1
|
de Assis GG, de Sousa MBC, Murawska-Ciałowicz E. Sex Steroids and Brain-Derived Neurotrophic Factor Interactions in the Nervous System: A Comprehensive Review of Scientific Data. Int J Mol Sci 2025; 26:2532. [PMID: 40141172 PMCID: PMC11942429 DOI: 10.3390/ijms26062532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Sex steroids and the neurotrophin brain-derived neurotrophic factor (BDNF) participate in neural tissue formation, phenotypic differentiation, and neuroplasticity. These processes are essential for the health and maintenance of the central nervous system. AIM The aim of our review is to elucidate the interaction mechanisms between BDNF and sex steroids in neuronal function. METHOD A series of searches were performed using Mesh terms for androgen/receptors, estrogen/receptors, and BDNF/receptors, and a collection of the scientific data available on PubMed up to February 2025 about mechanical interactions between BDNF and sex steroids was included in this literature review. DISCUSSION This review discussed the influence of sex steroids on the formation and/or maintenance of neural circuits via different mechanisms, including the regulation of BDNF expression and signaling. Estrogens exert a time- and region-specific effect on BDNF synthesis. The nuclear estrogen receptor can directly regulate BDNF expression, independently of the presence of estrogen, in neuronal cells, whereas progesterone and testosterone upregulate BDNF expression via their specific nuclear receptors. In addition, testosterone has a positive effect on BDNF release by glial cells, which lack androgen receptors.
Collapse
Affiliation(s)
- Gilmara Gomes de Assis
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, 4900-347 Viana do Castelo, Portugal
- Sport Physical Activity and Health Research & Innovation Center, 4900-347 Viana do Castelo, Portugal
| | | | - Eugenia Murawska-Ciałowicz
- Department of Physiology and Biomechanics, Wroclaw University of Health and Sport Sciences, 51-612 Wrocław, Poland;
| |
Collapse
|
2
|
Rahaei N, Buynack LM, Kires L, Movasseghi Y, Chapman CA. Progesterone and allopregnanolone facilitate excitatory synaptic transmission in the infralimbic cortex via activation of membrane progesterone receptors. Neuroscience 2025; 567:9-17. [PMID: 39722289 DOI: 10.1016/j.neuroscience.2024.12.042] [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: 08/15/2024] [Revised: 09/29/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
Estrogens and progesterone can have rapid effects on neuronal function and can modify the use of spatial navigation strategies dependent upon the prefrontal cortex, striatum, and hippocampus. Here, we assessed the effects of 17β-estradiol (E2), progesterone, and its metabolite allopregnanolone, on evoked excitatory postsynaptic potentials in the infralimbic region of the female rat prefrontal cortex. Field excitatory postsynaptic potentials (fEPSPs) evoked by stimulation of layer I were first characterized by recording responses at multiple depths between the cortical surface and the underlying white matter. Current source density analysis showed that the short-latency negative component was generated by activation of synaptic currents within layer I, and that putative polysynaptic responses were generated in layers III to V. The amplitude of evoked field EPSPs in layer I was not significantly affected by 20 min application of 17β-estradiol (10 nM), but both 100 nM progesterone and 1 µM allopregnanolone caused lasting increases in field EPSP amplitude. The effects of progesterone were not blocked by the nuclear progesterone receptor antagonist RU486 (1 µM). Both progesterone and allopregnanolone are known to activate membrane progesterone receptors, and we found that the membrane progesterone receptor agonist Org OD 02-0 facilitated EPSPs, and also occluded further increases induced by either progesterone or allopregnanolone. These results provide evidence that both progesterone and allopregnanolone facilitate synaptic responses in layer I of the infralimbic cortex by activating membrane progesterone receptors.
Collapse
Affiliation(s)
- Nima Rahaei
- Department of Psychology, Concordia University, Montreal, Canada
| | - Lauren M Buynack
- Department of Psychology, Concordia University, Montreal, Canada
| | - Lukas Kires
- Department of Psychology, Concordia University, Montreal, Canada
| | | | - C Andrew Chapman
- Department of Psychology, Concordia University, Montreal, Canada.
| |
Collapse
|
3
|
Valero-Ochando J, Cantó A, López-Pedrajas R, Almansa I, Miranda M. Role of Gonadal Steroid Hormones in the Eye: Therapeutic Implications. Biomolecules 2024; 14:1262. [PMID: 39456195 PMCID: PMC11506707 DOI: 10.3390/biom14101262] [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: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Gonadal steroid hormones are critical regulatory substances involved in various developmental and physiological processes from fetal development through adulthood. These hormones, derived from cholesterol, are synthesized primarily by the gonads, adrenal cortex, and placenta. The synthesis of these hormones involves a series of enzymatic steps starting in the mitochondria and includes enzymes such as cytochrome P450 and aromatase. Beyond their genomic actions, which involve altering gene transcription over hours, gonadal steroids also exhibit rapid, nongenomic effects through receptors located on the cell membrane. Additionally, recent research has highlighted the role of these hormones in the central nervous system (CNS). However, the interactions between gonadal steroid hormones and the retina have received limited attention, though it has been suggested that they may play a protective role in retinal diseases. This review explores the synthesis of gonadal hormones, their mechanisms of action, and their potential implications in various retinal and optic nerve diseases, such as glaucoma, age-related macular degeneration (AMD), diabetic retinopathy (DR), or retinitis pigmentosa (RP), discussing both protective and risk factors associated with hormone levels and their therapeutic potential.
Collapse
Affiliation(s)
| | | | | | | | - María Miranda
- Department of Biomedical Sciences, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, 46115 Valencia, Spain; (J.V.-O.); (A.C.); (R.L.-P.); (I.A.)
| |
Collapse
|
4
|
Handschuh PA, Reed MB, Murgaš M, Vraka C, Kaufmann U, Nics L, Klöbl M, Ozenil M, Konadu ME, Patronas EM, Spurny-Dworak B, Hahn A, Hacker M, Spies M, Baldinger-Melich P, Kranz GS, Lanzenberger R. Effects of gender-affirming hormone therapy on gray matter density, microstructure and monoamine oxidase A levels in transgender subjects. Neuroimage 2024; 297:120716. [PMID: 38955254 DOI: 10.1016/j.neuroimage.2024.120716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 07/04/2024] Open
Abstract
MAO-A catalyzes the oxidative degradation of monoamines and is thus implicated in sex-specific neuroplastic processes that influence gray matter (GM) density (GMD) and microstructure (GMM). Given the exact monitoring of plasma hormone levels and sex steroid intake, transgender individuals undergoing gender-affirming hormone therapy (GHT) represent a valuable cohort to potentially investigate sex steroid-induced changes of GM and concomitant MAO-A density. Here, we investigated the effects of GHT over a median time period of 4.5 months on GMD and GMM as well as MAO-A distribution volume. To this end, 20 cisgender women, 11 cisgender men, 20 transgender women and 10 transgender men underwent two MRI scans in a longitudinal design. PET scans using [11C]harmine were performed before each MRI session in a subset of 35 individuals. GM changes determined by diffusion weighted imaging (DWI) metrics for GMM and voxel based morphometry (VBM) for GMD were estimated using repeated measures ANOVA. Regions showing significant changes of both GMM and GMD were used for the subsequent analysis of MAO-A density. These involved the fusiform gyrus, rolandic operculum, inferior occipital cortex, middle and anterior cingulum, bilateral insula, cerebellum and the lingual gyrus (post-hoc tests: pFWE+Bonferroni < 0.025). In terms of MAO-A distribution volume, no significant effects were found. Additionally, the sexual desire inventory (SDI) was applied to assess GHT-induced changes in sexual desire, showing an increase of SDI scores among transgender men. Changes in the GMD of the bilateral insula showed a moderate correlation to SDI scores (rho = - 0.62, pBonferroni = 0.047). The present results are indicative of a reliable influence of gender-affirming hormone therapy on 1) GMD and GMM following an interregional pattern and 2) sexual desire specifically among transgender men.
Collapse
Affiliation(s)
- P A Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M B Reed
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M Murgaš
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - C Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - U Kaufmann
- Department of Obstetrics and Gynecology, Medical University of Vienna, Austria
| | - L Nics
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - M Klöbl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M Ozenil
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - M E Konadu
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - E M Patronas
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - B Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - A Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - M Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - P Baldinger-Melich
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - G S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria; Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria.
| |
Collapse
|
5
|
Szczepanska-Sadowska E, Czarzasta K, Bogacki-Rychlik W, Kowara M. The Interaction of Vasopressin with Hormones of the Hypothalamo-Pituitary-Adrenal Axis: The Significance for Therapeutic Strategies in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2024; 25:7394. [PMID: 39000501 PMCID: PMC11242374 DOI: 10.3390/ijms25137394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
A large body of evidence indicates that vasopressin (AVP) and steroid hormones are frequently secreted together and closely cooperate in the regulation of blood pressure, metabolism, water-electrolyte balance, and behavior, thereby securing survival and the comfort of life. Vasopressin cooperates with hormones of the hypothalamo-pituitary-adrenal axis (HPA) at several levels through regulation of the release of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and multiple steroid hormones, as well as through interactions with steroids in the target organs. These interactions are facilitated by positive and negative feedback between specific components of the HPA. Altogether, AVP and the HPA cooperate closely as a coordinated functional AVP-HPA system. It has been shown that cooperation between AVP and steroid hormones may be affected by cellular stress combined with hypoxia, and by metabolic, cardiovascular, and respiratory disorders; neurogenic stress; and inflammation. Growing evidence indicates that central and peripheral interactions between AVP and steroid hormones are reprogrammed in cardiovascular and metabolic diseases and that these rearrangements exert either beneficial or harmful effects. The present review highlights specific mechanisms of the interactions between AVP and steroids at cellular and systemic levels and analyses the consequences of the inappropriate cooperation of various components of the AVP-HPA system for the pathogenesis of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | | | | | | |
Collapse
|
6
|
Dardente H, Lomet D, Robert V, Lasserre O, Gonzalez AA, Mialhe X, Beltramo M. Photoperiod, but not progesterone, has a strong impact upon the transcriptome of the medio-basal hypothalamus in female goats and ewes. Mol Cell Endocrinol 2024; 588:112216. [PMID: 38556161 DOI: 10.1016/j.mce.2024.112216] [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: 11/23/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Photoperiod is the main environmental driver of seasonal responses in organisms living at temperate and polar latitudes. Other external cues such as food and temperature, and internal cues including hormones, intervene to fine-tune phasing of physiological functions to the solar year. In mammals, the medio-basal hypothalamus (MBH) is the key integrator of these cues, which orchestrates a wide array of seasonal functions, including breeding. Here, using RNAseq and RT-qPCR, we demonstrate that molecular components of the photoperiodic response previously identified in ewes are broadly conserved in does (female goats, Capra hircus), with a common core of ∼50 genes. This core group can be defined as the "MBH seasonal trancriptome", which includes key players of the pars tuberalis-tanycytes neuroendocrine retrograde pathway that governs intra-MBH photoperiodic switches of triiodothyronine (T3) production (Tshb, Eya3, Dio2 and SlcO1c1), the two histone methyltransferases Suv39H2 and Ezh2 and the secreted protein Vmo1. Prior data in ewes revealed that T3 and estradiol (E2), both key hormones for the proper timing of seasonal breeding, differentially impact the MBH seasonal transcriptome, and identified cellular and molecular targets through which these hormones might act. In contrast, information regarding the potential impact of progesterone (P4) upon the MBH transcriptome was nonexistent. Here, we demonstrate that P4 has no discernible transcriptional impact in either does or ewes. Taken together, our data show that does and ewes possess a common core set of photoperiod-responsive genes in the MBH and conclusively demonstrate that P4 is not a key regulator of the MBH transcriptome.
