1
|
Le MP, Burggren W, Martinez-Bautista G. Development and sex affect respiratory responses to temperature and dissolved oxygen in the air-breathing fishes Betta splendens and Trichopodus trichopterus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:27. [PMID: 39680326 DOI: 10.1007/s10695-024-01411-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Abstract
Ventilation frequencies of the gills (fG) and the air-breathing organ (fABO) were measured in juveniles and adults of the air-breathing betta (Betta splendens) and the blue gourami (Trichopodus trichopterus) in response to temperature and hypoxia. Ventilatory rates were evaluated after 1 h of exposure to 27 °C (control), 23 and 31 °C (PO2 = 21.0 kPa), after acute temperature changes (ATC) from 23 to 27, and 27 to 31 °C, and under progressive hypoxia (PH; PO2 = ~ 21 to 2.5 kPa). Complex, multi-phased ventilatory alterations were evident across species and experimental groups revealing different stress responses and shock reactions (e.g., changes in temperature sensitivity (Q10) of fG between 1-h exposure and ACT in both species). Female and male gourami showed differences in Q10 over the temperature range 23-31 °C. No such Q10 differences occurred in betta. Juveniles of both species showed higher Q10 for fABO (~ 3.7) than fG (~ 2.2). Adult fish exhibited variable Q10s for fG (~ 1.5 to ~ 4.3) and fABO (~ 0.8 to ~ 15.5) as a function of temperature, suggesting a switch from aquatic towards aerial ventilation in response to thermal stress. During PH, juveniles from both species showed higher fG than adults at all oxygen levels. Females from both species showed higher fG compared with males. Collectively, our results suggest that environmental cues modulate ventilatory responses in both species throughout ontogeny, but the actual responses reflect species-specific differences in natural habitat and ecology. Finally, we strongly suggest assessing physiological differences between male and female fish to avoid masking relevant findings and to facilitate results interpretation.
Collapse
Affiliation(s)
- My Phuong Le
- Department of Agriculture, Bac Lieu University, Bac Lieu, Vietnam
- Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Warren Burggren
- Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | | |
Collapse
|
2
|
Hua X, Hu R, Chen C, Sun J, Feng X, Zhang X. Sex specificity in associations between exposure to a mixture of per and poly-fluoroalkyl substances and anxiety among US adults. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2025:10.1038/s41370-024-00741-3. [PMID: 39775104 DOI: 10.1038/s41370-024-00741-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Exposure to per and poly-fluoroalkyl substances (PFAS) is suggested to interfere with the central nervous system that may affect mental health. Studies on the relationships between exposure to PFAS mixtures and anxiety in humans are rare. This study aimed to evaluate the associations between single and combined exposure to PFAS and anxiety among adults. METHODS Data were extracted from the National Health and Nutrition Examination Survey (NHANES, 2007-2012). Six serum PFAS concentrations were accessed including perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), PFOS (perfluorooctanesulfonic acid), PFHxS (perfluorohexane sulfonate), PFDA (perfluorodecanoic acid), Me-PFOSA-AcOH (2-(N-methylperfluorooctanesulfonamide) acetic acid). The anxiety state was defined through the questionnaire responses of the participants. Weighted logistics regression was used to calculate their odds ratio (OR) and corresponding confidence interval (95% CI) that assessed the relationship between PFAS exposure and anxiety. Moreover, Two different statistical methods including quantile-based g-computation (Qgcomp), and Bayesian kernel machine regression (BKMR) were employed to investigate the overall effects of PFAS mixtures on anxiety. RESULTS The effects of specific PFAS exposure on anxiety varied by sex. In male participants, one-unit increase in PFDA (OR = 0.62; 95%CI: 0.44, 0.88), PFOA (OR = 0.60; 95%CI: 0.41, 0.87), PFNA (OR = 0.68; 95%CI: 0.46, 0.96) concentrations were inversely linked to anxiety. In female participants, a one-unit increase in PFOA (OR = 1.50; 95%CI: 1.05, 2.14) concentration was associated with anxiety. Analysis of Qgcomp demonstrated that PFAS mixtures were negatively associated with anxiety in males (OR = 0.85; 95%CI: 0.74, 0.99), and were positively associated with anxiety in females(OR = 1.16; 95%CI: 1.01, 1.33). Analysis of BKMR suggested that PFAS mixtures were negatively associated with anxiety in the males, while its associations with anxiety were positive in the females. IMPACT Although a growing number of studies have focused on the relationship between PFAS and anxiety, most have been performed based on animal observations rather than human populations, and the combined effects of PFAS mixtures on anxiety have not been evaluated. To address these gaps, this study first explored the associations between individual PFAS and PFAS mixture exposures and anxiety among US adults. Using data from the National Health and Nutrition Examination Survey, we demonstrated that co-exposure to a mixture of PFAS was negatively associated with anxiety in males, and its association was contrary in females.
Collapse
Affiliation(s)
- Xiaoguo Hua
- Office of Medical Insurance Management, The Second Affiliated Hospital, Anhui Medical University, Hefei, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Rui Hu
- Department of Clinical Teaching Management, The First Affiliated Hospital, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Cai Chen
- Department of Emergency, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiangjie Sun
- Department of Health Data Science, School of Health Care Management, Anhui Medical University, Hefei, China
| | - Xiqiu Feng
- Office of Medical Insurance Management, The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Xiujun Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Varodayan FP, Erikson CM, Scroger MV, Roberto M. Noradrenergic Mechanisms and Circuitry of Hyperkatifeia in Alcohol Use Disorder. Biol Psychiatry 2024:S0006-3223(24)01609-3. [PMID: 39304172 DOI: 10.1016/j.biopsych.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Hyperkatifeia, the manifestation of emotional distress or pain, is a conceptual framework gaining traction throughout the alcohol and other substance use fields as an important driver of addiction. It is well known that previous or current negative life experiences can serve as powerful motivators for excessive alcohol consumption and precipitate the development of an alcohol use disorder (AUD). A major hallmark of later stages of AUD is the emergence of hyperkatifeia during withdrawal, which can persist well into protracted abstinence to drive relapse. Given these complex interactions, understanding the specific neuroadaptations that lie at the intersection of hyperkatifeia and AUD can inform ongoing therapeutic development. The monoamine norepinephrine is of particular interest. Noradrenergic dysfunction is implicated in AUD, anxiety, chronic stress, depression, and emotional and physical pain. Importantly, there are key sexual dimorphisms within the noradrenergic system that are thought to differentially impact the development and trajectory of AUD in women and men. In the current review, we discuss past and recent work on noradrenergic influences at each stage of the AUD cycle (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) through the lens of hyperkatifeia. Evidence from these studies support the prioritization of norepinephrine-specific drug development to treat AUD and the identification of AUD subpopulations that may benefit the most from these therapies (e.g., women or people with comorbid chronic pain or anxiety/stress disorders).
Collapse
Affiliation(s)
- Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University, SUNY, Binghamton, New York.
| | - Chloe M Erikson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Marcis V Scroger
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University, SUNY, Binghamton, New York
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California.
| |
Collapse
|
4
|
Mariscal P, Bravo L, Llorca-Torralba M, Razquin J, Miguelez C, Suárez-Pereira I, Berrocoso E. Sexual differences in locus coeruleus neurons and related behavior in C57BL/6J mice. Biol Sex Differ 2023; 14:64. [PMID: 37770907 PMCID: PMC10540344 DOI: 10.1186/s13293-023-00550-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND In addition to social and cultural factors, sex differences in the central nervous system have a critical influence on behavior, although the neurobiology underlying these differences remains unclear. Interestingly, the Locus Coeruleus (LC), a noradrenergic nucleus that exhibits sexual dimorphism, integrates signals that are related to diverse activities, including emotions, cognition and pain. Therefore, we set-out to evaluate sex differences in behaviors related to LC nucleus, and subsequently, to assess the sex differences in LC morphology and function. METHODS Female and male C57BL/6J mice were studied to explore the role of the LC in anxiety, depressive-like behavior, well-being, pain, and learning and memory. We also explored the number of noradrenergic LC cells, their somatodendritic volume, as well as the electrophysiological properties of LC neurons in each sex. RESULTS While both male and female mice displayed similar depressive-like behavior, female mice exhibited more anxiety-related behaviors. Interestingly, females outperformed males in memory tasks that involved distinguishing objects with small differences and they also showed greater thermal pain sensitivity. Immunohistological analysis revealed that females had fewer noradrenergic cells yet they showed a larger dendritic volume than males. Patch clamp electrophysiology studies demonstrated that LC neurons in female mice had a lower capacitance and that they were more excitable than male LC neurons, albeit with similar action potential properties. CONCLUSIONS Overall, this study provides new insights into the sex differences related to LC nucleus and associated behaviors, which may explain the heightened emotional arousal response observed in females.
Collapse
Affiliation(s)
- Patricia Mariscal
- Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Lidia Bravo
- Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain.
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain.
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain.
| | - Meritxell Llorca-Torralba
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
- Neuropsychopharmacology & Psychobiology Research Group, Department of Cell Biology & Histology, University of Cádiz, 11003, Cádiz, Spain
| | - Jone Razquin
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48940, Barakaldo, Spain
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48940, Barakaldo, Spain
| | - Irene Suárez-Pereira
- Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Esther Berrocoso
- Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain.
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain.
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain.
| |
Collapse
|
5
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
6
|
Reisz C, Figenshau K, Cheng AL, Elfagir A. Phenotypic and Clinical Traits That Correlate with Cognitive Impairment in Caucasian Females. WOMEN'S HEALTH REPORTS 2021; 2:528-532. [PMID: 34841399 PMCID: PMC8617581 DOI: 10.1089/whr.2021.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 11/23/2022]
Abstract
Background: Dementia affects more women than men. This suggests sex steroid-dependent structural and functional differences between male and female brains. Natural and iatrogenic changes to women's reproductive health may correlate with risk for dementia. Objective: To identify surrogate markers of key transitions within the reproductive axis that could correlate with dementia pathology in women. Specific Research Question: Could examination of the reproductive axis from birth to senescence expand our understanding of the gender predominance of dementia in women? Proxy measurements for fetal origins, reproduction, and age-related effects on estrogen-dependent tissues were collected to study dementia risk in women. Methods: Deidentified data were collected from 289 older Caucasian female patients from an out-patient clinic in Kansas City, Missouri. Women patients 65 years and older were offered the opportunity to join the study and written consent was obtained from all participants. Data were collected from 2017 to 2019. Results: Our subjects ranged in age from 65 to 98 years old, with a mean of 76 years old. Spearman correlation analysis showed significant correlation between dementia status and age (r = 0.219, p = 0.000), Fitzpatrick skin phototype (r = −0.141, p = 0.019), birth order (r = 0.151, p = 0.028), current height as measured in the office (r = −0.215, p = 0.001), and maximum height per patient recall (r = −0.173, p = 0.005). Results from the logistic regression model show that specific predictors of risk for dementia were age (odds ratio [OR] = 1.082 [1.034–1.132]; p = 0.0007), Fitzpatrick skin phototype 1 versus 3 (OR = 8.508 [1.075–67.313]; p = 0.0227), and current height (OR = 0.766 [0.642–0.915]; p = 0.0032). Of the four variables related to fetal origins: maternal age, number of siblings, birth order, and age difference between the subject and the next older sibling, none were found to be statistically significant. Since age is a significant predictor of risk for dementia, it was included as a covariate in the aforementioned logistic regression models. Conclusions: Our results showed that dementia in Caucasian women was associated with age, lower Fitzpatrick phototype, and current height. Dementia-related pathological processes in the brain may accrue over a woman's lifetime.
