1
|
Traiffort E, Kassoussi A, Zahaf A. Revisiting the role of sexual hormones in the demyelinated central nervous system. Front Neuroendocrinol 2024; 76:101172. [PMID: 39694337 DOI: 10.1016/j.yfrne.2024.101172] [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: 07/03/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Sex-related differences characterize multiple sclerosis, an autoimmune, inflammatory and neurodegenerative disease displaying higher incidence in females as well as discrepancies in susceptibility and progression. Besides clinical specificities, molecular and cellular differences related to sex hormones were progressively uncovered improving our understanding of the mechanisms involved in this disabling disease. The most recent findings may give rise to the identification of novel therapeutic perspectives that could meet the urgent need for a treatment preventing the transition from the recurrent- to the progressive form of the disease. The present review is an update of our current knowledge about progestagens, androgens and estrogens in the context of CNS demyelination including their synthesis, the impact of their dysregulation, the preclinical and clinical data presently available, the main molecular dimorphisms related to these hormones and their age-related changes and relationship with failure of spontaneous remyelination, likely impacting the inexorable progression of multiple sclerosis towards irreversible disabilities.
Collapse
Affiliation(s)
| | | | - Amina Zahaf
- U1195 Inserm, Paris-Saclay University, Kremlin-Bicêtre, France
| |
Collapse
|
2
|
Wang M, Hu S, Fu X, Zhou H, Yang S, Yang C. Neurosteroids: A potential target for neuropsychiatric disorders. J Steroid Biochem Mol Biol 2024; 239:106485. [PMID: 38369032 DOI: 10.1016/j.jsbmb.2024.106485] [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: 01/25/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Neurosteroids are steroids produced by endocrine glands and subsequently entering the brain, and also include steroids synthesis in the brain. It has been widely known that neurosteroids influence many neurological functions, including neuronal signaling, synaptic adaptations, and neuroprotective effects. In addition, abnormality in the synthesis and function of neurosteroids has been closely linked to neuropsychiatric disorders, such as Alzheimer's disease (AD), schizophrenia (SZ), and epilepsy. Given their important role in brain pathophysiology and disorders, neurosteroids offer potential therapeutic targets for a variety of neuropsychiatric diseases, and that therapeutic strategies targeting neurosteroids probably exert beneficial effects. We therefore summarized the role of neurosteroids in brain physiology and neuropsychiatric disorders, and introduced the recent findings of synthetic neurosteroid analogues for potential treatment of neuropsychiatric disorders, thereby providing insights for further research in the future.
Collapse
Affiliation(s)
- Mengyu Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinghuo Fu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Huixuan Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
3
|
Esperante IJ, Meyer M, Banzan C, Kruse MS, Lima A, Roig P, Guennoun R, Schumacher M, De Nicola AF, Gonzalez Deniselle MC. Testosterone Reduces Myelin Abnormalities in the Wobbler Mouse Model of Amyotrophic Lateral Sclerosis. Biomolecules 2024; 14:428. [PMID: 38672445 PMCID: PMC11048492 DOI: 10.3390/biom14040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motoneuron degenerative disease that is associated with demyelination. The Wobbler (WR) mouse exhibits motoneuron degeneration, gliosis and myelin deterioration in the cervical spinal cord. Since male WRs display low testosterone (T) levels in the nervous system, we investigated if T modified myelin-relative parameters in WRs in the absence or presence of the aromatase inhibitor, anastrozole (A). We studied myelin by using luxol-fast-blue (LFB) staining, semithin sections, electron microscopy and myelin protein expression, density of IBA1+ microglia and mRNA expression of inflammatory factors, and the glutamatergic parameters glutamine synthetase (GS) and the transporter GLT1. Controls and WR + T showed higher LFB, MBP and PLP staining, lower g-ratios and compact myelin than WRs and WR + T + A, and groups showing the rupture of myelin lamellae. WRs showed increased IBA1+ cells and mRNA for CD11b and inflammatory factors (IL-18, TLR4, TNFαR1 and P2Y12R) vs. controls or WR + T. IBA1+ cells, and CD11b were not reduced in WR + T + A, but inflammatory factors' mRNA remained low. A reduction of GS+ cells and GLT-1 immunoreactivity was observed in WRs and WR + T + A vs. controls and WR + T. Clinically, WR + T but not WR + T + A showed enhanced muscle mass, grip strength and reduced paw abnormalities. Therefore, T effects involve myelin protection, a finding of potential clinical translation.
Collapse
Affiliation(s)
- Ivan J. Esperante
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Carolina Banzan
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Maria Sol Kruse
- Laboratory of Neurobiology, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina;
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Rachida Guennoun
- U1195 INSERM and University Paris Sud: “Neuroprotective, Neuroregenerative and Remyelinating Small Molecules”, 94276 Kremlin-Bicêtre, France; (R.G.); (M.S.)
| | - Michael Schumacher
- U1195 INSERM and University Paris Sud: “Neuroprotective, Neuroregenerative and Remyelinating Small Molecules”, 94276 Kremlin-Bicêtre, France; (R.G.); (M.S.)
| | - Alejandro F. De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
- Departamento de Ciencias Fisiológicas, UA1, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
| |
Collapse
|
4
|
Alvarez Quintero GS, Lima A, Roig P, Meyer M, de Kloet ER, De Nicola AF, Garay LI. Effects of the mineralocorticoid receptor antagonist eplerenone in experimental autoimmune encephalomyelitis. J Steroid Biochem Mol Biol 2024; 238:106461. [PMID: 38219844 DOI: 10.1016/j.jsbmb.2024.106461] [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: 10/02/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
There is growing evidence indicating that mineralocorticoid receptor (MR) expression influences a wide variety of functions in metabolic and immune response. The present study explored if antagonism of the MR reduces neuroinflammation in the spinal cord of mice with experimental autoimmune encephalomyelitis (EAE). Eplerenone (EPLE) (100 mg/kg dissolved in 30% 2-hydroxypropyl-β-cyclodextrin) was administered intraperitoneally (i.p.) daily from EAE induction (day 0) until sacrificed on day 17 post-induction. The MR blocker (a) significantly decreased the inflammatory parameters TLR4, MYD88, IL-1β, and iNOS mRNAs; (b) attenuated HMGB1, NLRP3, TGF-β mRNAs, microglia, and aquaporin4 immunoreaction without modifying GFAP. Serum IL-1β was also decreased in the EAE+EPLE group. Moreover, EPLE treatment prevented demyelination and improved clinical signs of EAE mice. Interestingly, MR was decreased and GR remained unchanged in EAE mice while EPLE treatment restored MR expression, suggesting that a dysbalanced MR/GR was associated with the development of neuroinflammation. Our results indicated that MR blockage with EPLE attenuated inflammation-related spinal cord pathology in the EAE mouse model of Multiple Sclerosis, supporting a novel therapeutic approach for immune-related diseases.
