1
|
Baron M, Devor M. Neurosteroids foster sedation by engaging tonic GABA A-Rs within the mesopontine tegmental anesthesia area (MPTA). Neurosci Lett 2024; 843:138030. [PMID: 39490574 DOI: 10.1016/j.neulet.2024.138030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Neurosteroids are endogenous molecules with anxiolytic, anticonvulsant, sleep-promoting and sedative effects. They are biosynthesized de novo within the brain, among other tissues, and are thought to act primarily as positive allosteric modulators of high-affinity extrasynaptic GABAAδ-receptors. The location of action of neurosteroids in the brain, however, remains unknown. We have demonstrated that GABAergic anesthetics act within the brainstem mesopontine tegmental anesthesia area (MPTA) to induce and maintain anesthetic loss-of-consciousness. Here we asked whether endogenous and synthetic neurosteroids might also act in the MPTA to induce their suppressive effects. Direct exposure of the MPTA to the endogenous neurosteroids pregnenolone and progesterone, their metabolites testosterone, allopregnanolone and 3α5α-THDOC, and the synthetic neurosteroids ganaxolone and alphaxalone, was found to be pro-anesthetic. Although we cannot rule out additional sites of action, results of this study suggest that the suppressive effects of neurosteroids are due, at least in part, to actions within the MPTA, presumably by recruitment of dedicated neuronal circuitry. This undermines the usual presumption that neurosteroids, like other sedatives, endogenous somnogens and anesthetics, act by nonspecific global distribution.
Collapse
Affiliation(s)
- Mark Baron
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Marshall Devor
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
2
|
Reddy DS, Vadassery A, Ramakrishnan S, Singh T, Clossen B, Wu X. Kindling Models of Epileptogenesis for Developing Disease-Modifying Drugs for Epilepsy. Curr Protoc 2024; 4:e70020. [PMID: 39436626 PMCID: PMC11498896 DOI: 10.1002/cpz1.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Kindling models are widely used animal models to study the pathobiology of epilepsy and epileptogenesis. These models exhibit distinctive features whereby sub-threshold stimuli instigate the initial induction of brief focal seizures. Over time, the severity and duration of these seizures progressively increase, leading to a fully epileptic state, which is marked by consistent development of generalized tonic-clonic seizures. Kindling involves focal stimulation via implanted depth electrodes or repeated administration of chemoconvulsants such as pentylenetetrazol. Comparative analysis of preclinical and clinical findings has confirmed a high predictive validity of fully kindled animals for testing novel antiseizure medications. Thus, kindling models remain an essential component of anticonvulsant drug development programs. This article provides a comprehensive guide to working protocols, testing of therapeutic drugs, outcome parameters, troubleshooting, and data analysis for various electrical and chemical kindling epileptogenesis models for new therapeutic development and optimization. The use of pharmacological agents or genetically modified mice in kindling experiments is valuable, offering insights into the impact of a specific target on various aspects of seizures, including thresholds, initiation, spread, termination, and the generation of a hyperexcitable network. These kindling epileptogenesis paradigms are helpful in identifying mechanisms and disease-modifying interventions for epilepsy. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Hippocampal kindling Basic Protocol 2: Amygdala kindling Basic Protocol 3: Rapid hippocampal kindling Basic Protocol 4: Chemical kindling.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
- Institute of Pharmacology and Neurotherapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Abhinav Vadassery
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Bryan Clossen
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
3
|
Zhang J, Lai W, Tang Y, Gao J, Zhou X, Chen L. Hyperandrogenism Decreases Seizure Threshold in a Rat Model of Polycystic Ovary Syndrome. Neuroendocrinology 2024; 114:1005-1017. [PMID: 39053446 DOI: 10.1159/000540523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION In women of childbearing age with epilepsy, 30% experience the comorbidity of polycystic ovary syndrome (PCOS), which is marked by a higher prevalence of hyperandrogenism. Our recent clinical observations indicate the potential contribution of hyperandrogenism-induced PCOS to epilepsy susceptibility, and this study aimed to unravel the underlying factors that increase the susceptibility of females to epilepsy. METHODS A letrozole-induced PCOS rat model was employed to simulate endogenous hyperandrogenism. The threshold of seizure was assessed through seizure kindling rates using pentetrazol and electroencephalogram recordings. Additionally, the role of androgens in epilepsy was verified through interventions using Diane-35. RESULTS This study revealed that letrozole-induced elevated testosterone levels and PCOS-related changes in female rats. PCOS rats, through pentetrazol-kindling, exhibited a reduced seizure threshold compared with controls. Elevated testosterone levels were observed in both the hippocampal and frontal brain tissues, accompanied by changes in circulation. Two weeks of Diane-35 intervention showed a tendency to alleviate these changes, modifying testosterone levels in both the plasma and brain tissue. Western blotting and immunohistochemistry revealed increased expression of GABA-A receptor in the hippocampus and decreased AMPA receptor expression in the frontal cortex, correlating with antiepileptic status in PCOS rats. CONCLUSION This study delves into the impact of elevated androgen levels on seizure threshold, providing crucial insights into the underpinnings of the comorbidity between PCOS and epilepsy. These findings significantly contribute to the evolving field of epilepsy research, emphasizing the imperative consideration of hormonal influences for the development of targeted therapeutic interventions in individuals with epilepsy and PCOS.
Collapse
Affiliation(s)
- Jiaxian Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Wanlin Lai
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Yusha Tang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Gao
- Laboratory Neuro-diseases and Multi-morbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangyang Zhou
- Laboratory Neuro-diseases and Multi-morbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Demori I, Losacco S, Giordano G, Mucci V, Blanchini F, Burlando B. Fibromyalgia pathogenesis explained by a neuroendocrine multistable model. PLoS One 2024; 19:e0303573. [PMID: 38990866 PMCID: PMC11238986 DOI: 10.1371/journal.pone.0303573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/26/2024] [Indexed: 07/13/2024] Open
Abstract
Fibromyalgia (FM) is a central disorder characterized by chronic pain, fatigue, insomnia, depression, and other minor symptoms. Knowledge about pathogenesis is lacking, diagnosis difficult, clinical approach puzzling, and patient management disappointing. We conducted a theoretical study based on literature data and computational analysis, aimed at developing a comprehensive model of FM pathogenesis and addressing suitable therapeutic targets. We started from the evidence that FM must involve a dysregulation of central pain processing, is female prevalent, suggesting a role for the hypothalamus-pituitary-gonadal (HPG) axis, and is stress-related, suggesting a role for the HP-adrenocortical (HPA) axis. Central pathogenesis was supposed to involve a pain processing loop system including the thalamic ventroposterolateral nucleus (VPL), the primary somatosensory cortex (SSC), and the thalamic reticular nucleus (TRN). For decreasing GABAergic and/or increasing glutamatergic transmission, the loop system crosses a bifurcation point, switching from monostable to bistable, and converging on a high-firing-rate steady state supposed to be the pathogenic condition. Thereafter, we showed that GABAergic transmission is positively correlated with gonadal-hormone-derived neurosteroids, notably allopregnanolone, whereas glutamatergic transmission is positively correlated with stress-induced glucocorticoids, notably cortisol. Finally, we built a dynamic model describing a multistable, double-inhibitory loop between HPG and HPA axes. This system has a high-HPA/low-HPG steady state, allegedly reached in females under combined premenstrual/postpartum brain allopregnanolone withdrawal and stress condition, driving the thalamocortical loop to the high-firing-rate steady state, and explaining the connection between endocrine and neural mechanisms in FM pathogenesis. Our model accounts for FM female prevalence and stress correlation, suggesting the use of neurosteroid drugs as a possible solution to currently unsolved problems in the clinical treatment of the disease.
Collapse
Affiliation(s)
- Ilaria Demori
- Department of Pharmacy, DIFAR, University of Genova, Genova, Italy
| | - Serena Losacco
- Department of Pharmacy, DIFAR, University of Genova, Genova, Italy
| | - Giulia Giordano
- Department of Industrial Engineering, University of Trento, Trento, (TN), Italy
- Delft Center for Systems and Control, Delft University of Technology, Delft, The Netherlands
| | - Viviana Mucci
- School of Science, Western Sydney University, Penrith, Australia
| | - Franco Blanchini
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| | - Bruno Burlando
- Department of Pharmacy, DIFAR, University of Genova, Genova, Italy
| |
Collapse
|
5
|
Cui R, Ye L, Qiao X, Wang S, Zheng K, Yang J, Ge RS, Lin H, Wang Y. Carbon-chain length determines the binding affinity and inhibitory strength of per- and polyfluoroalkyl substances on human and rat steroid 5α-reductase 1 activity. Chem Biol Interact 2024; 394:110987. [PMID: 38574835 DOI: 10.1016/j.cbi.2024.110987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/06/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are widely used synthetic chemicals that persist in the environment and bioaccumulate in animals and humans. There is growing evidence that PFAS exposure adversely impacts neurodevelopment and neurological health. Steroid 5α-reductase 1 (SRD5A1) plays a key role in neurosteroidogenesis by catalyzing the conversion of testosterone or pregnenolone to neuroactive steroids, which influence neural development, cognition, mood, and behavior. This study investigated the inhibitory strength and binding interactions of 18 PFAS on human and rat SRD5A1 activity using enzyme assays, molecular docking, and structure-activity relationship analysis. Results revealed that C9-C14 PFAS carboxylic acid at 100 μM significantly inhibited human SRD5A1, with IC50 values ranged from 10.99 μM (C11) to 105.01 μM (C14), and only one PFAS sulfonic acid (C8S) significantly inhibited human SRD5A1 activity, with IC50 value of 8.15 μM. For rat SRD5A1, C9-C14 PFAS inhibited rat SRD5A1, showing the similar trend, depending on carbon number of the carbon chain. PFAS inhibit human and rat SRD5A1 in a carbon chain length-dependent manner, with optimal inhibition around C11. Kinetic studies indicated PFAS acted through mixed inhibition. Molecular docking revealed PFAS bind to the domain between NADPH and testosterone binding site of both SRD5A1 enzymes. Inhibitory potency correlated with physicochemical properties like carbon number of the carbon chain. These findings suggest PFAS may disrupt neurosteroid synthesis and provide insight into structure-based inhibition of SRD5A1.