Collapse
Affiliation(s)
- Colleen Reisz
- Department of Medicine and University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Karen Figenshau
- Department of Medicine and University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - An-Lin Cheng
- Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Abdelmoneim Elfagir
- Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| |
Collapse
|
7
|
Priyanka HP, Nair RS. Neuroimmunomodulation by estrogen in health and disease. AIMS Neurosci 2020; 7:401-417. [PMID: 33263078 PMCID: PMC7701372 DOI: 10.3934/neuroscience.2020025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Systemic homeostasis is maintained by the robust bidirectional regulation of the neuroendocrine-immune network by the active involvement of neural, endocrine and immune mediators. Throughout female reproductive life, gonadal hormones undergo cyclic variations and mediate concomitant modulations of the neuroendocrine-immune network. Dysregulation of the neuroendocrine-immune network occurs during aging as a cumulative effect of declining neural, endocrine and immune functions and loss of compensatory mechanisms including antioxidant enzymes, growth factors and co-factors. This leads to disruption of homeostasis and sets the stage for the development of female-specific age-associated diseases such as autoimmunity, osteoporosis, cardiovascular diseases and hormone-dependent cancers. Ovarian hormones especially estrogen, play a key role in the maintenance of health and homeostasis by modulating the nervous, endocrine and immune functions and thereby altering neuroendocrine-immune homeostasis. Immunologically estrogen's role in the modulation of Th1/Th2 immune functions and contributing to pro-inflammatory conditions and autoimmunity has been widely studied. Centrally, hypothalamic and pituitary hormones influence gonadal hormone secretion in murine models during onset of estrous cycles and are implicated in reproductive aging-associated acyclicity. Loss of estrogen affects neuronal plasticity and the ensuing decline in cognitive functions during reproductive aging in females implicates estrogen in the incidence and progression of neurodegenerative diseases. Peripherally, sympathetic noradrenergic (NA) innervations of lymphoid organs and the presence of both adrenergic (AR) and estrogen receptors (ER) on lymphocytes poise estrogen as a potent neuroimmunomodulator during health and disease. Cyclic variations in estrogen levels throughout reproductive life, perimenopausal surge in estrogen levels followed by its precipitous decline, concomitant with decline in central hypothalamic catecholaminergic activity, peripheral sympathetic NA innervation and associated immunosuppression present an interesting study to explore female-specific age-associated diseases in a new light.
Collapse
Affiliation(s)
- Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai-600002, India
| | | |
Collapse
|
8
|
Ponzi D, Gioiosa L, Parmigiani S, Palanza P. Effects of Prenatal Exposure to a Low-Dose of Bisphenol A on Sex Differences in Emotional Behavior and Central Alpha 2-Adrenergic Receptor Binding. Int J Mol Sci 2020; 21:ijms21093269. [PMID: 32380724 PMCID: PMC7246441 DOI: 10.3390/ijms21093269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/27/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Prenatal exposure to bisphenol A (BPA) influences the development of sex differences neurologically and behaviorally across many species of vertebrates. These effects are a consequence of BPA’s estrogenic activity and its ability to act as an endocrine disrupter even, at very low doses. When exposure to BPA occurs during critical periods of development, it can interfere with the normal activity of sex steroids, impacting the fate of neurons, neural connectivity and the development of brain regions sensitive to steroid activity. Among the most sensitive behavioral targets of BPA action are behaviors that are characterized by a sexual dimorphism, especially emotion and anxiety related behaviors, such as the amount of time spent investigating a novel environment, locomotive activity and arousal. Moreover, in some species of rodents, BPA exposure affected males’ sexual behaviors. Interestingly, these behaviors are at least in part modulated by the catecholaminergic system, which has been reported to be a target of BPA action. In the present study we investigated the influence of prenatal exposure of mice to a very low single dose of BPA on emotional and sexual behaviors and on the density and binding characteristics of alpha2 adrenergic receptors. Alpha2 adrenergic receptors are widespread in the central nervous system and they can act as autoreceptors, inhibiting the release of noradrenaline and other neurotransmitters from presynaptic terminals. BPA exposure disrupted sex differences in behavioral responses to a novel environment, but did not affect male mice sexual behavior. Importantly, BPA exposure caused a change in the binding affinity of alpha2 adrenergic receptors in the locus coeruleus and medial preoptic area (mPOA) and it eliminated the sexual dimorphism in the density of the receptors in the mPOA.
Collapse
Affiliation(s)
- Davide Ponzi
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (L.G.); (P.P.)
- Correspondence: ; Tel.: +39-0521904776
| | - Laura Gioiosa
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (L.G.); (P.P.)
| | - Stefano Parmigiani
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43121 Parma, Italy;
| | - Paola Palanza
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (L.G.); (P.P.)
| |
Collapse
|
9
|
Mulvey B, Bhatti DL, Gyawali S, Lake AM, Kriaucionis S, Ford CP, Bruchas MR, Heintz N, Dougherty JD. Molecular and Functional Sex Differences of Noradrenergic Neurons in the Mouse Locus Coeruleus. Cell Rep 2019; 23:2225-2235. [PMID: 29791834 DOI: 10.1016/j.celrep.2018.04.054] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 02/23/2018] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
Preclinical work has long focused on male animals, though biological sex clearly influences risk for certain diseases, including many psychiatric disorders. Such disorders are often treated by drugs targeting the CNS norepinephrine system. Despite roles for noradrenergic neurons in behavior and neuropsychiatric disease models, their molecular characterization has lagged. We profiled mouse noradrenergic neurons in vivo, defining over 3,000 high-confidence transcripts expressed therein, including druggable receptors. We uncovered remarkable sex differences in gene expression, including elevated expression of the EP3 receptor in females-which we leverage to illustrate the behavioral and pharmacologic relevance of these findings-and of Slc6a15 and Lin28b, both major depressive disorder (MDD)-associated genes. Broadly, we present a means of transcriptionally profiling locus coeruleus under baseline and experimental conditions. Our findings underscore the need for preclinical work to include both sexes and suggest that sex differences in noradrenergic neurons may underlie behavioral differences relevant to disease.
Collapse
Affiliation(s)
- Bernard Mulvey
- Department of Genetics and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Dionnet L Bhatti
- Department of Anesthesiology and Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Sandeep Gyawali
- Laboratory of Molecular Biology, Rockefeller University, and Howard Hughes Medical Institute, New York, NY, USA
| | - Allison M Lake
- Department of Genetics and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, Rockefeller University, and Howard Hughes Medical Institute, New York, NY, USA
| | - Joseph D Dougherty
- Department of Genetics and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
10
|
Sex differences in breathing. Comp Biochem Physiol A Mol Integr Physiol 2019; 238:110543. [PMID: 31445081 DOI: 10.1016/j.cbpa.2019.110543] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 01/15/2023]
Abstract
Breathing is a vital behavior that ensures both the adequate supply of oxygen and the elimination of CO2, and it is influenced by many factors. Despite that most of the studies in respiratory physiology rely heavily on male subjects, there is much evidence to suggest that sex is an important factor in the respiratory control system, including the susceptibility for some diseases. These different respiratory responses in males and females may be related to the actions of sex hormones, especially in adulthood. These hormones affect neuromodulatory systems that influence the central medullary rhythm/pontine pattern generator and integrator, sensory inputs to the integrator and motor output to the respiratory muscles. In this article, we will first review the sex dependence on the prevalence of some respiratory-related diseases. Then, we will discuss the role of sex and gonadal hormones in respiratory control under resting conditions and during respiratory challenges, such as hypoxia and hypercapnia, and whether hormonal fluctuations during the estrous/menstrual cycle affect breathing control. We will then discuss the role of the locus coeruleus, a sexually dimorphic CO2/pH-chemosensitive nucleus, on breathing regulation in males and females. Next, we will highlight the studies that exist regarding sex differences in respiratory control during development. Finally, the few existing studies regarding the influence of sex on breathing control in non-mammalian vertebrates will be discussed.
Collapse
|
11
|
Peltier MR, Verplaetse TL, Mineur YS, Petrakis IL, Cosgrove KP, Picciotto MR, McKee SA. Sex differences in stress-related alcohol use. Neurobiol Stress 2019; 10:100149. [PMID: 30949562 PMCID: PMC6430711 DOI: 10.1016/j.ynstr.2019.100149] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 01/12/2023] Open
Abstract
Rates of alcohol use disorder (AUD) have increased in women by 84% over the past ten years relative to a 35% increase in men. This substantive increase in female drinking is alarming given that women experience greater alcohol-related health consequences compared to men. Stress is strongly associated with all phases of alcohol addiction, including drinking initiation, maintenance, and relapse for both women and men, but plays an especially critical role for women. The purpose of the present narrative review is to highlight what is known about sex differences in the relationship between stress and drinking. The critical role stress reactivity and negative affect play in initiating and maintaining alcohol use in women is addressed, and the available evidence for sex differences in drinking for negative reinforcement as it relates to brain stress systems is presented. This review discusses the critical structures and neurotransmitters that may underlie sex differences in stress-related alcohol use (e.g., prefrontal cortex, amygdala, norepinephrine, corticotropin releasing factor, and dynorphin), the involvement of sex and stress in alcohol-induced neurodegeneration, and the role of ovarian hormones in stress-related drinking. Finally, the potential avenues for the development of sex-appropriate pharmacological and behavioral treatments for AUD are identified. Overall, women are generally more likely to drink to regulate negative affect and stress reactivity. Sex differences in the onset and maintenance of alcohol use begin to develop during adolescence, coinciding with exposure to early life stress. These factors continue to affect alcohol use into adulthood, when reduced responsivity to stress, increased affect-related psychiatric comorbidities and alcohol-induced neurodegeneration contribute to chronic and problematic alcohol use, particularly for women. However, current research is limited regarding the examination of sex in the initiation and maintenance of alcohol use. Probing brain stress systems and associated brain regions is an important future direction for developing sex-appropriate treatments to address the role of stress in AUD.