Collapse
Affiliation(s)
- Hugues Dardente
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France.
| | - Didier Lomet
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | - Vincent Robert
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | | |
Collapse
|
7
|
Sitruk-Ware R, Sussman H, Brinton R, Schumacher M, Singer P, Kumar N, De Nicola AF, El-Etr M, Guennoun R, V Borlongan C. Nestorone (segesterone acetate) effects on neuroregeneration. Front Neuroendocrinol 2024; 73:101136. [PMID: 38670433 DOI: 10.1016/j.yfrne.2024.101136] [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: 02/23/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Nestorone® (segesterone acetate) is a progestin with a chemical structure closely related to progesterone with high affinity and selectivity for the progesterone receptor without significant interaction with other steroid receptors. It has been developed for female and male contraception and is FDA-approved in a first long-acting contraceptive vaginal system for female contraception. Its safety has been extensively demonstrated in both preclinical and clinical studies for contraceptive indications. Nestorone was found to display neuroprotective and neuroregenerative activity in animal models of various central nervous system diseases, including multiple sclerosis, stroke, and amyotrophic lateral sclerosis. Reviewed herein are neuroprotective and myelin- regenerating properties of Nestorone in various animal models and its translational potential as a therapeutic agent for debilitating neurological diseases for which limited therapeutic options are available (Table 1).
Collapse
Affiliation(s)
| | | | - Roberta Brinton
- Center for Innovation in Brain Science, Tucson, AZ, United States
| | | | | | | | | | - Martine El-Etr
- U1195 Inserm and University Paris-Saclay Le Kremlin Bicêtre, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Saclay Le Kremlin Bicêtre, France
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
8
|
Reddy DS, Singh T, Ramakrishnan S, Huber M, Wu X. Neuroprotectant Activity of Novel Water-Soluble Synthetic Neurosteroids on Organophosphate Intoxication and Status Epilepticus-Induced Long-Term Neurological Dysfunction, Neurodegeneration, and Neuroinflammation. J Pharmacol Exp Ther 2024; 388:399-415. [PMID: 38071567 PMCID: PMC10801736 DOI: 10.1124/jpet.123.001819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/18/2023] [Indexed: 01/19/2024] Open
Abstract
Organophosphates (OPs) and nerve agents are potent neurotoxic compounds that cause seizures, status epilepticus (SE), brain injury, or death. There are persistent long-term neurologic and neurodegenerative effects that manifest months to years after the initial exposure. Current antidotes are ineffective in preventing these long-term neurobehavioral and neuropathological changes. Additionally, there are few effective neuroprotectants for mitigating the long-term effects of acute OP intoxication. We have pioneered neurosteroids as novel anticonvulsants and neuroprotectants for OP intoxication and seizures. In this study, we evaluated the efficacy of two novel synthetic, water-soluble neurosteroids, valaxanolone (VX) and lysaxanolone (LX), in combating the long-term behavioral and neuropathological impairments caused by acute OP intoxication and SE. Animals were exposed to the OP nerve agent surrogate diisopropylfluorophosphate (DFP) and were treated with VX or LX in addition to midazolam at 40 minutes postexposure. The extent of neurodegeneration, along with various behavioral and memory deficits, were assessed at 3 months postexposure. VX significantly reduced deficits of aggressive behavior, anxiety, memory, and depressive-like traits in control (DFP-exposed, midazolam-treated) animals; VX also significantly prevented the DFP-induced chronic loss of NeuN(+) principal neurons and PV(+) inhibitory neurons in the hippocampus and other regions. Additionally, VX-treated animals exhibited a reduced inflammatory response with decreased GFAP(+) astrogliosis and IBA1(+) microgliosis in the hippocampus, amygdala, and other regions. Similarly, LX showed significant improvement in behavioral and memory deficits, and reduced neurodegeneration and cellular neuroinflammation. Together, these results demonstrate the neuroprotectant effects of the novel synthetic neurosteroids in mitigating the long-term neurologic dysfunction and neurodegeneration associated with OP exposure. SIGNIFICANCE STATEMENT: Survivors of nerve agents and organophosphate (OP) exposures suffer from long-term neurological deficits. Currently, there is no specific drug therapy for mitigating the impact of OP exposure. However, novel synthetic neurosteroids that activate tonic inhibition provide a viable option for treating OP intoxication. The data from this study indicates the neuroprotective effects of synthetic, water-soluble neurosteroids for attenuation of long-term neurological deficits after OP intoxication. These findings establish valaxanolone and lysaxanolone as potent and efficacious neuroprotectants suitable for injectable dosing.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Madeline Huber
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| |
Collapse
|
9
|
Kaltsouni E, Schmidt F, Zsido RG, Eriksson A, Sacher J, Sundström-Poromaa I, Sumner RL, Comasco E. Electroencephalography findings in menstrually-related mood disorders: A critical review. Front Neuroendocrinol 2024; 72:101120. [PMID: 38176542 DOI: 10.1016/j.yfrne.2023.101120] [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: 05/09/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/06/2024]
Abstract
The female reproductive years are characterized by fluctuations in ovarian hormones across the menstrual cycle, which have the potential to modulate neurophysiological and behavioral dynamics. Menstrually-related mood disorders (MRMDs) comprise cognitive-affective or somatic symptoms that are thought to be triggered by the rapid fluctuations in ovarian hormones in the luteal phase of the menstrual cycle. MRMDs include premenstrual syndrome (PMS), premenstrual dysphoric disorder (PMDD), and premenstrual exacerbation (PME) of other psychiatric disorders. Electroencephalography (EEG) non-invasively records in vivo synchronous activity from populations of neurons with high temporal resolution. The present overview sought to systematically review the current state of task-related and resting-state EEG investigations on MRMDs. Preliminary evidence indicates lower alpha asymmetry at rest being associated with MRMDs, while one study points to the effect being luteal-phase specific. Moreover, higher luteal spontaneous frontal brain activity (slow/fast wave ratio as measured by the delta/beta power ratio) has been observed in persons with MRMDs, while sleep architecture results point to potential circadian rhythm disturbances. In this review, we discuss the quality of study designs as well as future perspectives and challenges of supplementing the diagnostic and scientific toolbox for MRMDs with EEG.
Collapse
Affiliation(s)
- Elisavet Kaltsouni
- Department of Womeńs and Childreńs Health, Science for Life Laboratory, Uppsala University, Sweden
| | - Felix Schmidt
- Department of Womeńs and Childreńs Health, Science for Life Laboratory, Uppsala University, Sweden; Centre for Women's Mental Health during the Reproductive Lifespan, Uppsala University, 751 85 Uppsala, Sweden
| | - Rachel G Zsido
- Cognitive Neuroendocrinology, Max Planck Institute for Human Cognitive and Brain Sciences, Germany; Department of Psychiatry, Clinical Neuroscience Laboratory for Sex Differences in the Brain, Massachusetts General Hospital, Harvard Medical School, USA
| | - Allison Eriksson
- Centre for Women's Mental Health during the Reproductive Lifespan, Uppsala University, 751 85 Uppsala, Sweden; Department of Womeńs and Childreńs Health, Uppsala University, Sweden
| | - Julia Sacher
- Cognitive Neuroendocrinology, Max Planck Institute for Human Cognitive and Brain Sciences, Germany; Clinic of Cognitive Neurology, University of Leipzig, Germany
| | | | | | - Erika Comasco
- Department of Womeńs and Childreńs Health, Science for Life Laboratory, Uppsala University, Sweden.
| |
Collapse
|
10
|
Amerio A, Magnani L, Arduino G, Fesce F, de Filippis R, Parise A, Costanza A, Nguyen KD, Saverino D, De Berardis D, Aguglia A, Escelsior A, Serafini G, De Fazio P, Amore M. Immunomodulatory Effects of Clozapine: More Than Just a Side Effect in Schizophrenia. Curr Neuropharmacol 2024; 22:1233-1247. [PMID: 38031778 PMCID: PMC10964093 DOI: 10.2174/1570159x22666231128101725] [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: 11/30/2022] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 12/01/2023] Open
Abstract
Recent evidence suggests a possible relationship between the immune system and schizophrenia spectrum disorders (SSDs), as neuroinflammation appears to play a role in major psychiatric conditions. Neuroinflammation is as a broad concept representing a physiological protective response to infection or injury, but in some cases, especially if chronic, it may represent an expression of maladaptive processes, potentially driving to clinical dysfunction and neurodegeneration. Several studies are concurrently highlighting the importance of microglia, the resident immune cells of the central nervous system, in a huge number of neurodegenerative diseases, including multiple sclerosis, Alzheimer's and Parkinson's diseases, as well as SSDs. A more fundamental phenomenon of maladaptive coupling of microglia may contribute to the genesis of dysfunctional brain inflammation involved in SSDs, from the onset of their neurophenomenological evolution. Clozapine and other antipsychotic drugs seem to express a provable immunomodulant effect and a more specific action on microglia, while neuroactive steroids and nonsteroidal anti-inflammatory drugs may reduce some SSDs symptoms in add-on therapy. Given these theoretical premises, this article aims to summarize and interpret the available scientific evidence about psychotropic and anti-inflammatory drugs that could express an immunomodulant activity on microglia.
Collapse
Affiliation(s)
- Andrea Amerio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Magnani
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
| | - Gabriele Arduino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
| | - Fabio Fesce
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
| | - Renato de Filippis
- Psychiatry Unit, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Alberto Parise
- Department of Geriatric-Rehabilitation,, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI) Lugano, Switzerland
| | - Khoa D. Nguyen
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, USA
- Tranquis Therapeutics, Palo Alto, CA, USA
| | - Daniele Saverino
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DiMeS), Section of Human Anatomy, University of Genoa, Genoa, Italy
| | - Domenico De Berardis
- NHS, Department of Mental Health, Psychiatric Service for Diagnosis and Treatment, Hospital “G. Mazzini”, Teramo, Italy
| | - Andrea Aguglia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Escelsior
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Pasquale De Fazio
- Psychiatry Unit, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
11
|
Abramicheva PA, Semenovich DS, Zorova LD, Pevzner IB, Sokolov IA, Popkov VA, Kazakov EP, Zorov DB, Plotnikov EY. Decreased renal expression of PAQR5 is associated with the absence of a nephroprotective effect of progesterone in a rat UUO model. Sci Rep 2023; 13:12871. [PMID: 37553369 PMCID: PMC10409855 DOI: 10.1038/s41598-023-39848-2] [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: 06/11/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023] Open
Abstract
Fibrosis is a severe complication of chronic kidney disease (CKD). Progesterone, like other sex hormones, plays an important role in renal physiology, but its role in CKD is poorly understood. We investigated progesterone effect on renal fibrosis progression in the rat model of unilateral ureteral obstruction (UUO). Female rats were exposed to UUO, ovariectomy and progesterone administration after UUO with ovariectomy. Expression of key fibrosis markers, proinflammatory cytokines, levels of membrane-bound (PAQR5) and nuclear (PGR) progesterone receptors, and matrix metalloproteinase (MMP) activity were analyzed in the obstructed and intact rat kidney. In all groups exposed to UUO, decreased PAQR5 expression was observed in the obstructed kidney while in the contralateral kidney, it remained unaffected. We found increased mRNA levels for profibrotic COL1A1, FN1, MMP2, TIMP1, TIMP2, proinflammatory IL1α, IL1β, and IL18, as well as elevated α-SMA and MMP9 proteins, collagen deposition, and MMP2 activity in all UUO kidneys. Progesterone had slight or no effect on the change in these markers. Thus, we demonstrate for the first time diminished sensitivity of the kidney to progesterone associated with renal fibrosis due to a severe decrease in PAQR5 expression that was accompanied by the lack of nephroprotection in a rat UUO model.
