1
|
Lu L, Jifu C, Pei B, Gao X, Xu Z, Yin X, Xia J, Wang J. USP18 is a key regulator of immune function in mouse midbrain microglia. Life Sci 2025; 374:123692. [PMID: 40348176 DOI: 10.1016/j.lfs.2025.123692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/21/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
AIMS Ubiquitin-specific peptidase 18 (USP18) is an important member of the deubiquitinating enzyme family, which has received much attention in recent years for its role in microglia regulation. The aim of this study was to investigate the role of USP18 in midbrain and its potential molecular mechanisms. METHODS In this study, we assessed behavioural phenotypes and pathological changes by adeno-associated virus (AAV)-mediated midbrain-specific USP18 high-expression mouse model. RNA sequencing and untargeted metabolomics were used for multi-omics analysis, and protein expression was detected by Western blot, and ELISA was used to detect neuroinflammatory factor levels. RESULTS Our analyses suggest that USP18 is a key regulator of immune activity in the midbrain. USP18 helps maintain the resting state of microglia and exerts neuroprotective effects by promoting TH protein expression. In the midbrain, interference with USP18 expression resulted in significant changes in neuroimmune responses, gene expression associated with inflammation, and metabolite levels. Notably, the TLR signalling pathway was significantly enriched. Loss of USP18 led to a significant increase in the expression of TLR2, Iba-1, and GFAP proteins and a significant decrease in TH levels, and this change was particularly pronounced in microglia. Importantly, the changes observed in USP18 silencing were also reflected in brain tissues of human neurodegenerative diseases. SIGNIFICANCE This study reveals the critical role of USP18 in midbrain and microglia, and suggests it can finely regulate neuroinflammatory and immune responses by modulating TLR2 protein levels. The findings provide new ideas for understanding mechanisms of neurodegenerative diseases and developing therapeutic strategies.
Collapse
Affiliation(s)
- Linxia Lu
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, China
| | - Cili Jifu
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, China
| | - Baoxin Pei
- College of Public Health, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Xuming Gao
- College of Biology and Agriculture, Jiamusi University, Jiamusi, Heilongjiang Province 154007, China
| | - Zhenyu Xu
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, China
| | - Xianglin Yin
- College of Public Health, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Jun Xia
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, China
| | - Jingtao Wang
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, China; College of Public Health, Jiamusi University, Jiamusi, Heilongjiang 154007, China.
| |
Collapse
|
2
|
Servi R, Akkoç RF, Aksu F, Servi S. Therapeutic potential of enzymes, neurosteroids, and synthetic steroids in neurodegenerative disorders: A critical review. J Steroid Biochem Mol Biol 2025; 251:106766. [PMID: 40288591 DOI: 10.1016/j.jsbmb.2025.106766] [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: 03/10/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Neurodegenerative disorders present a significant challenge to healthcare systems, mainly due to the limited availability of effective treatment options to halt or reverse disease progression. Endogenous steroids synthesized in the central nervous system, such as pregnenolone (PREG), dehydroepiandrosterone (DHEA), progesterone (PROG), and allopregnanolone (ALLO), have been identified as potential therapeutic agents for neurodegenerative diseases. Neurosteroids such as ALLO, DHEA, and PROG, as well as their synthetic analogs like Ganaxolene, Fluasterone, and Olexoxime, offer promising effects for conditions such as Alzheimer's disease (AD) and depression. Moreover, Brexanolone and Ganaxolone are synthetic steroids approved for the treatment of postpartum depression and epilepsy, respectively. Neurosteroids such as ALLO are crucial in modulating GABAergic neurotransmission and reducing neuroinflammation. These compounds enhance the activity of GABA-A receptors, leading to increased inhibitory signaling in the brain, which can help regulate mood, cognition, and neuroprotection. Small clinical trials and observational studies indicate that ALLO may have cognitive benefits, but no large-scale, definitive meta-analysis confirms a 20 % improvement in AD patients. Mitochondrial dysfunction plays a vital role in the pathogenesis of numerous neurological diseases due to the high-energy demand and sensitivity of neurons to oxidative stress. Reduced mitochondrial function leads to amyloid-beta plaques and tau tangles accumulation in AD. In Parkinson's disease (PD), mitochondrial dysfunction resulting from the PINK1 or Parkin genes leads to energy deficiencies and the accumulation of toxic byproducts. Mutations in genes such as SOD1, C9orf72, and TDP-43 have been associated with mitochondrial dysfunction in amyotrophic lateral sclerosis (ALS). Moreover, studies on these neurodegenerative diseases suggest that inflammation is not merely a consequence of neurodegeneration but is also an essential factor in this process. Many neurological disorders involve multifaceted interactions between genetics, the environment, and immune responses, making it difficult to pinpoint their exact causes. Future research aims to overcome these hurdles through genetic advances, regenerative medicine, and personalized therapies. Cutting-edge technologies such as artificial intelligence and high-throughput screening are expected to accelerate drug discovery and improve diagnostic accuracy. Increasing collaboration between interdisciplinary fields such as bioinformatics, neuroscience, and immunology will lead to innovative treatment strategies. This comprehensive review discusses the therapeutic effects of enzymes, neurosteroids, and synthetic steroids in different neurodegenerative diseases, particularly AD, PD, ALS, and MS. Potential challenges in the therapeutic use of neurosteroids, such as the limited bioavailability and off-target effects of synthetic steroids, are also discussed, and an up-to-date and comprehensive review of the impact of these steroids on neurodegenerative disorders is presented.