Collapse
Affiliation(s)
| | | | - Yann S. Mineur
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Ismene L. Petrakis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Kelly P. Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
- Department of Diagnostic Radiology, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Sherry A. McKee
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
| |
Collapse
|
12
|
Noradrenaline through β-adrenoceptor contributes to sexual dimorphism in primary CD4+ T-cell response in DA rat EAE model? Cell Immunol 2018; 336:48-57. [PMID: 30600100 DOI: 10.1016/j.cellimm.2018.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022]
Abstract
Males exhibit stronger sympathetic nervous system (SNS) activity, but weaker primary CD4+ T-cell (auto)immune responses. To test the role of catecholamines, major end-point SNS mediators, in this dimorphism, influence of propranolol (β-adrenoceptor blocker) on mitogen/neuroantigen-stimulated CD4+ T cells from female and male EAE rat draining lymph node (dLN) cell cultures was examined. Male rat dLNs exhibited higher noradrenaline concentration and frequency of β2-adrenoceptor-expressing CD4+ T lymphocytes and antigen presenting cells. Propranolol, irrespective of exogenous noradrenaline presence, more prominently augmented IL-2 production and proliferation of CD4+ lymphocytes in male than female rat dLN cell cultures. In neuroantigen-stimulated dLN cells of both sexes propranolol increased IL-1β and IL-23/p19 expression and IL-17+ CD4+ cell frequency, but enhanced IL-17 production only in male rat CD4+ lymphocytes, thereby abrogating sexual dimorphism in IL-17 concentration observed in propranolol-free cultures. Thus, β-adrenoceptor-mediated signalling may contribute to sex bias in rat IL-17-producing cell secretory capacity.
Collapse
|
13
|
Torres-Rojas C, Jones BC. Sex Differences in Neurotoxicogenetics. Front Genet 2018; 9:196. [PMID: 29922331 PMCID: PMC5996082 DOI: 10.3389/fgene.2018.00196] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/15/2018] [Indexed: 12/02/2022] Open
Abstract
A major development in biomedical research is the recognition that the sex of an individual plays a key role in susceptibility, treatment, and outcomes of most diseases. In this contribution, we present evidence that sex is also important in the toxicity of many environmental toxicants and contributes to the effect of genetics. Thus, individual differences in response to toxicants includes genetic makeup, the environment and sex; in fact, sex differences may be considered a part of genetic constitution. In this review, we present evidence for sex contribution to susceptibility for a number of toxicants.
Collapse
Affiliation(s)
- Carolina Torres-Rojas
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C Jones
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
14
|
Thalamic Cortical Error-Related Responses in Adult Social Drinkers: Sex Differences and Problem Alcohol Use. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 3:868-877. [PMID: 29859929 DOI: 10.1016/j.bpsc.2018.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Error-related brain activities are altered in individuals with substance use disorders. Here we examined error-related activities in relation to problem drinking in nondependent alcohol drinkers. In particular, we investigated sex differences and whether altered error responses are related to post-error behavioral control. METHODS A sample of 145 nondependent drinkers (77 women) performed the stop-signal task during functional magnetic resonance imaging. Imaging data were processed and modeled using statistical parametric mapping. Independent sample t test and linear regression were employed to examine sex differences in error response and relationship between error response and problem drinking. RESULTS Compared with men, women showed greater error-related (stop error > go success) activations in the bilateral thalamus, right middle/superior temporal cortex, and bilateral dorsal anterior cingulate cortex. In whole-brain linear regression of error responses against the Alcohol Use Disorders Identification Test score, a wide swath of cortical and subcortical regions, including the thalamus, showed decreased activation in association with problem drinking in women but not in men. However, men and women were not different in the extent of post-error slowing and decreased thalamic error response in association with problem drinking was not related to the extent of post-error slowing in women. CONCLUSIONS The results suggest sex differences in error-related activations with heavier drinking associated with reduced error activations in women but not in men. These differences in cerebral activations may reflect higher physiological arousal in response to errors and greater vulnerability of saliency-related arousal response to problem drinking in female as compared with male social drinkers.
Collapse
|
15
|
Leposavić GM, Pilipović IM. Intrinsic and Extrinsic Thymic Adrenergic Networks: Sex Steroid-Dependent Plasticity. Front Endocrinol (Lausanne) 2018; 9:13. [PMID: 29441042 PMCID: PMC5797573 DOI: 10.3389/fendo.2018.00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/11/2018] [Indexed: 12/30/2022] Open
Abstract
The thymus is sexually differentiated organ providing microenvironment for T-cell precursor differentiation/maturation in the major histocompatibility complex-restricted self-tolerant T cells. With increasing age, the thymus undergoes involution leading to the decline in efficacy of thymopoiesis. Noradrenaline from thymic nerve fibers and "(nor)adrenergic" cells is involved in the regulation of thymopoiesis. In rodents, noradrenaline concentration in thymus and adrenoceptor (AR) expression on thymic cells depend on sex and age. These differences are suggested to be implicated in the development of sexual diergism and the age-related decline in thymopoiesis. The programming of both thymic sexual differentiation and its involution occurs during the critical early perinatal period and may be reprogrammed during peripubertal development. The thymic (re)programming is critically dependent on circulating levels of gonadal steroids. Although the underlying molecular mechanisms have not yet been elucidated fully, it is assumed that the gonadal steroid action during the critical perinatal/peripubertal developmental periods leads to long-lasting changes in the efficacy of thymopoiesis partly through (re)programming of "(nor)adrenergic" cell networks and AR expression on thymic cells.
Collapse
Affiliation(s)
- Gordana Momčilo Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
- *Correspondence: Gordana Momčilo Leposavić,
| | - Ivan M. Pilipović
- Immunology Research Centre “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
| |
Collapse
|
16
|
Reproductive steroid receptors and actions in the locus coeruleus of male macaques: Part of an aggression circuit? Prog Neuropsychopharmacol Biol Psychiatry 2016; 71:210-22. [PMID: 27083854 PMCID: PMC4996758 DOI: 10.1016/j.pnpbp.2016.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/29/2016] [Accepted: 04/06/2016] [Indexed: 11/23/2022]
Abstract
This study was initiated to determine whether the noradrenergic (NE) neurons of the locus coeruleus (LC) could mediate the stimulatory action of androgens on serotonin-related gene expression in male macaques. These experiments follow our observations that serotonin neurons lack androgen receptors (ARs), and yet respond to androgens. Male Japanese macaques (Macaca fuscata) were castrated for 5-7months and then treated for 3months with [1] placebo, [2] T (testosterone), [3] DHT (dihydrotestosterone; non-aromatizable androgen) plus ATD (steroidal aromatase inhibitor), or [4] FLUT (Flutamide; androgen antagonist) plus ATD (n=5/group). The noradrenergic (NE) innervation of the raphe was determined with immunolabeling of axons with an antibody to dopamine-β-hydroxylase (DBH). Immunolabeling of tyrosine hydroxylase (TH) dendrites and corticotropin releasing hormone (CRH) axons innervating the LC was also determined. Due to the longer treatment period employed, the expression of the cognate nuclear receptors was sought. Androgen receptor (AR), estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) immunostaining was accomplished. Quantitative image analysis was applied and immunopositive neurons or axons with boutons were measured. Double-label of NE neurons for each receptor plus TH determined whether the receptors were localized in NE neurons. Androgens with or without aromatase activity significantly stimulated DBH axon density in the raphe (ANOVA, p=0.006), and LC dendritic TH (ANOVA, p<0.0001), similar to serotonin-related mRNA expression in the raphe. There were significantly more AR-positive neurons in T- and DHT+ATD-treated groups compared to placebo or FLUT+ATD-treated groups (ANOVA, p=0.0014). There was no difference in the number of positive-neurons stained for ERα or ERβ. The CRH axon density in the LC was significantly reduced with aromatase inhibition, suggesting that CRH depends on estrogen, not androgens (ANOVA, p=0.0023). Double-immunohistochemistry revealed that NE neurons did not contain AR. Rather, AR-positive nuclei were found in neighboring cells that are likely neurons. However, >80% of LC NE neurons contained ERα or ERβ. In conclusion, the LC NE neurons may transduce the stimulatory effect of androgens on serotonin-related gene expression. Since LC NE neurons lack AR, the androgenic stimulation of dendritic TH and axonal DBH may be indirectly mediated by other neurons. Estrogen, either from metabolism of T or from de novo synthesis, appears necessary for robust CRH innervation of the LC, which differs from female macaques.
Collapse
|
17
|
Pilipović I, Vujnović I, Arsenović-Ranin N, Dimitrijević M, Kosec D, Stojić-Vukanić Z, Leposavić G. Peripubertal ovariectomy influences thymic adrenergic network plasticity in adult rats. J Neuroimmunol 2016; 297:103-16. [PMID: 27397083 DOI: 10.1016/j.jneuroim.2016.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/07/2016] [Accepted: 05/22/2016] [Indexed: 11/19/2022]
Abstract
The study investigated the influence of peripubertal ovariectomy on the thymic noradrenaline (NA) concentration, and the thymocyte NA content and β2- and α1-adrenoceptor (AR) expression in adult 2- and 11-month-old rats. In control rats, the thymic NA concentration increased with age. This increase reflected rise in the density of catecholamine (CA)-containing fluorescent nerve fibers and cells and their CA content. Additionally, the average β2- and α1-AR thymocyte surface density changed in the opposite direction with age; the density of β2-AR decreased, whereas that of α1-AR increased. Ovariectomy diminished the thymic NA concentration in 2-month-old rats. This reflected the decrease in the density of fluorescent nerve fibers, and CA content in fluorescent nerve fibers and non-lymphoid cells, since the thymocyte NA content was increased in ovariectomized (Ox) rats. Estrogen supplementation prevented the ovariectomy-induced changes. In Ox rats, the density of CA-synthesizing nerve fibers and non-lymphoid cells diminished with age. To the contrary, NA content in thymocytes increased with age, but it did not exceed that in 11-month-old controls. Additionally, ovariectomy diminished the average thymocyte surface density of β2-ARs, but it increased that of α1-ARs in 2-month-old-rats (due to estrogen, and estrogen and progesterone deficiency, respectively). These changes, despite of the rise in circulating estrogen level post-ovariectomy, remained stable with age. This most likely reflected a decreased sensitivity to estrogen action, as a consequence of the hormone misprinting in peripubertal age. The analysis of thymocyte proliferation in culture suggested that age- and ovariectomy-induced alterations in thymocyte NA synthesis and AR expression altered NA autocrine/paracrine action on thymocytes. In conclusion, the study indicates that the ovarian hormone deficiency in peripubertal age affects ovarian steroid-dependent remodeling of thymic adrenergic regulatory network in adult rats.