Collapse
Affiliation(s)
- Rong Cui
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China
| | - Lei Ye
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China
| | - Xinyi Qiao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China
| | - Shaowei Wang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China
| | - Ke Zheng
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China
| | - Jin Yang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China
| | - Ren-Shan Ge
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China.
| | - Han Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Yiyan Wang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
6
|
Li Q, Zhang Z, Fang J. Hormonal Changes in Women with Epilepsy. Neuropsychiatr Dis Treat 2024; 20:373-388. [PMID: 38436042 PMCID: PMC10906279 DOI: 10.2147/ndt.s453532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/17/2024] [Indexed: 03/05/2024] Open
Abstract
Epilepsy is a prevalent neurological disorder among women globally, often requiring long-term treatment. Hormonal fluctuations in women with epilepsy (WWE) can have reciprocal effects on epilepsy and antiseizure medications (ASMs), posing significant challenges for WWE. Notably, WWE commonly experience endocrine alterations such as thyroid dysfunctions, low bone metabolism, and reproductive hormone irregularities. On the one hand, the presence of hormones in women with epilepsy affects their susceptibility to epilepsy as well as the metabolism of antiseizure medications in various ways. On the other hand, epilepsy itself and the use of antiseizure medications impact the production, secretion, and metabolism of hormones, resulting in low fertility, increased risk of pregnancy complications, negative offspring outcomes, and so on. In order to develop more precise treatment strategies in the future, it is necessary to comprehend the explicit relationships between hormones, epilepsy, and antiseizure medications, as well as to elucidate the currently known mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Qiwei Li
- Department of Neurology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, People’s Republic of China
| | - Zhiyun Zhang
- Department of Neurology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, People’s Republic of China
- Department of Neurology, The Mianyang Central Hospital, Mianyang, Sichuan Province, 621000, People’s Republic of China
| | - Jiajia Fang
- Department of Neurology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, People’s Republic of China
| |
Collapse
|
7
|
Reddy DS. Neurosteroids as Novel Anticonvulsants for Refractory Status Epilepticus and Medical Countermeasures for Nerve Agents: A 15-Year Journey to Bring Ganaxolone from Bench to Clinic. J Pharmacol Exp Ther 2024; 388:273-300. [PMID: 37977814 PMCID: PMC10801762 DOI: 10.1124/jpet.123.001816] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023] Open
Abstract
This article describes recent advances in the use of neurosteroids as novel anticonvulsants for refractory status epilepticus (RSE) and as medical countermeasures (MCs) for organophosphates and chemical nerve agents (OPNAs). We highlight a comprehensive 15-year journey to bring the synthetic neurosteroid ganaxolone (GX) from bench to clinic. RSE, including when caused by nerve agents, is associated with devastating morbidity and permanent long-term neurologic dysfunction. Although recent approval of benzodiazepines such as intranasal midazolam and intranasal midazolam offers improved control of acute seizures, novel anticonvulsants are needed to suppress RSE and improve neurologic function outcomes. Currently, few anticonvulsant MCs exist for victims of OPNA exposure and RSE. Standard-of-care MCs for postexposure treatment include benzodiazepines, which do not effectively prevent or mitigate seizures resulting from nerve agent intoxication, leaving an urgent unmet medical need for new anticonvulsants for RSE. Recently, we pioneered neurosteroids as next-generation anticonvulsants that are superior to benzodiazepines for treatment of OPNA intoxication and RSE. Because GX and related neurosteroids that activate extrasynaptic GABA-A receptors rapidly control seizures and offer robust neuroprotection by reducing neuronal damage and neuroinflammation, they effectively improve neurologic outcomes after acute OPNA exposure and RSE. GX has been selected for advanced, Biomedical Advanced Research and Development Authority-supported phase 3 trials of RSE and nerve agent seizures. In addition, in mechanistic studies of neurosteroids at extrasynaptic receptors, we identified novel synthetic analogs with features that are superior to GX for current medical needs. Development of new MCs for RSE is complex, tedious, and uncertain due to scientific and regulatory challenges. Thus, further research will be critical to fill key gaps in evaluating RSE and anticonvulsants in vulnerable (pediatric and geriatric) populations and military persons. SIGNIFICANCE STATEMENT: Following organophosphate and nerve agent intoxication, refractory status epilepticus (RSE) occurs despite benzodiazepine treatment. RSE occurs in 40% of status epilepticus patients, with a 35% mortality rate and significant neurological morbidity in survivors. To treat RSE, neurosteroids are better anticonvulsants than benzodiazepines. Our pioneering use of neurosteroids for RSE and nerve agents led us to develop ganaxolone as a novel anticonvulsant and neuroprotectant with significantly improved neurological outcomes. This article describes the bench-to-bedside journey of bringing neurosteroid therapy to patients, with ganaxolone leading the way.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
8
|
Singh T, Ramakrishnan S, Wu X, Reddy DS. Sex Differences in Organophosphate Model of Benzodiazepine-Refractory Status Epilepticus and Neuronal Damage. J Pharmacol Exp Ther 2024; 388:313-324. [PMID: 37770202 PMCID: PMC10801723 DOI: 10.1124/jpet.123.001747] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 10/03/2023] Open
Abstract
Sex differences are common in human epilepsy. Although men are more susceptible to seizure than women, the mechanisms underlying sex-specific vulnerabilities to seizure are unclear. The organophosphate (OP) diisopropylfluorophosphate (DFP) is known to cause neurotoxicity and status epilepticus (SE), a serious neurologic condition that causes prolonged seizures and brain damage. Current therapies for OP poisoning and SE do not consider neuronal variations between male and female brains. Therefore, we investigated sex-dependent differences in electrographic seizure activity and neuronal injury using the DFP model of refractory SE in rats. Electroencephalogram recordings were used to monitor DFP-induced SE, and the extent of brain injury was determined using fluoro-jade-B staining to detect cellular necrosis. After DFP exposure, we observed striking sex-dependent differences in SE and seizure activity patterns as well as protective responses to midazolam treatment. Following acute DFP exposure, male animals displayed more severe SE with intense epileptiform spiking and greater mortality than females. In contrast, we observed significantly more injured cells and cellular necrosis in the hippocampus and other brain regions in females than in males. We also observed extensive neuronal injury in the somatosensory cortex of males. The anticonvulsant effect of midazolam against SE was limited in this model and found to be similar in males and females. However, unlike males, females exhibited substantially more protection against neuronal damage after midazolam treatment. Overall, these results demonstrate significant sex-dependent differences in DFP-induced refractory SE and neuronal damage patterns, suggesting that it may be possible to develop sex-specific neuroprotective strategies for OP intoxication and refractory SE. SIGNIFICANCE STATEMENT: Sex-dependent differences in neurotoxicity and status epilepticus (SE) are key biological variables after organophosphate (OP) exposure. Here, we investigated sex-dependent differences in SE and brain injury after acute diisopropylfluorophosphate exposure. Male rats had more severe SE and less survival than females, while females had more neuronal damage. Females had more neuroprotection to midazolam than males, while both sexes had similar but partial anticonvulsant effects. These findings suggest that a sex-specific therapeutic approach may prevent neurological complications of OP-induced SE.
Collapse
Affiliation(s)
- Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| |
Collapse
|
9
|
Gore IR, Gould E. Developmental and adult stress: effects of steroids and neurosteroids. Stress 2024; 27:2317856. [PMID: 38563163 PMCID: PMC11046567 DOI: 10.1080/10253890.2024.2317856] [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: 07/03/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024] Open
Abstract
In humans, exposure to early life adversity has profound implications for susceptibility to developing neuropsychiatric disorders later in life. Studies in rodents have shown that stress experienced during early postnatal life can have lasting effects on brain development. Glucocorticoids and sex steroids are produced in endocrine glands and the brain from cholesterol; these molecules bind to nuclear and membrane-associated steroid receptors. Unlike other steroids that can also be made in the brain, neurosteroids bind specifically to neurotransmitter receptors, not steroid receptors. The relationships among steroids, neurosteroids, and stress are multifaceted and not yet fully understood. However, studies demonstrating altered levels of progestogens, androgens, estrogens, glucocorticoids, and their neuroactive metabolites in both developmental and adult stress paradigms strongly suggest that these molecules may be important players in stress effects on brain circuits and behavior. In this review, we discuss the influence of developmental and adult stress on various components of the brain, including neurons, glia, and perineuronal nets, with a focus on sex steroids and neurosteroids. Gaining an enhanced understanding of how early adversity impacts the intricate systems of brain steroid and neurosteroid regulation could prove instrumental in identifying novel therapeutic targets for stress-related conditions.
Collapse
Affiliation(s)
- Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| |
Collapse
|
10
|
Kim S, Kubelka NK, LaPorte HM, Krishnamoorthy VR, Singh M. Estradiol and 3β-diol protect female cortical astrocytes by regulating connexin 43 Gap Junctions. Mol Cell Endocrinol 2023; 578:112045. [PMID: 37595662 PMCID: PMC10592012 DOI: 10.1016/j.mce.2023.112045] [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: 03/04/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
While estrogens have been described to protect or preserve neuronal function in the face of insults such as oxidative stress, the prevailing mechanistic model would suggest that these steroids exert direct effects on the neurons. However, there is growing evidence that glial cells, such as astrocytes, are key cellular mediators of protection. Noting that connexin 43 (Cx43), a protein highly expressed in astrocytes, plays a key role in mediating inter-cellular communication, we hypothesized that Cx43 is a target of estradiol (E2), and the estrogenic metabolite of DHT, 3β-diol. Additionally, we sought to determine if either or both of these hormones attenuate oxidative stress-induced cytotoxicity by eliciting a reduction in Cx43 expression or inhibition of Cx43 channel permeability. Using primary cortical astrocytes, we found that E2 and 3β-diol were each protective against the mixed metabolic/oxidative insult, iodoacetic acid (IAA). Moreover, these effects were blocked by estrogen receptor antagonists. However, E2 and 3β-diol did not alter Cx43 mRNA levels in astrocytes but did inhibit IAA-induced Cx43 gap junction opening/permeability. Taken together, these data implicate astrocyte Cx43 gap junction as an understudied mediator of the cytoprotective effects of estrogens in the brain. Given the wide breadth of disease states associated with Cx43 function/dysfunction, further understanding the relationship between gonadal steroids and Cx43 channels may contribute to a better understanding of the biological basis for sex differences in various diseases.
Collapse
Affiliation(s)
- Seongcheol Kim
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Nicholas Knesek Kubelka
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, United States
| | - Heather M LaPorte
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Vignesh R Krishnamoorthy
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States.
| |
Collapse
|
11
|
Ziemka-Nalecz M, Pawelec P, Ziabska K, Zalewska T. Sex Differences in Brain Disorders. Int J Mol Sci 2023; 24:14571. [PMID: 37834018 PMCID: PMC10572175 DOI: 10.3390/ijms241914571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
A remarkable feature of the brain is its sexual dimorphism. Sexual dimorphism in brain structure and function is associated with clinical implications documented previously in healthy individuals but also in those who suffer from various brain disorders. Sex-based differences concerning some features such as the risk, prevalence, age of onset, and symptomatology have been confirmed in a range of neurological and neuropsychiatric diseases. The mechanisms responsible for the establishment of sex-based differences between men and women are not fully understood. The present paper provides up-to-date data on sex-related dissimilarities observed in brain disorders and highlights the most relevant features that differ between males and females. The topic is very important as the recognition of disparities between the sexes might allow for the identification of therapeutic targets and pharmacological approaches for intractable neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106 Warsaw, Poland; (M.Z.-N.); (P.P.); (K.Z.)
| |
Collapse
|
12
|
Reddy DS, Mbilinyi RH, Estes E. Preclinical and clinical pharmacology of brexanolone (allopregnanolone) for postpartum depression: a landmark journey from concept to clinic in neurosteroid replacement therapy. Psychopharmacology (Berl) 2023; 240:1841-1863. [PMID: 37566239 PMCID: PMC10471722 DOI: 10.1007/s00213-023-06427-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
This article describes the critical role of neurosteroids in postpartum depression (PPD) and outlines the landmark pharmacological journey of brexanolone as a first-in-class neurosteroid antidepressant with significant advantages over traditional antidepressants. PPD is a neuroendocrine disorder that affects about 20% of mothers after childbirth and is characterized by symptoms including persistent sadness, fatigue, dysphoria, as well as disturbances in cognition, emotion, appetite, and sleep. The main pathology behind PPD is the postpartum reduction of neurosteroids, referred to as neurosteroid withdrawal, a concept pioneered by our preclinical studies. We developed neurosteroid replacement therapy (NRT) as a rational approach for treating PPD and other conditions related to neurosteroid deficiency, unveiling the power of neurosteroids as novel anxiolytic-antidepressants. The neurosteroid, brexanolone (BX), is a progesterone-derived allopregnanolone that rapidly relieves anxiety and mood deficits by activating GABA-A receptors, making it a transformational treatment for PPD. In 2019, the FDA approved BX, an intravenous formulation of allopregnanolone, as an NRT to treat PPD. In clinical studies, BX significantly improved PPD symptoms within hours of administration, with tolerable side effects including headache, dizziness, and somnolence. We identified the molecular mechanism of BX in a neuronal PPD-like milieu. The mechanism of BX involves activation of both synaptic and extrasynaptic GABA-A receptors, which promote tonic inhibition and serve as a key target for PPD and related conditions. Neurosteroids offer several advantages over traditional antidepressants, including rapid onset, unique mechanism, and lack of tolerance upon repeated use. Some limitations of BX therapy include lack of aqueous solubility, limited accessibility, hospitalization for treatment, lack of oral product, and serious adverse events at high doses. However, the unmet need for synthetic neurosteroids to address this critical condition supersedes these limitations. Recently, we developed novel hydrophilic neurosteroids with a superior profile and improved drug delivery. Overall, approval of BX is a major milestone in the field of neurotherapeutics, paving the way for the development of novel synthetic neurosteroids to treat depression, epilepsy, and status epilepticus.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, 77807, USA.
- Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
| | - Robert H Mbilinyi
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, 77807, USA
| | - Emily Estes
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, 77807, USA
| |
Collapse
|
13
|
Bäckström T, Turkmen S, Das R, Doverskog M, Blackburn TP. The GABA system, a new target for medications against cognitive impairment-Associated with neuroactive steroids. J Intern Med 2023; 294:281-294. [PMID: 37518841 DOI: 10.1111/joim.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
The prevalence of cognitive dysfunction, dementia, and neurodegenerative disorders such as Alzheimer's disease (AD) is increasing in parallel with an aging population. Distinct types of chronic stress are thought to be instrumental in the development of cognitive impairment in central nervous system (CNS) disorders where cognitive impairment is a major unmet medical need. Increased GABAergic tone is a mediator of stress effects but is also a result of other factors in CNS disorders. Positive GABA-A receptor modulating stress and sex steroids (steroid-PAMs) such as allopregnanolone (ALLO) and medroxyprogesterone acetate can provoke impaired cognition. As such, ALLO impairs memory and learning in both animals and humans. In transgenic AD animal studies, continuous exposure to ALLO at physiological levels impairs cognition and increases degenerative AD pathology, whereas intermittent ALLO injections enhance cognition, indicating pleiotropic functions of ALLO. We have shown that GABA-A receptor modulating steroid antagonists (GAMSAs) can block the acute negative cognitive impairment of ALLO on memory in animal studies and in patients with cognitive impairment due to hepatic encephalopathy. Here we describe disorders affected by steroid-PAMs and opportunities to treat these adverse effects of steroid-PAMs with novel GAMSAs.
Collapse
Affiliation(s)
| | - Sahruh Turkmen
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
| | - Roshni Das
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
- Umecrine Cognition AB, Solna, Sweden
| | | | | |
Collapse
|
14
|
Killanin AD, Taylor BK, Embury CM, Picci G, Wang YP, Calhoun VD, Stephen JM, Heinrichs-Graham E, Wilson TW. Testosterone levels mediate the dynamics of motor oscillatory coding and behavior in developing youth. Dev Cogn Neurosci 2023; 61:101257. [PMID: 37236034 PMCID: PMC10232658 DOI: 10.1016/j.dcn.2023.101257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/17/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Recent investigations have studied the development of motor-related oscillatory responses to delineate maturational changes from childhood to young adulthood. While these studies included youth during the pubertal transition period, none have probed the impact of testosterone levels on motor cortical dynamics and performance. We collected salivary testosterone samples and recorded magnetoencephalography during a complex motor sequencing task in 58 youth aged 9-15 years old. The relationships between testosterone, age, task behavior, and beta (15-23 Hz) oscillatory dynamics were examined using multiple mediation modeling. We found that testosterone mediated the effect of age on movement-related beta activity. We also found that the effect of age on movement duration was mediated by testosterone and reaction time. Interestingly, the relationships between testosterone and motor performance were not mediated by beta activity in the left primary motor cortex, which may indicate the importance of higher-order motor regions. Overall, our results suggest that testosterone has unique associations with neural and behavioral indices of complex motor performance, beyond those already characterized in the literature. These findings are the first to link developmental changes in testosterone levels to maturation of beta oscillatory dynamics serving complex motor planning and execution, and specific measures of motor performance.
Collapse
Affiliation(s)
- Abraham D Killanin
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brittany K Taylor
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE, USA
| | - Christine M Embury
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Giorgia Picci
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Yu-Ping Wang
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Vince D Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA, USA
| | | | - Elizabeth Heinrichs-Graham
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE, USA
| | - Tony W Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE, USA.
| |
Collapse
|
15
|
Sivcev S, Kudova E, Zemkova H. Neurosteroids as positive and negative allosteric modulators of ligand-gated ion channels: P2X receptor perspective. Neuropharmacology 2023; 234:109542. [PMID: 37040816 DOI: 10.1016/j.neuropharm.2023.109542] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/06/2023] [Accepted: 04/07/2023] [Indexed: 04/13/2023]
Abstract
Neurosteroids are steroids synthesized de novo in the brain from cholesterol in an independent manner from peripheral steroid sources. The term "neuroactive steroid" includes all steroids independent of their origin, and newly synthesized analogs of neurosteroids that modify neuronal activities. In vivo application of neuroactive steroids induces potent anxiolytic, antidepressant, anticonvulsant, sedative, analgesic and amnesic effects, mainly through interaction with the γ-aminobutyric acid type-A receptor (GABAAR). However, neuroactive steroids also act as positive or negative allosteric regulators on several ligand-gated channels including N-methyl-d-aspartate receptors (NMDARs), nicotinic acetylcholine receptors (nAChRs) and ATP-gated purinergic P2X receptors. Seven different P2X subunits (P2X1-7) can assemble to form homotrimeric or heterotrimeric ion channels permeable for monovalent cations and calcium. Among them, P2X2, P2X4, and P2X7 are the most abundant within the brain and can be regulated by neurosteroids. Transmembrane domains are necessary for neurosteroid binding, however, no generic motif of amino acids can accurately predict the neurosteroid binding site for any of the ligand-gated ion channels including P2X. Here, we will review what is currently known about the modulation of rat and human P2X by neuroactive steroids and the possible structural determinants underlying neurosteroid-induced potentiation and inhibition of the P2X2 and P2X4 receptors.
Collapse
Affiliation(s)
- Sonja Sivcev
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Hana Zemkova
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
16
|
Adenosine receptors participate in anabolic-androgenic steroid-induced changes on risk assessment/anxiety-like behaviors in male and female rats. Physiol Behav 2023; 261:114071. [PMID: 36584765 DOI: 10.1016/j.physbeh.2022.114071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
Anabolic-androgenic steroids (AAS) and caffeine can induce several behavioral alterations in humans and rodents. Administration of nandrolone decanoate is known to affect defensive responses to aversive stimuli, generally decreasing inhibitory control and increasing aggressivity but whether caffeine intake influences behavioral changes induced by AAS is unknown. The present study aimed to investigate behavioral effects of caffeine (a non-selective antagonist of adenosine receptors) alone or combined with nandrolone decanoate (one of the most commonly AAS abused) in female and male Lister Hooded rats. Our results indicated that chronic administration of nandrolone decanoate (10 mg/kg, i.m., once a week for 8 weeks) decreased risk assessment/anxiety-like behaviors (in the elevated plus maze test), regardless of sex. These effects were prevented by combined caffeine intake (0.1 g/L, p.o., ad libitum). Overall, the present study heralds a key role for caffeine intake in the modulation of nandrolone decanoate-induced behavioral changes in rats, suggesting adenosine receptors as candidate targets to manage impact of AAS on brain function and behavior.
Collapse
|
17
|
Fox LC, Scholl JL, Watt MJ, Forster GL. GABA A Receptor and Serotonin Transporter Expression Changes Dissociate Following Mild Traumatic Brain Injury: Influence of Sex and Estrus Cycle Phase in Rats. Neuroscience 2023; 514:38-55. [PMID: 36736883 DOI: 10.1016/j.neuroscience.2023.01.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
Mild traumatic brain injuries (mild TBIs) can affect both males and females, but females are more likely to report long-term psychological complications, including changes in mood and generalized anxiety. Additionally, reproductive cycle phase has been shown to affect mild TBI symptom expression within females. These variances may result from sex differences in mild TBI-induced alterations to neurotransmission in brain regions that influence mood and emotion, possibly mediated by sex steroids. The hippocampus and amygdala are implicated in stress responses and anxiety, and within these regions, gamma-aminobutyric acid (GABA) and serotonin modulate output and behavioral expression. Metabolites of progesterone can allosterically enhance GABAergic signaling, and sex steroids are suggested to regulate the expression of the serotonin transporter (SERT). To determine how mild TBI might alter GABA receptor and SERT expression in males and females, immunocytochemistry was used to quantify expression of the alpha-1 subunit of the GABAA receptor (α1-GABAA), SERT, and a neuronal marker (NeuN) in the brains of adult male and naturally-cycling female rats, both with and without mild TBI, 17 days after injury. Mild TBI altered the expression of α1-GABAA in the amygdala and hippocampus in both sexes, but the direction of change observed depended on sex and reproductive cycle phase. In contrast, mild TBI had little effect on SERT expression. However, SERT expression differed between sexes and varied with the cycle phase. These findings demonstrate that regulation of neurotransmission following mild TBI differs between males and females, with implications for behavioral outcomes and the efficacy of therapeutic strategies.
