that are not easily observable (latent states). In this manuscript, we contextualize the observed effects of estrogen (decline) on neural pathways implicated in psychosis. We then propose how estrogen could drive changes in latent states giving rise to cognitive and psychotic symptoms associated with psychosis. Using computational frameworks to inform research in MAP may provide a systematic method for identifying patient-specific pathways driving symptoms and simultaneously refine models describing the pathogenesis of psychosis across all age groups.
Collapse
Affiliation(s)
- Victoria L Fisher
- Yale University School of Medicine and the Connecticut Mental Health Center, New Haven, CT, United States
| | - Liara S Ortiz
- Yale University School of Medicine and the Connecticut Mental Health Center, New Haven, CT, United States
| | - Albert R Powers
- Yale University School of Medicine and the Connecticut Mental Health Center, New Haven, CT, United States
| |
Collapse
|
28
|
Trofimova IN, Gaykalova AA. Emotionality vs. Other Biobehavioural Traits: A Look at Neurochemical Biomarkers for Their Differentiation. Front Psychol 2021; 12:781631. [PMID: 34987450 PMCID: PMC8720768 DOI: 10.3389/fpsyg.2021.781631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
This review highlights the differential contributions of multiple neurochemical systems to temperament traits related and those that are unrelated to emotionality, even though these systems have a significant overlap. The difference in neurochemical biomarkers of these traits is analysed from the perspective of the neurochemical model, Functional Ensemble of Temperament (FET) that uses multi-marker and constructivism principles. Special attention is given to a differential contribution of hypothalamic-pituitary hormones and opioid neuropeptides implicated in both emotional and non-emotional regulation. The review highlights the role of the mu-opioid receptor system in dispositional emotional valence and the role of the kappa-opioid system in dispositional perceptual and behavioural alertness. These opioid receptor (OR) systems, microbiota and cytokines are produced in three neuroanatomically distinct complexes in the brain and the body, which all together integrate dispositional emotionality. In contrast, hormones could be seen as neurochemical biomarkers of non-emotional aspects of behavioural regulation related to the construction of behaviour in fast-changing and current situations. As examples of the role of hormones, the review summarised their contribution to temperament traits of Sensation Seeking (SS) and Empathy (EMP), which FET considers as non-emotionality traits related to behavioural orientation. SS is presented here as based on (higher) testosterone (fluctuating), adrenaline and (low) cortisol systems, and EMP, as based on (higher) oxytocin, reciprocally coupled with vasopressin and (lower) testosterone. Due to the involvement of gonadal hormones, there are sex and age differences in these traits that could be explained by evolutionary theory. There are, therefore, specific neurochemical biomarkers differentiating (OR-based) dispositional emotionality and (hormones-based) body's regulation in fast-changing events. Here we propose to consider dispositional emotionality associated with OR systems as emotionality in a true sense, whereas to consider hormonal ensembles regulating SS and EMP as systems of behavioural orientation and not emotionality.
Collapse
Affiliation(s)
- Irina N. Trofimova
- Laboratory of Collective Intelligence, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
29
|
Peters R, Breitner J, James S, Jicha GA, Meyer P, Richards M, Smith AD, Yassine HN, Abner E, Hainsworth AH, Kehoe PG, Beckett N, Weber C, Anderson C, Anstey KJ, Dodge HH. Dementia risk reduction: why haven't the pharmacological risk reduction trials worked? An in-depth exploration of seven established risk factors. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12202. [PMID: 34934803 PMCID: PMC8655351 DOI: 10.1002/trc2.12202] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/03/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022]
Abstract
Identifying the leading health and lifestyle factors for the risk of incident dementia and Alzheimer's disease has yet to translate to risk reduction. To understand why, we examined the discrepancies between observational and clinical trial evidence for seven modifiable risk factors: type 2 diabetes, dyslipidemia, hypertension, estrogens, inflammation, omega-3 fatty acids, and hyperhomocysteinemia. Sample heterogeneity and paucity of intervention details (dose, timing, formulation) were common themes. Epidemiological evidence is more mature for some interventions (eg, non-steroidal anti-inflammatory drugs [NSAIDs]) than others. Trial data are promising for anti-hypertensives and B vitamin supplementation. Taken together, these risk factors highlight a future need for more targeted sample selection in clinical trials, a better understanding of interventions, and deeper analysis of existing data.
Collapse
Affiliation(s)
- Ruth Peters
- Neuroscience ResearchSydneyNew South WalesAustralia
- Department of Psychology University of New South WalesSydneyNew South WalesAustralia
| | - John Breitner
- Douglas Hospital Research Center and McGill UniversityQuebecCanada
| | - Sarah James
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | | | - Pierre‐Francois Meyer
- Center for Studies on the Prevention of Alzheimer's Disease (PREVENT‐AD)VerdunQuebecCanada
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - A. David Smith
- OPTIMADepartment of PharmacologyUniversity of OxfordOxfordUK
| | - Hussein N. Yassine
- Departments of Medicine and NeurologyUniversity of Southern CaliforniaCaliforniaUSA
| | - Erin Abner
- University of KentuckyLexingtonKentuckyUSA
| | - Atticus H. Hainsworth
- Molecular and Clinical Sciences Research InstituteSt GeorgesUniversity of LondonLondonUK
- Department of NeurologySt George's HospitalLondonUK
| | | | | | | | - Craig Anderson
- The George Institute for Global HealthSydneyNew South WalesAustralia
| | - Kaarin J. Anstey
- Neuroscience ResearchSydneyNew South WalesAustralia
- Department of Psychology University of New South WalesSydneyNew South WalesAustralia
| | | |
Collapse
|
30
|
Joue G, Chakroun K, Bayer J, Gläscher J, Zhang L, Fuss J, Hennies N, Sommer T. Sex Differences and Exogenous Estrogen Influence Learning and Brain Responses to Prediction Errors. Cereb Cortex 2021; 32:2022-2036. [PMID: 34649284 DOI: 10.1093/cercor/bhab334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/19/2021] [Accepted: 08/22/2021] [Indexed: 11/14/2022] Open
Abstract
Animal studies show marked sex differences as well as effects of estrogen (E2) in the mesocorticolimbic dopaminergic (DA) pathways, which play a critical role in reward processing and reinforcement learning and are also implicated in drug addiction. In this computational pharmacological fMRI study, we investigate the effects of both factors, sex and estrogen, on reinforcement learning and the dopaminergic system in humans; 67 male and 64 naturally cycling female volunteers, the latter in their low-hormone phase, were randomly assigned, double-blind, to take E2 or placebo. They completed a reinforcement learning task in the MRI scanner for which we have previously shown reward prediction error (RPE)-related activity to be dopaminergic. We found RPE-related brain activity to be enhanced in women compared with men and to a greater extent when E2 levels were elevated in both sexes. However, both factors, female sex and E2, slowed adaptation to RPEs (smaller learning rate). This discrepancy of larger RPE-related activity yet smaller learning rates can be explained by organizational sex differences and activational effects of circulating E2, which both affect DA release differently to DA receptor binding capacities.