Collapse
Affiliation(s)
- Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
18
|
Panzica G, Melcangi RC. Structural and molecular brain sexual differences: A tool to understand sex differences in health and disease. Neurosci Biobehav Rev 2016; 67:2-8. [DOI: 10.1016/j.neubiorev.2016.04.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 02/07/2023]
|
19
|
Pratap UP, Patil A, Sharma HR, Hima L, Chockalingam R, Hariharan MM, Shitoot S, Priyanka HP, ThyagaRajan S. Estrogen-induced neuroprotective and anti-inflammatory effects are dependent on the brain areas of middle-aged female rats. Brain Res Bull 2016; 124:238-53. [PMID: 27242078 DOI: 10.1016/j.brainresbull.2016.05.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Reproductive aging in females is characterized by fluctuations and precipitous decline in estrogen levels, which may lead to reduction in cognitive function and age-associated neurodegenerative disorders. The nature of estrogen-mediated neuronal plasticity is unknown during reproductive aging. We hypothesize that estrogen treatment of early middle-aged ovariectomized rats may exert specific effects in the brain by modulating signaling pathways regulating metabolic enzymes, inflammatory markers, antioxidant status, cholinergic function and survival signals. PURPOSE To investigate the mechanisms of estrogen-induced effects on neuroprotection and neuroinflammation through the involvement of intracellular signaling pathways in brain areas of ovariectomized (OVX) middle-aged (MA) female rats. METHODS Ovariectomized early MA female Sprague-Dawley rats (n=8/group) were implanted with 17β-estradiol (E2) 30-day release pellets (0.6μg and 300μg). At the end of the treatment period, frontal cortex (FC), striatum (STR), medial basal hypothalamus (MBH), and hippocampus (HP) were isolated and examined for the expression of tyrosine hydroxylase (p-TH), nerve growth factor (NGF), p-NF-κB (p50 and p65)and p-ERK, p-CREB, p-Akt, and activities of cholinesterases and antioxidant enzymes, key regulatory enzymes of metabolic pathways, and nitric oxide production. RESULTS E2 enhanced p-TH expression in FC and HP, reduced NGF expression in HP, and suppressed p-NF-κB expression in FC and STR. It also increased the expression of molecular markers (p-ERK, p-CREB and p-Akt), and nitric oxide production in various brain areas, while differentially regulating the activities of metabolic enzymes and cholinesterases. CONCLUSION Estrogen modulates the neural and inflammatory factors, and intracellular markers depending on the brain areas that may influence differential remodeling of neuronal circuitry which can be used to develop therapeutic strategies in cognitive impairment and neurodegenerative disorders in aging.
Collapse
Affiliation(s)
- Uday P Pratap
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Anushree Patil
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Himanshu R Sharma
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Lalgi Hima
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Ramanathan Chockalingam
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Murali M Hariharan
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Sushrut Shitoot
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Hannah P Priyanka
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
20
|
Brown ECZ, Steadman CJ, Lee TM, Padmanabhan V, Lehman MN, Coolen LM. Sex differences and effects of prenatal exposure to excess testosterone on ventral tegmental area dopamine neurons in adult sheep. Eur J Neurosci 2015; 41:1157-66. [PMID: 25784297 DOI: 10.1111/ejn.12871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/19/2015] [Accepted: 02/10/2015] [Indexed: 10/23/2022]
Abstract
Prenatal testosterone (T) excess in sheep results in a wide array of reproductive neuroendocrine deficits and alterations in motivated behavior. The ventral tegmental area (VTA) plays a critical role in reward and motivated behaviors and is hypothesised to be targeted by prenatal T. Here we report a sex difference in the number VTA dopamine cells in the adult sheep, with higher numbers of tyrosine hydroxylase (TH)-immunoreactive (-ir) cells in males than females. Moreover, prenatal exposure to excess T during either gestational days 30-90 or 60-90 resulted in increased numbers of VTA TH-ir cells in adult ewes compared to control females. Stereological analysis confirmed significantly greater numbers of neurons in the VTA of males and prenatal T-treated ewes, which was primarily accounted for by greater numbers of TH-ir cells. In addition, immunoreactivity for TH in the cells was denser in males and prenatal T-treated females, suggesting that sex differences and prenatal exposure to excess T affects both numbers of cells expressing TH and the protein levels within dopamine cells. Sex differences were also noted in numbers of TH-ir cells in the substantia nigra, with more cells in males than females. However, prenatal exposure to excess T did not affect numbers of TH-ir cells in the substantia nigra, suggesting that this sex difference is organised independently of prenatal actions of T. Together, these results demonstrate sex differences in the sheep VTA dopamine system which are mimicked by prenatal treatment with excess T.
Collapse
Affiliation(s)
- Erinna C Z Brown
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, N6A 5C1, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Parkinson's disease (PD) displays a greater prevalence and earlier age at onset in men. This review addresses the concept that sex differences in PD are determined, largely, by biological sex differences in the NSDA system which, in turn, arise from hormonal, genetic and environmental influences. Current therapies for PD rely on dopamine replacement strategies to treat symptoms, and there is an urgent, unmet need for disease modifying agents. As a significant degree of neuroprotection against the early stages of clinical or experimental PD is seen, respectively, in human and rodent females compared with males, a better understanding of brain sex dimorphisms in the intact and injured NSDA system will shed light on mechanisms which have the potential to delay, or even halt, the progression of PD. Available evidence suggests that sex-specific, hormone-based therapeutic agents hold particular promise for developing treatments with optimal efficacy in men and women.
Collapse
|
22
|
Tando S, Itoh K, Yaoi T, Ogi H, Goto S, Mori M, Fushiki S. Bisphenol A exposure disrupts the development of the locus coeruleus-noradrenergic system in mice. Neuropathology 2014; 34:527-34. [DOI: 10.1111/neup.12137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 05/13/2014] [Indexed: 01/08/2023]
Affiliation(s)
- So Tando
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| | - Takeshi Yaoi
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| | - Hiroshi Ogi
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| | - Shoko Goto
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| | - Miyuki Mori
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| | - Shinji Fushiki
- Department of Pathology and Applied Neurobiology; Graduate School of Medical Science; Kyoto Prefectural University of Medicine; Kyoto Japan
| |
Collapse
|
23
|
Hughes EL, Cover PO, Buckingham JC, Gavins FNE. Role and interactions of annexin A1 and oestrogens in the manifestation of sexual dimorphisms in cerebral and systemic inflammation. Br J Pharmacol 2013; 169:539-53. [PMID: 22897118 PMCID: PMC3682703 DOI: 10.1111/j.1476-5381.2012.02146.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 06/11/2012] [Accepted: 06/22/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Gender differences in inflammation are well described, with females often showing more robust, oestrogen-associated responses. Here, we investigated the influence of gender, oestrogen and the anti-inflammatory protein annexin A1 (AnxA1) on lipopolysaccharide (LPS)-induced leukocyte-endothelial cell interactions in murine cerebral and mesenteric microvascular beds. EXPERIMENTAL APPROACH Intravital microscopy was used to visualize and quantify the effects of LPS (10 μg·per mouse i.p.) on leukocyte-endothelial interactions in male and female wild-type (WT) mice. The effects of ovariectomy ± oestrogen replacement were examined in WT and AnxA1-null (AnxA1(-/-) ) female mice. KEY RESULTS LPS increased leukocyte adherence in the cerebral and mesenteric beds of both male and female WT mice; females showed exacerbated responses in the brain versus males, but not the mesentery. Ovariectomy further enhanced LPS-induced adhesion in the brain but not the mesentery; its effects were reversed by oestrogen treatment. OVX AnxA1(-/-) mice also showed exaggerated adhesive responses to LPS in the brain. However, these were unresponsive to ovariectomy and, paradoxically, responded to oestrogen with a pronounced increase in basal and LPS-induced leukocyte adhesion in the cerebrovasculature. CONCLUSIONS AND IMPLICATIONS Our data confirm the fundamental role of AnxA1 in limiting the inflammatory response in the central and peripheral microvasculature. They also (i) show that oestrogen acts via an AnxA1-dependent mechanism to protect the cerebral, but not the mesenteric, vasculature from the damaging effects of LPS and (ii) reveal a paradoxical and potentially toxic effect of the steroid in potentiating the central response to LPS in the absence of AnxA1.
Collapse
Affiliation(s)
- Ellen L Hughes
- Wolfson Neuroscience Laboratories, Imperial College LondonLondon, UK
| | - Patricia O Cover
- Wolfson Neuroscience Laboratories, Imperial College LondonLondon, UK
| | - Julia C Buckingham
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College LondonLondon, UK
| | | |
Collapse
|
24
|
Carkaci-Salli N, Battula S, Wang X, Connor JR, Vrana KE. Gender-specific regulation of tyrosine hydroxylase in thymocyte differentiation antigen-1 knockout mice. J Neurosci Res 2012; 90:1583-8. [PMID: 22430860 DOI: 10.1002/jnr.23046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 01/19/2012] [Accepted: 01/21/2012] [Indexed: 11/09/2022]
Abstract
Thymocyte differentiation antigen-1 (Thy-1) is a cell surface glycoprotein found on T cells and neurons and is involved in cell-to-cell interactions. In addition, Thy-1 knockouts (KO) are a potential mouse model of restless legs syndrome (RLS) based on clinical observations and the role of dopamine in the disease. In this study, we analyzed the activity and quantity of tyrosine hydroxylase (TH; the rate-limiting enzyme in dopamine production) and determined phosphorylation levels for the enzyme phosphoserine-40 (pSer-40). There was no significant difference in the total TH activity and pSer-40 TH levels between Thy-1 KO and control groups; however, TH specific activity was significantly lower (by 26%) in Thy-1 KO mice. This difference is due in part to increased TH protein levels in this group (increased by 29%). When analyzed by gender, Thy-1 KO female mouse striata contained less TH specific activity compared with control females (decreased by 41%) and male control or Thy-1 KO animals (decreased by 30%). TH specific activity and pSer-40 TH levels in male Thy-1 KO and control displayed no differences. However, pSer-40 TH was significantly higher in control females (38%) compared with control or Thy-1 KO males. The Thy-1 KO females exhibited significantly lower (28%) pSer-40 TH (normalized to GAPDH or TH) than control females. Indeed, the Thy-1 KO females had 50% of the pSer-40 TH found in controls. Our results suggest a gender effect on TH specific activity, TH protein levels, and serine-40 phosphorylation of TH in Thy-1 KO female mice.