Collapse
Affiliation(s)
- P A Abramicheva
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234.
| | - D S Semenovich
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
| | - L D Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia, 117997
| | - I B Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia, 117997
| | - I A Sokolov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
- Mendeleev University of Chemical Technology of Russia, Moscow, Russia, 125047
| | - V A Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia, 117997
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, Moscow, Russia, 119234
| | - E P Kazakov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
| | - D B Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia, 117997
| | - E Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119234.
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia, 117997.
| |
Collapse
|
12
|
Terrin F, Tesoriere A, Plotegher N, Dalla Valle L. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson's Disease. Cells 2023; 12:1486. [PMID: 37296608 PMCID: PMC10252697 DOI: 10.3390/cells12111486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Sex hormones and genes on the sex chromosomes are not only key factors in the regulation of sexual differentiation and reproduction but they are also deeply involved in brain homeostasis. Their action is crucial for the development of the brain, which presents different characteristics depending on the sex of individuals. The role of these players in the brain is fundamental in the maintenance of brain function during adulthood as well, thus being important also with respect to age-related neurodegenerative diseases. In this review, we explore the role of biological sex in the development of the brain and analyze its impact on the predisposition toward and the progression of neurodegenerative diseases. In particular, we focus on Parkinson's disease, a neurodegenerative disorder that has a higher incidence in the male population. We report how sex hormones and genes encoded by the sex chromosomes could protect from the disease or alternatively predispose toward its development. We finally underline the importance of considering sex when studying brain physiology and pathology in cellular and animal models in order to better understand disease etiology and develop novel tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| | - Luisa Dalla Valle
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| |
Collapse
|
13
|
Sourouni M, Götte M, Kiesel L, von Wahlde MK. Effect of 3α-dihydroprogesterone and 5α-dihydroprogesterone on DCIS cells and possible impact for postmenopausal women. Climacteric 2023; 26:275-283. [PMID: 36880551 DOI: 10.1080/13697137.2023.2182678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
OBJECTIVE Progesterone metabolites 5α-dihydroprogesterone (5αP) and 3α-dihydroprogesterone (3αP) have opposite effects on proliferation, apoptosis and metastasis in the breast. Evidence regarding their influence on ductal carcinoma in situ (DCIS) lesions is lacking. METHODS MCF10DCIS.com cells were cultured in a 3D culture system and treated with 5αP or 3αP. After 5 and 12 days of treatment, polymerase chain reaction (PCR) of proliferation, invasion/metastasis, anti-apoptotic or other markers was performed. Cells treated with the tumor-promoting 5αP were observed under the light and confocal microscopes to reveal possible morphological changes that could indicate a transition from an in situ to an invasive phenotype. As a control, the morphology of the MDA-MB-231 invasive cell line was examined. The invasive potential after exposure to 5αP was also assessed using a detachment assay. RESULTS The PCR analysis of the chosen markers showed no statistically significant difference between naive cells and cells treated with 5αP or 3αP. DCIS spheroids retained their in situ morphology after treatment with 5αP. The detachment assay showed no increased potential for invasion after exposure to 5αP. Progesterone metabolites 5αP and 3αP do not facilitate or prohibit tumor promotion/invasion in MCF10DCIS.com cells, respectively. CONCLUSION As oral micronized progesterone has been proved effective for hot flushes in postmenopausal women, first in vitro data propose that progesterone-only therapy could possibly be considered for women after DCIS suffering from hot flushes.
Collapse
Affiliation(s)
- M Sourouni
- Department of Obstetrics and Gynecology, University Hospital Muenster, Muenster, Germany.,Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, Heidelberg, Germany
| | - M Götte
- Department of Obstetrics and Gynecology, University Hospital Muenster, Muenster, Germany
| | - L Kiesel
- Department of Obstetrics and Gynecology, University Hospital Muenster, Muenster, Germany
| | - M-K von Wahlde
- Department of Obstetrics and Gynecology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
14
|
Abtin S, Ghasemi R, Manaheji H. Progesterone modulates the expression of spinal ephrin-B2 after peripheral nerve injury: New insights into progesterone mechanisms. Steroids 2023; 190:109155. [PMID: 36529276 DOI: 10.1016/j.steroids.2022.109155] [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: 08/29/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Recent studies have shown that the ephrin/Eph signaling pathway may contribute to the pathology of neuropathic pain. Drugs like progesterone may be used to counteract both thermal hyperalgesia and mechanical allodynia in different models of neuropathic pain. The present study was designed to determine progesterone's modulatory role on neuropathic pain and spinal expression of ephrin-B2 following chronic constriction nerve injury (CCI). Thirty-six adult male Wistar rats were used. The sciatic nerve was chronically constricted. Progesterone (5 mg/kg and 15 mg/kg) was administrated for 10 days (from day 1 up to day10) following sciatic constriction. Behavioral tests were performed before surgery (day 0) and on days 1, 3, 7, and 14 after CCI and before progesterone administration on the same days. Western blotting was performed on days 3, 7, and 14th post-surgery. The findings showed that after CCI, the expression of spinal cord ephrin-B2 increased significantly in parallel with mechanical allodynia and thermal hyperalgesia. Post-injury administration of progesterone (15 mg/kg but not 5) decreased mechanical allodynia, thermal hyperalgesia, and the expression of spinal ephrin-B2. It is concluded that post-injury repeated administration of progesterone could be an effective way of alleviating neuropathic pain by suppressing ephrin-B2 activation and helps to make the better design of steroid-based therapies to inhibit pain after peripheral injury.
Collapse
Affiliation(s)
- Shima Abtin
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homa Manaheji
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Abaffy T, Lu HY, Matsunami H. Sex steroid hormone synthesis, metabolism, and the effects on the mammalian olfactory system. Cell Tissue Res 2023; 391:19-42. [PMID: 36401093 PMCID: PMC9676892 DOI: 10.1007/s00441-022-03707-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
Abstract
Sex steroid hormones influence olfactory-mediated social behaviors, and it is generally hypothesized that these effects result from circulating hormones and/or neurosteroids synthesized in the brain. However, it is unclear whether sex steroid hormones are synthesized in the olfactory epithelium or the olfactory bulb, and if they can modulate the activity of the olfactory sensory neurons. Here, we review important discoveries related to the metabolism of sex steroids in the mouse olfactory epithelium and olfactory bulb, along with potential areas of future research. We summarize current knowledge regarding the expression, neuroanatomical distribution, and biological activity of the steroidogenic enzymes, sex steroid receptors, and proteins that are important to the metabolism of these hormones and reflect on their potential to influence early olfactory processing. We also review evidence related to the effects of sex steroid hormones on the development and activity of olfactory sensory neurons. By better understanding how these hormones are metabolized and how they act both at the periphery and olfactory bulb level, we can better appreciate the complexity of the olfactory system and discover potential similarities and differences in early olfactory processing between sexes.
Collapse
Affiliation(s)
- Tatjana Abaffy
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| | - Hsiu-Yi Lu
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| | - Hiroaki Matsunami
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
16
|
Bulletti C, Bulletti FM, Sciorio R, Guido M. Progesterone: The Key Factor of the Beginning of Life. Int J Mol Sci 2022; 23:ijms232214138. [PMID: 36430614 PMCID: PMC9692968 DOI: 10.3390/ijms232214138] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/17/2022] Open
Abstract
Progesterone is the ovarian steroid produced by the granulosa cells of follicles after the LH peak at mid-cycle. Its role is to sustain embryo endometrial implantation and ongoing pregnancy. Other biological effects of progesterone may exert a protective function in supporting pregnancy up to birth. Luteal phase support (LPS) with progesterone is the standard of care for assisted reproductive technology. Progesterone vaginal administration is currently the most widely used treatment for LPS. Physicians and patients have been reluctant to change an administration route that has proven to be effective. However, some questions remain open, namely the need for LPS in fresh and frozen embryo transfer, the route of administration, the optimal duration of LPS, dosage, and the benefit of combination therapies. The aim of this review is to provide an overview of the uterine and extra-uterine effects of progesterone that may play a role in embryo implantation and pregnancy, and to discuss the advantages of the use of progesterone for LPS in the context of Good Medical Practice.
Collapse
Affiliation(s)
- Carlo Bulletti
- Extra Omnes, Assisted Reproductive Technology, ART Center, Via Gallinelli, 8, 47841 Cattolica, Italy
- Department of Obstetrics, Gynecology, and Reproductive Science, Yale University, New Haven, CT 06510, USA
- Correspondence:
| | | | - Romualdo Sciorio
- Edinburgh Assisted Conception Programme, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
| | - Maurizio Guido
- Obstetrics and Gynecology Unit, Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
17
|
Thomas P, Pang Y, Camilletti MA, Castelnovo LF. Functions of Membrane Progesterone Receptors (mPRs, PAQRs) in Nonreproductive Tissues. Endocrinology 2022; 163:6679267. [PMID: 36041040 DOI: 10.1210/endocr/bqac147] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Indexed: 11/19/2022]
Abstract
Gender differences in a wide variety of physiological parameters have implicated the ovarian hormones, estrogens and progesterone, in the regulation of numerous nonreproductive tissue functions. Rapid, nongenomic (nonclassical) progesterone actions mediated by membrane progesterone receptors (mPRs), which belong to the progestin and adipoQ receptor family, have been extensively investigated in reproductive and nonreproductive tissues since their discovery in fish ovaries 20 years ago. The 5 mPR subtypes (α, β, γ, δ, ε) are widely distributed in vertebrate tissues and are often expressed in the same cells as the nuclear progesterone receptor (PR) and progesterone receptor membrane component 1, thereby complicating investigations of mPR-specific functions. Nevertheless, mPR-mediated progesterone actions have been identified in a wide range of reproductive and nonreproductive tissues and distinguished from nuclear PR-mediated ones by knockdown of these receptors with siRNA in combination with a pharmacological approach using mPR- and PR-specific agonists. There are several recent reviews on the roles of the mPRs in vertebrate reproduction and cancer, but there have been no comprehensive assessments of mPR functions in nonreproductive tissues. Therefore, this article briefly reviews mPR functions in a broad range of nonreproductive tissues. The evidence that mPRs mediate progesterone and progestogen effects on neuroprotection, lordosis behavior, respiratory control of apnea, olfactory responses to pheromones, peripheral nerve regeneration, regulation of prolactin secretion in prolactinoma, immune functions, and protective functions in vascular endothelial and smooth muscle cells is critically reviewed. The ubiquitous expression of mPRs in vertebrate tissues suggests mPRs regulate many additional nonreproductive functions that remain to be identified.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | - Yefei Pang
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | | | - Luca F Castelnovo
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| |
Collapse
|
18
|
Griksiene R, Monciunskaite R, Ruksenas O. What is there to know about the effects of progestins on the human brain and cognition? Front Neuroendocrinol 2022; 67:101032. [PMID: 36029852 DOI: 10.1016/j.yfrne.2022.101032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/24/2022] [Accepted: 08/19/2022] [Indexed: 12/27/2022]
Abstract
Progestins are an important component of hormonal contraceptives (HCs) and hormone replacement therapies (HRTs). Despite an increasing number of studies elucidating the effects of HCs and HRTs, little is known about the effects of different types of progestins included in these medications on the brain. Animal studies suggest that various progestins interact differently with sex steroid, mineralocorticoid and glucocorticoid receptors and have specific modulatory effects on neurotransmitter systems and on the expression of neuropeptides, suggesting differential impacts on cognition and behavior. This review focuses on the currently available knowledge from human behavioral and neuroimaging studies pooled with evidence from animal research regarding the effects of progestins on the brain. The reviewed information is highly relevant for improving women's mental health and making informed choices regarding specific types of contraception or treatment.