Collapse
Affiliation(s)
- Gina Joue
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Karima Chakroun
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Janine Bayer
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jan Gläscher
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lei Zhang
- Social, Cognitive and Affective Neuroscience Unit, Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, 1010 Vienna, Austria
| | - Johannes Fuss
- Institute for Sex Research, Sexual Medicine and Forensic Psychiatry, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nora Hennies
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tobias Sommer
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
31
|
Tsutsumi T, Imai S, Kashiwagi H, Sato Y, Sugawara M, Takekuma Y. Investigation of the risk factors of vomiting during linezolid therapy: a retrospective observational study. Eur J Clin Pharmacol 2021; 78:279-286. [PMID: 34581841 DOI: 10.1007/s00228-021-03221-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/12/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE Some clinical studies have reported the occurrence of nausea and vomiting with linezolid (LZD) administration. However, no studies have evaluated nausea and vomiting as primary endpoints. In a previous study, we noted a possible relationship between LZD and vomiting, but risk factors were not identified. Therefore, the aim of the present study was to identify them. METHODS Patients who received LZD 600 mg twice daily at Hokkaido University Hospital from September 2008 to April 2019 were enrolled in this retrospective observational study. Patient characteristics, concomitant medications, laboratory data, and the occurrence of vomiting were obtained from electronic medical records. Logistic regression analysis was performed to identify risk factors for vomiting, including age, sex, body weight, concomitant medications, and surgeries. RESULTS A total of 496 patients were included in this study, of which 90 experienced vomiting. By multivariate logistic regression analysis, female sex (adjusted odds ratio [aOR], 2.69; 95% confidence interval [CI], 1.62-4.47), ≥ 10 days of LZD administration (aOR, 2.57; CI, 1.46-4.50), and hyponatraemia (aOR, 2.96; CI, 1.72-5.10) were identified as independent risk factors for vomiting; administration of serotonergic agents (aOR, 0.23; CI, 0.07-0.82) was negatively associated. CONCLUSIONS This study is the first to successfully identify risk factors for LZD-induced vomiting. Careful monitoring of patients with these risk factors may lead to safer and sustainable LZD administration.
Collapse
Affiliation(s)
- Takezo Tsutsumi
- Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido, Japan.,Department of Pharmacy, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Shungo Imai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hitoshi Kashiwagi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuki Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mitsuru Sugawara
- Department of Pharmacy, Hokkaido University Hospital, Sapporo, Hokkaido, Japan.,Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan.,Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Kita 12-jo Nishi 6-chome, Kita-ku, Sapporo, 060-0812, Japan
| | - Yoh Takekuma
- Department of Pharmacy, Hokkaido University Hospital, Sapporo, Hokkaido, Japan.
| |
Collapse
|
32
|
Gougoulaki M, Lewis G, Nutt DJ, Peters TJ, Wiles NJ, Lewis G. Sex differences in depressive symptoms and tolerability after treatment with selective serotonin reuptake inhibitor antidepressants: Secondary analyses of the GENPOD trial. J Psychopharmacol 2021; 35:919-927. [PMID: 33637001 PMCID: PMC8358567 DOI: 10.1177/0269881120986417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Differences in serotonergic neurotransmission could lead to sex differences in depressive symptoms and tolerability after treatment with selective serotonin reuptake inhibitors (SSRIs). AIMS We investigated whether women have greater reductions in depressive symptoms than men after treatment with an SSRI (citalopram) compared with a noradrenaline reuptake inhibitor (reboxetine) control, and after antidepressant treatment irrespective of class. We also investigated tolerability and the influence of menopausal status. METHODS Secondary analyses of the GENPOD (GENetic and clinical Predictors Of treatment response in Depression) trial. Six hundred and one people with depression were recruited from UK primary care and randomized to citalopram or reboxetine. Beck Depression Inventory (BDI-II) score at 6 weeks was the primary outcome. Secondary outcomes included BDI-II score at 12 weeks, and physical symptoms and treatment discontinuation. We calculated main effects and interaction terms using linear and logistic regression models. RESULTS There was no evidence that women experienced greater reductions in depressive symptoms than men when treated with citalopram compared with reboxetine. We also found no evidence of sex differences at six or 12 weeks (irrespective of antidepressant class): men scored -0.31 (95% confidence interval (CI) -2.23 to 1.62) BDI-II points lower than women at six weeks and -0.44 (95% CI -2.62 to 1.74) points lower at 12 weeks. There was no evidence of sex differences in physical symptoms or treatment discontinuation and no evidence for an influence of menopausal status. CONCLUSION Citalopram was not more effective in women compared with men and there was no difference in tolerability. Women and men had similar prognosis after SSRI treatment and similar prognosis regardless of antidepressant class. Findings were unaltered by menopausal status.