Collapse
Affiliation(s)
- Guido S Alvarez Quintero
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - E R de Kloet
- Department of Clinical Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Department of Human Biochemistry, University of Buenos Aires, Paraguay 2155, 1121 Buenos Aires, Argentina
| | - Laura I Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Department of Human Biochemistry, University of Buenos Aires, Paraguay 2155, 1121 Buenos Aires, Argentina.
| |
Collapse
|
5
|
Meyer M, Meijer O, Hunt H, Belanoff J, Lima A, de Kloet ER, Gonzalez Deniselle MC, De Nicola AF. Stress-induced Neuroinflammation of the Spinal Cord is Restrained by Cort113176 (Dazucorilant), A Specific Glucocorticoid Receptor Modulator. Mol Neurobiol 2024; 61:1-14. [PMID: 37566177 DOI: 10.1007/s12035-023-03554-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
Glucocorticoids exert antiinflammatory, antiproliferative and immunosupressive effects. Paradoxically they may also enhance inflammation particularly in the nervous system, as shown in Cushing´ syndrome and neurodegenerative disorders of humans and models of human diseases. ."The Wobbler mouse model of amyotrophic lateral sclerosis shows hypercorticoidism and neuroinflammation which subsided by treatment with the glucocorticoid receptor (GR) modulator Dazucorilant (CORT113176). This effect suggests that GR mediates the chronic glucocorticoid unwanted effects. We now tested this hypothesis using a chronic stress model resembling the condition of the Wobbler mouse Male NFR/NFR mice remained as controls or were subjected to a restraining / rotation stress protocol for 3 weeks, with a group of stressed mice receiving CORT113176 also for 3 weeks. We determined the mRNAS or reactive protein for the proinflamatory factors HMGB1, TLR4, NFkB, TNFα, markers of astrogliosis (GFAP, SOX9 and acquaporin 4), of microgliosis (Iba, CD11b, P2RY12 purinergic receptor) as well as serum IL1β and corticosterone. We showed that chronic stress produced high levels of serum corticosterone and IL1β, decreased body and spleen weight, produced microgliosis and astrogliosis and increased proinflammatory mediators. In stressed mice, modulation of the GR with CORT113176 reduced Iba + microgliosis, CD11b and P2RY12 mRNAs, immunoreactive HMGB1 + cells, GFAP + astrogliosis, SOX9 and acquaporin expression and TLR4 and NFkB mRNAs vs. stress-only mice. The effects of CORT113176 indicate that glucocorticoids are probably involved in neuroinflammation. Thus, modulation of the GR would become useful to dampen the inflammatory component of neurodegenerative disorders.
Collapse
Affiliation(s)
- Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - Onno Meijer
- Dept. of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hazel Hunt
- Corcept Therapeutics, Menlo Park, Ca, USA
| | | | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - E Ronald de Kloet
- Dept. of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina
- Dept. of Physiology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina.
- Dept. of Human Biochemiistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Pletzer B, Winkler-Crepaz K, Hillerer K. Progesterone and contraceptive progestin actions on the brain: A systematic review of animal studies and comparison to human neuroimaging studies. Front Neuroendocrinol 2023; 69:101060. [PMID: 36758768 DOI: 10.1016/j.yfrne.2023.101060] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/25/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
In this review we systematically summarize the effects of progesterone and synthetic progestins on neurogenesis, synaptogenesis, myelination and six neurotransmitter systems. Several parallels between progesterone and older generation progestin actions emerged, suggesting actions via progesterone receptors. However, existing results suggest a general lack of knowledge regarding the effects of currently used progestins in hormonal contraception regarding these cellular and molecular brain parameters. Human neuroimaging studies were reviewed with a focus on randomized placebo-controlled trials and cross-sectional studies controlling for progestin type. The prefrontal cortex, amygdala, salience network and hippocampus were identified as regions of interest for future preclinical studies. This review proposes a series of experiments to elucidate the cellular and molecular actions of contraceptive progestins in these areas and link these actions to behavioral markers of emotional and cognitive functioning. Emotional effects of contraceptive progestins appear to be related to 1) alterations in the serotonergic system, 2) direct/indirect modulations of inhibitory GABA-ergic signalling via effects on the allopregnanolone content of the brain, which differ between androgenic and anti-androgenic progestins. Cognitive effects of combined oral contraceptives appear to depend on the ethinylestradiol dose.
Collapse
Affiliation(s)
- Belinda Pletzer
- Department of Psychology & Centre for Cognitive Neuroscience, Paris-Lodron-University Salzburg, Salzburg Austria.
| | | | - Katharina Hillerer
- Department of Gynaecology & Obstetrics, Private Medical University, Salzburg, Austria
| |
Collapse
|
7
|
Early Signs of Neuroinflammation in the Postnatal Wobbler Mouse Model of Amyotrophic Lateral Sclerosis. Cell Mol Neurobiol 2022:10.1007/s10571-022-01294-5. [PMID: 36219378 DOI: 10.1007/s10571-022-01294-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/02/2022] [Indexed: 11/03/2022]
Abstract
The Wobbler mouse is an accepted model of sporadic amyotrophic lateral sclerosis. The spinal cord of clinically symptomatic animals (3-5 months old) shows vacuolar motoneuron degeneration, inflammation, and gliosis accompanied by motor impairment. However, data are not conclusive concerning pathological changes appearing early after birth. To answer this question, we used postnatal day (PND) 6 genotyped Wobbler pups to determine abnormalities of glia and neurons at this early age period in the spinal cord. We found astrogliosis, microgliosis with morphophenotypic changes pointing to active ameboid microglia, enhanced expression of the proinflammatory markers TLR4, NFkB, TNF, and inducible nitric oxide synthase. The astrocytic enzyme glutamine synthase and the glutamate-aspartate transporter GLAST were also reduced in PND 6 Wobbler pups, suggesting excitotoxicity due to impaired glutamate homeostasis. At the neuronal level, PND 6 Wobblers showed swollen soma, increased choline acetyltransferase immunofluorescence staining, and low expression of the neuronal nuclear antigen NeuN. However, vacuolated motoneurons, a typical signature of older clinically symptomatic Wobbler mice, were absent in the spinal cord of PND 6 Wobblers. The results suggest predominance of neuroinflammation and abnormalities of microglia and astrocytes at this early period of Wobbler life, accompanied by some neuronal changes. Data support the non-cell autonomous hypothesis of the Wobbler disorder, and bring useful information with regard to intervening molecular inflammatory mechanisms at the beginning stage of human motoneuron degenerative diseases.