Collapse
Affiliation(s)
- Laura C Fox
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark St, Vermillion, SD, USA.
| | - Jamie L Scholl
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark St, Vermillion, SD, USA.
| | - Michael J Watt
- Center for Brain and Behavior Research, Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Gina L Forster
- Center for Brain and Behavior Research, Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
18
|
Golub V, Ramakrishnan S, Reddy DS. Isobolographic analysis of adjunct antiseizure activity of the FDA-approved cannabidiol with neurosteroids and benzodiazepines in adult refractory focal onset epilepsy. Exp Neurol 2023; 360:114294. [PMID: 36493860 PMCID: PMC9884179 DOI: 10.1016/j.expneurol.2022.114294] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/27/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Epilepsy is a serious neurological disorder associated with recurrent and unpredictable seizures and extensive neuropsychiatric comorbidities. There is no cure for epilepsy, and over one third of epileptic patients have been diagnosed with drug-refractory epilepsy, indicating the critical need for novel antiseizure medications (ASMs). Cannabidiol (CBD) has been shown to decrease seizures in pediatric epilepsies, such as Dravet and Lennox-Gastaut syndromes; however, it has not been rigorously tested for adult seizures or in models of refractory focal epilepsy. Although the exact mechanism is unknown, it is likely to act in a way that is unique to certain GABA-A receptor-modulating drugs, such as neurosteroids and benzodiazepines. In this study, we sought to determine the adjunct antiseizure activity of a clinical CBD product in an adult 6-Hz model of focal refractory epilepsy. CBD was evaluated alone in both a dose-response and time-course manner and in an adjunct combination with two ASMs ganaxolone (neurosteroid) and midazolam (benzodiazepine) against 6-Hz-induced refractory focal onset, generalized seizures. In pharmacological studies, CBD produced dose-dependent protection against seizures (ED50, 53 mg/kg, i.p.) without any side effects. CBD significantly reduced both electrographic activity and behavioral ictal responses with no apparent sex differences. CBD was evaluated in an isobologram design in conjunction with ganaxolone or midazolam at three standard ratios (1:1, 1:3, 3:1). Isobolographic analysis shows the combination regimens of CBD + ganaxolone and CBD + midazolam exerted combination index of 0.313 and 0.164, indicating strong synergism for seizure protection, with little to no toxicity. Together, these results demonstrate the therapeutic potential of CBD monotherapy and as an adjunct therapy for adult focal refractory epilepsy in combination with GABAergic ASMs.
Collapse
Affiliation(s)
- Victoria Golub
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA; Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University, Bryan, TX, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA; Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
19
|
Efficacy of the FDA-approved cannabidiol on the development and persistence of temporal lobe epilepsy and complex focal onset seizures. Exp Neurol 2023; 359:114240. [PMID: 36216124 DOI: 10.1016/j.expneurol.2022.114240] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/09/2022]
Abstract
Presently there is no drug therapy for curing epilepsy. Despite many advancements in epilepsy research, nearly 30% of people with epilepsy remain refractory to current antiseizure medications (ASM). Cannabidiol (CBD) has recently been approved as an ASM for pediatric refractory seizures, but it has not been widely tested for adult epileptogenesis and focal onset seizures. In this study, we investigated the efficacy of the FDA-approved CBD in controlling epileptogenesis and complex focal onset seizures using the mouse kindling model of human temporal lobe epilepsy. We also tested combination regimens of CBD with other ASMs. The two primary outcome measures were disease modification and suppression of generalized seizures. In the epileptogenesis study, CBD had a striking effect in attenuating kindling development, with a dose-dependent decrease in behavioral and electrographic seizure activity. In the retention study, mice previously treated with CBD had significantly reduced overall seizure burden, suggesting disease modification. In a fully-kindled seizure study, CBD produced rapid and atypical U-shaped dose-dependent protection against generalized seizures (ED50, 52 mg/kg, i.p.). In a time-course study, CBD showed a maximal protective effect within 1 h of injection, and it declined within 4 h with a biphasic response. In the combination study, CBD produced synergistic/ additive protection when given with midazolam and ganaxolone but not with tiagabine, indicating its strong potential as an adjunct ASM. Finally, the protective effects of CBD were not associated with motor and functional impairments. These preclinical findings demonstrate the potential of adjunct CBD for controlling adult complex focal onset seizure conditions.
Collapse
|
20
|
Bakalar D, O’Reilly JJ, Lacaille H, Salzbank J, Ellegood J, Lerch JP, Sasaki T, Imamura Y, Hashimoto-Torii K, Vacher CM, Penn AA. Lack of placental neurosteroid alters cortical development and female somatosensory function. Front Endocrinol (Lausanne) 2022; 13:972033. [PMID: 36313771 PMCID: PMC9606442 DOI: 10.3389/fendo.2022.972033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 01/24/2023] Open
Abstract
Placental endocrine function is essential to fetal brain development. Placental hormones include neurosteroids such as allopregnanolone (ALLO), a regulator of neurodevelopmental processes via positive allosteric modulation of the GABAA receptor (GABAA-R). Using a mouse model (plKO) in which the gene encoding the ALLO synthesis enzyme is specifically deleted in trophoblasts, we previously showed that placental ALLO insufficiency alters cerebellar white matter development and leads to male-specific autistic-like behavior. We now demonstrate that the lack of placental ALLO causes female-predominant alterations of cortical development and function. Placental ALLO insufficiency disrupts cell proliferation in the primary somatosensory cortex (S1) in a sex-linked manner. Early changes are seen in plKO embryos of both sexes, but persist primarily in female offspring after birth. Adolescent plKO females show significant reduction in pyramidal neuron density, as well as somatosensory behavioral deficits as compared with plKO males and control littermates. Assessment of layer-specific markers in human postmortem cortices suggests that preterm infants may also have female-biased abnormalities in cortical layer specification as compared with term infants. This study establishes a novel and fundamental link between placental function and sex-linked long-term neurological outcomes, emphasizing the importance of the growing field of neuroplacentology.
Collapse
Affiliation(s)
- Dana Bakalar
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Health System, Washington, DC, United States
| | - Jiaqi J. O’Reilly
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Helene Lacaille
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Jacquelyn Salzbank
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON, Canada
| | - Jason P. Lerch
- Wellcome Centre for Integrative Neuroimaging (WIN), Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Toru Sasaki
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Health System, Washington, DC, United States
| | - Yuka Imamura
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Health System, Washington, DC, United States
| | - Claire-Marie Vacher
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Anna A. Penn
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
21
|
Medel-Matus JS, Orozco-Suárez S, Escalante RG. Factors not considered in the study of drug-resistant epilepsy: Psychiatric comorbidities, age, and gender. Epilepsia Open 2022. [PMID: 34967149 DOI: 10.1002/epi4.12576.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
In basic research and clinical practice, the control of seizures has been the most important goal, but it should not be the only one. There are factors that remain poorly understood in the study of refractory epilepsy such as the age and gender of patients and the presence of psychiatric comorbidities. It is known that in patients with drug-resistant epilepsy (DRE), the comorbidities contribute to the deterioration of the quality of life, increase the severity, and worsen the prognosis of epilepsy. Some studies have demonstrated that patients diagnosed with a co-occurrence of epilepsy and psychiatric disorders are more likely to present refractory seizures and the probability of seizure remission after pharmacotherapy is reduced. The evidence of this association suggests the presence of shared pathogenic mechanisms that may include endocrine disorders, neuroinflammatory processes, disturbances of neurotransmitters, and mechanisms triggered by stress. Additionally, significant demographic, clinical, and electrographic differences have been observed between women and men with epilepsy. Epilepsy affects the female gender in a greater proportion, although there are no studies that report whether refractoriness affects more females. The reasons behind these sex differences are unclear; however, it is likely that sex hormones and sex brain differences related to chromosomal genes play an important role. On the other hand, it has been shown in industrialized countries that prevalence of DRE is higher in the elderly when compared to youngsters. Conversely, this phenomenon is not observed in developing regions, where more cases are found in children and young adults. The correct identification and management of these factors is crucial in order to improve the quality of life of the patients.
Collapse
Affiliation(s)
- Jesús Servando Medel-Matus
- Department of Pediatrics, Neurology Division, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Sandra Orozco-Suárez
- Unit of Medical Research in Neurological Diseases, Specialty Hospital "Dr. Bernardo Sepúlveda", National Medical Center S.XXI, Mexico City, Mexico
| | - Ruby G Escalante
- Department of Pediatrics, Neurology Division, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
22
|
Medel‐Matus JS, Orozco‐Suárez S, Escalante RG. Factors not considered in the study of drug-resistant epilepsy: Psychiatric comorbidities, age, and gender. Epilepsia Open 2022; 7 Suppl 1:S81-S93. [PMID: 34967149 PMCID: PMC9340311 DOI: 10.1002/epi4.12576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 11/10/2022] Open
Abstract
In basic research and clinical practice, the control of seizures has been the most important goal, but it should not be the only one. There are factors that remain poorly understood in the study of refractory epilepsy such as the age and gender of patients and the presence of psychiatric comorbidities. It is known that in patients with drug-resistant epilepsy (DRE), the comorbidities contribute to the deterioration of the quality of life, increase the severity, and worsen the prognosis of epilepsy. Some studies have demonstrated that patients diagnosed with a co-occurrence of epilepsy and psychiatric disorders are more likely to present refractory seizures and the probability of seizure remission after pharmacotherapy is reduced. The evidence of this association suggests the presence of shared pathogenic mechanisms that may include endocrine disorders, neuroinflammatory processes, disturbances of neurotransmitters, and mechanisms triggered by stress. Additionally, significant demographic, clinical, and electrographic differences have been observed between women and men with epilepsy. Epilepsy affects the female gender in a greater proportion, although there are no studies that report whether refractoriness affects more females. The reasons behind these sex differences are unclear; however, it is likely that sex hormones and sex brain differences related to chromosomal genes play an important role. On the other hand, it has been shown in industrialized countries that prevalence of DRE is higher in the elderly when compared to youngsters. Conversely, this phenomenon is not observed in developing regions, where more cases are found in children and young adults. The correct identification and management of these factors is crucial in order to improve the quality of life of the patients.
Collapse
Affiliation(s)
- Jesús Servando Medel‐Matus
- Department of PediatricsNeurology DivisionDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| | - Sandra Orozco‐Suárez
- Unit of Medical Research in Neurological DiseasesSpecialty Hospital “Dr. Bernardo Sepúlveda”National Medical Center S.XXIMexico CityMexico
| | - Ruby G. Escalante
- Department of PediatricsNeurology DivisionDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
23
|
Waldman G, Benson R. Epilepsy Care in Transgender Patients. Curr Neurol Neurosci Rep 2022; 22:451-458. [PMID: 35666366 DOI: 10.1007/s11910-022-01208-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW The goal of this review is to outline the main considerations when treating transgender patients with epilepsy. Points to be addressed include the gender affirming hormone therapy regimens and how they interact with anti-seizure medications and seizure control, as well as common co-morbidities in the transgender epilepsy population. RECENT FINDINGS Gender affirming hormone therapy (GAHT) may affect seizure control directly, due to proconvulsant or anticonvulsant properties. GAHT may interact with anti-seizure medications; most notably, estrogen will decrease serum concentration of lamotrigine. Enzyme-inducing anti-seizure medications may decrease hormone levels, potentially interfering with goals of GAHT. Transgender epilepsy patients are at risk for co-morbidities such as decreased bone mineral density and depression. There are minimal direct studies on treatment or outcomes in the transgender epilepsy population. Providers must be knowledgeable about the bi-directional interactions between gender affirming hormone therapy and anti-seizure medications, as well as direct hormonal influences on seizure control. Future research should directly evaluate outcomes in transgender epilepsy patients with regard to seizure control, success of hormone therapy, and management of co-morbidities, to further educate providers and patients how to best manage their healthcare.