Collapse
Affiliation(s)
- Refik Servi
- Fırat University, Faculty of Medicine, Department of Anatomy, Elazığ, Turkey.
| | - Ramazan Fazıl Akkoç
- Fırat University, Faculty of Medicine, Department of Anatomy, Elazığ, Turkey.
| | - Feyza Aksu
- Fırat University, Faculty of Medicine, Department of Anatomy, Elazığ, Turkey.
| | - Süleyman Servi
- Fırat University, Faculty of Science, Department of Chemistry, Elazığ, Turkey.
| |
Collapse
|
3
|
Cattaneo C, Pagonabarraga J. Sex Differences in Parkinson's Disease: A Narrative Review. Neurol Ther 2025; 14:57-70. [PMID: 39630386 PMCID: PMC11762054 DOI: 10.1007/s40120-024-00687-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 01/27/2025] Open
Abstract
Sex differences in epidemiology, clinical features, and therapeutical responses are emerging in several movement disorders, even though they are still not widely recognized. Parkinson's disease (PD) is not an exception: men and women suffering from PD have different levels of disability. Research has been performed using multiple databases and scientific journals; this review summarizes the available evidence on sex differences in PD regarding epidemiology, risk factors, genetics, clinical phenotype, social impact, and therapeutic management. The role of hormones in determining such differences is also briefly discussed. The results confirm the existence of differences between men and women in PD; women have a higher risk of developing disabling motor complications and non-motor fluctuations compared to men, while men have a higher risk of developing cognitive impairment, postural instability, and gait disorders. Improving our knowledge in these differences may result in the implementation of strategies for disease-tailored treatment and management.
Collapse
|
4
|
Scuto M, Majzúnová M, Torcitto G, Antonuzzo S, Rampulla F, Di Fatta E, Trovato Salinaro A. Functional Food Nutrients, Redox Resilience Signaling and Neurosteroids for Brain Health. Int J Mol Sci 2024; 25:12155. [PMID: 39596221 PMCID: PMC11594618 DOI: 10.3390/ijms252212155] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
The interplay between functional food nutrients and neurosteroids has garnered significant attention for its potential to enhance stress resilience in health and/or disease. Several bioactive nutrients, including medicinal herbs, flavonoids, and bioavailable polyphenol-combined nanoparticles, as well as probiotics, vitamin D and omega-3 fatty acids, have been shown to improve blood-brain barrier (BBB) dysfunction, endogenous neurosteroid homeostasis and brain function. These nutrients can inhibit oxidative stress and neuroinflammation, which are linked to the pathogenesis of various neurological disorders. Interestingly, flavonoids exhibit dose-dependent effects, activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway at the physiological/low dose (neurohormesis). This leads to the upregulation of antioxidant phase II genes and proteins such as heme oxygenase-1 (HO-1) and sirtuin-1 (Sirt1), which are activated by curcumin and resveratrol, respectively. These adaptive neuronal response mechanisms help protect against reactive oxygen species (ROS) and neurotoxicity. Impaired Nrf2 and neurosteroid hormone signaling in the brain can exacerbate selective vulnerability to neuroinflammatory conditions, contributing to the onset and progression of neurodegenerative and psychiatric disorders, including Alzheimer's disease, anxiety and depression and other neurological disorders, due to the vulnerability of neurons to stress. This review focuses on functional food nutrients targeting Nrf2 antioxidant pathway and redox resilience genes to regulate the neurosteroid homeostasis and BBB damage associated with altered GABAergic neurotransmission. By exploring the underlying molecular mechanisms using innovative technologies, we aim to develop promising neuroprotective strategies and personalized nutritional and neuroregenerative therapies to prevent or attenuate oxidative stress and neuroinflammation, ultimately promoting brain health.