Collapse
Affiliation(s)
| | - Glyn Lewis
- Division of Psychiatry, Faculty of Brain Sciences, University College London, London, UK
| | - David J Nutt
- Department of Psychiatry, Division of Brain Science, Imperial College, London, UK
| | - Tim J Peters
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicola J Wiles
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gemma Lewis
- Division of Psychiatry, Faculty of Brain Sciences, University College London, London, UK,Gemma Lewis, Division of Psychiatry, UCL, 6th Floor, Maple House, 149 Tottenham Court Road, London, W1T 7NF, UK.
| |
Collapse
|
33
|
Petersen N, Rapkin AJ, Okita K, Kinney KR, Mizuno T, Mandelkern MA, London ED. Striatal dopamine D 2-type receptor availability and peripheral 17β-estradiol. Mol Psychiatry 2021; 26:2038-2047. [PMID: 33420479 PMCID: PMC9196143 DOI: 10.1038/s41380-020-01000-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/16/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022]
Abstract
Research using rodent models has established a relationship between the steroid hormone estrogen and dopamine function, by revealing changes throughout the estrous cycle and by directly manipulating neuroendocrine signaling through ovariectomy and administration of estrogen. However, a direct link between estrogen levels and dopamine signaling had not been established in humans. The goal of this study, therefore, was to assess the relationship between circulating 17β-estradiol and dopamine signaling in the human brain by testing for a relationship between two proxies for these variables: peripheral 17β-estradiol and striatal dopamine D2-type receptor availability, measured with [18F]fallypride and positron emission tomography (PET). Sixteen (23-45 years of age) women were tested on 2 days of the menstrual cycle estimated prospectively to occur during (a) the early follicular phase, when estrogen levels are near their nadir, and (b) the periovulatory phase, when estrogen levels peak. PET scans with [18F]fallypride were performed on these 2 days, and serum 17β-estradiol was measured using radioimmunoassay. Dopamine D2-type receptor availability did not differ significantly in the whole striatum or the caudate, putamen, or accumbens subregions during the high-estrogen vs. the low-estrogen phases of the menstrual cycle. We conclude that circulating estrogen levels do not affect dopamine D2-type receptor availability in the human striatum although other indices of dopaminergic function may be affected.
Collapse
Affiliation(s)
- Nicole Petersen
- Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, 90095, USA. .,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, 90095, USA. .,Veterans Administration of Greater Los Angeles Health System, Los Angeles, CA, 90073, USA.
| | - Andrea J. Rapkin
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Kyoji Okita
- Department of Clinical Neuroimaging, Integrative Brain Imaging Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan,Department of Drug Dependence, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Kaitlin R. Kinney
- Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA 90095, USA,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA 90095, USA,Veterans Administration of Greater Los Angeles Health System, Los Angeles, CA 90073, USA
| | - Tomi Mizuno
- Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA 90095, USA,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA 90095, USA
| | - Mark A. Mandelkern
- Veterans Administration of Greater Los Angeles Health System, Los Angeles, CA 90073, USA,Department of Physics, University of California at Irvine, Irvine, CA 92697, USA
| | - Edythe D. London
- Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA 90095, USA,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA 90095, USA,Veterans Administration of Greater Los Angeles Health System, Los Angeles, CA 90073, USA,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
34
|
Sezer E, Köşger F, Altınöz AE, Yiğitaslan S. Relationship Between Gonadal Hormone Levels and Symptom Severity in Female Patients With Schizophrenia. ALPHA PSYCHIATRY 2021; 22:130-135. [PMID: 36425447 PMCID: PMC9590640 DOI: 10.5455/apd.119468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/23/2020] [Indexed: 06/16/2023]
Abstract
OBJECTIVE It is thought that sex-specific differences in schizophrenia may be associated with gonadal hormones, especially estrogen. This study aimed to investigate the relationship between follicle stimulating hormone (FSH), luteinizing hormone (LH), prolactin, estradiol, and progesterone serum levels and symptom severity during the menstrual cycle in female patients with schizophrenia. METHODS Serum samples were taken in the follicular and periovulatory phases from 32 female patients with schizophrenia; and FSH, LH, prolactin, estradiol, and progesterone levels were performed. Simultaneously, the patients were administered positive and negative symptom scale (PANSS), Calgary depression scale for schizophrenia (CDSS), and Hamilton anxiety rating scale (HAM-A). RESULTS PANSS (z = -2.52, P < .001), HAM-A (z = -3.60, P < .001), and CDSS (z = -2.52, P = .012) scores were lower in the periovulatory phase than in the follicular phase. Negative correlations between FSH and PANSS positive symptom subscale (r = -0.393, P = .035), and between prolactin and PANSS total score (r = -0.406, P = .029) were detected. CONCLUSION Hypoestrogenism should be studied more in patients with schizophrenia. Studies with large samples evaluating FSH, LH, prolactin, and progesterone together with estrogen are needed to be able to safely use gonadal hormones, which may be related to schizophrenia symptom severity, especially in patients who do not respond adequately to treatment.