Collapse
Affiliation(s)
- Genna Waldman
- Department of Neurology, Columbia University, 710 W. 168th Street, 7th floor, New York, NY, 10032, USA.,New York Presbyterian Hospital, 630 West 168th Street , New York, NY, 10032, USA
| | - Rachael Benson
- Department of Neurology, Columbia University, 710 W. 168th Street, 7th floor, New York, NY, 10032, USA.
| |
Collapse
|
24
|
Evaluation of the effect of nicotine and O-acetyl-L-carnitine on testosterone-induced spatial learning impairment in Morris water maze and assessment of protein markers. LEARNING AND MOTIVATION 2022. [DOI: 10.1016/j.lmot.2022.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Panichi R, Dieni CV, Sullivan JA, Biscarini A, Contemori S, Faralli M, Pettorossi VE. Inhibition of androgenic pathway impairs encoding of cerebellar‐dependent motor learning in male rats. J Comp Neurol 2022; 530:2014-2032. [DOI: 10.1002/cne.25318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Roberto Panichi
- Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Cristina V. Dieni
- Department of Neurobiology and Evelyn McKnight Brain Institute University of Alabama at Birmingham Birmingham Alabama USA
| | | | - Andrea Biscarini
- Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Samuele Contemori
- Center for Sensorimotor Performance, School of Human Movement and Nutrition Sciences The University of Queensland Brisbane Queensland Australia
| | - Mario Faralli
- Department of Medical‐Surgical Specialization, Otolaryngology and Cervicofacial Surgery Division University of Perugia Perugia Italy
| | | |
Collapse
|
26
|
Reddy DS. Neurosteroid replacement therapy for catamenial epilepsy, postpartum depression and neuroendocrine disorders in women. J Neuroendocrinol 2022; 34:e13028. [PMID: 34506047 PMCID: PMC9247111 DOI: 10.1111/jne.13028] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022]
Abstract
Neurosteroids are involved in the pathophysiology of many neuroendocrine disorders in women. This review describes recent advancements in pharmacology of neurosteroids and emphasizes the benefits of neurosteroid replacement therapy for the management of neuroendocrine disorders such as catamenial epilepsy (CE), postpartum depression (PPD) and premenstrual brain conditions. Neurosteroids are endogenous modulators of neuronal excitability. A variety of neurosteroids are present in the brain including allopregnanolone (AP), allotetrahydro-deoxycorticosterone and androstanediol. Neurosteroids interact with synaptic and extrasynaptic GABAA receptors in the brain. AP and related neurosteroids, which are positive allosteric modulators of GABAA receptors, are powerful anticonvulsants, anxiolytic, antistress and neuroprotectant agents. In CE, seizures are most often clustered around a specific menstrual period in women. Neurosteroid withdrawal-linked plasticity in extrasynaptic receptors has been shown to play a key role in catamenial seizures, anxiety and other mood disorders. Based on our extensive research spanning two decades, we have proposed and championed neurosteroid replacement therapy as a rational strategy for treating disorders marked by neurosteroid-deficiency, such as CE and other related ovarian or menstrual disorders. In 2019, AP (renamed as brexanolone) was approved for treating PPD. A variety of synthetic neurosteroids are in clinical trials for epilepsy, depression and other brain disorders. Recent advancements in our understanding of neurosteroids have entered a new era of drug discovery and one that offers a high therapeutic potential for treating complex brain disorders.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA
| |
Collapse
|
27
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
28
|
Pahwa P, Singh T, Goel RK. Anticonvulsant Effect of Asparagus racemosus Willd. in a Mouse Model of Catamenial Epilepsy. Neurochem Res 2021; 47:422-433. [PMID: 34623565 DOI: 10.1007/s11064-021-03455-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/05/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Asparagus racemosus Willd. (Family Liliaceae), also known as female reproductive tonic, is traditionally used across the Sub-Himalayan region in Uttarakhand, India for treatment of epilepsy and disorders of female reproductive system. Therefore, in this study, we investigated the anticonvulsant effect of A. racemosus in a mouse model of catamenial epilepsy. We artificially increased progesterone and neurosteroid levels (a state of pseudo-pregnancy) in adult Swiss albino female mice by injecting pregnant mares' serum gonadotropin (PMSG) (5 IU s.c.), followed by human chorionic gonadotropin (HCG) (5 IU s.c.) after 46 h. In the following 10 days, A. racemosus treatment was given along with measurement of progesterone, estradiol, and corticosterone levels in the blood. Neurosteroid withdrawal was induced by finasteride (50 mg/kg, i.p.) on treatment day 9. Twenty-four hours after finasteride administration (day 10 of treatment), seizure susceptibility was evaluated with the sub-convulsant pentylenetetrazole (PTZ) dose (40 mg/kg i.p.). Four hours after PTZ, animals were assessed for depression like phenotypes followed by euthanasia and separation of brain parts (cortex and hippocampus). The results showed that PMSG and HCG treatment elevated progesterone and estradiol levels. Treatment with finasteride increased seizure susceptibility and depression due to decreased progesterone and elevated estrogen levels coupled with decreased monoamine and elevated corticosterone levels. A. racemosus treatment, on the other hand, significantly decreased seizure susceptibility and depression like behaviors, possibly because of increased progesterone, restored estradiol, corticosterone, and monoamine levels. We concluded that herbal formulations using A. racemosus root extracts may be used as monotherapy or adjuvant therapy along with available AEDs for the better and safe management of catamenial epilepsy as well as comorbid depression.
Collapse
Affiliation(s)
- Priyanka Pahwa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Tanveer Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.,Department of Neuroscience and Experimental Therapeutics, Health Science Center, College of Medicine, Texas A&M University, Bryan, TX, 77807, USA
| | - Rajesh Kumar Goel
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
29
|
Le déficit en testostérone en pratique sexologique. Populations cibles et signes cliniques évocateurs. SEXOLOGIES 2021. [DOI: 10.1016/j.sexol.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
30
|
Reddy DS. Brain structural and neuroendocrine basis of sex differences in epilepsy. HANDBOOK OF CLINICAL NEUROLOGY 2021; 175:223-233. [PMID: 33008527 DOI: 10.1016/b978-0-444-64123-6.00016-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
This chapter reviews the current information about sex differences in epilepsy and potential mechanisms underlying sex differences in seizure susceptibility and epilepsy. The susceptibility to and occurrence of seizures are generally higher in men than women. There is gender-specific epilepsies such as catamenial epilepsy, a neuroendocrine condition in which seizures are most often clustered around the perimenstrual or periovulatory period in adult women. Structural differences in cerebral morphology, the structural and functional circuits may render men and women differentially vulnerable to seizure disorders and epileptogenic processes. Changes in seizure sensitivity are evident at puberty, pregnancy, and menopause, often attributed to circulating steroid hormones and neurosteroids as well as neuroplasticity in receptor systems. An improved understanding of the sexual dimorphism in neural circuits and the neuroendocrine basis of sex differences or resistance to protective drugs is essential to develop sex-specific therapies for seizure conditions.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, United States.
| |
Collapse
|
31
|
Reddy DS, Thompson W, Calderara G. Molecular mechanisms of sex differences in epilepsy and seizure susceptibility in chemical, genetic and acquired epileptogenesis. Neurosci Lett 2021; 750:135753. [PMID: 33610673 PMCID: PMC7994197 DOI: 10.1016/j.neulet.2021.135753] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
This article provides a succinct overview of sex differences in epilepsy and putative molecular mechanisms underlying sex differences in seizure susceptibility in chemical, genetic, and acquired epileptogenesis. The susceptibility to excitability episodes and occurrence of epileptic seizures are generally higher in men than women. The precise molecular mechanisms remain unclear, but differences in regional morphology and neural circuits in men and women may explain differential vulnerability to seizures and epileptogenic cascades. Changes in seizure sensitivity can be attributed to steroid hormones, including fluctuations in neurosteroids as well as neuroplasticity in their receptor signaling systems. Other potential neurobiological bases for sex differences in epilepsies include differences in brain development, neurogenesis, neuronal chloride homeostasis, and neurotrophic and glial responses. In catamenial epilepsy, a gender-specific neuroendocrine condition, epileptic seizures are most often clustered around a specific menstrual period in adult women. A deeper understanding of the molecular and neural network basis of sex differences in seizures and response to antiepileptic drugs is highly warranted for designing effective, sex-specific therapies for epilepsy, epileptogenesis, and seizure disorders.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States.
| | - Wesley Thompson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| | - Gianmarco Calderara
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| |
Collapse
|
32
|
Janković SM, Dješević M, Janković SV. Experimental GABA A Receptor Agonists and Allosteric Modulators for the Treatment of Focal Epilepsy. J Exp Pharmacol 2021; 13:235-244. [PMID: 33727865 PMCID: PMC7954424 DOI: 10.2147/jep.s242964] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/23/2021] [Indexed: 12/16/2022] Open
Abstract
GABA A receptors are ubiquitous in the central nervous system and there is a huge diversity of receptor subtypes in almost all regions of the brain. However, the expression of GABA A receptor subtypes is altered in both the gray and white matter of patients with focal epilepsy. Although there is a number of anticonvulsants with marketing authorization for the treatment of focal epilepsy which act through GABA A receptors, potentiating the inhibitory effects of GABA, it is necessary to develop more potent and more specific GABAergic anticonvulsants that are effective in drug-resistant patients with focal epilepsy. There are three orthosteric and at least seven allosteric agonist binding sites at the GABA A receptor. In experimental and clinical studies, full agonists of GABA A receptors showed a tendency to cause desensitization of the receptors, tolerance, and physical dependence; therefore, partial orthosteric agonists and positive allosteric modulators of GABA A receptors were further developed. Preclinical studies demonstrated the anticonvulsant efficacy of positive allosteric modulators with selective action on GABA A receptors with α2/α3 subunits, but only a handful of them were further tested in clinical trials. The best results were obtained for clobazam (already marketed), ganaxolone (in phase III trials), CVL-865 (in phase II trials), and padsevonil (in phase III trials). Several compounds with more selective action on GABA A receptors, perhaps only in certain brain regions, have the potential to become effective drugs against specific subtypes of focal-onset epilepsy. However, their development needs time, and in the near future we can expect only one or two new GABA A agonists to obtain marketing authorization for focal epilepsy, an advance that would be of use for just a fraction of patients with drug-resistant epilepsy.
Collapse
Affiliation(s)
| | - Miralem Dješević
- Cardiology Department, Private Policlinic Center Eurofarm, Sarajevo, Bosnia and Hercegovina
| | - Snežana V Janković
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
33
|
Androgen Affects the Inhibitory Avoidance Memory by Primarily Acting on Androgen Receptor in the Brain in Adolescent Male Rats. Brain Sci 2021; 11:brainsci11020239. [PMID: 33672867 PMCID: PMC7918178 DOI: 10.3390/brainsci11020239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 11/17/2022] Open
Abstract
Adolescence is the critical postnatal stage for the action of androgen in multiple brain regions. Androgens can regulate the learning/memory functions in the brain. It is known that the inhibitory avoidance test can evaluate emotional memory and is believed to be dependent largely on the amygdala and hippocampus. However, the effects of androgen on inhibitory avoidance memory have never been reported in adolescent male rats. In the present study, the effects of androgen on inhibitory avoidance memory and on androgen receptor (AR)-immunoreactivity in the amygdala and hippocampus were studied using behavioral analysis, Western blotting and immunohistochemistry in sham-operated, orchiectomized, orchiectomized + testosterone or orchiectomized + dihydrotestosterone-administered male adolescent rats. Orchiectomized rats showed significantly reduced time spent in the illuminated box after 30 min (test 1) or 24 h (test 2) of electrical foot-shock (training) and reduced AR-immunoreactivity in amygdala/hippocampal cornu Ammonis (CA1) in comparison to those in sham-operated rats. Treatment of orchiectomized rats with either non-aromatizable dihydrotestosterone or aromatizable testosterone were successfully reinstated these effects. Application of flutamide (AR-antagonist) in intact adolescent rats exhibited identical changes to those in orchiectomized rats. These suggest that androgens enhance the inhibitory avoidance memory plausibly by binding with AR in the amygdala and hippocampus.