Collapse
Affiliation(s)
- N Carkaci-Salli
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | | | | | |
Collapse
|
25
|
Sirivelu MP, MohanKumar PS, MohanKumar SMJ. Differential effects of systemic interleukin-1β on gene expression in brainstem noradrenergic nuclei. Life Sci 2012; 90:77-81. [PMID: 22036618 PMCID: PMC3268688 DOI: 10.1016/j.lfs.2011.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 08/18/2011] [Accepted: 10/10/2011] [Indexed: 11/26/2022]
Abstract
AIMS The cytokine, interleukin-1β (IL-1β), is known to produce specific effects on the neuroendocrine system such as suppression of the reproductive axis and stimulation of the stress axis. The mechanism by which IL-1β produces these differential effects is not clear. Since norepinephrine (NE) is involved in these effects, we hypothesized that IL-1β acts on brainstem noradrenergic nuclei to affect gene transcription of NE synthesizing enzymes, cytokines and associated transcription factors. MAIN METHODS Adult female Sprague Dawley rats in proestrus were divided into two groups. Control animals received PBS-BSA and the treatment group received 5 μg of rat recombinant IL-1β i.p. at noon. They were sacrificed in groups at 1, 3 and 5 pm (n=6/group) for measurement of tyrosine hydroxylase (TH) mRNA by qPCR or at 3 pm for mRNA analysis by qPCR array. KEY FINDINGS TH mRNA levels decreased gradually with time in both control and IL-1β-treated rats in the ventrolateral medulla. In the nucleus of solitary tract, TH mRNA levels were significantly reduced by IL-1β treatment at 5 pm. In the locus coeruleus, TH mRNA levels increased significantly at 5 pm with IL-1β treatment compared to controls. In the second set of animals analyzed by qPCR array, there were several fold increases in the expression of certain cytokines, chemokines, and transcription factors in specific noradrenergic nuclei. SIGNIFICANCE Systemic administration of IL-1β causes significant changes in the expression of tyrosine hydroxylase and several chemokines in brain stem noradrenergic nuclei, thereby mediating its neuroendocrine effects.
Collapse
Affiliation(s)
- Madhu P. Sirivelu
- Neuroendocrine Research Laboratory, Comparative Medicine & Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - P. S. MohanKumar
- Neuroendocrine Research Laboratory, Comparative Medicine & Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Neuroendocrine Research Laboratory, Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Sheba M. J. MohanKumar
- Neuroendocrine Research Laboratory, Comparative Medicine & Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Neuroendocrine Research Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
26
|
Marwarha G, Rhen T, Schommer T, Ghribi O. The oxysterol 27-hydroxycholesterol regulates α-synuclein and tyrosine hydroxylase expression levels in human neuroblastoma cells through modulation of liver X receptors and estrogen receptors--relevance to Parkinson's disease. J Neurochem 2011; 119:1119-36. [PMID: 21951066 PMCID: PMC3217121 DOI: 10.1111/j.1471-4159.2011.07497.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Loss of dopaminergic neurons and α-synuclein accumulation are the two major pathological hallmarks of Parkinson's disease. Currently, the mechanisms governing depletion of dopamine content and α-synuclein accumulation are not well understood. We showed that the oxysterol 27-hydroxycholesterol (27-OHC) reduces the expression of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis, and increases α-synuclein levels in SH-SY5Y cells. However, the cellular mechanisms involved in 27-OHC effects were not elucidated. In this study, we demonstrate that 27-OHC regulates TH and α-synuclein expression levels through the estrogen receptors (ER) and liver X receptors (LXR). We specifically show that inhibition of ERβ mediates 27-OHC-induced decrease in TH expression, an effect reversed by the ER agonist estradiol. We also show that 27-OHC and the LXR agonist GW3965 increase α-synuclein while the LXR antagonist 5α-6α-epoxycholesterol-3-sulfate significantly attenuated the 27-OHC-induced increase in α-synuclein expression. We further demonstrate that LXRβ positively regulates α-synuclein expression and 27-OHC increases LXRβ-mediated α-synuclein transcription. Our results demonstrate the involvement of two distinct pathways that are involved in the 27-OHC regulation of TH and α-synuclein levels. Concomitant activation of ERβ and inhibition of LXRβ prevent 27-OHC effects and may therefore reduce the progression of Parkinson's disease by precluding TH reduction and α-synuclein accumulation.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202
| | - Turk Rhen
- Department of Biology, University of North Dakota, Grand Forks, North Dakota, 58202
| | | | - Othman Ghribi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202
| |
Collapse
|
27
|
McArthur S, Gillies GE. Peripheral vs. Central Sex Steroid Hormones in Experimental Parkinson's Disease. Front Endocrinol (Lausanne) 2011; 2:82. [PMID: 22649388 PMCID: PMC3355917 DOI: 10.3389/fendo.2011.00082] [Citation(s) in RCA: 7] [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] [Received: 10/05/2011] [Accepted: 11/10/2011] [Indexed: 01/01/2023] Open
Abstract
The nigrostriatal dopaminergic (NSDA) pathway degenerates in Parkinson's disease (PD), which occurs with approximately twice the incidence in men than women. Studies of the influence of systemic estrogens in females suggest sex hormones contribute to these differences. In this review we analyze the evidence revealing great complexity in the response of the healthy and injured NSDA system to hormonal influences, and emphasize the importance of centrally generated estrogens. At physiological levels, circulating estrogen (in females) or estrogen precursors (testosterone in males, aromatized to estrogen centrally) have negligible effects on dopaminergic neuron survival in experimental PD, but can modify striatal dopamine levels via actions on the activity or adaptive responses of surviving cells. However, these effects are sexually dimorphic. In females, estradiol promotes adaptive responses in the partially injured NSDA pathway, preserving striatal dopamine, whereas in males gonadal steroids and exogenous estradiol have a negligible or even suppressive effect, effectively exacerbating dopamine loss. On balance, the different effects of gonadal factors in males and females contribute to sex differences in experimental PD. Fundamental sex differences in brain organization, including the sexually dimorphic networks regulating NSDA activity are likely to underpin these responses. In contrast, estrogen generated locally appears to preserve striatal dopamine in both sexes. The available data therefore highlight the need to understand the biological basis of sex-specific responses of the NSDA system to peripheral hormones, so as to realize the potential for sex-specific, hormone-based therapies in PD. Furthermore, they suggest that targeting central steroid generation could be equally effective in preserving striatal dopamine in both sexes. Clarification of the relative roles of peripheral and central sex steroid hormones is thus an important challenge for future studies.
Collapse
Affiliation(s)
- Simon McArthur
- Department of Medicine, Centre for Neuroscience, Imperial College LondonLondon, UK
- *Correspondence: Simon McArthur, Department of Medicine, Centre for Neuroscience, Imperial College London, London SW7 2AZ, UK. e-mail:
| | - Glenda E. Gillies
- Department of Medicine, Centre for Neuroscience, Imperial College LondonLondon, UK
| |
Collapse
|
28
|
Schulz K, Korz V. Hippocampal testosterone relates to reference memory performance and synaptic plasticity in male rats. Front Behav Neurosci 2010; 4:187. [PMID: 21188275 PMCID: PMC3006668 DOI: 10.3389/fnbeh.2010.00187] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 12/01/2010] [Indexed: 12/12/2022] Open
Abstract
Steroids are important neuromodulators influencing cognitive performance and synaptic plasticity. While the majority of literature concerns adrenal- and gonadectomized animals, very little is known about the “natural” endogenous release of hormones during learning. Therefore, we measured blood and brain (hippocampus, prefrontal cortex) testosterone, estradiol, and corticosterone concentrations of intact male rats undergoing a spatial learning paradigm which is known to reinforce hippocampal plasticity. We found significant modulations of all investigated hormones over the training course. Corticosterone and testosterone were correlated manifold with behavior, while estradiol expressed fewer correlations. In the recall session, testosterone was tightly coupled to reference memory (RM) performance, which is crucial for reinforcement of synaptic plasticity in the dentate gyrus. Intriguingly, prefrontal cortex and hippocampal levels related differentially to RM performance. Correlations of testosterone and corticosterone switched from unspecific activity to specific cognitive functions over training. Correspondingly, exogenous application of testosterone revealed different effects on synaptic and neuronal plasticity in trained versus untrained animals. While hippocampal long-term potentiation (LTP) of the field excitatory postsynaptic potential (fEPSP) was prolonged in untrained rats, both the fEPSP- and the population spike amplitude (PSA)-LTP was impaired in trained rats. Behavioral performance was unaffected, but correlations of hippocampal field potentials with behavior were decoupled in treated rats. The data provide important evidence that besides adrenal, also gonadal steroids play a mechanistic role in linking synaptic plasticity to cognitive performance.
Collapse
|
29
|
Fan L, Smith CE, Curtis KS. Regional differences in estradiol effects on numbers of HSD2-containing neurons in the nucleus of the solitary tract of rats. Brain Res 2010; 1358:89-101. [PMID: 20728435 DOI: 10.1016/j.brainres.2010.08.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 08/11/2010] [Accepted: 08/12/2010] [Indexed: 12/15/2022]
Abstract
Estrogens affect body fluid balance, including sodium ingestion. Recent findings of a population of neurons in the hindbrain nucleus of the solitary tract (NTS) of rats that are activated during sodium need suggest a possible central substrate for this effect of estrogens. We used immunohistochemistry to label neurons in the NTS that express 11-β-hydroxysteroid dehydrogenase type 2 (HSD2), an enzyme that promotes aldosterone binding, in male rats, and in ovariectomized (OVX) rats given estradiol benzoate (EB) or oil vehicle (OIL). During baseline conditions, the number of HSD2 immunoreactive neurons in the NTS immediately rostral to the area postrema was greater in EB-treated OVX rats compared to those in OIL-treated OVX and male rats. A small number of HSD2 immunoreactive neurons was also labeled for dopamine-β-hydroxylase (DBH), an enzyme involved in norepinephrine biosynthesis. Double-labeled neurons in the NTS were located primarily in the more lateral portion of the HSD2 population, at the level of the area postrema in all three groups, with no sex or estrogen-mediated differences in the number of double-labeled neurons. These results suggest that two subpopulations of HSD2 neurons are present in the NTS. One subpopulation, which does not colocalize with DBH and is increased during conditions of elevated estradiol, may contribute to the effects of estrogens on sodium ingestion. The role of the other, smaller subpopulation, which colocalizes with DBH and is not affected by estradiol, remains to be determined, but one possibility is that these latter neurons are part of a larger network of catecholaminergic input to neuroendocrine neurons in the hypothalamus.