Collapse
Affiliation(s)
- Erdi Sezer
- Clinic of Psychiatry, Health Sciences University İzmir Tepecik Training and Research Hospital,
İzmir,
Turkey
| | - Ferdi Köşger
- Department of Psychiatry, Eskişehir Osmangazi University School of Medicine,
Eskişehir,
Turkey
| | - Ali Ercan Altınöz
- Department of Psychiatry, Eskişehir Osmangazi University School of Medicine,
Eskişehir,
Turkey
| | - Semra Yiğitaslan
- Department of Medical Pharmacology, Eskişehir Osmangazi University School of Medicine,
Eskişehir,
Turkey
| |
Collapse
|
35
|
Dean B, Gogos A. The impact of ovariectomy and chronic estrogen treatment on gene expression in the rat cortex: Implications for psychiatric disorders. Psychoneuroendocrinology 2021; 127:105192. [PMID: 33730612 DOI: 10.1016/j.psyneuen.2021.105192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 01/01/2023]
Abstract
Estrogens, via estrogen-mediated changes in CNS function, have been suggested to be beneficial in the treatment of several psychiatric disorders. Few studies have used transcriptomic technologies to determine the effect of estrogen on gene expression in the CNS. Thus, we aimed to examine the impact of ovariectomy (the removal of all ovarian hormones) and estrogen replacement on rat frontal cortical gene expression. We used the Agilent SurePrint G3 Gene Expression Rat Array to measure levels of RNA in intact (cycling) female rats and in ovariectomized rats that were, or were not, given 17β-estradiol in implants for 4 weeks. Compared to untreated ovariectomized rats, intact rats (effect of ovarian hormones; comparison 1) and rats receiving 17β-estradiol replacement (estrogen-specific effects; comparison 2) showed significant changes in cortical gene expression (58 and 36 genes, respectively). These changes in gene expression would be expected to affect pathways that regulate neurotransmitters, glutathione and sphingolipids; pathways known to be implicated in the pathophysiologies of psychiatric disorders. When we compared the levels of gene expression in the two comparisons that had a significance of p < 0.01 independent of magnitude of change, there was a strong correlation between fold changes in gene expression for 127 genes. We posit that this correlation is due to the level of expression of these genes being strongly influenced by both cycling and replacement estrogen. Further exploration of ovarian hormone- and estrogen-sensitive gene expression may provide new insight into the aetiology of aspects of psychiatric disorders that show sex differences.
Collapse
Affiliation(s)
- Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
36
|
Sepulveda M, Manning EE, Gogos A, Hale M, van den Buuse M. Long-term effects of young-adult methamphetamine on dorsal raphe serotonin systems in mice: Role of brain-derived neurotrophic factor. Brain Res 2021; 1762:147428. [PMID: 33737066 DOI: 10.1016/j.brainres.2021.147428] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/15/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023]
Abstract
To assess the long-term effects of chronic adolescent methamphetamine (METH) treatment on the serotonin system in the brain, we used serotonin-1A receptor (5-HT1A) and serotonin transporter (SERT) autoradiography, and quantitative tryptophan-hydroxylase 2 (TPH2) immunohistochemistry in the raphe nuclei of mice. Because of the modulatory role of brain-derived neurotrophic factor (BDNF) on the serotonin system and the effects of METH, we included both BDNF heterozygous (HET) mice and wildtype (WT) controls. Male and female mice of both genotypes were treated with an escalating METH dose regimen from the age of 6-9 weeks. At least two weeks later, acute locomotor hyperactivity induced by a 5 mg/kg D-amphetamine challenge was significantly enhanced in METH-pretreated mice, showing long-term sensitisation. METH pretreatment caused a small, but significant decrease of 5-HT1A receptor binding in the dorsal raphe nucleus (DRN) of males independent of genotype, but there were no changes in the median raphe nucleus (MRN) or in SERT binding density. METH treatment reduced the number of TPH2 positive cells in ventral subregions of the rostral and medial DRN independent of genotype. METH treatment selectively reduced DRN cell counts in BDNF HET mice compared to wildtype mice in medial and caudal ventrolateral subregions previously associated with panic-like behaviour. The data increase our understanding of the long-term and selective effects of METH on brain serotonin systems. These findings could be relevant for some of the psychosis-like symptoms associated with long-term METH use.
Collapse
Affiliation(s)
- Mauricio Sepulveda
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Elizabeth E Manning
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Andrea Gogos
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Matthew Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia.
| |
Collapse
|
37
|
Shin S, Nam HY, Lee MJ, Pak K, Kim K, Kim IJ. Effect of sex on aging-related decline of dopamine transporter in healthy subjects. Ann Nucl Med 2021; 35:76-82. [PMID: 33052524 DOI: 10.1007/s12149-020-01538-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Aging decreases dopamine transporter (DAT) availability of striatum both in humans and rodents. We aimed to investigate the relationship of DAT availabilities from ventral striatum, caudate nucleus, and putamen with aging in healthy subjects. METHODS 123I-FP-CIT single photon emission computed tomography (SPECT) was performed in all subjects. Specific binding of 123I-FP-CIT regarding DAT was calculated using a volume-of-interest-based analysis of ventral striatum, caudate nucleus, putamen. The cerebellum was chosen as a reference region. Specific binding ratios (SBRs) were calculated as follows: SBR = (target- cerebellum)/cerebellum. RESULTS A total of 166 healthy subjects (109 males and 57 females) were included in this study. SBRs of ventral striatum, caudate nucleus, and putamen were negatively correlated with age. In young males, SBRs of ventral striatum and putamen were not correlated with aging. However, SBRs of caudate nucleus showed the trend toward negative correlation with age in the young group. In old males, SBR of caudate nucleus was negatively correlated with age and SBR of ventral striatum showed a trend toward negative correlation with age. Slopes of regression lines were not significantly different according to age groups in ventral striatum, caudate nucleus, or putamen. SBRs of ventral striatum, caudate nucleus, and putamen were negatively correlated with age in young females, but not in old females. Interestingly, slopes of regression line were significantly different between young and old females in ventral striatum, caudate nucleus, and putamen. CONCLUSIONS We have shown that slopes of regression lines of DAT availabilities and age were significantly different between young and old subjects in females, not in males. Therefore, sex has an impact on aging-related decline of striatal DAT availability.
Collapse
Affiliation(s)
- Seunghyeon Shin
- Department of Nuclear Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Hyun-Yeol Nam
- Department of Nuclear Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea.
| | - Myung Jun Lee
- Departments of Neurology and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| | - Kyoungjune Pak
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| | - Keunyoung Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - In Joo Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
38
|
Hwang WJ, Lee TY, Kim NS, Kwon JS. The Role of Estrogen Receptors and Their Signaling across Psychiatric Disorders. Int J Mol Sci 2020; 22:ijms22010373. [PMID: 33396472 PMCID: PMC7794990 DOI: 10.3390/ijms22010373] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests estrogen and estrogen signaling pathway disturbances across psychiatric disorders. Estrogens are not only crucial in sexual maturation and reproduction but are also highly involved in a wide range of brain functions, such as cognition, memory, neurodevelopment, and neuroplasticity. To add more, the recent findings of its neuroprotective and anti-inflammatory effects have grown interested in investigating its potential therapeutic use to psychiatric disorders. In this review, we analyze the emerging literature on estrogen receptors and psychiatric disorders in cellular, preclinical, and clinical studies. Specifically, we discuss the contribution of estrogen receptor and estrogen signaling to cognition and neuroprotection via mediating multiple neural systems, such as dopaminergic, serotonergic, and glutamatergic systems. Then, we assess their disruptions and their potential implications for pathophysiologies in psychiatric disorders. Further, in this review, current treatment strategies involving estrogen and estrogen signaling are evaluated to suggest a future direction in identifying novel treatment strategies in psychiatric disorders.