Collapse
|
34
|
Christian CA, Reddy DS, Maguire J, Forcelli PA. Sex Differences in the Epilepsies and Associated Comorbidities: Implications for Use and Development of Pharmacotherapies. Pharmacol Rev 2021; 72:767-800. [PMID: 32817274 DOI: 10.1124/pr.119.017392] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epilepsies are common neurologic disorders characterized by spontaneous recurrent seizures. Boys, girls, men, and women of all ages are affected by epilepsy and, in many cases, by associated comorbidities as well. The primary courses of treatment are pharmacological, dietary, and/or surgical, depending on several factors, including the areas of the brain affected and the severity of the epilepsy. There is a growing appreciation that sex differences in underlying brain function and in the neurobiology of epilepsy are important factors that should be accounted for in the design and development of new therapies. In this review, we discuss the current knowledge on sex differences in epilepsy and associated comorbidities, with emphasis on those aspects most informative for the development of new pharmacotherapies. Particular focus is placed on sex differences in the prevalence and presentation of various focal and generalized epilepsies; psychiatric, cognitive, and physiologic comorbidities; catamenial epilepsy in women; sex differences in brain development; the neural actions of sex and stress hormones and their metabolites; and cellular mechanisms, including brain-derived neurotrophic factor signaling and neuronal-glial interactions. Further attention placed on potential sex differences in epilepsies, comorbidities, and drug effects will enhance therapeutic options and efficacy for all patients with epilepsy. SIGNIFICANCE STATEMENT: Epilepsy is a common neurological disorder that often presents together with various comorbidities. The features of epilepsy and seizure activity as well as comorbid afflictions can vary between men and women. In this review, we discuss sex differences in types of epilepsies, associated comorbidities, pathophysiological mechanisms, and antiepileptic drug efficacy in both clinical patient populations and preclinical animal models.
Collapse
Affiliation(s)
- Catherine A Christian
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Doodipala Samba Reddy
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Jamie Maguire
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Patrick A Forcelli
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| |
Collapse
|
35
|
Rehbein E, Hornung J, Sundström Poromaa I, Derntl B. Shaping of the Female Human Brain by Sex Hormones: A Review. Neuroendocrinology 2021; 111:183-206. [PMID: 32155633 DOI: 10.1159/000507083] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/09/2020] [Indexed: 12/26/2022]
Abstract
Traditionally sex hormones have been associated with reproductive and developmental processes only. Since the 1950s we know that hormones can have organizational effects on the developing brain and initiate hormonal transition periods such as puberty. However, recent evidence shows that sex hormones additionally structure the brain during important hormonal transition periods across a woman's life including short-term fluctuations during the menstrual cycle. However, a comprehensive review focusing on structural changes during all hormonal transition phases of women is still missing. Therefore, in this review structural changes across hormonal transition periods (i.e., puberty, menstrual cycle, oral contraceptive intake, pregnancy and menopause) were investigated in a structured way and correlations with sex hormones evaluated. Results show an overall reduction in grey matter and region-specific decreases in prefrontal, parietal and middle temporal areas during puberty. Across the menstrual cycle grey matter plasticity in the hippocampus, the amygdala as well as temporal and parietal regions were most consistently reported. Studies reporting on pre- and post-pregnancy measurements revealed volume reductions in midline structures as well as prefrontal and temporal cortices. During perimenopause, the decline in sex hormones was paralleled with a reduction in hippocampal and parietal cortex volume. Brain volume changes were significantly correlated with estradiol, testosterone and progesterone levels in some studies, but directionality remains inconclusive between studies. These results indicate that sex hormones play an important role in shaping women's brain structure during different transition periods and are not restricted to specific developmental periods.
Collapse
Affiliation(s)
- Elisa Rehbein
- Department of Psychiatry and Psychotherapy, Innovative Neuroimaging, University of Tübingen, Tübingen, Germany,
| | - Jonas Hornung
- Department of Psychiatry and Psychotherapy, Innovative Neuroimaging, University of Tübingen, Tübingen, Germany
| | | | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, Innovative Neuroimaging, University of Tübingen, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- Lead Graduate School, University of Tübingen, Tübingen, Germany
| |
Collapse
|
36
|
Chew L, Sun KL, Sun W, Wang Z, Rajadas J, Flores RE, Arnold E, Jo B, Fung LK. Association of serum allopregnanolone with restricted and repetitive behaviors in adult males with autism. Psychoneuroendocrinology 2021; 123:105039. [PMID: 33161257 PMCID: PMC8428554 DOI: 10.1016/j.psyneuen.2020.105039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/02/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022]
Abstract
Autism spectrum disorder (ASD) has been associated with imbalance between excitatory and inhibitory (E/I) neurotransmission systems, as well as with neuroinflammation. Sitting at the crossroads between E/I imbalance and neuroinflammation is a class of endogenous hormones known as neurosteroids. Current literature points to dysregulated steroid metabolism and atypical neurosteroid levels in ASD as early as in utero. However, due to the complexity of neurosteroid metabolomics, including possible sex differences, the impact of neurosteroids on ASD symptomatology remains unclear. In this study, we assessed neurosteroid levels and ASD symptom severity of 21 males with ASD and 20 full-scale-IQ-matched typically developing (TD) males, all aged 18-39. Using liquid chromatography-tandem mass spectrometry, concentrations of allopregnanolone, cortisol, dehydroepiandrosterone, progesterone, and testosterone were measured in saliva and serum. With the exception of cortisol's, all neurosteroids' concentrations were found to have ASD vs. TD group differences in distribution, where one group was normally distributed and the other non-normally distributed. Serum allopregnanolone levels in males with ASD were found to negatively correlate with clinician-rated measures of restricted and repetitive behavior measures (ADOS-2 RRB and ADI-R RRSB domain scores). Additionally, lower serum allopregnanolone levels were found to predict more negative camouflaging scores, which represent greater differences in self- and clinician-rated symptom severity, of both ASD symptomatology overall and repetitive behaviors in particular. Taken together, our findings demonstrate that in adult males with ASD, decreased serum allopregnanolone levels are associated with more severe restricted and repetitive behaviors and with less insight into the severity of these behaviors.
Collapse
Affiliation(s)
- Leila Chew
- Department of Psychiatry & Behavioral Sciences, Stanford University, California, USA; David Geffen School of Medicine, University of California at Los Angeles, California, USA
| | - Kevin L Sun
- Department of Psychiatry & Behavioral Sciences, Stanford University, California, USA
| | - Wenchao Sun
- Biomaterial and Advanced Drug Delivery Lab, Stanford University, California, USA
| | - Zhe Wang
- Biomaterial and Advanced Drug Delivery Lab, Stanford University, California, USA
| | - Jayakumar Rajadas
- Biomaterial and Advanced Drug Delivery Lab, Stanford University, California, USA
| | - Ryan E Flores
- Department of Psychiatry & Behavioral Sciences, Stanford University, California, USA; Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Emily Arnold
- Department of Psychiatry & Behavioral Sciences, Stanford University, California, USA
| | - Booil Jo
- Department of Psychiatry & Behavioral Sciences, Stanford University, California, USA
| | - Lawrence K Fung
- Department of Psychiatry & Behavioral Sciences, Stanford University, California, USA.
| |
Collapse
|
37
|
Taubøll E, Isojärvi JIT, Herzog AG. The interactions between reproductive hormones and epilepsy. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:155-174. [PMID: 34266590 DOI: 10.1016/b978-0-12-819973-2.00011-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There are complex interactions between hormones, epilepsy, and antiepileptic drugs (AEDs). While there is ample evidence that hormones influence epilepsy, it is also apparent that epileptic activity influences hormones in both women and men. In addition, AEDs may disturb endocrine function. The clinical importance of these interactions is primarily related to the effects on reproductive hormones, which is the focus of this article. Reproductive endocrine dysfunction is common among women and men with epilepsy. Menstrual disorders, polycystic ovaries, and infertility have been described among women with epilepsy, while reduced potency and sperm abnormalities have been found in men. Sexual problems and endocrine changes have been frequently described in both sexes. Epilepsy and AEDs can target a number of substrates to impact hormone levels. These include the limbic system, hypothalamus, pituitary, peripheral endocrine glands, liver, and adipose tissue. AEDs may also alter the synthesis of steroids and binding proteins, as well as hormone metabolism, and produce direct gonadal effects.
Collapse
Affiliation(s)
- Erik Taubøll
- Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway.
| | | | - Andrew G Herzog
- Harvard Neuroendocrine Unit, Beth Israel Deaconess Medical Center, Boston, MA, United States; Faculty of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
38
|
Li S, Zhang L, Wei N, Tai Z, Yu C, Xu Z. Research Progress on the Effect of Epilepsy and Antiseizure Medications on PCOS Through HPO Axis. Front Endocrinol (Lausanne) 2021; 12:787854. [PMID: 34992582 PMCID: PMC8726549 DOI: 10.3389/fendo.2021.787854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Epilepsy is a common chronic neurological disease that manifests as recurrent seizures. The incidence and prevalence of epilepsy in women are slightly lower than those in men. Polycystic ovary syndrome (PCOS), a reproductive endocrine system disease, is a complication that women with epilepsy are susceptible to, and its total prevalence is 8%-13% in the female population and sometimes as high as 26% in female epilepsy patients. The rate of PCOS increased markedly in female patients who chose valproate (VPA), to 1.95 times higher than that of other drugs. In addition, patients receiving other anti-seizure medications (ASMs), such as lamotrigine (LTG), oxcarbazepine (OXC), and carbamazepine (CBZ), also have reproductive endocrine abnormalities. Some scholars believe that the increase in incidence is related not only to epilepsy itself but also to ASMs. Epileptiform discharges can affect the activity of the pulse generator and then interfere with the reproductive endocrine system by breaking the balance of the hypothalamic-pituitary-ovarian (HPO) axis. ASMs may also cause PCOS-like disorders of the reproductive endocrine system through the HPO axis. Moreover, other factors such as hormone metabolism and related signalling pathways also play a role in it.
Collapse
Affiliation(s)
| | | | | | | | | | - Zucai Xu
- *Correspondence: Changyin Yu, ; Zucai Xu,
| |
Collapse
|
39
|
Advances in Knowledge of Androgens: How Intentional and Accidental Neurosteroid Changes Inform Us of Their Action and Role. CURRENT SEXUAL HEALTH REPORTS 2020. [DOI: 10.1007/s11930-020-00276-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Abstract
Purpose of Review
Here, we summarize current knowledge of androgens’ action gained over the recent years.
Recent Findings
Neurosteroids are produced in the brain and peripheral nerves, independent of endocrine glands have been investigated for how they are regulated, and have actions via non-steroid receptor targets to mediate social, affective, and cognitive behavior and to protect the brain. Androgens’ organizing actions in the peri-natal period have effects throughout the lifetime that may be recapitulated later in life during critical periods and at times of challenge. Developmental changes in androgens occur during mid-childhood, adrenarche, puberty, adolescence, young adulthood, middle age, and andropause. Changes in androgens with a 5α-reductase inhibitor, such as finasteride, result in disruptions in organizational and activational functions of androgens that can be unremitting.