Collapse
Affiliation(s)
- Ramune Griksiene
- Department of Neurobiology and Biophysics, Life Sciences Center, Vilnius University, Lithuania
| | - Rasa Monciunskaite
- Department of Neurobiology and Biophysics, Life Sciences Center, Vilnius University, Lithuania
| | - Osvaldas Ruksenas
- Department of Neurobiology and Biophysics, Life Sciences Center, Vilnius University, Lithuania
| |
Collapse
|
19
|
Kolatorova L, Vitku J, Suchopar J, Hill M, Parizek A. Progesterone: A Steroid with Wide Range of Effects in Physiology as Well as Human Medicine. Int J Mol Sci 2022; 23:7989. [PMID: 35887338 PMCID: PMC9322133 DOI: 10.3390/ijms23147989] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022] Open
Abstract
Progesterone is a steroid hormone traditionally linked with female fertility and pregnancy. In current reproductive medicine, progesterone and its analogues play crucial roles. While the discovery of its effects has a long history, over recent decades, various novel actions of this interesting steroid have been documented, of which its neuro- and immunoprotective activities are the most widely discussed. Discoveries of the novel biological activities of progesterone have also driven research and development in the field of progesterone analogues used in human medicine. Progestogen treatment has traditionally and predominately been used in maintaining pregnancy, the prevention of preterm labor, various gynecological pathologies, and in lowering the negative effects of menopause. However, there are also various other medical fields where progesterone and its analogues could find application in the future. The aim of this work is to show the mechanisms of action of progesterone and its metabolites, the physiological and pharmacological actions of progesterone and its synthetic analogues in human medicine, as well as the impacts of its production and use on the environment.
Collapse
Affiliation(s)
- Lucie Kolatorova
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic; (J.V.); (M.H.)
| | - Jana Vitku
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic; (J.V.); (M.H.)
| | - Josef Suchopar
- DrugAgency, a.s., Klokotska 833/1a, 142 00 Prague, Czech Republic;
| | - Martin Hill
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic; (J.V.); (M.H.)
| | - Antonin Parizek
- Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General Teaching Hospital, Apolinarska 18, 128 51 Prague, Czech Republic;
| |
Collapse
|
20
|
Sarkar M, Sharma H, Singh P, Ranu R, Sharma RD, Agrawal U, Pal R. Progesterone limits the tumor-promoting effects of the beta-subunit of human chorionic gonadotropin via non-nuclear receptors. iScience 2022; 25:104527. [PMID: 35754725 PMCID: PMC9218381 DOI: 10.1016/j.isci.2022.104527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/13/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
The post-menopausal state in women is associated with increased cancer incidence, the reasons for which remain obscure. Curiously, increased circulating levels of beta-hCG (human chorionic gonadotropin) (a hormonal subunit linked with tumors of several lineages) are also often observed post-menopause. This study describes a previously unidentified interplay between beta-hCG and progesterone in tumorigenesis. Progesterone mediated apoptosis in beta-hCG responsive tumor cells via non-nuclear receptors. The transgenic expression of beta-hCG, particularly in the absence of the ovaries (a mimic of the post-menopausal state) constituted a potent pro-tumorigenic signal. Significantly, the administration of progesterone had significant anti-tumor effects. RNA-seq profiling identified molecular signatures associated with these processes. TCGA analysis revealed correlates between the expression of several newly identified genes and poor prognosis in post-menopausal patients of lung adenocarcinoma, colon adenocarcinoma, and glioblastoma. Specifically in these women, the detection of intra-tumoral/extra-tumoral beta-hCG may serve as a useful prognostic indicator, and treatment with progesterone on its detection may prove beneficial.
Collapse
Affiliation(s)
- Moumita Sarkar
- Immunoendocrinology Lab, National Institute of Immunology, New Delhi, Delhi 110067, India
| | - Harsh Sharma
- Amity Institute of Integrative Sciences and Health, Amity University, Gurugram, Haryana 122413, India
| | - Parminder Singh
- Larry L. Hillblom Center, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Ranbala Ranu
- Cancer Research Imaging and Biobanking Lab, National Institute of Pathology, New Delhi, Delhi 110029, India
| | - Ravi Datta Sharma
- Amity Institute of Integrative Sciences and Health, Amity University, Gurugram, Haryana 122413, India
| | - Usha Agrawal
- Cancer Research Imaging and Biobanking Lab, National Institute of Pathology, New Delhi, Delhi 110029, India
| | - Rahul Pal
- Immunoendocrinology Lab, National Institute of Immunology, New Delhi, Delhi 110067, India
| |
Collapse
|
21
|
Bello-Alvarez C, Zamora-Sánchez CJ, Camacho-Arroyo I. Rapid Actions of the Nuclear Progesterone Receptor through cSrc in Cancer. Cells 2022; 11:cells11121964. [PMID: 35741094 PMCID: PMC9221966 DOI: 10.3390/cells11121964] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 12/30/2022] Open
Abstract
The nuclear progesterone receptor (PR) is mainly known for its role as a ligand-regulated transcription factor. However, in the last ten years, this receptor’s extranuclear or rapid actions have gained importance in the context of physiological and pathophysiological conditions such as cancer. The PR’s polyproline (PXPP) motif allows protein–protein interaction through SH3 domains of several cytoplasmatic proteins, including the Src family kinases (SFKs). Among members of this family, cSrc is the most well-characterized protein in the scenario of rapid actions of the PR in cancer. Studies in breast cancer have provided the most detailed information on the signaling and effects triggered by the cSrc–PR interaction. Nevertheless, the study of this phenomenon and its consequences has been underestimated in other types of malignancies, especially those not associated with the reproductive system, such as glioblastomas (GBs). This review will provide a detailed analysis of the impact of the PR–cSrc interplay in the progression of some non-reproductive cancers, particularly, in GBs.
Collapse
Affiliation(s)
- Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México C.P. 0451, Mexico
| | - Carmen J Zamora-Sánchez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México C.P. 0451, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México C.P. 0451, Mexico
| |
Collapse
|
22
|
Atli MO, Akbalık ME, Kose M, Alak I, Atli Z, Hitit M. Expression pattern and cellular localization of two critical non-nuclear progesterone receptors in the ovine corpus luteum during the estrous cycle and early pregnancy. Anim Reprod Sci 2022; 243:107026. [PMID: 35752032 DOI: 10.1016/j.anireprosci.2022.107026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022]
Abstract
The study aimed to investigate the expression and cellular localization of two critical non-nuclear progesterone receptors, including membrane-associated-progesterone-receptor-component-1 (PGRMC1) and progestin and adipoQ receptor family member 7 (PAQR7) throughout the estrous cycle and early pregnancy in ovine corpus luteum (CL). Ewes were randomly grouped into cyclic (C, n = 4 per group) or pregnant (P, n = 4 per group) groups. Following slaughtering, the CL was obtained from both cyclic and pregnant ewes on days 12 (C12 and P12), 16 (C16 and P16), and 22 (C22 and P22). Western blotting and RT-qPCR were utilized to assess the expression levels of PGRMC1 and PAQR7, whereas immunohistochemistry was performed to determine the localization of PGRMC1 and PAQR7 in CL. Data were evaluated by one-way ANOVA, and the P < 0.05 was considered a significant difference. PGRMC1 was shown to be expressed in both small and large luteal cells and endothelial cells in CL, while PAQR7 expression was only found in small and large luteal cells. Compared to cycle days, pregnancy increased the expression of PGRMC1. PAQR7 did not differ during early pregnancy but reduced during the functional luteolysis stage (C16). mRNA and protein expression patterns for PGRMC1 and PAQR7 were similar on the studied days. This is the first study that demonstrates the expression and cellular localization of PGRMC1 and PAQR7 in ovine CL. We suggest that these receptors could execute a significant role in the ovine CL life span in both cyclic changes and the establishment of pregnancy.
Collapse
Affiliation(s)
- Mehmet O Atli
- Department of Reproduction, Faculty of Veterinary Medicine, Harran University, Şanlıurfa, Turkey.
| | - Mehmet Erdem Akbalık
- Department of Histology and Embiyology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey.
| | - Mehmet Kose
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey.
| | - Ilyas Alak
- Department of Biochemistry, Scholl Veterinary, Yidirim beyazit University, Ankara, Turkey.
| | - Zehra Atli
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey.
| | - Mustafa Hitit
- Department of Genetics, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey.
| |
Collapse
|
23
|
Etonogestrel Administration Reduces the Expression of PHOX2B and Its Target Genes in the Solitary Tract Nucleus. Int J Mol Sci 2022; 23:ijms23094816. [PMID: 35563209 PMCID: PMC9101578 DOI: 10.3390/ijms23094816] [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: 03/11/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Heterozygous mutations of the transcription factor PHOX2B are responsible for Congenital Central Hypoventilation Syndrome, a neurological disorder characterized by inadequate respiratory response to hypercapnia and life-threatening hypoventilation during sleep. Although no cure is currently available, it was suggested that a potent progestin drug provides partial recovery of chemoreflex response. Previous in vitro data show a direct molecular link between progestins and PHOX2B expression. However, the mechanism through which these drugs ameliorate breathing in vivo remains unknown. Here, we investigated the effects of chronic administration of the potent progestin drug Etonogestrel (ETO) on respiratory function and transcriptional activity in adult female rats. We assessed respiratory function with whole-body plethysmography and measured genomic changes in brain regions important for respiratory control. Our results show that ETO reduced metabolic activity, leading to an enhanced chemoreflex response and concurrent increased breathing cycle variability at rest. Furthermore, ETO-treated brains showed reduced mRNA and protein expression of PHOX2B and its target genes selectively in the dorsal vagal complex, while other areas were unaffected. Histological analysis suggests that changes occurred in the solitary tract nucleus (NTS). Thus, we propose that the NTS, rich in both progesterone receptors and PHOX2B, is a good candidate for ETO-induced respiratory modulation.