Collapse
Affiliation(s)
- Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | - Miroslava Majzúnová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 84215 Bratislava, Slovakia;
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovakia
| | - Gessica Torcitto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | - Silvia Antonuzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | - Francesco Rampulla
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | | | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| |
Collapse
|
5
|
Bourque M, Morissette M, Isenbrandt A, Giatti S, Melcangi RC, Carta M, Frau R, Bortolato M, Soulet D, Di Paolo T. Effect of 5-alpha reductase inhibitors in animal models of Parkinson's disease. Front Neuroendocrinol 2024; 75:101156. [PMID: 39353534 DOI: 10.1016/j.yfrne.2024.101156] [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: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD) is characterized by motor symptoms due to loss of brain dopamine and non-motor symptoms, including gastrointestinal disorders. Although there is no cure for PD, symptomatic treatments are available. L-Dopa is the gold standard PD therapy, but most patients develop dyskinesias (LID), which are challenging to manage. Amantadine is recognized as the most effective drug for LID, but its adverse effects limit the use in patients. Here we review how 5α-reductase inhibitors (5ARIs), drugs used to treat benign prostatic hyperplasia and alopecia, exhibit beneficial effects in PD animal models. 5ARIs show neuroprotective properties in brain and gut dopaminergic systems, and reduce dyskinesias in rodent model of PD. Additionally, the 5ARI finasteride dampened dopaminergic-induced drug gambling in PD patients. Neuroprotection and antidyskinetic activities of 5ARIs in animal models of PD suggest their potential repurposing in men with PD to address gut dysfunction, protect brain DA and inhibit dyskinesias.
Collapse
Affiliation(s)
- Mélanie Bourque
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Amandine Isenbrandt
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, USA
| | - Denis Soulet
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
| |
Collapse
|
6
|
Scheggi S, Concas L, Corsi S, Carta M, Melis M, Frau R. Expanding the therapeutic potential of neuro(active)steroids: a promising strategy for hyperdopaminergic behavioral phenotypes. Neurosci Biobehav Rev 2024; 164:105842. [PMID: 39103066 DOI: 10.1016/j.neubiorev.2024.105842] [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: 05/23/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
Imbalances in dopamine activity significantly contribute to the pathophysiology of several neuropsychiatric disorders, including addiction, ADHD, schizophrenia, impulse control disorders, and Parkinson's Disease. Neuro(active)steroids, comprising endogenous steroids that finely modulate neuronal activity, are considered crucial regulators of brain function and behavior, with implications in various physiological processes and pathological conditions. Specifically, subclasses of Neuro(active)steroids belonging to the 5α reductase pathway are prominently involved in brain disorders characterized by dopaminergic signaling imbalances. This review highlights the neuromodulatory effects of Neuro(active)steroids on the dopamine system and related aberrant behavioral phenotypes. We critically appraise the role of pregnenolone, progesterone, and allopregnanolone on dopamine signaling. Additionally, we discuss the impact of pharmacological interventions targeting 5α reductase activity in neuropsychiatric conditions characterized by excessive activation of the dopaminergic system, ranging from psychotic (endo)phenotypes and motor complications to decision-making problems and addiction.