Collapse
|
8
|
Liu C, Gao W, Zhao L, Cao Y. Progesterone attenuates neurological deficits and exerts a protective effect on damaged axons via the PI3K/AKT/mTOR-dependent pathway in a mouse model of intracerebral hemorrhage. Aging (Albany NY) 2022; 14:2574-2589. [PMID: 35305084 PMCID: PMC9004566 DOI: 10.18632/aging.203954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating event with high disability and fatality rates. However, there is a lack of effective treatments for this condition. We aimed to investigate the neuroprotective and axonal regenerative effects of progesterone after ICH. For this purpose, an ICH model was established in adult mice by injecting type VII collagenase into the striatum; the mice were then treated with progesterone (8 mg/kg). Hematoma absorption, neurological scores, and brain water content were evaluated on days one, three, and seven after the ICH. The effect of progesterone on inflammation and axonal regeneration was examined on day three after the ICH using western blotting, immunohistochemistry, immunofluorescence, as well as hematoxylin-eosin, Nissl, and Luxol fast blue staining. In addition, we combined progesterone with the phosphoinositide 3-kinase/serine/threonine-specific protein kinase (PI3K/AKT) inhibitor, LY294002, to explore its potential neuroprotective mechanisms. Administration of progesterone attenuated the neurological deficits and expression of inflammatory cytokines and promoted axonal regeneration after ICH, this effect was blocked by LY294002. Collectively, these results suggest that progesterone could reduce axonal damage and produced partial neuroprotective effects after ICH through the PI3K/AKT/mTOR pathway, providing a new therapeutic target and basis for the treatment of ICH.
Collapse
Affiliation(s)
- Chang Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, P.R. China
| | - Weina Gao
- Department of Intensive Care Unit, The Affiliated Chengdu 363 Hospital of Southwest Medical University, Chengdu 610041, Sichuan Province, P.R. China
| | - Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637002, Sichuan Province, P.R. China
| | - Yi Cao
- Department of Neurosurgery, Chengdu Second People's Hospital, Chengdu 610021, Sichuan Province, P.R. China
| |
Collapse
|
9
|
Rundquist O, Nestor CE, Jenmalm MC, Hellberg S, Gustafsson M. Progesterone Inhibits the Establishment of Activation-Associated Chromatin During T H1 Differentiation. Front Immunol 2022; 13:835625. [PMID: 35185927 PMCID: PMC8848251 DOI: 10.3389/fimmu.2022.835625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/14/2022] [Indexed: 01/08/2023] Open
Abstract
TH1-mediated diseases such as multiple sclerosis (MS) and rheumatoid arthritis (RA) improve during pregnancy, coinciding with increasing levels of the pregnancy hormone progesterone (P4), highlighting P4 as a potential mediator of this immunomodulation. Here, we performed detailed characterization of how P4 affects the chromatin and transcriptomic landscape during early human TH1 differentiation, utilizing both ATAC-seq and RNA-seq. Time series analysis of the earlier events (0.5-24 hrs) during TH1 differentiation revealed that P4 counteracted many of the changes induced during normal differentiation, mainly by downregulating key regulatory genes and their upstream transcription factors (TFs) involved in the initial T-cell activation. Members of the AP-1 complex such as FOSL1, FOSL2, JUN and JUNB were particularly affected, in both in promoters and in distal regulatory elements. Moreover, the changes induced by P4 were significantly enriched for disease-associated changes related to both MS and RA, revealing several shared upstream TFs, where again JUN was highlighted to be of central importance. Our findings support an immune regulatory role for P4 during pregnancy by impeding T-cell activation, a crucial checkpoint during pregnancy and in T-cell mediated diseases, and a central event prior to T-cell lineage commitment. Indeed, P4 is emerging as a likely candidate involved in disease modulation during pregnancy and further studies evaluating P4 as a potential treatment option are needed.
Collapse
Affiliation(s)
- Olof Rundquist
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Colm E. Nestor
- Crown Princess Victoria Children’s Hospital, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria C. Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sandra Hellberg
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
10
|
Breton JM, Long KLP, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis II: Implications for Myelin Repair. Biomolecules 2021; 11:290. [PMID: 33669242 PMCID: PMC7919830 DOI: 10.3390/biom11020290] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in myelin, the protective and insulating sheath surrounding axons, affect brain function, as is evident in demyelinating diseases where the loss of myelin leads to cognitive and motor dysfunction. Recent evidence suggests that changes in myelination, including both hyper- and hypo-myelination, may also play a role in numerous neurological and psychiatric diseases. Protecting myelin and promoting remyelination is thus crucial for a wide range of disorders. Oligodendrocytes (OLs) are the cells that generate myelin, and oligodendrogenesis, the creation of new OLs, continues throughout life and is necessary for myelin plasticity and remyelination. Understanding the regulation of oligodendrogenesis and myelin plasticity within disease contexts is, therefore, critical for the development of novel therapeutic targets. In our companion manuscript, we review literature demonstrating that multiple hormone classes are involved in the regulation of oligodendrogenesis under physiological conditions. The majority of hormones enhance oligodendrogenesis, increasing oligodendrocyte precursor cell differentiation and inducing maturation and myelin production in OLs. Thus, hormonal treatments present a promising route to promote remyelination. Here, we review the literature on hormonal regulation of oligodendrogenesis within the context of disorders. We focus on steroid hormones, including glucocorticoids and sex hormones, peptide hormones such as insulin-like growth factor 1, and thyroid hormones. For each hormone, we describe whether they aid in OL survival, differentiation, or remyelination, and we discuss their mechanisms of action, if known. Several of these hormones have yielded promising results in both animal models and in human conditions; however, a better understanding of hormonal effects, interactions, and their mechanisms will ultimately lead to more targeted therapeutics for myelin repair.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Matthew K Barraza
- Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Olga S Perloff
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
- Integrative Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
11
|
Hornung RS, Benton WL, Tongkhuya S, Uphouse L, Kramer PR, Averitt DL. Progesterone and Allopregnanolone Rapidly Attenuate Estrogen-Associated Mechanical Allodynia in Rats with Persistent Temporomandibular Joint Inflammation. Front Integr Neurosci 2020; 14:26. [PMID: 32457584 PMCID: PMC7225267 DOI: 10.3389/fnint.2020.00026] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/15/2020] [Indexed: 12/16/2022] Open