Collapse
|
24
|
Lee J, Troike K, Fodor R, Lathia JD. Unexplored Functions of Sex Hormones in Glioblastoma Cancer Stem Cells. Endocrinology 2022; 163:bqac002. [PMID: 35023543 PMCID: PMC8807164 DOI: 10.1210/endocr/bqac002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Indexed: 01/14/2023]
Abstract
Biological sex impacts a wide array of molecular and cellular functions that impact organismal development and can influence disease trajectory in a variety of pathophysiological states. In nonreproductive cancers, epidemiological sex differences have been observed in a series of tumors, and recent work has identified previously unappreciated sex differences in molecular genetics and immune response. However, the extent of these sex differences in terms of drivers of tumor growth and therapeutic response is less clear. In glioblastoma (GBM), the most common primary malignant brain tumor, there is a male bias in incidence and outcome, and key genetic and epigenetic differences, as well as differences in immune response driven by immune-suppressive myeloid populations, have recently been revealed. GBM is a prototypic tumor in which cellular heterogeneity is driven by populations of therapeutically resistant cancer stem cells (CSCs) that underlie tumor growth and recurrence. There is emerging evidence that GBM CSCs may show a sex difference, with male tumor cells showing enhanced self-renewal, but how sex differences impact CSC function is not clear. In this mini-review, we focus on how sex hormones may impact CSCs in GBM and implications for other cancers with a pronounced CSC population. We also explore opportunities to leverage new models to better understand the contribution of sex hormones vs sex chromosomes to CSC function. With the rising interest in sex differences in cancer, there is an immediate need to understand the extent to which sex differences impact tumor growth, including effects on CSC function.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic
| | - Katie Troike
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University
| | - R’ay Fodor
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University
| | - Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic
- Case Comprehensive Cancer Center
| |
Collapse
|
25
|
Ceccherini I, Kurek KC, Weese-Mayer DE. Developmental disorders affecting the respiratory system: CCHS and ROHHAD. HANDBOOK OF CLINICAL NEUROLOGY 2022; 189:53-91. [PMID: 36031316 DOI: 10.1016/b978-0-323-91532-8.00005-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Rapid-onset Obesity with Hypothalamic dysfunction, Hypoventilation, and Autonomic Dysregulation (ROHHAD) and Congenital Central Hypoventilation Syndrome (CCHS) are ultra-rare distinct clinical disorders with overlapping symptoms including altered respiratory control and autonomic regulation. Although both disorders have been considered for decades to be on the same spectrum with necessity of artificial ventilation as life-support, recent acquisition of specific knowledge concerning the genetic basis of CCHS coupled with an elusive etiology for ROHHAD have definitely established that the two disorders are different. CCHS is an autosomal dominant neurocristopathy characterized by alveolar hypoventilation resulting in hypoxemia/hypercarbia and features of autonomic nervous system dysregulation (ANSD), with presentation typically in the newborn period. It is caused by paired-like homeobox 2B (PHOX2B) variants, with known genotype-phenotype correlation but pathogenic mechanism(s) are yet unknown. ROHHAD is characterized by rapid weight gain, followed by hypothalamic dysfunction, then hypoventilation followed by ANSD, in seemingly normal children ages 1.5-7 years. Postmortem neuroanatomical studies, thorough clinical characterization, pathophysiological assessment, and extensive genetic inquiry have failed to identify a cause attributable to a traditional genetic basis, somatic mosaicism, epigenetic mechanism, environmental trigger, or other. To find the key to the ROHHAD pathogenesis and to improve its clinical management, in the present chapter, we have carefully compared CCHS and ROHHAD.
Collapse
Affiliation(s)
- Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Kyle C Kurek
- Department of Pathology & Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Debra E Weese-Mayer
- Division of Autonomic Medicine, Department of Pediatrics, Ann & Robert H Lurie Children's Hospital of Chicago and Stanley Manne Children's Research Institute; and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
26
|
Young CH, Snow B, DeVore SB, Mohandass A, Nemmara VV, Thompson PR, Thyagarajan B, Navratil AM, Cherrington BD. Progesterone stimulates histone citrullination to increase IGFBP1 expression in uterine cells. Reproduction 2021; 162:117-127. [PMID: 34034233 PMCID: PMC8284904 DOI: 10.1530/rep-21-0132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/25/2021] [Indexed: 12/31/2022]
Abstract
Peptidylarginine deiminases (PAD) enzymes were initially characterized in uteri, but since then little research has examined their function in this tissue. PADs post-translationally convert arginine residues in target proteins to citrulline and are highly expressed in ovine caruncle epithelia and ovine uterine luminal epithelial (OLE)-derived cell line. Progesterone (P4) not only maintains the uterine epithelia but also regulates the expression of endometrial genes that code for proteins that comprise the histotroph and are critical during early pregnancy. Given this, we tested whether P4 stimulates PAD-catalyzed histone citrullination to epigenetically regulate expression of the histotroph gene insulin-like growth factor binding protein 1 (IGFBP1) in OLE cells. 100 nM P4 significantly increases IGFBP1 mRNA expression; however, this increase is attenuated by pre-treating OLE cells with 100 nM progesterone receptor antagonist RU486 or 2 µM of a pan-PAD inhibitor. P4 treatment of OLE cells also stimulates citrullination of histone H3 arginine residues 2, 8, and 17 leading to enrichment of the ovine IGFBP1 gene promoter. Since PAD2 nuclear translocation and catalytic activity require calcium, we next investigated whether P4 triggers calcium influx in OLE cells. OLE cells were pre-treated with 10 nM nicardipine, an L-type calcium channel blocker, followed by stimulation with P4. Using fura2-AM imaging, we found that P4 initiates a rapid calcium influx through L-type calcium channels in OLE cells. Furthermore, this influx is necessary for PAD2 nuclear translocation and resulting citrullination of histone H3 arginine residues 2, 8, and 17. Our work suggests that P4 stimulates rapid calcium influx through L-type calcium channels initiating PAD-catalyzed histone citrullination and an increase in IGFBP1 expression.
Collapse
Affiliation(s)
- Coleman H Young
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| | - Bryce Snow
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| | - Stanley B DeVore
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Venkatesh V Nemmara
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Amy M Navratil
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| | - Brian D Cherrington
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
27
|
Chiarella SE, Cardet JC, Prakash YS. Sex, Cells, and Asthma. Mayo Clin Proc 2021; 96:1955-1969. [PMID: 34218868 PMCID: PMC8262071 DOI: 10.1016/j.mayocp.2020.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/19/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
There are marked sex differences in asthma prevalence and severity. Sex hormones play a central role in these sex biases and directly interact with multiple key cells involved in the pathogenesis of asthma. Here we review the known effects of estrogen, progesterone, and testosterone on airway epithelial cells, airway smooth muscle cells, the mononuclear phagocyte system, innate lymphoid cells, eosinophils, mast cells, T cells, and B cells, all in the context of asthma. Furthermore, we explore unresolved clinical questions, such as the role of sex hormones in the link between asthma and obesity.
Collapse
Affiliation(s)
- Sergio E Chiarella
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Juan Carlos Cardet
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida, Tampa
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN.
| |
Collapse
|
28
|
Marino L, Messina A, S Acierno J, Phan-Hug F, J Niederländer N, Santoni F, La Rosa S, Pitteloud N. Testosterone-induced increase in libido in a patient with a loss-of-function mutation in the AR gene. Endocrinol Diabetes Metab Case Rep 2021; 2021:EDM21-0031. [PMID: 34152287 PMCID: PMC8240814 DOI: 10.1530/edm-21-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/13/2021] [Indexed: 11/08/2022] Open
Abstract
SUMMARY Complete androgen-insensitivity syndrome (CAIS), a disorder of sex development (46,XY DSD), is caused primarily by mutations in the androgen receptor (AR). Gonadectomy is recommended due to the increased risk of gonadoblastoma, however, surgical intervention is often followed by loss of libido. We present a 26-year-old patient with CAIS who underwent gonadectomy followed by a significant decrease in libido, which was improved with testosterone treatment but not with estradiol. Genetic testing was performed and followed by molecular characterization. We found that this patient carried a previously unidentified start loss mutation in the androgen receptor. This variant resulted in an N-terminal truncated protein with an intact DNA binding domain and was confirmed to be loss-of-function in vitro. This unique CAIS case and detailed functional studies raise intriguing questions regarding the relative roles of testosterone and estrogen in libido, and in particular, the potential non-genomic actions of androgens. LEARNING POINTS N-terminal truncation of androgen receptor can cause androgen-insensitivity syndrome. Surgical removal of testosterone-producing gonads can result in loss of libido. Libido may be improved with testosterone treatment but not with estradiol in some forms of CAIS. A previously unreported AR mutation - p.Glu2_Met190del (c.2T>C) - is found in a CAIS patient and results in blunted AR transcriptional activity under testosterone treatment.
Collapse
Affiliation(s)
- Laura Marino
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| | - Andrea Messina
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| | - James S Acierno
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| | - Franziska Phan-Hug
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| | - Nicolas J Niederländer
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| | - Federico Santoni
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| | - Stefano La Rosa
- Department of Laboratory Medicine and Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Vaud, Switzerland
| | - Nelly Pitteloud
- Department of Service of Endocrinology, Diabetes, and Metabolism, Faculty of Biology and Medicine, University of Lausanne, Lausanne University Hospital, Lausanne, Vaud, Switzerland
| |
Collapse
|
29
|
Amirkhosravi L, Khaksari M, Sheibani V, Shahrokhi N, Ebrahimi MN, Amiresmaili S, Salmani N. Improved spatial memory, neurobehavioral outcomes, and neuroprotective effect after progesterone administration in ovariectomized rats with traumatic brain injury: Role of RU486 progesterone receptor antagonist. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:349-359. [PMID: 33995946 PMCID: PMC8087858 DOI: 10.22038/ijbms.2021.50973.11591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVES The contribution of classic progesterone receptors (PR) in interceding the neuroprotective efficacy of progesterone (P4) on the prevention of brain edema and long-time behavioral disturbances was assessed in traumatic brain injury (TBI). MATERIALS AND METHODS Female Wistar rats were ovariectomized and apportioned into 6 groups: sham, TBI, oil, P4, vehicle, and RU486. P4 or oil was injected following TBI. The antagonist of PR (RU486) or DMSO was administered before TBI. The brain edema and destruction of the blood-brain barrier (BBB) were determined. Intracranial pressure (ICP), cerebral perfusion pressure (CPP), and beam walk (BW) task were evaluated previously and at various times post-trauma. Long-time locomotor and cognitive consequences were measured one day before and on days 3, 7, 14, and 21 after the trauma. RESULTS RU486 eliminated the inhibitory effects of P4 on brain edema and BBB leakage (P<0.05, P<0.001, respectively). RU486 inhibited the decremental effect of P4 on ICP as well as the increasing effect of P4 on CPP (P<0.001) after TBI. Also, RU486 inhibited the effect of P4 on the increase in traversal time and reduction in vestibulomotor score in the BW task (P<0.001). TBI induced motor, cognitive, and anxiety-like disorders, which lasted for 3 weeks after TBI; but, P4 prevented these cognitive and behavioral abnormalities (P<0.05), and RU486 opposed this P4 effect (P<0.001). CONCLUSION The classic progesterone receptors have neuroprotective effects and prevent long-time behavioral and memory deficiency after brain trauma.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Neuroscience Research and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Nader Shahrokhi
- Physiology Research Centers, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Navid Ebrahimi
- Neuroscience Research and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Neda Salmani
- Department of Psychology, Genetic Institute, Islamic Azad University- Zarand Branch, Kerman, Iran
| |
Collapse
|
30
|
Contoreggi NH, Mazid S, Goldstein LB, Park J, Ovalles AC, Waters EM, Glass MJ, Milner TA. Sex and age influence gonadal steroid hormone receptor distributions relative to estrogen receptor β-containing neurons in the mouse hypothalamic paraventricular nucleus. J Comp Neurol 2021; 529:2283-2310. [PMID: 33341960 DOI: 10.1002/cne.25093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022]
Abstract
Within the hypothalamic paraventricular nucleus (PVN), estrogen receptor (ER) β and other gonadal hormone receptors play a role in central cardiovascular processes. However, the influence of sex and age on the cellular and subcellular relationships of ERβ with ERα, G-protein ER (GPER1), as well as progestin and androgen receptors (PR and AR) in the PVN is uncertain. In young (2- to 3-month-old) females and males, ERβ-enhanced green fluorescent protein (EGFP) containing neurons were approximately four times greater than ERα-labeled and PR-labeled nuclei in the PVN. In subdivisions of the PVN, young females, compared to males, had: (1) more ERβ-EGFP neurons in neuroendocrine rostral regions; (2) fewer ERα-labeled nuclei in neuroendocrine and autonomic projecting medial subregions; and (3) more ERα-labeled nuclei in an autonomic projecting caudal region. In contrast, young males, compared to females, had approximately 20 times more AR-labeled nuclei, which often colocalized with ERβ-EGFP in neuroendocrine (approximately 70%) and autonomic (approximately 50%) projecting subregions. Ultrastructurally, in soma and dendrites, PVN ERβ-EGFP colocalized primarily with extranuclear AR (approximately 85% soma) and GPER1 (approximately 70% soma). Aged (12- to 24-month-old) males had more ERβ-EGFP neurons in a rostral neuroendocrine subregion compared to aged females and females with accelerated ovarian failure (AOF) and in a caudal autonomic subregion compared to post-AOF females. Late-aged (18- to 24-month-old) females compared to early-aged (12- to 14-month-old) females and AOF females had fewer AR-labeled nuclei in neuroendrocrine and autonomic projecting subregions. These findings indicate that gonadal steroids may directly and indirectly influence PVN neurons via nuclear and extranuclear gonadal hormone receptors in a sex-specific manner.