Collapse
Affiliation(s)
- Wu Jeong Hwang
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea; (W.J.H.); (J.S.K.)
| | - Tae Young Lee
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan 50612, Korea;
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
- Correspondence: ; Tel.: +82-55-360-2468
| | - Nahrie Suk Kim
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan 50612, Korea;
| | - Jun Soo Kwon
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea; (W.J.H.); (J.S.K.)
- Department of Psychiatry, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
39
|
Nadalin S, Flego V, Pavlić SD, Volarić D, Radojčić Badovinac A, Kapović M, Ristić S. Association between the ACE-I/D polymorphism and nicotine dependence amongst patients with lung cancer. Biomed Rep 2020; 13:58. [PMID: 33123372 DOI: 10.3892/br.2020.1365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/14/2020] [Indexed: 11/06/2022] Open
Abstract
The biologically active peptide angiotensin II is cleaved from angiotensinogen by the renin and the angiotensin-converting enzyme (ACE), an enzymatic cascade known as the renin-angiotensin system (RAS). RAS may be important in the etiology of nicotine dependence by influencing dopaminergic signaling. In the present study, the association between an insertion/deletion (I/D) polymorphism of ACE and nicotine dependence amongst patients with lung cancer was assessed. To date, several studies have shown the relevance of this polymorphic variant in both nicotine dependence and lung cancer. However, the present study is the first to address the potential role of the ACE-I/D polymorphism in nicotine dependence among patients with lung cancer. Genotyping was performed in 305 patients with lung cancer (males/females, 214/91). Significantly more male smokers had the ACE-I allele compared with male non-smokers (44.9 vs. 20.0%; P<0.05). The risk of smoking was ~5-fold higher for males with the ACE-I allele (ACE-II homozygous and ACE-ID heterozygous) vs. ACE-DD homozygous (odds ratio, 5.47; 95% confidence interval, 1.4-21.9; P=0.016). The pack-year smoking history in a subgroup of females with squamous cell carcinoma carrying the ACE-I allele was significantly lower compared with ACE-DD (37.1±14.1 vs. 57.0±29.1; F=4.5; P=0.046). The ACE-I/D polymorphism accounted for 17.6% of the smoking severity in this patient group (β, -0.42; multiple R2 change, 0.176; P=0.046). These results suggest that the ACE-I/D polymorphism contributes to the risk of nicotine dependence and smoking severity in lung cancer patients in a sex-specific manner.
Collapse
Affiliation(s)
- Sergej Nadalin
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Veljko Flego
- Department of Pulmonology, Clinical Hospital Center Rijeka, University of Rijeka, 51000 Rijeka, Croatia
| | - Sanja Dević Pavlić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Darian Volarić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Anđelka Radojčić Badovinac
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia.,Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Miljenko Kapović
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Smiljana Ristić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
40
|
Wong FY, Gogos A, Hale N, Ingelse SA, Brew N, Shepherd KL, van den Buuse M, Walker DW. Impact of hypoxia-ischemia and dopamine treatment on dopamine receptor binding density in the preterm fetal sheep brain. J Appl Physiol (1985) 2020; 129:1431-1438. [PMID: 33054660 DOI: 10.1152/japplphysiol.00677.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Dopamine is often used to treat hypotension in preterm infants who are at risk of hypoxic-ischemic (HI) brain injury due to cerebral hypoperfusion and impaired autoregulation. There is evidence that systemically administered dopamine crosses the preterm blood-brain barrier. However, the effects of exogenous dopamine and cerebral HI on dopaminergic signaling in the immature brain are unknown. We determined the effect of HI and dopamine on D1 and D2 receptor binding and expressions of dopamine transporter (DAT) and tyrosine hydroxylase (TH) in the striatum of the preterm fetal sheep. Fetal sheep (99 days of gestation, term = 147days) were unoperated controls (n = 6) or exposed to severe HI using umbilical cord occlusion and saline infusion (UCO + saline, n = 8) or to HI with dopamine infusion (UCO + dopamine, 10 µg/kg/min, n = 7) for 74 h. D1 and D2 receptor densities were measured by autoradiography in vitro. DAT, TH, and cell death were measured using immunohistochemistry. HI resulted in cell death in the caudate nucleus and putamen, and dopamine infusion started before HI did not exacerbate or ameliorate these effects. HI led to reduced D1 and D2 receptor densities in the caudate nucleus and reduction in DAT protein expression in the caudate and putamen. Fetal brains exposed to dopamine in addition to HI were not different from those exposed to HI alone in these changes in dopaminergic parameters. We conclude that dopamine infusion does not alter the striatal cell death or the reductions in D1 and D2 receptor densities and DAT protein expression induced by HI in the preterm brain.NEW & NOTEWORTHY This is the first study on the effects of hypoxia-ischemia and dopamine treatment on the dopaminergic pathway in the preterm brain. In the striatum of fetal sheep (equivalent to ∼26-28 wk of human gestation), we demonstrate that hypoxia-ischemia leads to cell death, reduces D1 and D2 receptors, and reduces dopamine transporter. Intravenous dopamine infusion at clinical dosage used in preterm human infants does not alter the striatal cell death, D1 and D2 receptor density levels, and DAT protein expressions after hypoxia-ischemia in the preterm brain.