Summary
Normal developmental or perturbation in androgens through other means can cause changes in androgen-sensitive phenotypes throughout the lifespan, in part through actions of neurosteroids.
Collapse
|
40
|
Swink JM, Rings LM, Snyder HA, McAuley RC, Burns TA, Dembek KA, Gilsenan WF, Browne N, Toribio RE. Dynamics of androgens in healthy and hospitalized newborn foals. J Vet Intern Med 2020; 35:538-549. [PMID: 33277956 PMCID: PMC7848305 DOI: 10.1111/jvim.15974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 01/25/2023] Open
Abstract
Background Information on steroids derived from the adrenal glands, gonads, or fetoplacental unit is minimal in newborn foals. Objective To measure androgen concentrations in serum and determine their association with disease severity and outcome in hospitalized foals. Animals Hospitalized (n = 145) and healthy (n = 80) foals. Methods Prospective, multicenter, cross‐sectional study. Foals of ≤3 days of age from 3 hospitals and horse farms were classified as healthy and hospitalized (septic, sick nonseptic, neonatal maladjustment syndrome [NMS]) based on physical exam, medical history, and laboratory findings. Serum androgen and plasma ACTH concentrations were measured with immunoassays. Data were analyzed by nonparametric methods and univariate analysis. Results Serum dehydroepiandrosterone (DHEA), androstenedione, testosterone, and dihydrotestosterone (DHT) concentrations were higher upon admission in hospitalized foals (P < .05), were associated with nonsurvival, decreased to 4.9‐10.8%, 5.7‐31%, and 30.8‐62.8% admission values in healthy, SNS, and septic foals, respectively (P < .05), but remained unchanged or increased in nonsurviving foals. ACTH:androgen ratios were higher in septic and NMS foals (P < .05). Foals with decreased androgen clearance were more likely to die (odds ratio > 3; P < .05). Conclusions and Clinical Importance Similar to glucocorticoids, mineralocorticoids, and progestagens, increased serum concentrations of androgens are associated with disease severity and adverse outcome in hospitalized newborn foals. In healthy foals, androgens decrease over time, however, remain elevated longer in septic and nonsurviving foals. Androgens could play a role in or reflect a response to disorders such as sepsis or NMS in newborn foals.
Collapse
Affiliation(s)
- Jacob M Swink
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Lindsey M Rings
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA.,Rood and Riddle Equine Hospital, Lexington, Kentucky, USA
| | - Hailey A Snyder
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Rachel C McAuley
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Teresa A Burns
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Katarzyna A Dembek
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | | | - Nimet Browne
- Hagyard Equine Medical Institute, Lexington, Kentucky, USA
| | - Ramiro E Toribio
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
41
|
Wilson HA, Creighton C, Scharfman H, Choleris E, MacLusky NJ. Endocrine Insights into the Pathophysiology of Autism Spectrum Disorder. Neuroscientist 2020; 27:650-667. [PMID: 32912048 DOI: 10.1177/1073858420952046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders that affects males more frequently than females. Numerous genetic and environmental risk factors have been suggested to contribute to the development of ASD. However, no one factor can adequately explain either the frequency of the disorder or the male bias in its prevalence. Gonadal, thyroid, and glucocorticoid hormones all contribute to normal development of the brain, hence perturbations in either their patterns of secretion or their actions may constitute risk factors for ASD. Environmental factors may contribute to ASD etiology by influencing the development of neuroendocrine and neuroimmune systems during early life. Emerging evidence suggests that the placenta may be particularly important as a mediator of the actions of environmental and endocrine risk factors on the developing brain, with the male being particularly sensitive to these effects. Understanding how various risk factors integrate to influence neural development may facilitate a clearer understanding of the etiology of ASD.
Collapse
Affiliation(s)
- Hayley A Wilson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.,Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Carolyn Creighton
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Helen Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY, USA.,Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Elena Choleris
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Neil J MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
42
|
Bianchi VE, Rizzi L, Bresciani E, Omeljaniuk RJ, Torsello A. Androgen Therapy in Neurodegenerative Diseases. J Endocr Soc 2020; 4:bvaa120. [PMID: 33094209 PMCID: PMC7568521 DOI: 10.1210/jendso/bvaa120] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington disease, are characterized by the loss of neurons as well as neuronal function in multiple regions of the central and peripheral nervous systems. Several studies in animal models have shown that androgens have neuroprotective effects in the brain and stimulate axonal regeneration. The presence of neuronal androgen receptors in the peripheral and central nervous system suggests that androgen therapy might be useful in the treatment of neurodegenerative diseases. To illustrate, androgen therapy reduced inflammation, amyloid-β deposition, and cognitive impairment in patients with AD. As well, improvements in remyelination in MS have been reported; by comparison, only variable results are observed in androgen treatment of PD. In ALS, androgen administration stimulated motoneuron recovery from progressive damage and regenerated both axons and dendrites. Only a few clinical studies are available in human individuals despite the safety and low cost of androgen therapy. Clinical evaluations of the effects of androgen therapy on these devastating diseases using large populations of patients are strongly needed.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta, Falciano, San Marino
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
43
|
Camacho-Arroyo I, Piña-Medina AG, Bello-Alvarez C, Zamora-Sánchez CJ. Sex hormones and proteins involved in brain plasticity. VITAMINS AND HORMONES 2020; 114:145-165. [PMID: 32723542 DOI: 10.1016/bs.vh.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
It is well known that peripheral sex steroid hormones cross the blood-brain barrier and control a broad spectrum of reproductive behaviors. However, their role in other essential brain functions was investigated since the 1980s, when the accumulation of pregnenolone and dehydroepiandrosterone in the brain of mammalian species was determined. Since then, numerous studies have demonstrated the participation of sex hormones in brain plasticity processes. Sex hormones through both genomic and non-genomic mechanisms of action are capable of inducing gene transcription or activating signaling cascades that result in the promotion of different physiological and pathological events of brain plasticity, such as remodeling or formation of dendritic spines, neurogenesis, synaptogenesis or myelination. In this chapter, we will present the effects of sex hormones and proteins involved in brain plasticity.
Collapse
Affiliation(s)
- Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| | - Ana Gabriela Piña-Medina
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Carmen J Zamora-Sánchez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
44
|
Gholaminejad A, Gholamipour-Badie H, Nasehi M, Naghdi N. Prelimbic of Medial Prefrontal Cortex GABA Modulation through Testosterone on Spatial Learning and Memory. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:1429-1444. [PMID: 32641952 PMCID: PMC6934985 DOI: 10.22037/ijpr.2019.1100745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Prefrontal cortex (PFC) is involved in multiple functions including attentional processes, spatial orientation, short-term memory, and long-term memory. Our previous study indicated that microinjection of testosterone in CA1 impaired spatial learning and memory. Some evidence suggests that impairment effect of testosterone is mediated by GABAergic system. In the present study, we investigated the interaction of testosterone (androgenic receptor agonist) and bicuculline (GABAA receptor antagonist) on spatial learning and memory performance in the prelimbic (PL) of male Wistar rats. Cannulae were bilaterally implanted into the PL region of PFC and drugs were daily microinjected for two minutes in each side. There are 4 experiments. In the first experiment, three sham groups were operated (solvent of testosterone, bicuculline, testosterone plus bicuculline). In the second experiment, different doses of testosterone (40, 80 μg /0.5 μL DMSO/each side) were injected into the PL before each session. In the third experiment, intra PL injections of bicuculline (2, 4 μg/0.5 μL DMSO/each side) were given before every session. In the last experiment, testosterone (80μg/0.5 μL DMSO/each side) along with bicuculline (2 μg/0.5 μL DMSO/each side) was injected into the PL. The results showed there is no difference between control group and sham operated group. Testosterone 80 μg and bicuculline 2 μg, each given separately, and also in combination increased escape latency to find the platform compared to the sham operated and cause to impaired spatial learning and memory. It is shown that intra PL microinjection of bicuculline after testosterone treatment could not rescue the spatial learning and memory impaired induced by testosterone.
Collapse
Affiliation(s)
- Azadeh Gholaminejad
- Department of Physiology and Pharmacology, Pasteur Institute of Iran (IPI), Tehran, Iran.,Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | | | - Mohammad Nasehi
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran.,Cognitive and neuroscience research center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nasser Naghdi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran (IPI), Tehran, Iran.,Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| |
Collapse
|
45
|
Jayakar SS, Chiara DC, Zhou X, Wu B, Bruzik KS, Miller KW, Cohen JB. Photoaffinity labeling identifies an intersubunit steroid-binding site in heteromeric GABA type A (GABA A) receptors. J Biol Chem 2020; 295:11495-11512. [PMID: 32540960 DOI: 10.1074/jbc.ra120.013452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Allopregnanolone (3α5α-P), pregnanolone, and their synthetic derivatives are potent positive allosteric modulators (PAMs) of GABAA receptors (GABAARs) with in vivo anesthetic, anxiolytic, and anti-convulsant effects. Mutational analysis, photoaffinity labeling, and structural studies have provided evidence for intersubunit and intrasubunit steroid-binding sites in the GABAAR transmembrane domain, but revealed only little definition of their binding properties. Here, we identified steroid-binding sites in purified human α1β3 and α1β3γ2 GABAARs by photoaffinity labeling with [3H]21-[4-(3-(trifluoromethyl)-3H-diazirine-3-yl)benzoxy]allopregnanolone ([3H]21-pTFDBzox-AP), a potent GABAAR PAM. Protein microsequencing established 3α5α-P inhibitable photolabeling of amino acids near the cytoplasmic end of the β subunit M4 (β3Pro-415, β3Leu-417, and β3Thr-418) and M3 (β3Arg-309) helices located at the base of a pocket in the β+-α- subunit interface that extends to the level of αGln-242, a steroid sensitivity determinant in the αM1 helix. Competition photolabeling established that this site binds with high affinity a structurally diverse group of 3α-OH steroids that act as anesthetics, anti-epileptics, and anti-depressants. The presence of a 3α-OH was crucial: 3-acetylated, 3-deoxy, and 3-oxo analogs of 3α5α-P, as well as 3β-OH analogs that are GABAAR antagonists, bound with at least 1000-fold lower affinity than 3α5α-P. Similarly, for GABAAR PAMs with the C-20 carbonyl of 3α5α-P or pregnanolone reduced to a hydroxyl, binding affinity is reduced by 1,000-fold, whereas binding is retained after deoxygenation at the C-20 position. These results provide a first insight into the structure-activity relationship at the GABAAR β+-α- subunit interface steroid-binding site and identify several steroid PAMs that act via other sites.