Collapse
Affiliation(s)
| | - Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Lily B Goldstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - John Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Astrid C Ovalles
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, NY
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY.,Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, NY
| |
Collapse
|
31
|
Di Lascio S, Benfante R, Cardani S, Fornasari D. Research Advances on Therapeutic Approaches to Congenital Central Hypoventilation Syndrome (CCHS). Front Neurosci 2021; 14:615666. [PMID: 33510615 PMCID: PMC7835644 DOI: 10.3389/fnins.2020.615666] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Congenital central hypoventilation syndrome (CCHS) is a genetic disorder of neurodevelopment, with an autosomal dominant transmission, caused by heterozygous mutations in the PHOX2B gene. CCHS is a rare disorder characterized by hypoventilation due to the failure of autonomic control of breathing. Until now no curative treatment has been found. PHOX2B is a transcription factor that plays a crucial role in the development (and maintenance) of the autonomic nervous system, and in particular the neuronal structures involved in respiratory reflexes. The underlying pathogenetic mechanism is still unclear, although studies in vivo and in CCHS patients indicate that some neuronal structures may be damaged. Moreover, in vitro experimental data suggest that transcriptional dysregulation and protein misfolding may be key pathogenic mechanisms. This review summarizes latest researches that improved the comprehension of the molecular pathogenetic mechanisms responsible for CCHS and discusses the search for therapeutic intervention in light of the current knowledge about PHOX2B function.
Collapse
Affiliation(s)
- Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Roberta Benfante
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy.,CNR-Institute of Neuroscience, Milan, Italy.,NeuroMi-Milan Center for Neuroscience, University of Milano Bicocca, Milan, Italy
| | - Silvia Cardani
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy.,CNR-Institute of Neuroscience, Milan, Italy
| |
Collapse
|
32
|
Sitruk-Ware R, Bonsack B, Brinton R, Schumacher M, Kumar N, Lee JY, Castelli V, Corey S, Coats A, Sadanandan N, Gonzales-Portillo B, Heyck M, Shear A, Blaise C, Zhang H, Sheyner M, García-Sánchez J, Navarro L, El-Etr M, De Nicola AF, Borlongan CV. Progress in progestin-based therapies for neurological disorders. Neurosci Biobehav Rev 2020; 122:38-65. [PMID: 33359391 DOI: 10.1016/j.neubiorev.2020.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/26/2020] [Accepted: 12/12/2020] [Indexed: 12/16/2022]
Abstract
Hormone therapy, primarily progesterone and progestins, for central nervous system (CNS) disorders represents an emerging field of regenerative medicine. Following a failed clinical trial of progesterone for traumatic brain injury treatment, attention has shifted to the progestin Nestorone for its ability to potently and selectively transactivate progesterone receptors at relatively low doses, resulting in robust neurogenetic, remyelinating, and anti-inflammatory effects. That CNS disorders, including multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), spinal cord injury (SCI), and stroke, develop via demyelinating, cell death, and/or inflammatory pathological pathways advances Nestorone as an auspicious candidate for these disorders. Here, we assess the scientific and clinical progress over decades of research into progesterone, progestins, and Nestorone as neuroprotective agents in MS, ALS, SCI, and stroke. We also offer recommendations for optimizing timing, dosage, and route of the drug regimen, and identifying candidate patient populations, in advancing Nestorone to the clinic.
Collapse
Affiliation(s)
| | - Brooke Bonsack
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vanessa Castelli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alexandreya Coats
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bella Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matt Heyck
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alex Shear
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cozene Blaise
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Henry Zhang
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michael Sheyner
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Julián García-Sánchez
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Lisset Navarro
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
33
|
Astrocytoma: A Hormone-Sensitive Tumor? Int J Mol Sci 2020; 21:ijms21239114. [PMID: 33266110 PMCID: PMC7730176 DOI: 10.3390/ijms21239114] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Astrocytomas and, in particular, their most severe form, glioblastoma, are the most aggressive primary brain tumors and those with the poorest vital prognosis. Standard treatment only slightly improves patient survival. Therefore, new therapies are needed. Very few risk factors have been clearly identified but many epidemiological studies have reported a higher incidence in men than women with a sex ratio of 1:4. Based on these observations, it has been proposed that the neurosteroids and especially the estrogens found in higher concentrations in women's brains could, in part, explain this difference. Estrogens can bind to nuclear or membrane receptors and potentially stimulate many different interconnected signaling pathways. The study of these receptors is even more complex since many isoforms are produced from each estrogen receptor encoding gene through alternative promoter usage or splicing, with each of them potentially having a specific role in the cell. The purpose of this review is to discuss recent data supporting the involvement of steroids during gliomagenesis and to focus on the potential neuroprotective role as well as the mechanisms of action of estrogens in gliomas.
Collapse
|
34
|
Nader N, Dib M, Hodeify R, Courjaret R, Elmi A, Hammad AS, Dey R, Huang XY, Machaca K. Membrane progesterone receptor induces meiosis in Xenopus oocytes through endocytosis into signaling endosomes and interaction with APPL1 and Akt2. PLoS Biol 2020; 18:e3000901. [PMID: 33137110 PMCID: PMC7660923 DOI: 10.1371/journal.pbio.3000901] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 11/12/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
The steroid hormone progesterone (P4) mediates many physiological processes through either nuclear receptors that modulate gene expression or membrane P4 receptors (mPRs) that mediate nongenomic signaling. mPR signaling remains poorly understood. Here we show that the topology of mPRβ is similar to adiponectin receptors and opposite to that of G-protein-coupled receptors (GPCRs). Using Xenopus oocyte meiosis as a well-established physiological readout of nongenomic P4 signaling, we demonstrate that mPRβ signaling requires the adaptor protein APPL1 and the kinase Akt2. We further show that P4 induces clathrin-dependent endocytosis of mPRβ into signaling endosome, where mPR interacts transiently with APPL1 and Akt2 to induce meiosis. Our findings outline the early steps involved in mPR signaling and expand the spectrum of mPR signaling through the multitude of pathways involving APPL1. The steroid hormone progesterone mediates many physiological processes through either nuclear receptors that modulate gene expression, or membrane progesterone receptors (mPRs) that mediate non-genomic signaling. This study shows that non-genomic mPRβ signaling progresses through clathrin-dependent endocytosis into signaling endosomes where it interacts with and activates APPL1 and Akt2 to induce oocyte meiosis.
Collapse
Affiliation(s)
- Nancy Nader
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Maya Dib
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Rawad Hodeify
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Asha Elmi
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
- College of Health and Life Science, Hamad bin Khalifa University, Doha, Qatar
| | - Ayat S. Hammad
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
- College of Health and Life Science, Hamad bin Khalifa University, Doha, Qatar
| | - Raja Dey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
- * E-mail:
| |
Collapse
|
35
|
Shakkour Z, Habashy KJ, Berro M, Takkoush S, Abdelhady S, Koleilat N, Eid AH, Zibara K, Obeid M, Shear D, Mondello S, Wang KK, Kobeissy F. Drug Repurposing in Neurological Disorders: Implications for Neurotherapy in Traumatic Brain Injury. Neuroscientist 2020; 27:620-649. [PMID: 33089741 DOI: 10.1177/1073858420961078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) remains a significant leading cause of death and disability among adults and children globally. To date, there are no Food and Drug Administration-approved drugs that can substantially attenuate the sequelae of TBI. The innumerable challenges faced by the conventional de novo discovery of new pharmacological agents led to the emergence of alternative paradigm, which is drug repurposing. Repurposing of existing drugs with well-characterized mechanisms of action and human safety profiles is believed to be a promising strategy for novel drug use. Compared to the conventional discovery pathways, drug repurposing is less costly, relatively rapid, and poses minimal risk of the adverse outcomes to study on participants. In recent years, drug repurposing has covered a wide range of neurodegenerative diseases and neurological disorders including brain injury. This review highlights the advances in drug repurposing and presents some of the promising candidate drugs for potential TBI treatment along with their possible mechanisms of neuroprotection. Edaravone, glyburide, ceftriaxone, levetiracetam, and progesterone have been selected due to their potential role as putative TBI neurotherapeutic agents. These drugs are Food and Drug Administration-approved for purposes other than brain injuries; however, preclinical and clinical studies have shown their efficacy in ameliorating the various detrimental outcomes of TBI.
Collapse
Affiliation(s)
- Zaynab Shakkour
- Department of Biochemistry & Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Moussa Berro
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samira Takkoush
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nadia Koleilat
- Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Makram Obeid
- Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Deborah Shear
- Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Sicilia, Italy
| | - Kevin K Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
36
|
Hornung J, Lewis CA, Derntl B. Sex hormones and human brain function. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:195-207. [PMID: 33008525 DOI: 10.1016/b978-0-444-64123-6.00014-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sex hormones have organizational and activational effects on the human brain and can interact with the neurotransmitter systems. These biologic mechanisms may have a far-reaching impact, with both behavioral consequences and structural as well as functional brain modulation. The impact of cycling hormone levels throughout the menstrual cycle on cognitive and emotion processing has especially received some attention recently. Therefore, the aim of this chapter is to give an overview of findings regarding the effects of estradiol and progesterone, but also testosterone, on functional brain domains comprising cognition, emotion, and reward processing.