Abstract
Temporomandibular joint disorder (TMD) is associated with pain in the joint (temporomandibular joint, TMJ) and muscles involved in mastication. TMD pain dissipates following menopause but returns in some women undergoing estrogen replacement therapy. Progesterone has both anti-inflammatory and antinociceptive properties, while estrogen's effects on nociception are variable and highly dependent on both natural hormone fluctuations and estrogen dosage during pharmacological treatments, with high doses increasing pain. Allopregnanolone, a progesterone metabolite and positive allosteric modulator of the GABAA receptor, also has antinociceptive properties. While progesterone and allopregnanolone are antinociceptive, their effect on estrogen-exacerbated TMD pain has not been determined. We hypothesized that removing the source of endogenous ovarian hormones would reduce inflammatory allodynia in the TMJ of rats and both progesterone and allopregnanolone would attenuate the estrogen-provoked return of allodynia. Baseline mechanical sensitivity was measured in female Sprague-Dawley rats (150-175 g) using the von Frey filament method followed by a unilateral injection of complete Freund's adjuvant (CFA) into the TMJ. Mechanical allodynia was confirmed 24 h later; then rats were ovariectomized or received sham surgery. Two weeks later, allodynia was reassessed and rats received one of the following subcutaneous hormone treatments over 5 days: a daily pharmacological dose of estradiol benzoate (E2; 50 μg/kg), daily E2 and pharmacological to sub-physiological doses of progesterone (P4; 16 mg/kg, 16 μg/kg, or 16 ng/kg), E2 daily and interrupted P4 given every other day, daily P4, or daily vehicle control. A separate group of animals received allopregnanolone (0.16 mg/kg) instead of P4. Allodynia was reassessed 1 h following injections. Here, we report that CFA-evoked mechanical allodynia was attenuated following ovariectomy and daily high E2 treatment triggered the return of allodynia, which was rapidly attenuated when P4 was also administered either daily or every other day. Allopregnanolone treatment, whether daily or every other day, also attenuated estrogen-exacerbated allodynia within 1 h of treatment, but only on the first treatment day. These data indicate that when gonadal hormone levels have diminished, treatment with a lower dose of progesterone may be effective at rapidly reducing the estrogen-evoked recurrence of inflammatory mechanical allodynia in the TMJ.
Collapse
Affiliation(s)
- Rebecca S. Hornung
- Department of Biology, Texas Woman’s University, Denton, TX, United States
| | - William L. Benton
- Department of Biology, Texas Woman’s University, Denton, TX, United States
| | - Sirima Tongkhuya
- Department of Biology, Texas Woman’s University, Denton, TX, United States
| | - Lynda Uphouse
- Department of Biology, Texas Woman’s University, Denton, TX, United States
| | - Phillip R. Kramer
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Dayna Loyd Averitt
- Department of Biology, Texas Woman’s University, Denton, TX, United States
| |
Collapse
|
12
|
Frye CA, Chittur SV. Mating Enhances Expression of Hormonal and Trophic Factors in the Midbrain of Female Rats. Front Behav Neurosci 2020; 14:21. [PMID: 32351369 PMCID: PMC7176275 DOI: 10.3389/fnbeh.2020.00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/31/2020] [Indexed: 11/23/2022] Open
Abstract
Among female rats, mating enhances neurosteroid formation in the midbrain ventral tegmental area (VTA; independent of peripheral steroid-secreting glands, ovaries, and adrenals). The sources/targets for these actions are not well understood. In Experiment 1, proestrous rats engaged in a mating paradigm, or did not, and the midbrains had been assessed via the Affymetrix rat genome microarrays. In Experiment 2, the influence of gonadal and adrenal glands on the expression of these genes was assessed in rats that were proestrous, ovariectomized (OVX), or OVX and adrenalectomized (ADX). The microarrays revealed 53 target genes that were significantly up-regulated (>2.0-fold change) in response to mating. Mating significantly enhanced the midbrain mRNA expression of genes involved in hormonal and trophic actions: Gh1, S100g, and Klk1b3 in proestrous, but not OVX and/or ADX, rats; Fshb in all but OVX/ADX rats; and lutenizing hormone β and thyroid-stimulating hormone (TSH) β in all rats. Thus, mating enhances midbrain gene expression independent and dependent of peripheral glands.
Collapse
Affiliation(s)
- Cheryl A Frye
- Department of Psychology, The University at Albany-SUNY, Albany, NY, United States.,Department of Biological Sciences, The University at Albany-SUNY, Albany, NY, United States.,Center for Neuroscience Research, The University at Albany-SUNY, Albany, NY, United States.,Center for Life Sciences Research, The University at Albany-SUNY, Albany, NY, United States
| | - Sridar V Chittur
- Center for Functional Genomics, The University at Albany-SUNY, Albany, NY, United States
| |
Collapse
|
13
|
González-Orozco JC, Moral-Morales AD, Camacho-Arroyo I. Progesterone through Progesterone Receptor B Isoform Promotes Rodent Embryonic Oligodendrogenesis. Cells 2020; 9:cells9040960. [PMID: 32295179 PMCID: PMC7226962 DOI: 10.3390/cells9040960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/28/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022] Open
Abstract
Oligodendrocytes are the myelinating cells of the central nervous system (CNS). These cells arise during the embryonic development by the specification of the neural stem cells to oligodendroglial progenitor cells (OPC); newly formed OPC proliferate, migrate, differentiate, and mature to myelinating oligodendrocytes in the perinatal period. It is known that progesterone promotes the proliferation and differentiation of OPC in early postnatal life through the activation of the intracellular progesterone receptor (PR). Progesterone supports nerve myelination after spinal cord injury in adults. However, the role of progesterone in embryonic OPC differentiation as well as the specific PR isoform involved in progesterone actions in these cells is unknown. By using primary cultures obtained from the embryonic mouse spinal cord, we showed that embryonic OPC expresses both PR-A and PR-B isoforms. We found that progesterone increases the proliferation, differentiation, and myelination potential of embryonic OPC through its PR by upregulating the expression of oligodendroglial genes such as neuron/glia antigen 2 (NG2), sex determining region Y-box9 (SOX9), myelin basic protein (MBP), 2′,3′-cyclic-nucleotide 3′-phosphodiesterase (CNP1), and NK6 homeobox 1 (NKX 6.1). These effects are likely mediated by PR-B, as they are blocked by the silencing of this isoform. The results suggest that progesterone contributes to the process of oligodendrogenesis during prenatal life through specific activation of PR-B.