Collapse
Affiliation(s)
- F Y Wong
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.,Department of Paediatrics, Monash University, Melbourne, Australia.,Monash Newborn, Monash Medical Centre, Melbourne, Australia
| | - A Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - N Hale
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | - S A Ingelse
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | - N Brew
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | - K L Shepherd
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.,Department of Paediatrics, Monash University, Melbourne, Australia
| | - M van den Buuse
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia.,School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - D W Walker
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.,School of Health & Biomedical Sciences, RMIT University, Melbourne, Australia
| |
Collapse
|
41
|
Sbisa A, Kusljic S, Zethoven D, van den Buuse M, Gogos A. The effect of 17β-estradiol on maternal immune activation-induced changes in prepulse inhibition and dopamine receptor and transporter binding in female rats. Schizophr Res 2020; 223:249-257. [PMID: 32878698 DOI: 10.1016/j.schres.2020.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/09/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022]
Abstract
Maternal immune activation (MIA) during pregnancy is associated with an increased risk of development of schizophrenia in later life. 17β-estradiol treatment may improve schizophrenia symptoms, but little is known about its efficacy on MIA-induced psychosis-like behavioural deficits in animals. Therefore, in this study we used the poly(I:C) neurodevelopmental model of schizophrenia to examine whether MIA-induced psychosis-like behavioural and neurochemical changes can be attenuated by chronic treatment (2-6 weeks) with 17β-estradiol. Pregnant rats were treated with saline or the viral mimetic, poly(I:C), on gestational day 15 and adult female offspring were tested for changes in prepulse inhibition (PPI) and density of dopamine D1 and D2 receptors and dopamine transporters in the forebrain compared to control offspring. Poly(I:C)-treated offspring exhibited significantly disrupted PPI, an effect which was reversed by chronic treatment with 17β-estradiol. In control offspring, but not poly(I:C) offspring, PPI was significantly reduced by acute treatment with either the dopamine D1/D2 receptor agonist, apomorphine, or dopamine releaser, methamphetamine. 17β-estradiol restored the effect of apomorphine, but not methamphetamine, on PPI in poly(I:C) offspring. There was a strong trend for a dopamine D2 receptor binding density increase in the nucleus accumbens core region in poly(I:C) offspring, and this was reversed by chronic 17β-estradiol treatment. No changes were found in the nucleus accumbens shell, caudate putamen or frontal cortex or in the density of dopamine D1 receptors or transporters. These findings suggest that 17β-estradiol may improve some symptoms of schizophrenia, an effect that may be mediated by selective changes in dopamine D2 receptor density.
Collapse
Affiliation(s)
- Alyssa Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - Snezana Kusljic
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Nursing, University of Melbourne, Parkville, VIC, Australia
| | - Damon Zethoven
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia; Department of Pharmacology, University of Melbourne, Parkville, VIC, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
42
|
Barbosa-Méndez S, Osorio-Santiago KL, Salazar-Juárez A. Mirtazapine reduces the expression of cocaine-induced locomotor sensitization in male and female Wistar rats. Horm Behav 2020; 125:104817. [PMID: 32682854 DOI: 10.1016/j.yhbeh.2020.104817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/19/2020] [Accepted: 07/08/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Epidemiological studies have described that women are more vulnerable to the reinforcing effects of cocaine. In animals, the findings are similar: female rats show higher levels of cocaine self-administration and increased cocaine-induced locomotor activity. In contrast, women with depression respond better to treatment with antidepressants, however their therapeutic response to tetracyclic antidepressants is lower. Several studies have shown that mirtazapine-a tetracyclic antidepressant-decreases the behavioral effects of cocaine in male rats. The objective of this study was to evaluate the efficacy of daily dosing of mirtazapine on cocaine-induced locomotor activity and sensitization in naive female rats compared to male rats. METHODS Male and female Wistar rats were daily dosed with 10 mg/kg of cocaine. During extinction, cocaine was withdrawn and the groups received daily mirtazapine (30 mg/kg, i.p.) or saline. Tamoxifen was administered during the antagonism phase. After each administration, locomotor activity for each animal was recorded for 30 min in transparent Plexiglass activity chambers. RESULTS In this study, a higher cocaine locomotor response was found in females than in males and the mirtazapine was equally effective in decreasing cocaine-induced locomotor activity and the expression of locomotor sensitization in male and female rats. In addition, co-administration of mirtazapine and tamoxifen enhanced the efficacy of mirtazapine in female rats. CONCLUSION The results suggest that mirtazapine may be considered an effective therapeutic option for the treatment of cocaine use disorder in men and women.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, Ciudad de México 14370. Mexico
| | - Katya Lorena Osorio-Santiago
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, Ciudad de México 14370. Mexico
| | - Alberto Salazar-Juárez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, Ciudad de México 14370. Mexico.
| |
Collapse
|
43
|
Proaño SB, Meitzen J. Estradiol decreases medium spiny neuron excitability in female rat nucleus accumbens core. J Neurophysiol 2020; 123:2465-2475. [PMID: 32432511 DOI: 10.1152/jn.00210.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The menstrual cycle in humans and its analogous cycle in rodents, the estrous cycle, modulate brain function and behavior. Both cycles are characterized by the cyclical fluctuation of ovarian hormones including estrogens such as estradiol. Estradiol induces cycle- and sex-dependent differences in the phenotype and incidence of many behaviors, including those related to reward and motivation. The nucleus accumbens core (AcbC), a limbic and premotor system nexus region, directly regulates these behaviors. We previously showed that the estrous cycle modulates intrinsic excitability and excitatory synapse properties of medium spiny neurons (MSNs) in the AcbC. The identity of the underlying hormone mechanism is unknown, with estradiol being a prime candidate. The present study tests the hypothesis that estradiol induces estrous cycle-relevant differences in MSN electrophysiology. To accomplish this goal, a time- and dose-dependent estradiol replacement paradigm designed to simulate the rise of circulating estradiol levels across the estrous cycle was employed in ovariectomized adult female rats as well as a vehicle control group. Estradiol replacement decreased MSN excitability by modulating properties such as resting membrane potential, input resistance in both the linear and rectified ranges, and rheobase compared with vehicle-treated females. These differences in MSN excitability mimic those previously described regarding estrous cycle effects on MSN electrophysiology. Excitatory synapse properties were not modulated in response to this estradiol replacement paradigm. These data are the first to demonstrate that an estrous cycle-relevant estradiol exposure modulates MSN electrophysiology, providing evidence of the fundamental neuroendocrine mechanisms regulating the AcbC.NEW & NOTEWORTHY The present study shows, for the first time, that an estrous cycle-relevant estradiol exposure modulates nucleus accumbens neuron excitability. This evidence provides insight into the neuroendocrine mechanisms by which estradiol cyclically alters neuron properties during the estrous cycle. Overall, these data emphasize the significant influence of hormone action in the brain and especially individual neuron physiology.