Collapse
Affiliation(s)
- Liming Fan
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK 74107-1898, USA
| | | | | |
Collapse
|
30
|
Gillies GE, McArthur S. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol Rev 2010; 62:155-98. [PMID: 20392807 PMCID: PMC2879914 DOI: 10.1124/pr.109.002071] [Citation(s) in RCA: 480] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The classic view of estrogen actions in the brain was confined to regulation of ovulation and reproductive behavior in the female of all mammalian species studied, including humans. Burgeoning evidence now documents profound effects of estrogens on learning, memory, and mood as well as neurodevelopmental and neurodegenerative processes. Most data derive from studies in females, but there is mounting recognition that estrogens play important roles in the male brain, where they can be generated from circulating testosterone by local aromatase enzymes or synthesized de novo by neurons and glia. Estrogen-based therapy therefore holds considerable promise for brain disorders that affect both men and women. However, as investigations are beginning to consider the role of estrogens in the male brain more carefully, it emerges that they have different, even opposite, effects as well as similar effects in male and female brains. This review focuses on these differences, including sex dimorphisms in the ability of estradiol to influence synaptic plasticity, neurotransmission, neurodegeneration, and cognition, which, we argue, are due in a large part to sex differences in the organization of the underlying circuitry. There are notable sex differences in the incidence and manifestations of virtually all central nervous system disorders, including neurodegenerative disease (Parkinson's and Alzheimer's), drug abuse, anxiety, and depression. Understanding the cellular and molecular basis of sex differences in brain physiology and responses to estrogen and estrogen mimics is, therefore, vitally important for understanding the nature and origins of sex-specific pathological conditions and for designing novel hormone-based therapeutic agents that will have optimal effectiveness in men or women.
Collapse
Affiliation(s)
- Glenda E Gillies
- Centre for Neuroscience, Department of Medicine, Hammersmith Hospital, Imperial College Faculty of Medicine, DuCane Road, London W12ONN, UK.
| | | |
Collapse
|
31
|
Goel N, Bale TL. Sex differences in the serotonergic influence on the hypothalamic-pituitary-adrenal stress axis. Endocrinology 2010; 151:1784-94. [PMID: 20185764 PMCID: PMC2850237 DOI: 10.1210/en.2009-1180] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Appropriate interactions between serotonin (5-HT) and stress pathways are critical for maintaining homeostasis. Dysregulation of hypothalamic-pituitary-adrenal (HPA) stress axis is a common feature in affective disorders in which an involvement of 5-HT neurocircuitry has been implicated in disease vulnerability and treatment responsiveness. Because there is a greater prevalence of affective disorders in women, sex differences in the 5-HTergic influence on stress pathways may contribute to disease disparity. Therefore, our studies compared stress or citalopram-induced corticosterone levels in male and female mice. To determine whether sex-dependent HPA axis responsiveness was mediated by the difference in testosterone levels, testosterone-treated females were also examined. Gene expression patterns in 5-HTergic and stress neurocircuitry were analyzed to determine sites of potential sex differences and mechanisms of testosterone action. As expected, restraint stress corticosterone levels were higher in intact females and were masculinized by testosterone. Interestingly, citalopram administration independent of stress resulted in a greater corticosterone response in females, which was also masculinized by testosterone. Analyses along the 5-HT-HPA axis revealed sex differences including greater pituitary 5-HT receptors and adrenal weights in females. Moreover, in stress-regulatory regions, we found sex differences in glucocorticoid receptor and glutamic acid decarboxylase expression supportive of greater inhibitory modulation and feedback potential in males. Taken together, these data suggest that multiple sites related to 5-HTergic stimulation, corticosterone production, and negative feedback of HPA neurocircuitry combine to produce higher female stress responsiveness. These studies support a potential for sex-specific involvement of 5-HT and stress pathways in the etiology of affective disorders.
Collapse
Affiliation(s)
- Nirupa Goel
- School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
32
|
Sadahiro R, Suzuki A, Shibuya N, Kamata M, Matsumoto Y, Goto K, Otani K. Association study between a functional polymorphism of tyrosine hydroxylase gene promoter and personality traits in healthy subjects. Behav Brain Res 2010; 208:209-12. [PMID: 19958792 DOI: 10.1016/j.bbr.2009.11.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/17/2009] [Accepted: 11/22/2009] [Indexed: 02/05/2023]
Affiliation(s)
- Ryoichi Sadahiro
- Department of Psychiatry, Yamagata University School of Medicine, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Herbison AE, de Tassigny XD, Doran J, Colledge WH. Distribution and postnatal development of Gpr54 gene expression in mouse brain and gonadotropin-releasing hormone neurons. Endocrinology 2010; 151:312-21. [PMID: 19966188 DOI: 10.1210/en.2009-0552] [Citation(s) in RCA: 215] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kisspeptin and G protein-coupled receptor 54 (GPR54) are now acknowledged to play essential roles in the neural regulation of fertility. Using a transgenic Gpr54 LacZ knock-in mouse model, this study aimed to provide 1) a detailed map of cells expressing Gpr54 in the mouse brain and 2) an analysis of Gpr54 expression in GnRH neurons across postnatal development. The highest density of Gpr54-expressing cells in the mouse central nervous system was found in the dentate gyrus of the hippocampus beginning on postnatal d 6 (P6). Abundant Gpr54 expression was also noted in the septum, rostral preoptic area (rPOA), anteroventral nucleus of the thalamus, posterior hypothalamus, periaqueductal grey, supramammillary and pontine nuclei, and dorsal cochlear nucleus. No Gpr54 expression was detected in the arcuate and rostral periventricular nuclei of the hypothalamus. Dual-labeling experiments showed that essentially all Gpr54-expressing cells in the rPOA were GnRH neurons. Analyses of mice at birth, P1, P5, P20, and P30 and as adults revealed a gradual increase in the percentage of GnRH neurons expressing Gpr54 from approximately 40% at birth through to approximately 70% from P20 onward. Whereas GnRH neurons located in the septum displayed a consistent increase across this time, GnRH neurons in the rPOA showed a sharp reduction in Gpr54 expression after birth (to approximately 10% at P5) before increasing to the 70% expression levels by P20. Together these findings provide an anatomical basis for the exploration of Gpr54 actions outside the reproductive axis and reveal a complex temporal and spatial pattern of Gpr54 gene expression in developing GnRH neurons.
Collapse
Affiliation(s)
- Allan E Herbison
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, P.O. Box 913, Dunedin, New Zealand.
| | | | | | | |
Collapse
|
34
|
Jazin E, Cahill L. Sex differences in molecular neuroscience: from fruit flies to humans. Nat Rev Neurosci 2010; 11:9-17. [DOI: 10.1038/nrn2754] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Gonadectomy and hormone replacement exert region- and enzyme isoform-specific effects on monoamine oxidase and catechol-O-methyltransferase activity in prefrontal cortex and neostriatum of adult male rats. Neuroscience 2009; 165:850-62. [PMID: 19909795 DOI: 10.1016/j.neuroscience.2009.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/02/2009] [Accepted: 11/04/2009] [Indexed: 12/23/2022]
Abstract
Sex differences and gonadal hormone influences are well known for diverse aspects of forebrain amine and indolamine neurotransmitter systems, the cognitive and affective functions they govern and their malfunction in mental illness. This study explored whether hormone regulation/dysregulation of these systems could be related to gonadal steroid effects on catechol-O-methyltransferase and monoamine oxidase which are principal enzymatic controllers of forebrain dopamine, serotonin and norepinephrine levels. Driven by male over female differences in cortical enzyme activities, by male-specific associations between monoamine oxidase and catechol-O-methyltransferase gene polymorphisms and cognitive and dysfunction in disease and by male-specific consequences of gene knockouts in mice, the question of hormone sensitivity was addressed here using a male rat model where prefrontal dopamine levels and related behaviors are also known to be affected. Specifically, quantitative O-methylation and oxidative deamination assays were used to compare the activities of catechol-O-methyltransferase's soluble and membrane-bound isoforms and of monoamine oxidase's A and B isoforms in the pregenual medial prefrontal cortex and dorsal striatum of male rats that were sham operated, gonadectomized or gonadectomized and supplemented with testosterone propionate or with estradiol for 28 days. These studies revealed significant effects of hormone replacement but not gonadectomy on the soluble but not the membrane-bound isorfom of catechol-O-methyltransferase in both striatum and cortex. A significant, cortex-specific testosterone-but not estradiol-attenuated effect (increase) of gonadectomy on monoamine oxidase's A but not B isoform was also observed. Although none of these actions suggest potential roles in the regulation/dysregulation of prefrontal dopamine, the suppressive effects of testosterone on cortical monoamine oxidase-A that were observed could have bearing on the increased incidence of cognitive deficits and symptoms of depression and anxiety that are repeatedly observed in males in conditions of hypogonadalism related to aging, other biological factors or in prostate cancer where androgen deprivation is used as a neoadjuvant treatment.
Collapse
|
36
|
Maharjan S, Serova LI, Sabban EL. Membrane-initiated estradiol signaling increases tyrosine hydroxylase promoter activity with ER alpha in PC12 cells. J Neurochem 2009; 112:42-55. [PMID: 19818101 DOI: 10.1111/j.1471-4159.2009.06430.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tyrosine hydroxylase (TH) promoter activity is induced by 17beta-estradiol (E(2)) in PC12 cells expressing estradiol receptor-alpha (ERalpha) requiring a cAMP/calcium response element (CRE/CaRE) at -45. To examine whether membrane-initiated estradiol signaling is underlying this induction, cells co-transfected with TH reporter construct and ERalpha expression vector were exposed to membrane-impermeant estradiol conjugate (beta-estradiol-6-(O-carboxy-methyl) oxime-bovine serum albumin, E(2)BSA). TH promoter activity was elevated by E(2)BSA in dose- and time-dependent manner. E(2)BSA also elicited rapid phosphorylation of CRE binding protein (CREB) and increased CRE-driven promoter activity. Over-expression of dominant negative forms of CREB, with mutations in DNA binding or phosphorylation site, prevented TH promoter response to E(2)BSA. Pre-treatment with protein kinase A (PKA) and MEK inhibitors reduced E(2) dependent phosphorylation of CREB and ERK, and also decreased induction of TH promoter activity by E(2) or E(2)BSA. Blocking S-palmitoylation of ERalpha with C451A mutation and/or pre-treatment with 2-Bromopalmitate did not prevent but instead enhanced E(2) or E(2)BSA-elicited induction of TH promoter activity. These findings reveal, for the first time, that estradiol induction of TH gene transcription with ERalpha in PC12 cells involves membrane-initiated estradiol signaling, rapid activation of dual PKA/MEK signaling pathways, leading to CREB phosphorylation, acting at CRE/CaRE. The data demonstrate possible mechanism whereby estradiol affects catecholaminergic systems in vivo.