Collapse
|
14
|
González SL, Coronel MF, Raggio MC, Labombarda F. Progesterone receptor-mediated actions and the treatment of central nervous system disorders: An up-date of the known and the challenge of the unknown. Steroids 2020; 153:108525. [PMID: 31634489 DOI: 10.1016/j.steroids.2019.108525] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 01/04/2023]
Abstract
Progesterone has been shown to exert a wide range of remarkable protective actions in experimental models of central nervous system injury or disease. However, the intimate mechanisms involved in each of these beneficial effects are not fully depicted. In this review, we intend to give the readers a thorough revision on what is known about the participation of diverse receptors and signaling pathways in progesterone-mediated neuroprotective, pro-myelinating and anti-inflammatory outcomes, as well as point out to novel regulatory mechanisms that could open new perspectives in steroid-based therapies.
Collapse
Affiliation(s)
- Susana L González
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG Buenos Aires, Argentina.
| | - María F Coronel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina; Facultad de Ciencias Biomédicas, Universidad Austral, Presidente Perón 1500, B1629AHJ Pilar, Buenos Aires, Argentina
| | - María C Raggio
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratorio de Bioquímica Neuroendócrina, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
15
|
Meyer M, Kruse MS, Garay L, Lima A, Roig P, Hunt H, Belanoff J, de Kloet ER, Deniselle MCG, De Nicola AF. Long-term effects of the glucocorticoid receptor modulator CORT113176 in murine motoneuron degeneration. Brain Res 2019; 1727:146551. [PMID: 31726042 DOI: 10.1016/j.brainres.2019.146551] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/31/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023]
Abstract
The Wobbler mouse spinal cord shows vacuolated motoneurons, glial reaction, inflammation and abnormal glutamatergic parameters. Wobblers also show deficits of motor performance. These conditions resemble amyotrophic lateral sclerosis (ALS). Wobbler mice also show high levels of corticosterone in blood, adrenals and brain plus adrenal hypertrophy, suggesting that chronically elevated glucocorticoids prime spinal cord neuroinflammation. Therefore, we analyzed if treatment of Wobbler mice with the glucocorticoid receptor (GR) antagonist CORT113176 mitigated the mentioned abnormalities. 30 mg/kg CORT113176 given daily for 3 weeks reduced motoneuron vacuolation, decreased astro and microgliosis, lowered the inflammatory mediators high mobility group box 1 protein (HMGB1), toll-like receptor 4, myeloid differentiation primary response 88 (MyD88), p50 subunit of nuclear factor kappa B (NFκB), tumor necrosis factor (TNF) receptor, and interleukin 18 (IL18) compared to untreated Wobblers. CORT113176 increased the survival signal pAKT (serine-threonine kinase) and decreased the death signal phosphorylated Junk-N-terminal kinase (pJNK), symptomatic of antiapoptosis. There was a moderate positive effect on glutamine synthase and astrocyte glutamate transporters, suggesting decreased glutamate excitotoxicity. In this pre-clinical study, Wobblers receiving CORT113176 showed enhanced resistance to fatigue in the rota rod test and lower forelimb atrophy at weeks 2-3. Therefore, long-term treatment with CORT113176 attenuated degeneration and inflammation, increased motor performance and decreased paw deformity. Antagonism of the GR may be of potential therapeutic value for neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria Meyer
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Maria Sol Kruse
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Laura Garay
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina
| | - Analia Lima
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Paulina Roig
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Hazel Hunt
- CORCEPT Therapeutics, 149 Commonwealth Dr, Menlo Park, CA 94025, USA
| | - Joseph Belanoff
- CORCEPT Therapeutics, 149 Commonwealth Dr, Menlo Park, CA 94025, USA
| | - E Ronald de Kloet
- Division of Endocrinology, Dept. of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Maria Claudia Gonzalez Deniselle
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Physiology, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratories of Neuroendocrine Biochemistry and Neurobiology, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina.
| |
Collapse
|
16
|
Gargiulo-Monachelli G, Meyer M, Lara A, Garay L, Lima A, Roig P, De Nicola AF, Gonzalez Deniselle MC. Comparative effects of progesterone and the synthetic progestin norethindrone on neuroprotection in a model of spontaneous motoneuron degeneration. J Steroid Biochem Mol Biol 2019; 192:105385. [PMID: 31150830 DOI: 10.1016/j.jsbmb.2019.105385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/21/2019] [Accepted: 05/26/2019] [Indexed: 12/12/2022]
Abstract
The Wobbler mouse has been proposed as an experimental model of the sporadic form of amyotrophic lateral sclerosis (ALS). The administration of natural progesterone (PROG) to Wobbler mice attenuates neuropathology, inhibits oxidative stress, enhances the expression of genes involved in motoneuron function, increases survival and restores axonal transport. However, current pharmacological treatments for ALS patients are still partially effective. This encouraged us to investigate if the synthetic progestin norethindrone (NOR), showing higher potency than PROG and used for birth control and hormone therapy might also afford neuroprotection. Two-month-old Wobbler mice (wr/wr) were left untreated or received either a 20 mg pellet of PROG or a 1 mg pellet of NOR for 18 days. Untreated control NFR/NFR mice (background strain for Wobbler) were also employed. Wobblers showed typical clinical and spinal cord abnormalities, while these abnormalities were normalized with PROG treatment. Surprisingly, we found that NOR did not increase immunoreactivity and gene expression for choline-acetyltransferase, drastically decreased GFAP + astrogliosis, favored proinflammatory mediators, promoted the inflammatory phenotype of IBA1+ microglia, increased the receptor for advanced glycation end products (RAGE) mRNA and protein expression and the activity of nitric oxide synthase (NOS)/NADPH diaphorase in the cervical spinal cord. Additionally, NOR treatment produced atrophy of the thymus. The combined negative effects of NOR on clinical assessments (forelimb atrophy and rotarod performance) suggest a detrimental effect on muscle trophism and motor function. These findings reinforce the evidence that the type of progestin used for contraception, endometriosis or replacement therapy, may condition the outcome of preclinical and clinical studies targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Gisella Gargiulo-Monachelli
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Agustina Lara
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Laura Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Depto. de Bioquímica Humana, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1121 Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Depto. de Bioquímica Humana, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1121 Buenos Aires, Argentina
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental-CONICET, Obligado 2490, 1428 Buenos Aires, Argentina; Depto. de Ciencias Fisiológicas, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1121 Buenos Aires, Argentina.
| |
Collapse
|
17
|
Sex differences in central nervous system plasticity and pain in experimental autoimmune encephalomyelitis. Pain 2019; 160:1037-1049. [PMID: 30649100 DOI: 10.1097/j.pain.0000000000001483] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disease with many known structural and functional changes in the central nervous system. A well-recognized, but poorly understood, complication of MS is chronic pain. Little is known regarding the influence of sex on the development and maintenance of MS-related pain. This is important to consider, as MS is a predominantly female disease. Using the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, we demonstrate sex differences in measures of spinal cord inflammation and plasticity that accompany tactile hypersensitivity. Although we observed substantial inflammatory activity in both sexes, only male EAE mice exhibit robust staining of axonal injury markers and increased dendritic arborisation in morphology of deep dorsal horn neurons. We propose that tactile hypersensitivity in female EAE mice may be more immune-driven, whereas pain in male mice with EAE may rely more heavily on neurodegenerative and plasticity-related mechanisms. Morphological and inflammatory differences in the spinal cord associated with pain early in EAE progression supports the idea of differentially regulated pain pathways between the sexes. Results from this study may indicate future sex-specific targets that are worth investigating for their functional role in pain circuitry.