Collapse
Affiliation(s)
- Stephanie B Proaño
- Graduate Program in Biology, North Carolina State University, Raleigh, North Carolina.,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - John Meitzen
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
44
|
Woodcock EA, Zakiniaeiz Y, Morris ED, Cosgrove KP. Sex and the dopaminergic system: Insights from addiction studies. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:141-165. [PMID: 33008522 PMCID: PMC11267480 DOI: 10.1016/b978-0-444-64123-6.00011-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Sex differences are present in psychiatric disorders associated with disrupted dopamine function, and thus, sex differences in dopamine neurobiology may underlie these clinical disparities. In this chapter, we review sex differences in the dopaminergic system with a focus on substance use disorders, especially tobacco smoking, as our exemplar disorder. This chapter is organized into five sections describing sex differences in the dopaminergic system: (1) neurobiology, (2) role of sex hormones, (3) genetic underpinnings, (4) cognitive function, and (5) influence on addiction. In each section, we provide an overview of the topic area, summarize sex differences identified to date, highlight addiction research, especially clinical neuroimaging studies, and suggest avenues for future research.
Collapse
Affiliation(s)
- Eric A Woodcock
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States; Yale Positron Emission Tomography (PET) Center, Yale University, New Haven, CT, United States
| | - Yasmin Zakiniaeiz
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; Yale Positron Emission Tomography (PET) Center, Yale University, New Haven, CT, United States
| | - Evan D Morris
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States; Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Invicro, LLC, New Haven, CT, United States
| | - Kelly P Cosgrove
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States; Yale Positron Emission Tomography (PET) Center, Yale University, New Haven, CT, United States.
| |
Collapse
|
45
|
Nadalin S, Rebić J, Šendula Jengić V, Peitl V, Karlović D, Buretić-Tomljanović A. Association between PLA2G6 gene polymorphism for calcium-independent phospholipase A2 and nicotine dependence among males with schizophrenia. Prostaglandins Leukot Essent Fatty Acids 2019; 148:9-15. [PMID: 31492433 DOI: 10.1016/j.plefa.2019.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/03/2019] [Accepted: 07/02/2019] [Indexed: 01/08/2023]
Abstract
We investigated the relationship between the rs10798059 (BanI) and rs4375 polymorphisms in the phospholipase A2 (PLA2)G4A and PLA2G6 genes and the risk of nicotine dependence in 263 Croatian patients with schizophrenia. We also examined whether interactions between these polymorphisms and smoking contributed to schizophrenia onset and Positive and Negative Syndrome Scale (PANSS) psychopathology. We found no significant differences in the distribution of PLA2G4A genotypes and alleles according to smoking status, and no effect of the PLA2G4A genotype-smoking interaction on disease onset or PANSS. The PLA2G6-TT homozygous genotype was significantly overrepresented in male smokers compared to nonsmokers (34.7% vs. 17.1%, p < 0.05). These patients had ∼2.6-fold higher risk of becoming smokers than males with heterozygous PLA2G6-CT and homozygous PLA2G6-CC genotypes. In addition, male smokers without the PLA2G6-C allele (PLA2G6-TT homozygous) experienced earlier onset than nonsmoking homozygous PLA2G6-TT males. Thus, the PLA2G6 polymorphism affected the risk of nicotine dependence in male patients and the PLA2G6 genotype-smoking interaction was linked to the age of disease onset.
Collapse
Affiliation(s)
- Sergej Nadalin
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000, Rijeka, Croatia.
| | - Jelena Rebić
- Psychiatry Clinic, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | | | - Vjekoslav Peitl
- Department of Psychiatry, Sestre Milosrdnice University Hospital Center and Catholic University of Croatia, Zagreb, Croatia
| | - Dalibor Karlović
- Department of Psychiatry, Sestre Milosrdnice University Hospital Center and Catholic University of Croatia, Zagreb, Croatia
| | - Alena Buretić-Tomljanović
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000, Rijeka, Croatia
| |
Collapse
|
46
|
Molina-Jiménez T, Jiménez-Tlapa M, Brianza-Padilla M, Zepeda RC, Hernández-González M, Bonilla-Jaime H. The neonatal treatment with clomipramine decreases sexual motivation and increases estrogen receptors expression in the septum of male rats: Effects of the apomorphine. Pharmacol Biochem Behav 2019; 180:83-91. [DOI: 10.1016/j.pbb.2019.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/20/2019] [Accepted: 03/19/2019] [Indexed: 02/08/2023]
|
47
|
Estrogens and SERMS as adjunctive treatments for schizophrenia. Front Neuroendocrinol 2019; 53:100743. [PMID: 30922675 DOI: 10.1016/j.yfrne.2019.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 03/12/2019] [Accepted: 03/23/2019] [Indexed: 01/25/2023]
Abstract
More than thirty years have passed since sex and gender differences were noted in the age of onset, course and outcomes for schizophrenia. The 'estrogen hypothesis" was coined in the 1990's to describe neuroprotective effects of estrogen. Intervention studies in schizophrenia patients with estradiol and selective estrogen receptor modulators (SERMs) are promising but psychiatrists and other health practitioners do not generally take up this useful adjunctive treatment for their female patients with schizophrenia. The reasons for this are manifold, but overall a cultural shift in the practice of psychiatry is needed to recognise the specific needs of women with schizophrenia and tailor treatments, such as hormone adjuncts to improve the outcomes for this significant population. The two main aims of this article are to review the evidence and theory of estrogen treatments in schizophrenia and to recommend translation of adjunctive estrogen treatment into clinical practice for women with schizophrenia.