Collapse
Affiliation(s)
- Simona Scheggi
- Dept. of Molecular and Developmental Medicine, University of Siena, Italy
| | - Luca Concas
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Sara Corsi
- Dept. of Developmental and Regenerative Neurobiology, Lund University, Sweden
| | - Manolo Carta
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Miriam Melis
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Roberto Frau
- Dept. Of Biomedical Sciences, University of Cagliari, Italy; Guy Everett Laboratory, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
7
|
Cavallieri F, Lucchi C, Grisanti S, Monfrini E, Fioravanti V, Toschi G, Di Rauso G, Rossi J, Di Fonzo A, Biagini G, Valzania F. Neurosteroid Levels in GBA Mutated and Non-Mutated Parkinson's Disease: A Possible Factor Influencing Clinical Phenotype? Biomolecules 2024; 14:1022. [PMID: 39199409 PMCID: PMC11352262 DOI: 10.3390/biom14081022] [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: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024] Open
Abstract
Neurosteroids are pleiotropic molecules involved in various neurodegenerative diseases with neuroinflammation. We assessed neurosteroids' serum levels in a cohort of Parkinson's Disease (PD) patients with heterozygous glucocerebrosidase (GBA) mutations (GBA-PD) compared with matched cohorts of consecutive non-mutated PD (NM-PD) patients and healthy subjects with (GBA-HC) and without (NM-HC) GBA mutations. A consecutive cohort of GBA-PD was paired for age, sex, disease duration, Hoehn and Yahr stage, and comorbidities with a cohort of consecutive NM-PD. Two cohorts of GBA-HC and HC were also considered. Clinical assessment included the Movement Disorder Society revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and the Montreal Cognitive Assessment (MoCA). Serum samples were processed and analyzed by liquid chromatography coupled with the triple quadrupole mass spectrometry. Twenty-two GBA-PD (males: 11, age: 63.68), 22 NM-PD (males: 11, age: 63.05), 14 GBA-HC (males: 8; age: 49.36), and 15 HC (males: 4; age: 60.60) were studied. Compared to NM-PD, GBA-PD showed more hallucinations and psychosis (p < 0.05, Fisher's exact test) and higher MDS-UPDRS part-II (p < 0.05). Most of the serum neurosteroids were reduced in both GBA-PD and NM-PD compared to the respective control cohorts, except for 5α-dihydroprogesterone. Allopregnanolone was the only neurosteroid significantly lower (p < 0.01, Dunn's test) in NM-PD compared to GBA-PD patients. Only in GBA-PD, allopregnanolone, and pregnanolone levels correlated (Spearman) with a more severe MDS-UPDRS part-III. Allopregnanolone levels also negatively correlated with MoCA scores, and pregnanolone levels correlated with more pronounced bradykinesia. This pilot study provides the first observation of changes in neurosteroid peripheral levels in GBA-PD. The involvement of the observed changes in the development of neuropsychological and motor symptoms of GBA-PD deserves further attention.
Collapse
Affiliation(s)
- Francesco Cavallieri
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.C.); (V.F.); (G.T.); (G.D.R.); (J.R.); (F.V.)
| | - Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Sara Grisanti
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Edoardo Monfrini
- Neurology Unit, Fondazione IRCCS Ca’ Grande Ospedale Maggiore Policlinico, 20122 Milan, Italy; (E.M.); (A.D.F.)
| | - Valentina Fioravanti
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.C.); (V.F.); (G.T.); (G.D.R.); (J.R.); (F.V.)
| | - Giulia Toschi
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.C.); (V.F.); (G.T.); (G.D.R.); (J.R.); (F.V.)
| | - Giulia Di Rauso
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.C.); (V.F.); (G.T.); (G.D.R.); (J.R.); (F.V.)
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Jessica Rossi
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.C.); (V.F.); (G.T.); (G.D.R.); (J.R.); (F.V.)
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Alessio Di Fonzo
- Neurology Unit, Fondazione IRCCS Ca’ Grande Ospedale Maggiore Policlinico, 20122 Milan, Italy; (E.M.); (A.D.F.)
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Franco Valzania
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.C.); (V.F.); (G.T.); (G.D.R.); (J.R.); (F.V.)