Collapse
|
18
|
González SL, Meyer L, Raggio MC, Taleb O, Coronel MF, Patte-Mensah C, Mensah-Nyagan AG. Allopregnanolone and Progesterone in Experimental Neuropathic Pain: Former and New Insights with a Translational Perspective. Cell Mol Neurobiol 2019; 39:523-537. [PMID: 30187261 DOI: 10.1007/s10571-018-0618-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/31/2018] [Indexed: 02/06/2023]
Abstract
In the last decades, an active and stimulating area of research has been devoted to explore the role of neuroactive steroids in pain modulation. Despite challenges, these studies have clearly contributed to unravel the multiple and complex actions and potential mechanisms underlying steroid effects in several experimental conditions that mimic human chronic pain states. Based on the available data, this review focuses mainly on progesterone and its reduced derivative allopregnanolone (also called 3α,5α-tetrahydroprogesterone) which have been shown to prevent or even reverse the complex maladaptive changes and pain behaviors that arise in the nervous system after injury or disease. Because the characterization of new related molecules with improved specificity and enhanced pharmacological profiles may represent a crucial step to develop more efficient steroid-based therapies, we have also discussed the potential of novel synthetic analogs of allopregnanolone as valuable molecules for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Susana Laura González
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG, Buenos Aires, Argentina.
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| | - Laurence Meyer
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France
| | - María Celeste Raggio
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Omar Taleb
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France
| | - María Florencia Coronel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France
| | - Ayikoe Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France.
| |
Collapse
|
19
|
Zhu X, Fréchou M, Schumacher M, Guennoun R. Cerebroprotection by progesterone following ischemic stroke: Multiple effects and role of the neural progesterone receptors. J Steroid Biochem Mol Biol 2019; 185:90-102. [PMID: 30031789 DOI: 10.1016/j.jsbmb.2018.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022]
Abstract
Treatment with progesterone limits brain damage after stroke. However, the cellular bases of the cerebroprotective effects of progesterone are not well documented. The aims of this study were to determine neural cells and functions that are affected by progesterone treatment and the role of neural progesterone receptors (PR) after stroke. Adult male PRNesCre mice, selectively lacking PR in the central nervous system, and their control PRloxP/loxP littermates were subjected to transient ischemia by middle cerebral artery occlusion (MCAO) for 30 min. Mice received either progesterone (8 mg/kg) or vehicle at 1-, 6- and 24- hrs post-MCAO and outcomes were analyzed at 48 h post-MCAO. In PRloxP/loxP mice, progesterone exerted multiple effects on different neural cell types, improved motor functional outcomes and reduced total infarct volumes. In the peri-infarct, progesterone increased the density of neurons (NeuN+ cells), of cells of the oligodendroglial lineage (Olig2+ cells) and of oligodendrocyte progenitors (OP, NG2+ cells). Progesterone decreased the density of activated astrocytes (GFAP+ cells) and reactive microglia (Iba1+ cells) coexpressing the mannose receptor type 1 CD206 marker. Progesterone also reduced the expression of aquaporin 4 (AQP4), the water channel involved in both edema formation and resorption. The beneficial effects of progesterone were not observed in PRNesCre mice. Our findings show that progesterone treatment exerts beneficial effects on neurons, oligodendroglial cells and neuroinflammatory responses via PR. These findings demonstrate that progesterone is a pleiotropic cerebroprotective agent and that neural PR represent a therapeutic target for stroke cerebroprotection.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| | - Magalie Fréchou
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| |
Collapse
|
20
|
Jure I, De Nicola AF, Labombarda F. Progesterone effects on the oligodendrocyte linage: all roads lead to the progesterone receptor. Neural Regen Res 2019; 14:2029-2034. [PMID: 31397329 PMCID: PMC6788243 DOI: 10.4103/1673-5374.262570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A new role has emerged for progesterone after discovering its potent actions away from reproduction in both the central and the peripheral nervous system. The aim of the present report is to discuss progesterone’s mechanisms of action involved in myelination, remyelination and neuroinflammation. The pivotal role of the classic progesterone receptor is described and evidence is compiled about progesterone’s direct effects on oligodendrocyte linage and its indirect effects on oligodendrocyte precursor cell differentiation by decreasing the neuroinflammatory environment.
Collapse
Affiliation(s)
- Ignacio Jure
- Laboratorio de Bioquímica Neuroendocrina, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratorio de Bioquímica Neuroendocrina, Instituto de Biología y Medicina Experimental, CONICET; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratorio de Bioquímica Neuroendocrina, Instituto de Biología y Medicina Experimental, CONICET; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
21
|
Jure I, De Nicola AF, Labombarda F. Progesterone effects on oligodendrocyte differentiation in injured spinal cord. Brain Res 2018; 1708:36-46. [PMID: 30527678 DOI: 10.1016/j.brainres.2018.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/27/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022]
Abstract
Spinal cord lesions result in chronic demyelination as a consequence of secondary injury. Although oligodendrocyte precursor cells proliferate the differentiation program fails. Successful differentiation implies progressive decrease of transcriptional inhibitors followed by upregulation of activators. Progesterone emerges as an anti-inflammatory and pro-myelinating agent which improves locomotor outcome after spinal cord injury. In this study, we have demonstrated that spinal cord injury enhanced oligodendrocyte precursor cell number and decreased mRNA expression of transcriptional inhibitors (Id2, Id4, hes5). However, mRNA expression of transcriptional activators (Olig2, Nkx2.2, Sox10 and Mash1) was down-regulated 3 days post injury. Interestingly, a differentiation factor such as progesterone increased transcriptional activator mRNA levels and the density of Olig2- expressing oligodendrocyte precursor cells. The differentiation program is regulated by extracellular signals which modify transcriptional factors and epigenetic players. As TGFβ1 is a known oligodendrocyte differentiation factor which is regulated by progesterone in reproductive tissues, we assessed whether TGFβ1 could mediate progesterone remyelinating actions after the lesion. Notwithstanding that astrocyte, oligodendrocyte precursor and microglial cell density increased after spinal cord injury, the number of these cells which expressed TGFβ1 remained unchanged regarding sham operated rats. However, progesterone treatment increased TGFβ1 mRNA expression and the number of astrocytes and microglial TGFβ1 expressing cells which would indirectly enhance oligodendrocyte differentiation. Therefore, TGFβ1 arises as a potential mediator of progesterone differentiating effects on oligodendrocyte linage.