Collapse
Affiliation(s)
- Jonas Hornung
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Carolin A Lewis
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
37
|
Avallone R, Lucchi C, Puja G, Codeluppi A, Filaferro M, Vitale G, Rustichelli C, Biagini G. BV-2 Microglial Cells Respond to Rotenone Toxic Insult by Modifying Pregnenolone, 5α-Dihydroprogesterone and Pregnanolone Levels. Cells 2020; 9:E2091. [PMID: 32933155 PMCID: PMC7563827 DOI: 10.3390/cells9092091] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 01/05/2023] Open
Abstract
Neuroinflammation, whose distinctive sign is the activation of microglia, is supposed to play a key role in the development and progression of neurodegenerative diseases. The aim of this investigation was to determine levels of neurosteroids produced by resting and injured BV-2 microglial cells. BV-2 cells were exposed to increasing concentrations of rotenone to progressively reduce their viability by increasing reactive oxygen species (ROS) production. BV-2 cell viability was significantly reduced 24, 48 and 72 h after rotenone (50-1000 nM) exposure. Concomitantly, rotenone (50-100 nM) determined a dose-independent augmentation of ROS production. Then, BV-2 cells were exposed to a single, threshold dose of rotenone (75 nM) to evaluate the overtime release of neurosteroids. In particular, pregnenolone, pregnenolone sulfate, progesterone, 5α-dihydroprogesterone (5α-DHP), allopregnanolone, and pregnanolone, were quantified in the culture medium by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. BV-2 cells synthesized all the investigated neurosteroids and, after exposure to rotenone, 5αDHP and pregnanolone production was remarkably increased. In conclusion, we found that BV-2 cells not only synthesize several neurosteroids, but further increase this production following oxidative damage. Pregnanolone and 5α-DHP may play a role in modifying the progression of neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rossella Avallone
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| | - Giulia Puja
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Alessandro Codeluppi
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Monica Filaferro
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| | - Giovanni Vitale
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Cecilia Rustichelli
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| |
Collapse
|
38
|
Chen B, Ye P, Chen Y, Liu T, Cha JH, Yan X, Yang WH. Involvement of the Estrogen and Progesterone Axis in Cancer Stemness: Elucidating Molecular Mechanisms and Clinical Significance. Front Oncol 2020; 10:1657. [PMID: 33014829 PMCID: PMC7498570 DOI: 10.3389/fonc.2020.01657] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Estrogen and progesterone regulate the growth and development of human tissues, including the reproductive system and breasts, through estrogen and progesterone receptors, respectively. These receptors are also important indicators for the clinical prognosis of breast cancer and various reproductive cancers. Many studies have reported that cancer stem cells (CSCs) play a key role in tumor initiation, progression, metastasis, and recurrence. Although the role of estrogen and progesterone in human organs and various cancers has been studied, the molecular mechanisms underlying the action of these hormones on CSCs remain unclear. Therefore, further elucidation of the effects of estrogen and progesterone on CSCs should provide a new direction for developing pertinent therapies. In this review, we summarize the current knowledge on the estrogen and progesterone axis involved in cancer stemness and discuss potential therapeutic strategies to inhibit CSCs by targeting relevant pathways.
Collapse
Affiliation(s)
- Bi Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Peng Ye
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yeh Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Tong Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.,The Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
| | - Jong-Ho Cha
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, South Korea
| | - Xiuwen Yan
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Wen-Hao Yang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
39
|
Dwyer JB, Aftab A, Radhakrishnan R, Widge A, Rodriguez CI, Carpenter LL, Nemeroff CB, McDonald WM, Kalin NH. Hormonal Treatments for Major Depressive Disorder: State of the Art. Am J Psychiatry 2020; 177:686-705. [PMID: 32456504 PMCID: PMC7841732 DOI: 10.1176/appi.ajp.2020.19080848] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Major depressive disorder is a common psychiatric disorder associated with marked suffering, morbidity, mortality, and cost. The World Health Organization projects that by 2030, major depression will be the leading cause of disease burden worldwide. While numerous treatments for major depression exist, many patients do not respond adequately to traditional antidepressants. Thus, more effective treatments for major depression are needed, and targeting certain hormonal systems is a conceptually based approach that has shown promise in the treatment of this disorder. A number of hormones and hormone-manipulating compounds have been evaluated as monotherapies or adjunctive treatments for major depression, with therapeutic actions attributable not only to the modulation of endocrine systems in the periphery but also to the CNS effects of hormones on non-endocrine brain circuitry. The authors describe the physiology of the hypothalamic-pituitary-adrenal (HPA), hypothalamic-pituitary thyroid (HPT), and hypothalamic-pituitary-gonadal (HPG) axes and review the evidence for selected hormone-based interventions for the treatment of depression in order to provide an update on the state of this field for clinicians and researchers. The review focuses on the HPA axis-based interventions of corticotropin-releasing factor antagonists and the glucocorticoid receptor antagonist mifepristone, the HPT axis-based treatments of thyroid hormones (T3 and T4), and the HPG axis-based treatments of estrogen replacement therapy, the progesterone derivative allopregnanolone, and testosterone. While some treatments have largely failed to translate from preclinical studies, others have shown promising initial results and represent active fields of study in the search for novel effective treatments for major depression.
Collapse
Affiliation(s)
| | | | | | - Alik Widge
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis
| | - Carolyn I. Rodriguez
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and VA Palo Alto Health Care System, Palo Alto, Calif
| | - Linda L. Carpenter
- Department of Psychiatry and Human Behavior, Butler Hospital, Brown University, Providence, R.I
| | | | - William M. McDonald
- Department of Psychiatry and Human Behavior, Emory University School of Medicine, Atlanta
| | - Ned H. Kalin
- Department of Psychiatry, University of Wisconsin–Madison
| | -
- Child Study Center and Department of Radiology and Biomedical Imaging, Yale University, New Haven, Conn. (Dwyer); Department of Psychiatry, Case Western Reserve University, Cleveland, and Northcoast Behavioral Healthcare Hospital, Northfield, Ohio (Aftab); Yale School of Medicine, New Haven, Conn. (Radhakrishnan); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and VA Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Human Behavior, Butler Hospital, Brown University, Providence, R.I. (Carpenter); Department of Psychiatry, University of Texas at Austin (Nemeroff); Department of Psychiatry and Human Behavior, Emory University School of Medicine, Atlanta (McDonald); and Department of Psychiatry, University of Wisconsin-Madison (Kalin)
| |
Collapse
|
40
|
Guennoun R. Progesterone in the Brain: Hormone, Neurosteroid and Neuroprotectant. Int J Mol Sci 2020; 21:ijms21155271. [PMID: 32722286 PMCID: PMC7432434 DOI: 10.3390/ijms21155271] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/29/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
Progesterone has a broad spectrum of actions in the brain. Among these, the neuroprotective effects are well documented. Progesterone neural effects are mediated by multiple signaling pathways involving binding to specific receptors (intracellular progesterone receptors (PR); membrane-associated progesterone receptor membrane component 1 (PGRMC1); and membrane progesterone receptors (mPRs)) and local bioconversion to 3α,5α-tetrahydroprogesterone (3α,5α-THPROG), which modulates GABAA receptors. This brief review aims to give an overview of the synthesis, metabolism, neuroprotective effects, and mechanism of action of progesterone in the rodent and human brain. First, we succinctly describe the biosynthetic pathways and the expression of enzymes and receptors of progesterone; as well as the changes observed after brain injuries and in neurological diseases. Then, we summarize current data on the differential fluctuations in brain levels of progesterone and its neuroactive metabolites according to sex, age, and neuropathological conditions. The third part is devoted to the neuroprotective effects of progesterone and 3α,5α-THPROG in different experimental models, with a focus on traumatic brain injury and stroke. Finally, we highlight the key role of the classical progesterone receptors (PR) in mediating the neuroprotective effects of progesterone after stroke.
Collapse
Affiliation(s)
- Rachida Guennoun
- U 1195 Inserm and University Paris Saclay, University Paris Sud, 94276 Le kremlin Bicêtre, France
| |
Collapse
|
41
|
Stanojlović M, Guševac Stojanović I, Zarić M, Martinović J, Mitrović N, Grković I, Drakulić D. Progesterone Protects Prefrontal Cortex in Rat Model of Permanent Bilateral Common Carotid Occlusion via Progesterone Receptors and Akt/Erk/eNOS. Cell Mol Neurobiol 2020; 40:829-843. [PMID: 31865501 PMCID: PMC11448933 DOI: 10.1007/s10571-019-00777-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023]
Abstract
Sustained activation of pro-apoptotic signaling due to a sudden and prolonged disturbance of cerebral blood circulation governs the neurodegenerative processes in prefrontal cortex (PFC) of rats whose common carotid arteries are permanently occluded. The adequate neuroprotective therapy should minimize the activation of toxicity pathways and increase the activity of endogenous protective mechanisms. Several neuroprotectants have been proposed, including progesterone (P4). However, the underlying mechanism of its action in PFC following permanent bilateral occlusion of common carotid arteries is not completely investigated. We, thus herein, tested the impact of post-ischemic P4 treatment (1.7 mg/kg for seven consecutive days) on previously reported aberrant neuronal morphology and amount of DNA fragmentation, as well as the expression of progesterone receptors along with the key elements of Akt/Erk/eNOS signal transduction pathway (Bax, Bcl-2, cytochrome C, caspase 3, PARP, and the level of nitric oxide). The obtained results indicate that potential amelioration of histological changes in PFC might be associated with the absence of activation of Bax/caspase 3 signaling cascade and the decline of DNA fragmentation. The study also provides the evidence that P4 treatment in repeated regiment of administration might be effective in neuronal protection against ischemic insult due to re-establishment of the compromised action of Akt/Erk/eNOS-mediated signaling pathway and the upregulation of progesterone receptors.
Collapse
Affiliation(s)
- Miloš Stanojlović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Ivana Guševac Stojanović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Marina Zarić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Jelena Martinović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Dunja Drakulić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia.
| |
Collapse
|
42
|
Motta CM, Tammaro S, Di Lorenzo M, Panzuto R, Verderame M, Migliaccio V, Simoniello P. Spring and Fall recrudescence in Podarcis siculus ovaries: A role for progesterone. Gen Comp Endocrinol 2020; 290:113393. [PMID: 31982398 DOI: 10.1016/j.ygcen.2020.113393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/04/2020] [Accepted: 01/15/2020] [Indexed: 11/18/2022]
Abstract
In the lacertid Podarcis siculus the reproductive cycle is typically biphasic, with alternate recrudescence (Spring and Fall) and resting (Summer and Winter) phases. This study aimed to shed some light on the role exerted by progesterone during the two recrudescence periods; to this purpose, exogenous progesterone was administered intraperitoneally and the effects on oogonial proliferation, oocyte recruitment, and follicle cells apoptosis were determined. The presence and distribution of progesterone receptors was also investigated by immunohistochemistry and western blotting. Results indicate that progesterone would play different roles and follow different route of action in the two recrudescence periods thus confirming the complexity of the mechanisms controlling oogenesis in this species of vertebrate.