Collapse
Affiliation(s)
- Shreekrishna Maharjan
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| | | | | |
Collapse
|
37
|
Hazell GGJ, Yao ST, Roper JA, Prossnitz ER, O'Carroll AM, Lolait SJ. Localisation of GPR30, a novel G protein-coupled oestrogen receptor, suggests multiple functions in rodent brain and peripheral tissues. J Endocrinol 2009; 202:223-36. [PMID: 19420011 PMCID: PMC2710976 DOI: 10.1677/joe-09-0066] [Citation(s) in RCA: 279] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, the G protein-coupled receptor GPR30 has been identified as a novel oestrogen receptor (ER). The distribution of the receptor has been thus far mapped only in the rat central nervous system. This study was undertaken to map the distribution of GPR30 in the mouse brain and rodent peripheral tissues. Immunohistochemistry using an antibody against GPR30 revealed high levels of GPR30 immunoreactivity (ir) in the forebrain (e.g. cortex, hypothalamus and hippocampus), specific nuclei of the midbrain (e.g. the pontine nuclei and locus coeruleus) and the trigeminal nuclei and cerebellum Purkinje layer of the hindbrain in the adult mouse brain. In the rat and mouse periphery, GPR30-ir was detected in the anterior, intermediate and neural lobe of the pituitary, adrenal medulla, renal pelvis and ovary. In situ hybridisation histochemistry using GPR30 riboprobes, revealed intense hybridisation signal for GPR30 in the paraventricular nucleus and supraoptic nucleus (SON) of the hypothalamus, anterior and intermediate lobe of the pituitary, adrenal medulla, renal pelvis and ovary of both rat and mouse. Double immunofluorescence revealed GPR30 was present in both oxytocin and vasopressin neurones of the paraventricular nucleus and SON of the rat and mouse brain. The distribution of GPR30 is distinct from the other traditional ERs and offers an additional way in which oestrogen may mediate its effects in numerous brain regions and endocrine systems in the rodent.
Collapse
|
38
|
Sabban EL, Maharjan S, Nostramo R, Serova LI. Divergent effects of estradiol on gene expression of catecholamine biosynthetic enzymes. Physiol Behav 2009; 99:163-8. [PMID: 19638280 DOI: 10.1016/j.physbeh.2009.07.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 07/10/2009] [Accepted: 07/13/2009] [Indexed: 12/15/2022]
Abstract
Within the catecholaminergic systems, there are contradictory findings regarding ability of estradiol to regulate expression of genes related to catecholamine biosynthesis. Several parameters important for effects of estradiol on the catecholamine (CA) related enzyme gene expression were examined in two CA regions. Ovariectomized (OVX) female rats were given prolonged estradiol treatments, either in a pulsatile fashion by injections or continuously by pellets. The mode affected the response of tyrosine hydroxylase (TH) and GTP cyclohydrolase I (GTPCH) mRNAs differentially in the nucleus of solitary tract (NTS) and the locus coeruleus (LC). In rostral-medial NTS, TH mRNA levels were increased with injections, but declined in rats administered estradiol by pellets. In LC, a significant change was only observed in GTPCH with injections. These differences may reflect activation of different estrogen receptors (ER). The response to estradiol in the presence of ERalpha and ER beta was examined in PC12 cell culture. Estradiol directly regulated promoter activity of TH, GTPCH and dopamine beta-hydroxylase (DBH) genes. With ERalpha, 17 beta-estradiol elevated TH promoter activity, while there was a decline with ERbeta. In contrast, both DBH and GTPCH promoters were enhanced by 17 beta-estradiol over a wide range of concentrations with either ER subtype. Thus, mode of administration, location examined and ER subtype expressed are important considerations in the overall response of catecholamine related enzymes to estradiol.
Collapse
Affiliation(s)
- Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA.
| | | | | | | |
Collapse
|
39
|
Tyrosine hydroxylase deficit in the chemoafferent and the sympathoadrenergic pathways of the Mecp2 deficient mouse. Neurosci Lett 2008; 447:82-6. [DOI: 10.1016/j.neulet.2008.09.045] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 09/12/2008] [Accepted: 09/18/2008] [Indexed: 11/18/2022]
|
40
|
Leposavić G, Pilipović I, Radojević K, Pešić V, Perišić M, Kosec D. Catecholamines as immunomodulators: A role for adrenoceptor-mediated mechanisms in fine tuning of T-cell development. Auton Neurosci 2008; 144:1-12. [DOI: 10.1016/j.autneu.2008.09.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 09/16/2008] [Indexed: 01/28/2023]
|
41
|
Rantham Prabhakara JP, Feist G, Thomasson S, Thompson A, Schommer E, Ghribi O. Differential effects of 24-hydroxycholesterol and 27-hydroxycholesterol on tyrosine hydroxylase and alpha-synuclein in human neuroblastoma SH-SY5Y cells. J Neurochem 2008; 107:1722-9. [PMID: 19014385 PMCID: PMC3205449 DOI: 10.1111/j.1471-4159.2008.05736.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Evidence suggests that environmental and dietary factors may contribute to the pathogenesis of Parkinson's disease (PD). High dietary intake of cholesterol is such a factor that has been shown to increase or decrease the risk of PD. However, because circulating cholesterol does not cross the blood-brain barrier, the mechanisms linking dietary cholesterol to the pathogenesis of PD remain to be understood. In contrast to cholesterol, the oxidized cholesterol metabolites (oxysterols), 24S-hydroxycholesterol (24-OHC) and 27-hydroxycholesterol (27-OHC), can cross the blood-brain barrier and may place the brain at risk of degeneration. In this study, we incubated the human neuroblastoma SH-SY5Y cells for 24 h with 24-OHC, 27-OHC, or a mixture of 24-OHC plus 27-OHC, and have determined effects on tyrosine hydroxylase (the rate-limiting enzyme in dopamine synthesis) levels, alpha-synuclein levels, and apoptosis. We demonstrate that while 24-OHC increases the levels of tyrosine hydroxylase, 27-OHC increases levels of alpha-synuclein, and induces apoptosis. Our findings show for the first time that oxysterols trigger changes in levels of proteins that are associated with the pathogenesis of PD. As steady state levels of 24-OHC and 27-OHC are tightly regulated in the brain, disturbances in these levels may contribute to the pathogenesis of PD.
Collapse
Affiliation(s)
- Jaya Prasanthi Rantham Prabhakara
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | | | | | | | | | | |
Collapse
|
42
|
Pendergast JS, Tuesta LM, Bethea JR. Oestrogen receptor beta contributes to the transient sex difference in tyrosine hydroxylase expression in the mouse locus coeruleus. J Neuroendocrinol 2008; 20:1155-64. [PMID: 18680559 DOI: 10.1111/j.1365-2826.2008.01776.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oestrogen receptors (ERs) are important for sexual differentiation of the brain. Previous studies in rats have reported that the locus coeruleus (LC), a catecholaminergic nucleus in the brain stem, is sexually dimorphic such that females have more neurones than males. We hypothesised that ERs may be important for sexual differentiation of this nucleus in mice. Because previous studies reported conflicting results regarding ER protein expression in the mouse LC, we evaluated ER alpha and ER beta gene expression by in situ hybridisation and the real-time reverse transcription-polymerase chain reaction. We demonstrated that both ER alpha and ER beta mRNAs are present in tyrosine hydroxylase-immunoreactive (TH-ir) cells in the male LC. In the female LC, ER alpha mRNA is present at levels similar to males, whereas ER beta mRNA expression is significantly lower than in males. Similar to rats, male mice have fewer TH-ir cells in the LC than females at 60 days after birth, but the difference is absent at 120 days after birth when females exhibit a similar reduction in TH-ir cells. The transient sex difference is ER beta-dependent because is it absent in ER beta knockout mice, and is due to regulation of TH expression and not from death of TH-ir cells. Testicular hormones produced at adolescence are necessary for the regulation of TH expression in the male LC because orchidectomy of pre-pubertal males prevented the decrease in TH-ir cells, whereas treatment of gonadectomized males with testosterone or its metabolite, 5 alpha-androstan-3beta,17beta-diol, restored the intact male phenotype. Overall, these studies indicate that ER beta is important in regulating TH expression in the mouse LC.
Collapse
Affiliation(s)
- J S Pendergast
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, Miami, FL, USA
| | | | | |
Collapse
|
43
|
Serguera C, Triaca V, Kelly-Barrett J, Banchaabouchi MA, Minichiello L. Increased dopamine after mating impairs olfaction and prevents odor interference with pregnancy. Nat Neurosci 2008; 11:949-56. [PMID: 18641644 DOI: 10.1038/nn.2154] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 06/03/2008] [Indexed: 11/09/2022]
Abstract
In rodents, social odor sensing influences female reproductive status by affecting neuroendocrine cascades. The odor of male mouse urine can induce ovulation or block pregnancy within 3 d post coitus. Females avoid the action of such olfactory stimuli after embryonic implantation. The mechanisms underlying these changes are unknown. Here we report that shortly after mating, a surge in dopamine in the mouse main olfactory bulb impairs the perception of social odors contained in male urine. Treatment of females at 6.5 d post coitus with a dopamine D2 receptor antagonist restores social odor sensing and favors disruption of pregnancy by inhibition of prolactin release, when administered in the presence of alien male urine odors. These results show that an active sensory barrier blocks social olfactory cues detrimental to pregnancy, consistent with the main olfactory bulb being a major relay through which social odor modulates reproductive status.
Collapse
Affiliation(s)
- Che Serguera
- European Molecular Biology Laboratory, Mouse Biology Unit, Via Ramarini 32, 00015 Monterotondo, Italy
| | | | | | | | | |
Collapse
|
44
|
Pilipović I, Vidić-Danković B, Perisić M, Radojević K, Colić M, Todorović V, Leposavić G. Sexual dimorphism in the catecholamine-containing thymus microenvironment: a role for gonadal hormones. J Neuroimmunol 2008; 195:7-20. [PMID: 18262658 DOI: 10.1016/j.jneuroim.2007.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 12/14/2007] [Accepted: 12/19/2007] [Indexed: 01/15/2023]
Abstract
The study was undertaken to explore whether there were: i) apart from neural and circulatory, some other sources of catecholamines (CAs) in rat thymus and ii) gender-specific differences in thymic CA levels, and if so to elucidate the role of sex steroids in this phenomenon. Tyrosine hydroxylase (TH) immunoreactivity was found in thymocytes and thymic epithelial cells (some of which showed morphological features of nurse cells). The density of CA-synthesizing cells was greater in male than in female rats. Noradrenaline (NA), but not dopamine (DA), was detected in thymocytes. NA and DA levels in thymi, and the NA level in thymocytes, were higher in male rats. To explore the putative role of sex steroids in this dichotomy in the thymi of adult rats gonadectomized (Gx) or sham-Gx at the age of 30 days the density of TH+ cells and CA levels were measured. Gonadectomy abolished sexual dimorphism in the density of thymic TH+ cells (diminishing their density in male rats) and thymic CA levels (the NA levels were reduced in rats of both sexes and also the DA level in male rats). Therefore, it can be assumed that testicular and ovarian hormones control thymic NA and DA levels via different mechanisms. Moreover, in Gx rats, despite the decrease in the overall thymic NA level, an increase in the thymocyte NA level was found indicating that gonadal hormones exert differential effects on the NA level in distinct thymic cellular compartments.