| |
Collapse
|
8
|
Cioffi L, Grassi D, Diviccaro S, Caruso D, Pinto-Benito D, Arevalo MA, Garcia-Segura LM, Melcangi RC, Giatti S. Sex chromosome complement interacts with gonadal hormones in determining regional-specific neuroactive steroid levels in plasma, hippocampus, and hypothalamus. A study using the four core genotype mouse model. J Steroid Biochem Mol Biol 2024; 241:106514. [PMID: 38554982 DOI: 10.1016/j.jsbmb.2024.106514] [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: 02/02/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
An important aspect of the neuromodulatory and neuroprotective actions exerted by neuroactive steroids is that they are sex-specific, as determined by the sexually dimorphic levels of these molecules in plasma and the nervous tissue. Thus, the identification of the factors that generate the sex-dimorphic levels of neuroactive steroids may be crucial from a neuroprotectant perspective. The main driver for sex determination in mammals is the SRY gene and the subsequent presence of a specific gonad: testes for males and ovaries for females, thus producing hormonal compounds, primarily androgens and estrogens, respectively. Nowadays, it is well established that despite the relevance of gonads, other factors control sexual features, and, among them, sex chromosome complement is highly relevant. In this study, neuroactive steroids were evaluated by liquid chromatography-tandem mass spectrometry in the hypothalamus, the hippocampus, and plasma of the four core genotype mouse model, to determine the relative contribution of sex chromosome complement and gonads in determining their sex dimorphic levels. The data obtained reveal that although gonads are the main contributing factor for sex differences in neuroactive steroid levels, the levels of some neuroactive steroids, including testosterone, are also influenced in brain and plasma by tissue-specific actions of sex chromosomes. The data presented here adds a new piece to the puzzle of steroid level regulation, which may be useful in designing sex-specific neuroprotective approaches to pathological conditions affecting the nervous system.
Collapse
Affiliation(s)
- Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Daniela Grassi
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid 28029, Spain
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Daniel Pinto-Benito
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain and Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Maria-Angeles Arevalo
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain and Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Luis Miguel Garcia-Segura
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain and Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| |
Collapse
|
9
|
Bäckström T, Doverskog M, Blackburn TP, Scharschmidt BF, Felipo V. Allopregnanolone and its antagonist modulate neuroinflammation and neurological impairment. Neurosci Biobehav Rev 2024; 161:105668. [PMID: 38608826 DOI: 10.1016/j.neubiorev.2024.105668] [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: 10/19/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
Neuroinflammation accompanies several brain disorders, either as a secondary consequence or as a primary cause and may contribute importantly to disease pathogenesis. Neurosteroids which act as Positive Steroid Allosteric GABA-A receptor Modulators (Steroid-PAM) appear to modulate neuroinflammation and their levels in the brain may vary because of increased or decreased local production or import from the systemic circulation. The increased synthesis of steroid-PAMs is possibly due to increased expression of the mitochondrial cholesterol transporting protein (TSPO) in neuroinflammatory tissue, and reduced production may be due to changes in the enzymatic activity. Microglia and astrocytes play an important role in neuroinflammation, and their production of inflammatory mediators can be both activated and inhibited by steroid-PAMs and GABA. What is surprising is the finding that both allopregnanolone, a steroid-PAM, and golexanolone, a novel GABA-A receptor modulating steroid antagonist (GAMSA), can inhibit microglia and astrocyte activation and normalize their function. This review focuses on the role of steroid-PAMs in neuroinflammation and their importance in new therapeutic approaches to CNS and liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
10
|
Plug BC, Revers IM, Breur M, González GM, Timmerman JA, Meijns NRC, Hamberg D, Wagendorp J, Nutma E, Wolf NI, Luchicchi A, Mansvelder HD, van Til NP, van der Knaap MS, Bugiani M. Human post-mortem organotypic brain slice cultures: a tool to study pathomechanisms and test therapies. Acta Neuropathol Commun 2024; 12:83. [PMID: 38822428 PMCID: PMC11140981 DOI: 10.1186/s40478-024-01784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/16/2024] [Indexed: 06/03/2024] Open
Abstract
Human brain experimental models recapitulating age- and disease-related characteristics are lacking. There is urgent need for human-specific tools that model the complex molecular and cellular interplay between different cell types to assess underlying disease mechanisms and test therapies. Here we present an adapted ex vivo organotypic slice culture method using human post-mortem brain tissue cultured at an air-liquid interface to also study brain white matter. We assessed whether these human post-mortem brain slices recapitulate the in vivo neuropathology and if they are suitable for pathophysiological, experimental and pre-clinical treatment development purposes, specifically regarding leukodystrophies. Human post-mortem brain tissue and cerebrospinal fluid were obtained from control, psychiatric and leukodystrophy donors. Slices were cultured up to six weeks, in culture medium with or without human cerebrospinal fluid. Human post-mortem organotypic brain slice cultures remained viable for at least six weeks ex vivo and maintained tissue structure and diversity of (neural) cell types. Supplementation with cerebrospinal fluid could improve slice recovery. Patient-derived organotypic slice cultures recapitulated and maintained known in vivo neuropathology. The cultures also showed physiologic multicellular responses to lysolecithin-induced demyelination ex vivo, indicating their suitability to study intrinsic repair mechanisms upon injury. The slice cultures were applicable for various experimental studies, as multi-electrode neuronal recordings. Finally, the cultures showed successful cell-type dependent transduction with gene therapy vectors. These human post-mortem organotypic brain slice cultures represent an adapted ex vivo model suitable for multifaceted studies of brain disease mechanisms, boosting translation from human ex vivo to in vivo. This model also allows for assessing potential treatment options, including gene therapy applications. Human post-mortem brain slice cultures are thus a valuable tool in preclinical research to study the pathomechanisms of a wide variety of brain diseases in living human tissue.