Collapse
Affiliation(s)
- C M Motta
- Department of Biology, University of Naples Federico II, Italy
| | - S Tammaro
- Department of Biology, University of Naples Federico II, Italy
| | - M Di Lorenzo
- Department of Biology, University of Naples Federico II, Italy
| | - R Panzuto
- Department of Biology, University of Naples Federico II, Italy
| | - M Verderame
- Department of Biology, University of Naples Federico II, Italy
| | - V Migliaccio
- Department of Biology, University of Naples Federico II, Italy
| | - P Simoniello
- Department of Sciences and Technologies, University of Naples Parthenope, Italy.
| |
Collapse
|
43
|
Roles of Progesterone, Testosterone and Their Nuclear Receptors in Central Nervous System Myelination and Remyelination. Int J Mol Sci 2020; 21:ijms21093163. [PMID: 32365806 PMCID: PMC7246940 DOI: 10.3390/ijms21093163] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Progesterone and testosterone, beyond their roles as sex hormones, are neuroactive steroids, playing crucial regulatory functions within the nervous system. Among these, neuroprotection and myelin regeneration are important ones. The present review aims to discuss the stimulatory effects of progesterone and testosterone on the process of myelination and remyelination. These effects have been demonstrated in vitro (i.e., organotypic cultures) and in vivo (cuprizone- or lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE)). Both steroids stimulate myelin formation and regeneration by acting through their respective intracellular receptors: progesterone receptors (PR) and androgen receptors (AR). Activation of these receptors results in multiple events involving direct transcription and translation, regulating general homeostasis, cell proliferation, differentiation, growth and myelination. It also ameliorates immune response as seen in the EAE model, resulting in a significant decrease in inflammation leading to a fast recovery. Although natural progesterone and testosterone have a therapeutic potential, their synthetic derivatives—the 19-norprogesterone (nestorone) and 7α-methyl-nortestosterone (MENT), already used as hormonal contraception or in postmenopausal hormone replacement therapies, may offer enhanced benefits for myelin repair. We summarize here a recent advancement in the field of myelin biology, to treat demyelinating disorders using the natural as well as synthetic analogs of progesterone and testosterone.
Collapse
|
44
|
Houshdaran S, Chen JC, Vallvé-Juanico J, Balayan S, Vo KC, Smith-McCune K, Greenblatt RM, Irwin JC, Giudice LC. Progestins Related to Progesterone and Testosterone Elicit Divergent Human Endometrial Transcriptomes and Biofunctions. Int J Mol Sci 2020; 21:ijms21072625. [PMID: 32283828 PMCID: PMC7177488 DOI: 10.3390/ijms21072625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/30/2022] Open
Abstract
Progestins are widely used for the treatment of gynecologic disorders and alone, or combined with an estrogen, are used as contraceptives. While their potencies, efficacies and side effects vary due to differences in structures, doses and routes of administration, little is known about their effects on the endometrial transcriptome in the presence or absence of estrogen. Herein, we assessed the transcriptome and pathways induced by progesterone (P4) and the three most commonly used synthetic progestins, medroxyprogesterone acetate (MPA), levonorgestrel (LNG), and norethindrone acetate (NETA), on human endometrial stromal fibroblasts (eSF), key players in endometrial physiology and reproductive success. While there were similar transcriptional responses, each progestin induced unique genes and biofunctions, consistent with their structural similarities to progesterone (P4 and MPA) or testosterone (LNG and NETA), involving cellular proliferation, migration and invasion. Addition of estradiol (E2) to each progestin influenced the number of differentially expressed genes and biofunctions in P4 and MPA, while LNG and NETA signatures were more independent of E2. Together, these data suggest different mechanisms of action for different progestins, with progestin-specific altered signatures when combined with E2. Further investigation is warranted for a personalized approach in different gynecologic disorders, for contraception, and minimizing side effects associated with their use.
Collapse
Affiliation(s)
- Sahar Houshdaran
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
| | | | - Júlia Vallvé-Juanico
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
| | - Shayna Balayan
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
| | - Kim Chi Vo
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
| | - Karen Smith-McCune
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
| | - Ruth M. Greenblatt
- Departments of Clinical Pharmacy, Medicine, Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA;
| | - Juan C. Irwin
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
| | - Linda C. Giudice
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA; (S.H.); (J.V.-J.); (S.B.); (K.C.V.); (K.S.-M.); (J.C.I.)
- Correspondence: ; Tel.: +1-4154762039
| |
Collapse
|
45
|
Trabert B, Sherman ME, Kannan N, Stanczyk FZ. Progesterone and Breast Cancer. Endocr Rev 2020; 41:5568276. [PMID: 31512725 PMCID: PMC7156851 DOI: 10.1210/endrev/bnz001] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 09/06/2019] [Indexed: 12/31/2022]
Abstract
Synthetic progestogens (progestins) have been linked to increased breast cancer risk; however, the role of endogenous progesterone in breast physiology and carcinogenesis is less clearly defined. Mechanistic studies using cell culture, tissue culture, and preclinical models implicate progesterone in breast carcinogenesis. In contrast, limited epidemiologic data generally do not show an association of circulating progesterone levels with risk, and it is unclear whether this reflects methodologic limitations or a truly null relationship. Challenges related to defining the role of progesterone in breast physiology and neoplasia include: complex interactions with estrogens and other hormones (eg, androgens, prolactin, etc.), accounting for timing of blood collections for hormone measurements among cycling women, and limitations of assays to measure progesterone metabolites in blood and progesterone receptor isotypes (PRs) in tissues. Separating the individual effects of estrogens and progesterone is further complicated by the partial dependence of PR transcription on estrogen receptor (ER)α-mediated transcriptional events; indeed, interpreting the integrated interaction of the hormones may be more essential than isolating independent effects. Further, many of the actions of both estrogens and progesterone, particularly in "normal" breast tissues, are driven by paracrine mechanisms in which ligand binding to receptor-positive cells evokes secretion of factors that influence cell division of neighboring receptor-negative cells. Accordingly, blood and tissue levels may differ, and the latter are challenging to measure. Given conflicting data related to the potential role of progesterone in breast cancer etiology and interest in blocking progesterone action to prevent or treat breast cancer, we provide a review of the evidence that links progesterone to breast cancer risk and suggest future directions for filling current gaps in our knowledge.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Mark E Sherman
- Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - Nagarajan Kannan
- Laboratory of Stem Cell and Cancer Biology, Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Frank Z Stanczyk
- Departments of Obstetrics and Gynecology, and Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
46
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
47
|
Jarras H, Bourque M, Poirier AA, Morissette M, Coulombe K, Di Paolo T, Soulet D. Neuroprotection and immunomodulation of progesterone in the gut of a mouse model of Parkinson's disease. J Neuroendocrinol 2020; 32:e12782. [PMID: 31430407 DOI: 10.1111/jne.12782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal symptoms appear in Parkinson's disease patients many years before motor symptoms, suggesting the implication of dopaminergic neurones of the gut myenteric plexus. Inflammation is also known to be increased in PD. We previously reported neuroprotection with progesterone in the brain of mice lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and hypothesised that it also has neuroprotective and immunomodulatory activities in the gut. To test this hypothesis, we investigated progesterone administered to adult male C57BL/6 mice for 10 days and treated with MPTP on day 5. In an additional experiment, progesterone was administered for 5 days following MPTP treatment. Ilea were collected on day 10 of treatment and microdissected to isolate the myenteric plexus. Dopaminergic neurones were reduced by approximately 60% and pro-inflammatory macrophages were increased by approximately 50% in MPTP mice compared to intact controls. These changes were completely prevented by progesterone administered before and after MPTP treatment and were normalised by 8 mg kg-1 progesterone administered after MPTP. In the brain of MPTP mice, brain-derived neurotrophic peptide (BDNF) and glial fibrillary acidic protein (GFAP) were associated with progesterone neuroprotection. In the myenteric plexus, increased BDNF levels compared to controls were measured in MPTP mice treated with 8 mg kg-1 progesterone started post MPTP, whereas GFAP levels remained unchanged. In conclusion, the results obtained in the present study show neuroprotective and anti-inflammatory effects of progesterone in the myenteric plexus of MPTP mice that are similar to our previous findings in the brain. Progesterone is non-feminising and could be used for both men and women in the pre-symptomatic stages of the disease.
Collapse
Affiliation(s)
- Hend Jarras
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Mélanie Bourque
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Andrée-Anne Poirier
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Katherine Coulombe
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Denis Soulet
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| |
Collapse
|
48
|
Yilmaz C, Karali K, Fodelianaki G, Gravanis A, Chavakis T, Charalampopoulos I, Alexaki VI. Neurosteroids as regulators of neuroinflammation. Front Neuroendocrinol 2019; 55:100788. [PMID: 31513776 DOI: 10.1016/j.yfrne.2019.100788] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/12/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is a physiological protective response in the context of infection and injury. However, neuroinflammation, especially if chronic, may also drive neurodegeneration. Neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and traumatic brain injury (TBI), display inflammatory activation of microglia and astrocytes. Intriguingly, the central nervous system (CNS) is a highly steroidogenic environment synthesizing steroids de novo, as well as metabolizing steroids deriving from the circulation. Neurosteroid synthesis can be substantially affected by neuroinflammation, while, in turn, several steroids, such as 17β-estradiol, dehydroepiandrosterone (DHEA) and allopregnanolone, can regulate neuroinflammatory responses. Here, we review the role of neurosteroids in neuroinflammation in the context of MS, AD, PD and TBI and describe underlying molecular mechanisms. Moreover, we introduce the concept that synthetic neurosteroid analogues could be potentially utilized for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Canelif Yilmaz
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Kanelina Karali
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
49
|
Meng ID, Barton ST, Goodney I, Russell R, Mecum NE. Progesterone Application to the Rat Forehead Produces Corneal Antinociception. Invest Ophthalmol Vis Sci 2019; 60:1706-1713. [PMID: 31013343 PMCID: PMC6736375 DOI: 10.1167/iovs.18-26049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Ocular pain and discomfort are the most defining symptoms of dry eye disease. We determined the ability of topical progesterone to affect corneal sensitivity and brainstem processing of nociceptive inputs. Methods Progesterone or vehicle gel was applied to the shaved forehead in male Sprague Dawley rats. As a site control, gel also was applied to the cheek on the side contralateral to corneal stimulation. Corneal mechanical thresholds were determined using the Cochet-Bonnet esthesiometer in intact and lacrimal gland excision–induced dry eye animals. Eye wipe behaviors in response to hypertonic saline and capsaicin were examined, and corneal mustard oil-induced c-Fos immunohistochemistry was quantified in the brainstem spinal trigeminal nucleus. Results Progesterone gel application to the forehead, but not the contralateral cheek, increased corneal mechanical thresholds in intact and lacrimal gland excision animals beginning <30 minutes after treatment. Subcutaneous injection of the local anesthetic bupivacaine into the forehead region before application of progesterone prevented the increase in corneal mechanical thresholds. Furthermore, progesterone decreased capsaicin-evoked eye wipe behavior in intact animals and hypertonic saline evoked eye wipe behavior in dry eye animals. The number of Fos-positive neurons located in the caudal region of the spinal trigeminal nucleus after corneal mustard oil application was reduced in progesterone-treated animals. Conclusions Results from this study indicate that progesterone, when applied to the forehead, produces analgesia as indicated by increased corneal mechanical thresholds and decreased nociceptive responses to hypertonic saline and capsaicin.
Collapse
Affiliation(s)
- Ian D Meng
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States
| | - Stephen T Barton
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Ian Goodney
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Rachel Russell
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Neal E Mecum
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Molecular and Biomedical Sciences, University of Maine, Orono, Maine, United States
| |
Collapse
|
50
|
Progesterone, 5a-dihydropogesterone and allopregnanolone's effects on seizures: A review of animal and clinical studies. Seizure 2018; 63:26-36. [DOI: 10.1016/j.seizure.2018.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023] Open
|