Collapse
|
48
|
Lonstein JS. The dynamic serotonin system of the maternal brain. Arch Womens Ment Health 2019; 22:237-243. [PMID: 30032323 PMCID: PMC7001094 DOI: 10.1007/s00737-018-0887-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 06/28/2018] [Indexed: 12/16/2022]
Abstract
Many pregnant and postpartum women worldwide suffer from high anxiety and/or depression, which can have detrimental effects on maternal and infant well-being. The first-line pharmacotherapies for prepartum and postpartum affective disorders continue to be the selective serotonin reuptake inhibitors (SSRIs), despite the lack of large well-controlled studies demonstrating their efficacy in reproducing women and the potential for fetal/neonatal exposure to the drugs. Prepartum or postpartum use of SSRIs or other drugs that modulate the brain's serotonin system is also troubling because very little is known about the typical, let alone the atypical, changes that occur in the female central serotonin system across reproduction. We do know from a handful of studies of women and female laboratory rodents that numerous aspects of the central serotonin system are naturally dynamic across reproduction and are also affected by pregnancy stress (a major predisposing factor for maternal psychopathology). Thus, it should not be assumed that the maternal central serotonin system being targeted by SSRIs is identical to non-parous females or males. More information about the normative and stress-derailed changes in the maternal central serotonin system is essential for understanding how serotonin is involved in the etiology of, and the best use of SSRIs for potentially treating, affective disorders in the pregnant and postpartum populations.
Collapse
Affiliation(s)
- Joseph S. Lonstein
- Department of Psychology & Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI 48824, USA
| |
Collapse
|
49
|
Velásquez VB, Zamorano GA, Martínez-Pinto J, Bonansco C, Jara P, Torres GE, Renard GM, Sotomayor-Zárate R. Programming of Dopaminergic Neurons by Early Exposure to Sex Hormones: Effects on Morphine-Induced Accumbens Dopamine Release, Reward, and Locomotor Behavior in Male and Female Rats. Front Pharmacol 2019; 10:295. [PMID: 30971928 PMCID: PMC6443923 DOI: 10.3389/fphar.2019.00295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/11/2019] [Indexed: 11/13/2022] Open
Abstract
Neonatal programming with sex hormones produces long-term functional changes in various tissues, including the brain. Previously, we demonstrated a higher content of dopamine and an increase in potassium-induced dopamine release in the nucleus accumbens of adult rats exposed to estradiol valerate. On the other hand, sex hormones also affect the opioid system increasing the expression of the μ opioid receptor and β-endorphins. Here, we investigated if neonatal programming with sex hormones alters the response to morphine during adulthood in rats and predispose them to neurochemical, rewarding and behavioral activating effects. We examined the effects of neonatal exposure to a single dose of estradiol valerate or testosterone propionate on morphine-induced (5 mg/kg, i.v.) dopamine release in the nucleus accumbens and morphine-induced (3 mg/kg, s.c.) locomotor activity and conditioned place preference when these rats were adults. Our results showed a significant increase in morphine-induced dopamine release in the nucleus accumbens of rats that were exposed neonatally to estradiol compared with control rats. This effect was correlated with higher place preference and locomotor activity induced by morphine in adult rats neonatally exposed to estradiol valerate. However, the effect of morphine on dopamine release and behaviors was similar in rats treated with testosterone compared to control rats. Additionally, the expression of mu (μ) opioid receptor, dopamine receptor type 1 (D1) and dopamine receptor type 2 (D2) in the nucleus accumbens of adult rats was not different after treatment with sex hormones. Taken together, our results demonstrated an enhancement of pharmacological effects produced by morphine in rats neonatally programmed with estradiol valerate, suggesting that early exposure to sex hormones could represent a vulnerability factor in the development of addiction to opioid drugs such as morphine and heroin in adulthood.
Collapse
Affiliation(s)
| | | | | | - Christian Bonansco
- Laboratorio de Neurofisiología, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Jara
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Gonzalo E Torres
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Georgina M Renard
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | | |
Collapse
|
50
|
Cataldi M, Muscogiuri G, Savastano S, Barrea L, Guida B, Taglialatela M, Colao A. Gender-related issues in the pharmacology of new anti-obesity drugs. Obes Rev 2019; 20:375-384. [PMID: 30589980 DOI: 10.1111/obr.12805] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/01/2018] [Accepted: 10/13/2018] [Indexed: 12/14/2022]
Abstract
Four new medicines-liraglutide, lorcaserin, bupropion/naltrexone, and phentermine/topiramate-have been recently added to the pharmacological arsenal for obesity treatment and could represent important tools to manage this epidemic disease. To achieve satisfactory anti-obesity goals, the use of these new medicines should be optimized and tailored to specific patient subpopulations also by applying dose adjustments if needed. In the present review, we posit that gender could be among the factors influencing the activity of the new obesity drugs both because of pharmacokinetic and pharmacodynamic factors. Although evidence from premarketing clinical studies suggested that no dose adjustment by gender is necessary for any of these new medicines, these studies were not specifically designed to identify gender-related differences. This observation, together with the strong theoretical background supporting the hypothesis of a gender-dimorphic response, strongly call upon an urgent need of new real-life data on gender-related difference in the pharmacology of these new obesity drugs.
Collapse
Affiliation(s)
- Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Giovanna Muscogiuri
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Silvia Savastano
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Luigi Barrea
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Bruna Guida
- Division of Physiology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Annamaria Colao
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| |
Collapse
|