Collapse
Affiliation(s)
- Bonnie C Plug
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Ilma M Revers
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Marjolein Breur
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Gema Muñoz González
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Jaap A Timmerman
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niels R C Meijns
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Daniek Hamberg
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Jikke Wagendorp
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Erik Nutma
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
| | - Nicole I Wolf
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niek P van Til
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marjo S van der Knaap
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marianna Bugiani
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
| |
Collapse
|
11
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
12
|
Bourque M, Morissette M, Di Paolo T. Neuroactive steroids and Parkinson's disease: Review of human and animal studies. Neurosci Biobehav Rev 2024; 156:105479. [PMID: 38007170 DOI: 10.1016/j.neubiorev.2023.105479] [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: 06/01/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The greater prevalence and incidence of Parkinson's disease (PD) in men suggest a beneficial effect of sex hormones. Neuroactive steroids have neuroprotective activities thus offering interesting option for disease-modifying therapy for PD. Neuroactive steroids are also neuromodulators of neurotransmitter systems and may thus help to control PD symptoms and side effect of dopamine medication. Here, we review the effect on sex hormones (estrogen, androgen, progesterone and its metabolites) as well as androstenediol, pregnenolone and dehydroepiandrosterone) in human studies and in animal models of PD. The effect of neuroactive steroids is reviewed by considering sex and hormonal status to help identify specifically for women and men with PD what might be a preventive approach or a symptomatic treatment. PD is a complex disease and the pathogenesis likely involves multiple cellular processes. Thus it might be useful to target different cellular mechanisms that contribute to neuronal loss and neuroactive steroids provide therapeutics options as they have multiple mechanisms of action.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada; Faculté de pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec G1V 0A6, Canada.
| |
Collapse
|
13
|
di Michele F. Neurosteroids and their Therapeutic Potential: Remembering the Contribution of Dr. Sabina Luchetti, Netherlands Institute for Neuroscience, Amsterdam, Netherlands. Curr Med Chem 2024; 31:2895-2899. [PMID: 38904159 DOI: 10.2174/0109298673270157231120072205] [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: 07/14/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 06/22/2024]
Abstract
This article traces the career of Dr. Sabina Luchetti (1969-2021), a noted physician (medical doctor, specialized in Neurology at Tor Vergata University of Rome, Italy), a dedicated neuroscientist (Ph.D. in Neuroscience at Tor Vergata University and IRCCS Santa Lucia of Rome), and a member of a renowned Netherlands group (senior researcher at Professor Swaab Laboratory of the Netherlands Institute for Neuroscience, Amsterdam, Netherlands), working in the field of brain function and diseases. She is particularly involved in the study of natural compounds, such as neurosteroids and their biosynthetic pathways in neurodegenerative and neuroinflammation- related disorders, working on post-mortem human brains. This editorial outlines Dr. Luchetti's wide range of interests, discloses her superior fund of knowledge, and recollects her humanitarian spirit, all of which contribute to creating a great sense of belonging to any group of researchers whom she worked with. The impact of Dr. Luchetti's work will continue to be felt for many years. From the bench to the bedside, her work has indirectly contributed to shedding light on the neurosteroids' potential therapeutic effects, considering that neurosteroids and their analogues (some of which are over-the-counter) are now used to treat depression, epilepsy, and substance abuse disorders. Moreover, the potential therapeutic effects of allopregnanolone with respect to its capability to promote neuroregeneration and neuroprotection are a promising basis for future treatment of neurodegenerative diseases.
Collapse
|