Collapse
Affiliation(s)
- Ignacio Jure
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155 C1121, Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155 C1121, Buenos Aires, Argentina.
| |
Collapse
|
22
|
Leicaj ML, Pasquini LA, Lima A, Gonzalez Deniselle MC, Pasquini JM, De Nicola AF, Garay LI. Changes in neurosteroidogenesis during demyelination and remyelination in cuprizone-treated mice. J Neuroendocrinol 2018; 30:e12649. [PMID: 30303567 DOI: 10.1111/jne.12649] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/20/2018] [Accepted: 10/01/2018] [Indexed: 12/28/2022]
Abstract
Changes of neurosteroids may be involved in the pathophysiology of multiple sclerosis (MS). The present study investigated whether changes of neurosteroidogenesis also occurred in the grey and white matter regions of the brain in mice subjected to cuprizone-induced demyelination. Accordingly, we compared the expression of neurosteroidogenic proteins, including steroidogenic acute regulatory protein (StAR), voltage-dependent anion channel (VDAC) and 18 kDa translocator protein (TSPO), as well as neurosteroidogenic enzymes, including the side chain cleavage enzyme (P450scc), 3β-hydroxysteroid dehydrogenase/isomerase and 5α-reductase (5α-R), during the demyelination and remyelination periods. Using immunohistochemistry and a quantitative polymerase chain reaction, we demonstrated a decreased expression of StAR, P450scc and 5α-R with respect to an increase astrocytic and microglial reaction and elevated levels of tumor necrosis factor (TNF)α during the cuprizone demyelination period in the hippocampus, cortex and corpus callosum. These parameters, as well as the glial reaction, were normalised after 2 weeks of spontaneous remyelination in regions containing grey matter. Conversely, persistent elevated levels of TNFα and low levels of StAR and P450scc were observed during remyelination in corpus callosum white matter. We conclude that neurosteroidogenesis/myelination status and glial reactivity are inversely related in the hippocampus and neocortex. Establishing a cause and effect relationship for the measured variables remains a future challenge for understanding the pathophysiology of MS.
Collapse
Affiliation(s)
- María L Leicaj
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
| | - Laura A Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry and Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
| | - Maria C Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
- Department of Physiological Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Juana M Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry and Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
- Department of Human Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Laura I Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
- Department of Human Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
23
|
Dong S, Zhang Q, Kong D, Zhou C, Zhou J, Han J, Zhou Y, Jin G, Hua X, Wang J, Hua F. Gender difference in the effect of progesterone on neonatal hypoxic/ischemic brain injury in mouse. Exp Neurol 2018; 306:190-198. [DOI: 10.1016/j.expneurol.2018.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/29/2018] [Accepted: 05/12/2018] [Indexed: 12/27/2022]
|
24
|
Meyer M, Lara A, Hunt H, Belanoff J, de Kloet ER, Gonzalez Deniselle MC, De Nicola AF. The Selective Glucocorticoid Receptor Modulator Cort 113176 Reduces Neurodegeneration and Neuroinflammation in Wobbler Mice Spinal Cord. Neuroscience 2018; 384:384-396. [PMID: 29890290 DOI: 10.1016/j.neuroscience.2018.05.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/18/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022]
Abstract
Wobbler mice are experimental models for amyotrophic lateral sclerosis. As such they show motoneuron degeneration, motor deficits, and astrogliosis and microgliosis of the spinal cord. Additionally, Wobbler mice show increased plasma, spinal cord and brain corticosterone levels and focal adrenocortical hyperplasia, suggesting a pathogenic role for glucocorticoids in this disorder. Considering this endocrine background, we examined whether the glucocorticoid receptor (GR) modulator CORT 113176 prevents spinal cord neuropathology of Wobblers. CORT 113176 shows high affinity for the GR, with low or null affinity for other steroid receptors. We employed five-month-old genotyped Wobbler mice that received s.c. vehicle or 30 mg/kg/day for 4 days of CORT 113176 dissolved in sesame oil. The mice were used on the 4th day, 2 h after the last dose of CORT 113176. Vehicle-treated Wobbler mice presented vacuolated motoneurons, increased glial fibrillary acidic protein (GFAP)+ astrocytes and decreased glutamine synthase (GS)+ cells. There was strong neuroinflammation, shown by increased staining for IBA1+ microglia and CD11b mRNA, enhanced expression of tumor necrosis factor-α, its cognate receptor TNFR1, toll-like receptor 4, the inducible nitric oxide synthase, NFkB and the high-mobility group box 1 protein (HMGB1). Treatment of Wobbler mice with CORT 113176 reversed the abnormalities of motoneurons and down-regulated proinflammatory mediators and glial reactivity. Expression of glutamate transporters GLT1 and GLAST mRNAs and GLT1 protein was significantly enhanced over untreated Wobblers. In summary, antagonism of GR with CORT 113176 prevented neuropathology and showed anti-inflammatory and anti-glutamatergic effects in the spinal cord of Wobbler mice.
Collapse
Affiliation(s)
- Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Agustina Lara
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Hazel Hunt
- CORCEPT Therapeutics, Menlo Park, CA, USA
| | | | - E Ronald de Kloet
- Division of Endocrinology, Dept. of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Physiology, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina.