Collapse
Affiliation(s)
- Ivan Pilipović
- Immunology Research Centre Branislav Janković, Institute of Immunology and Virology Torlak, Belgrade, Serbia
| | | | | | | | | | | | | |
Collapse
|
45
|
Campbell RE, Herbison AE. Definition of brainstem afferents to gonadotropin-releasing hormone neurons in the mouse using conditional viral tract tracing. Endocrinology 2007; 148:5884-90. [PMID: 17823269 PMCID: PMC6101187 DOI: 10.1210/en.2007-0854] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Brainstem monoamines have long been considered to play a role in regulating the activity of GnRH neurons, although their neuroanatomical relationship with these cells has remained unclear. Using a Cre-dependent pseudorabies virus (Ba2001) technique that permits retrograde tracing selectively from GnRH neurons in the mouse, we have examined the organization of brainstem inputs to rostral preoptic area (rPOA) GnRH neurons. Two days after injection of Ba2001 into the rPOA of adult female GnRH-Cre transgenic mice, five to nine GnRH neurons located immediately adjacent to the injection site were found to express green fluorescent protein (GFP), the marker of virus infection, with no GFP expression anywhere else in the brain. In mice killed 24 h later (3 d after injection), GFP-expressing cells were identified (in order of density) in the raphe nuclei, periaqueductal grey, locus coeruleus, nucleus tractus solitarius, and area postrema. This time course is compatible with these neurons representing primary afferent inputs to the GnRH neurons. Four and 6 d after Ba2001 injection, GFP-expressing cells were found in additional brain regions. Dual-label immunofluorescence experiments in 3-d postinjection mice demonstrated that 100% of GFP-expressing neurons in the raphe were positive for tryptophan hydroxylase, whereas 100% and approximately 50% of GFP neurons in the locus coeruleus and nucleus tractus solitarius, respectively, expressed tyrosine hydroxylase. These observations demonstrate that rPOA GnRH neurons receive direct projections from brainstem A2 and A6 noradrenergic neurons and that, surprisingly, the largest afferent input from the brainstem originates from raphe serotonin neurons in the mouse.
Collapse
Affiliation(s)
- Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand 9001
| | | |
Collapse
|
46
|
Abstract
Recent studies in both animals and humans indicate that gonadal hormones have profound control over emotional states, and certainly contribute to the increased occurrence of psychiatric illness in women. Reports, as reviewed here, suggest that two important regions of the limbic system, the central nucleus of the amygdala (CeA) and the bed nucleus of the stria terminalis (BNST), control different aspects of emotional behaviour. Short-term cue-specific emotional responses, like Pavlovian fear conditioning, require activation of the CeA, while long-duration and contextual emotional responses, are dependant on the BNST. There is accumulating experimental evidence that gender and sex hormones specifically modulate BNST-mediated anxiety behaviours. Moreover, the functional separation between the CeA and the BNST may be exaggerated during lactation in the rat, a time of profound hormonal and behavioural change. In this study, the effects of sex hormones on fear and anxiety are reviewed with an emphasis on the differential effects of these hormones on functions subserved by the BNST as opposed to the CeA. Studies, as highlighted here, looking at sex hormone and gender effects on the ability of corticotrophin-releasing factor and bright ambient light to enhance startle, emphasise the importance of understanding both the effect of, and brain region where, gonadal hormones exert their control over emotional behaviour.
Collapse
Affiliation(s)
- D Toufexis
- Emory University, Department of Psychiatry, Yerkes National Primate Centre, Atlanta, GA 30329, USA.
| |
Collapse
|
47
|
Gómez C, Briñón JG, Valero J, Recio JS, Murias AR, Curto GG, Orio L, Colado MI, Alonso JR. Sex differences in catechol contents in the olfactory bulb of control and unilaterally deprived rats. Eur J Neurosci 2007; 25:1517-28. [PMID: 17425578 DOI: 10.1111/j.1460-9568.2007.05407.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The dopaminergic system plays important roles in the modulation of olfactory transmission. The present study examines the distribution of dopaminergic cells and the content of dopamine (DA) and its metabolites in control and deprived olfactory bulbs (OB), focusing on the differences between sexes. The content of DA and of its metabolites, dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), were measured by HPLC. The morphology and distribution of dopaminergic neurons were studied using tyrosine hydroxylase (TH) immunohistochemistry. Cells were typified with TH-parvalbumin, TH-cholecystokinin or TH-neurocalcin double-immunofluorescence assays. Biochemical analyses revealed sex differences in the content of DA and of its metabolites. In normal conditions, the OBs of male rats had higher concentrations of DA, DOPAC and HVA than the OBs of females. The immunohistochemical data pointed to sex differences in the number of TH-immunopositive cells (higher in male than in female rats). Colocalization analyses revealed that dopaminergic cells constitute a different cell subpopulation from those labelled after parvalbumin, cholecystokinin or neurocalcin immunostaining. Unilateral olfactory deprivation caused dramatic alterations in the dopaminergic system. The DA content and the density of dopaminergic cells decreased, the contents of DA and DOPAC as well as TH immunoreactivity were similar in deprived males and females and, finally, the metabolite/neurotransmitter ratio increased. Our results show that the dopaminergic modulation of olfactory transmission seems to differ between males and females and that it is regulated by peripheral olfactory activity. A possible role of the dopaminergic system in the sexually different olfactory sensitivity, discrimination and memory is discussed.
Collapse
Affiliation(s)
- C Gómez
- Laboratory Plasticidad Neuronal y Neurorreparación, Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, E-37007 Salamanca, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
McArthur S, Murray HE, Dhankot A, Dexter DT, Gillies GE. Striatal susceptibility to a dopaminergic neurotoxin is independent of sex hormone effects on cell survival and DAT expression but is exacerbated by central aromatase inhibition. J Neurochem 2007; 100:678-92. [PMID: 17116232 DOI: 10.1111/j.1471-4159.2006.04226.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate further the hormone-dependent processes underlying sex differences in neurotoxic responses within the rat nigrostriatal dopaminergic (NSDA) pathway after partial lesioning with 6-OHDA, a state thought to mimic the early stages of Parkinson's disease where, in humans and animal models alike, males appear to be more susceptible. Contrary to our hypotheses, hormone manipulations (gonadectomy +/- oestrogen or androgen treatment) failed to alter survival of tyrosine hydroxylase immunoreactive cells in the substantia nigra pars compacta (SNc) after lesioning; this indicates that, unlike inherent sex differences in toxin-induced striatal dopamine depletion, sex differences in cell loss were not hormonally generated, and that hormone-dependent changes in dopamine depletion can occur independently of cell survival. In addition, hormonally induced changes in striatal expression of the dopamine transporter (DAT), an important factor for 6-OHDA toxicity, did not correlate with hormonal influences on striatal dopamine loss and, in males, central inhibition of aromatase prior to 6-OHDA infusion exacerbated striatal dopamine loss with no effect on SNc tyrosine hydroxylase-immunoreactive survival, suggesting locally generated oestrogen is neuroprotective. These results support the novel view that sex steroid hormones produced peripherally and centrally play a significant, sex-specific role within the sexually dimorphic NSDA pathway to modulate plastic, compensatory responses aimed at restoring striatal dopamine functionality, without affecting cell loss.
Collapse
Affiliation(s)
- Simon McArthur
- Division of Neuroscience and Mental Health, Faculty of Medicine, Imperial College London, London, UK
| | | | | | | | | |
Collapse
|
49
|
Schlenker EH, Hansen SN. Sex-specific densities of estrogen receptors alpha and beta in the subnuclei of the nucleus tractus solitarius, hypoglossal nucleus and dorsal vagal motor nucleus weanling rats. Brain Res 2006; 1123:89-100. [PMID: 17045976 DOI: 10.1016/j.brainres.2006.09.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 09/12/2006] [Accepted: 09/13/2006] [Indexed: 11/24/2022]
Abstract
In rats ventilatory responses to N-methyl-d-aspartate (NMDA) receptor modulation are sexually dimorphic and may be altered by manipulating brain levels of estrogen receptors. Here we used image analysis and immunohistochemistry in weanling male and female rats to quantitate areas and densities of ER alpha and ER beta-positive neurons within medullary regions associated with cardiopulmonary regulation including the hypoglossal nucleus, subnuclei of the nucleus of the solitary tract (NTS), and the dorsal motor nucleus of the vagus. Weanling rats were selected because ventilation, metabolic rate, and body and brain weights are comparable at this age and there are no large fluctuations in plasma hormone levels. Females, relative to males, had smaller areas in the A2 region and parts of the NTS. Counts and densities for ER alpha were greater in females than males in almost all regions studied. In contrast sex differences in ER beta were found in fewer nuclei, but in those higher counts and densities were noted in females. In general, ER beta-positive neurons in the brainstem regions examined were less prevalent than ER alpha neurons. Thus, in weanling rats sex affected ER alpha and ER beta neuronal densities in brainstem regions associated with cardiopulmonary regulation that may be responsible for sex differences in control of breathing.
Collapse
Affiliation(s)
- Evelyn H Schlenker
- Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD 57069, USA.
| | | |
Collapse
|
50
|
Millan MJ. Multi-target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 2006; 110:135-370. [PMID: 16522330 DOI: 10.1016/j.pharmthera.2005.11.006] [Citation(s) in RCA: 389] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 12/20/2022]
Abstract
Major depression is a debilitating and recurrent disorder with a substantial lifetime risk and a high social cost. Depressed patients generally display co-morbid symptoms, and depression frequently accompanies other serious disorders. Currently available drugs display limited efficacy and a pronounced delay to onset of action, and all provoke distressing side effects. Cloning of the human genome has fuelled expectations that symptomatic treatment may soon become more rapid and effective, and that depressive states may ultimately be "prevented" or "cured". In pursuing these objectives, in particular for genome-derived, non-monoaminergic targets, "specificity" of drug actions is often emphasized. That is, priority is afforded to agents that interact exclusively with a single site hypothesized as critically involved in the pathogenesis and/or control of depression. Certain highly selective drugs may prove effective, and they remain indispensable in the experimental (and clinical) evaluation of the significance of novel mechanisms. However, by analogy to other multifactorial disorders, "multi-target" agents may be better adapted to the improved treatment of depressive states. Support for this contention is garnered from a broad palette of observations, ranging from mechanisms of action of adjunctive drug combinations and electroconvulsive therapy to "network theory" analysis of the etiology and management of depressive states. The review also outlines opportunities to be exploited, and challenges to be addressed, in the discovery and characterization of drugs recognizing multiple targets. Finally, a diversity of multi-target strategies is proposed for the more efficacious and rapid control of core and co-morbid symptoms of depression, together with improved tolerance relative to currently available agents.
Collapse
Affiliation(s)
- Mark J Millan
- Institut de Recherches Servier, Centre de Recherches de Croissy, Psychopharmacology Department, 125, Chemin de Ronde, 78290-Croissy/Seine, France.
| |
Collapse
|