Collapse
Affiliation(s)
- Selwyn S Jayakar
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| | - David C Chiara
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| | - Xiaojuan Zhou
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Bo Wu
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois
| | - Karol S Bruzik
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois
| | - Keith W Miller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan B Cohen
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Islam MN, Sakimoto Y, Jahan MR, Ishida M, Tarif AMM, Nozaki K, Masumoto KH, Yanai A, Mitsushima D, Shinoda K. Androgen Affects the Dynamics of Intrinsic Plasticity of Pyramidal Neurons in the CA1 Hippocampal Subfield in Adolescent Male Rats. Neuroscience 2020; 440:15-29. [PMID: 32450298 DOI: 10.1016/j.neuroscience.2020.05.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022]
Abstract
Androgen receptor (AR) is abundantly expressed in the preoptico-hypothalamic area, bed nucleus of stria terminalis, and medial amygdala of the brain where androgen plays an important role in regulating male sociosexual, emotional and aggressive behaviors. In addition to these brain regions, AR is also highly expressed in the hippocampus, suggesting that the hippocampus is another major target of androgenic modulation. It is known that androgen can modulate synaptic plasticity in the CA1 hippocampal subfield. However, to date, the effects of androgen on the intrinsic plasticity of hippocampal neurons have not been clearly elucidated. In this study, the effects of androgen on the expression of AR in the hippocampus and on the dynamics of intrinsic plasticity of CA1 pyramidal neurons were examined using immunohistochemistry, Western blotting and whole-cell current-clamp recording in unoperated, sham-operated, orchiectomized (OCX), OCX + testosterone (T) or OCX + dihydrotestosterone (DHT)-primed adolescent male rats. Orchiectomy significantly decreased AR-immunoreactivity, resting membrane potential, action potential numbers, afterhyperpolarization amplitude and membrane resistance, whereas it significantly increased action potential threshold and membrane capacitance. These effects were successfully reversed by treatment with either aromatizable androgen T or non-aromatizable androgen DHT. Furthermore, administration of the AR-antagonist flutamide in intact rats showed similar changes to those in OCX rats, suggesting that androgens affect the excitability of CA1 pyramidal neurons possibly by acting on the AR. Our current study potentially clarifies the role of androgen in enhancing the basal excitability of the CA1 pyramidal neurons, which may influence selective neuronal excitation/activation to modulate certain hippocampal functions.
Collapse
Affiliation(s)
- Md Nabiul Islam
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Yuya Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Mir Rubayet Jahan
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan; Department of Anatomy and Histology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Mako Ishida
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Abu Md Mamun Tarif
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Kanako Nozaki
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Koh-Hei Masumoto
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Akie Yanai
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan; Department of Basic Laboratory Sciences, Faculty of Medicine and Health Sciences, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Dai Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube 755-8505, Japan.
| |
Collapse
|
47
|
Kohtz AS, Frye CA. Learning and the Lifespan: What's Sex Got to Do With It? Front Neurosci 2020; 14:216. [PMID: 32265631 PMCID: PMC7099170 DOI: 10.3389/fnins.2020.00216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/27/2020] [Indexed: 11/23/2022] Open
Abstract
Engagement in sexual behavior can impact neurosteroidogenesis, in particular production of the prohormone testosterone (T) and likely its subsequent metabolism to 5α-androstane-3α-17β-Diol (3α-Diol) or aromatization to estradiol (E2). Androgens and their metabolites vary across the lifespan and impact many behaviors, including cognition, anxiety, and sexual behavior. Thus, we hypothesized that mating may alter cognitive performance via androstane neurosteroids in an age- and experience-dependent manner. We first investigated if exposure to mating during memory consolidation could enhance performance in the novel object recognition task (NOR). Male rats were trained in NOR and then immediately exposed to mating-relevant or control stimuli. Following a 4 h inter-trial interval (ITI), male rats were tested for object memory. Male rats that were exposed to a receptive female during the ITI had better performance in NOR. We then investigated if these effects were due to novelty associated with mating. Male rats were exposed to mating-relevant stimuli and identified as sexually responsive (SR) or sexually non-responsive (SNR) based on a median split of engagement in mating with the stimulus female. We found that a brief history (10 min session daily for five consecutive days) of sexual history substantially influenced performance in the NOR task, such that SR males had better performance in the NOR task, but only when presented with the opportunity to mate during the ITI. As T levels substantially decrease with age in male rodents, we investigated whether the effects of long-term sexual experience (10 months) influenced neurosteroids and NOR performance in mid-aged (12 months old) males. Mid-aged SR males maintain neural T; however, they have decreased neural E2 and decreased cognitive performance at 12 months compared to mid-aged SNR rats. In sexually experienced rats, those with better cognitive performance had greater levels of T metabolites (e.g., 3α-Diol in mated SR males, E2 in mid-aged SNR rats). While naïve males that were mated during the ITI had better cognitive performance, T metabolites were decreased compared to controls. These findings suggest that T metabolites, but not the prohormone, may influence learning dependent on sexual proclivity, experience, and proximate opportunity to mate.
Collapse
Affiliation(s)
- Amy Stave Kohtz
- Department of Psychology, University at Albany – State University of New York (SUNY), Albany, NY, United States
| | - Cheryl A. Frye
- Department of Psychology, University at Albany – State University of New York (SUNY), Albany, NY, United States
- Department of Biological Sciences, University at Albany – State University of New York (SUNY), Albany, NY, United States
- Center for Neuroscience Research, University at Albany – State University of New York (SUNY), Albany, NY, United States
- Center for Life Sciences Research, University at Albany – State University of New York (SUNY), Albany, NY, United States
| |
Collapse
|
48
|
Porey S, Zhang X, Bhowmick S, Kumar Singh V, Guin S, Paton RS, Maiti D. Alkyne Linchpin Strategy for Drug:Pharmacophore Conjugation: Experimental and Computational Realization of a Meta-Selective Inverse Sonogashira Coupling. J Am Chem Soc 2020; 142:3762-3774. [PMID: 31910623 DOI: 10.1021/jacs.9b10646] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The late-stage functionalization (LSF) of pharmaceutical and agrochemical compounds by the site-selective activation of C-H bonds provides access to diverse structural analogs and expands synthetically-accessible chemical space. We report a C-H functionalization LSF strategy that hinges on the use of an alkyne linchpin to assemble conjugates of sp2-rich marketed pharmaceuticals and agrochemicals with sp3-rich 3D fragments and natural products. This is accomplished through a template-assisted inverse Sonogashira reaction that displays high levels of selectivity for the meta position. This protocol is also amenable to distal structural modifications of α-amino acids. The transformation of alkyne functionality to other functional groups further highlights the applicative potential. Computational and experimental mechanistic studies shed light on the detailed mechanism. Turnover-limiting 1,2-migratory insertion of the bromoalkyne coupling partner occurs after relatively fast C-H activation. While this insertion occurs unselectively, regioconvergence results from one of the adducts undergoing a 1,2-trialkylsilyl migration to form the alkynylated product. A heterobimetallic Pd-Ag transition structure is essential for product formation in the β-bromide elimination step.
Collapse
Affiliation(s)
- Sandip Porey
- Department of Chemistry , Indian Institute of Technology Bombay , Powai, Mumbai 400076 , India
| | - Xinglong Zhang
- Chemistry Research Laboratory , University of Oxford , Mansfield Road , Oxford OX1 3TA , United Kingdom
| | - Suman Bhowmick
- Department of Chemistry , Indian Institute of Technology Bombay , Powai, Mumbai 400076 , India
| | - Vikas Kumar Singh
- Department of Chemistry , Indian Institute of Technology Bombay , Powai, Mumbai 400076 , India
| | - Srimanta Guin
- Department of Chemistry , Indian Institute of Technology Bombay , Powai, Mumbai 400076 , India
| | - Robert S Paton
- Chemistry Research Laboratory , University of Oxford , Mansfield Road , Oxford OX1 3TA , United Kingdom.,Department of Chemistry , Colorado State University , Fort Collins , Colorado 80523 , United States
| | - Debabrata Maiti
- Department of Chemistry , Indian Institute of Technology Bombay , Powai, Mumbai 400076 , India.,Tokyo Tech World Research Hub Initiative (WRHI), Laboratory for Chemistry and Life Science , Tokyo Institute of Technology , Tokyo 152-8550 , Japan
| |
Collapse
|
49
|
Spritzer MD, Roy EA. Testosterone and Adult Neurogenesis. Biomolecules 2020; 10:biom10020225. [PMID: 32028656 PMCID: PMC7072323 DOI: 10.3390/biom10020225] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
It is now well established that neurogenesis occurs throughout adulthood in select brain regions, but the functional significance of adult neurogenesis remains unclear. There is considerable evidence that steroid hormones modulate various stages of adult neurogenesis, and this review provides a focused summary of the effects of testosterone on adult neurogenesis. Initial evidence came from field studies with birds and wild rodent populations. Subsequent experiments with laboratory rodents have tested the effects of testosterone and its steroid metabolites upon adult neurogenesis, as well as the functional consequences of induced changes in neurogenesis. These experiments have provided clear evidence that testosterone increases adult neurogenesis within the dentate gyrus region of the hippocampus through an androgen-dependent pathway. Most evidence indicates that androgens selectively enhance the survival of newly generated neurons, while having little effect on cell proliferation. Whether this is a result of androgens acting directly on receptors of new neurons remains unclear, and indirect routes involving brain-derived neurotrophic factor (BDNF) and glucocorticoids may be involved. In vitro experiments suggest that testosterone has broad-ranging neuroprotective effects, which will be briefly reviewed. A better understanding of the effects of testosterone upon adult neurogenesis could shed light on neurological diseases that show sex differences.
Collapse
Affiliation(s)
- Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
- Correspondence: ; Tel.: 802-443-5676
| | - Ethan A. Roy
- Graduate School of Education, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
50
|
Mendell AL, Creighton SD, Wilson HA, Jardine KH, Isaacs L, Winters BD, MacLusky NJ. Inhibition of 5α Reductase Impairs Cognitive Performance, Alters Dendritic Morphology and Increases Tau Phosphorylation in the Hippocampus of Male 3xTg-AD Mice. Neuroscience 2020; 429:185-202. [PMID: 31954826 DOI: 10.1016/j.neuroscience.2020.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 10/25/2022]
Abstract
Recent work has suggested that 5α-reduced metabolites of testosterone may contribute to the neuroprotection conferred by their parent androgen, as well as to sex differences in the incidence and progression of Alzheimer's disease (AD). This study investigated the effects of inhibiting 5α-reductase on object recognition memory (ORM), hippocampal dendritic morphology and proteins involved in AD pathology, in male 3xTg-AD mice. Male 6-month old wild-type or 3xTg-AD mice received daily injections of finasteride (50 mg/kg i.p.) or vehicle (18% β-cyclodextrin, 1% v/b.w.) for 20 days. Female wild-type and 3xTg-AD mice received only the vehicle. Finasteride treatment differentially impaired ORM in males after short-term (3xTg-AD only) or long-term (3xTg-AD and wild-type) retention delays. Dendritic spine density and dendritic branching of pyramidal neurons in the CA3 hippocampal subfield were significantly lower in 3xTg-AD females than in males. Finasteride reduced CA3 dendritic branching and spine density in 3xTg-AD males, to within the range observed in vehicle-treated females. In the CA1 hippocampal subfield, dendritic branching and spine density were reduced in both male and female 3xTg-AD mice, compared to wild type controls. Hippocampal amyloid β levels were substantially higher in 3xTg-AD females compared to both vehicle and finasteride-treated 3xTg-AD males. Site-specific Tau phosphorylation was higher in 3xTg-AD mice compared to sex-matched wild-type controls, increasing slightly after finasteride treatment. These results suggest that 5α-reduced neurosteroids may play a role in testosterone-mediated neuroprotection and may contribute to sex differences in the development and severity of AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Boyer D Winters
- Psychology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | | |
Collapse
|