| |
Collapse
|
25
|
Recalde G, Moreno-Sosa T, Yúdica F, Quintero CA, Sánchez MB, Jahn GA, Kalergis AM, Mackern-Oberti JP. Contribution of sex steroids and prolactin to the modulation of T and B cells during autoimmunity. Autoimmun Rev 2018. [DOI: 10.1016/j.autrev.2018.03.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
26
|
De Nicola AF, Garay LI, Meyer M, Guennoun R, Sitruk-Ware R, Schumacher M, Gonzalez Deniselle MC. Neurosteroidogenesis and progesterone anti-inflammatory/neuroprotective effects. J Neuroendocrinol 2018; 30. [PMID: 28675779 DOI: 10.1111/jne.12502] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/19/2022]
Abstract
Progesterone shows anti-inflammatory and promyelinating effects in mice with experimental autoimmune encephalomyelitis (EAE), a commonly used model for multiple sclerosis (MS). Because neurosteroids have been implicated as protective factors for MS and EAE, we analysed the expression of neurosteroidogenic enzymes in the compromised spinal cord of EAE mice. EAE was induced in female C57Bl6 mice, which were then killed on day 16 after induction. Progesterone was given by pellet implantation 1 week before EAE induction. Untreated EAE mice showed decreased mRNAs for the steroidogenic acute regulatory protein (Star), voltage-dependent anion channel (VDAC), cholesterol side-chain cleavage (P450scc), 5α-reductase, 3α-hydroxysteroid dehydrogenase (3α-HSOR) and aromatase, whereas changes of 3β-hydroxysteroid dehydrogenase (3β-HSD) were not significant. mRNA translocator protein (18 kDa) (TSPO) was elevated, concomitantly with a reactive microgliosis. EAE mice also showed abnormal mitochondrial ultrastructure in axons and neuronal bodies, as well as reduced expression of fission and fusion protein mRNAs. Progesterone pretreatment before EAE induction increased Star, VDAC, P450scc, 5α-reductase type I, 3α-HSOR and aromatase mRNAs and did not modify 3β-HSD. TSPO mRNA was decreased, possibly as a result of reversal of microgliosis. Progesterone pretreatment also improved mitochondrial ultrastructure and increased fission/fusion protein mRNAs. These mitochondrial effects may be part of the progesterone recovery of neurosteroidogenesis. The enzymes 3β-HSD, 3α-HSOR and 5α-reductase are also responsible for the formation of androgens. Because MS patients and EAE rodents show changes of central androgen levels, it is likely that, together with progestins and oestrogens, neuroandrogens afford neuroprotection for EAE and MS. The data reviewed suggest that enhanced synthesis of neurosteroids contributes in an auto/paracrine manner to reinforce the neuroprotective and anti-inflammatory effects of exogenous progesterone given to EAE mice.
Collapse
Affiliation(s)
- A F De Nicola
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - L I Garay
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - M Meyer
- Instituto de Biologia y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - R Guennoun
- U1195 Inserm and Université Paris-Sud, Le Kremlin-Bicêtre, France
| | | | - M Schumacher
- U1195 Inserm and Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - M C Gonzalez Deniselle
- The Population Council, New York, NY, USA
- Department of Physiology and Biophysics, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
27
|
Rosen S, Ham B, Mogil JS. Sex differences in neuroimmunity and pain. J Neurosci Res 2017; 95:500-508. [PMID: 27870397 DOI: 10.1002/jnr.23831] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022]
Abstract
Differences in the prevalence of chronic pain in women vs. men are well known, and decades of laboratory experimentation have demonstrated that women are more sensitive to pain than are men. Attention has thus shifted to investigating mechanisms underlying such differences. Recent evidence suggests that neuroimmune modulation of pain may represent an important cause of sex differences. The current Review examines the evidence for gonadal hormone modulation of the immune system, immune system modulation of pain, and interactions that might help to explain sex differences in pain. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sarah Rosen
- Department of Psychology and Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Boram Ham
- Department of Psychology and Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Jeffrey S Mogil
- Department of Psychology and Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Multipotency and therapeutic potential of NG2 cells. Biochem Pharmacol 2017; 141:42-55. [DOI: 10.1016/j.bcp.2017.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022]
|
29
|
Leva G, Klein C, Benyounes J, Hallé F, Bihel F, Collongues N, De Seze J, Mensah-Nyagan AG, Patte-Mensah C. The translocator protein ligand XBD173 improves clinical symptoms and neuropathological markers in the SJL/J mouse model of multiple sclerosis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3016-3027. [PMID: 28899788 DOI: 10.1016/j.bbadis.2017.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/04/2017] [Accepted: 09/08/2017] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is a severe autoimmune disease characterized by inflammatory, demyelinating and neurodegenerative components causing motor, sensory, visual and/or cognitive symptoms. The relapsing-remitting MS affecting 85% of patients is reliably mimicked by the proteolipid-protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) SJL/J-mouse model. Significant progress was made for MS treatment but the development of effective therapies devoid of severe side-effects remains a great challenge. Here, we combine clinical, behavioral, histopathological, biochemical and molecular approaches to demonstrate that low and well tolerated doses (10-20mg/kg) of TSPO ligand XBD173 (Emapunil) efficiently ameliorate clinical signs and neuropathology of PLP-EAE mice. In addition to the conventional clinical scoring of symptoms, we applied the robust behavioral Catwalk-method to confirm that XBD173 (10mg/kg) increases the maximum contact area parameter at EAE-disease peak, indicating an improvement/recovery of motor functions. Consistently, histopathological studies coupled with microscope-cellSens quantification and RT-qPCR analyzes showed that XBD173 prevented demyelination by restoring normal protein and mRNA levels of myelin basic protein that was significantly repressed in PLP-EAE mice spinal cord and brain. Interestingly, ELISA-based measurement revealed that XBD173 increased allopregnanolone concentrations in PLP-EAE mice spinal and brain tissues. Furthermore, flow cytometry assessment demonstrated that XBD173 therapy decreased serum level of pro-inflammatory cytokines, including interleukin-17A, Interleukin-6 and tumor-necrosis-factor alpha in PLP-EAE mice. As the optimal XBD173 dosing exerting the maximal beneficial action in EAE mice is the lower 10mg/kg dose, the paper opens interesting perspectives for the development of efficient and safe therapies against MS with slight or no side-effects.
Collapse
Affiliation(s)
- Géraldine Leva
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - Jérémie Benyounes
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - François Hallé
- Laboratoire d'innovation thérapeutique (LIT) CNRS UMR 7200, Faculté de Pharmacie de Strasbourg, 74 route du Rhin, CS 60024, 67401 Illkirch Cedex, France
| | - Frédéric Bihel
- Laboratoire d'innovation thérapeutique (LIT) CNRS UMR 7200, Faculté de Pharmacie de Strasbourg, 74 route du Rhin, CS 60024, 67401 Illkirch Cedex, France
| | - Nicolas Collongues
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - Jérôme De Seze
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France.
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France.
| |
Collapse
|
30
|
Aryanpour R, Pasbakhsh P, Zibara K, Namjoo Z, Beigi Boroujeni F, Shahbeigi S, Kashani IR, Beyer C, Zendehdel A. Progesterone therapy induces an M1 to M2 switch in microglia phenotype and suppresses NLRP3 inflammasome in a cuprizone-induced demyelination mouse model. Int Immunopharmacol 2017; 51:131-139. [PMID: 28830026 DOI: 10.1016/j.intimp.2017.08.007] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/14/2017] [Accepted: 08/09/2017] [Indexed: 12/14/2022]
Abstract
Demyelination of the central nervous system (CNS) has been associated to reactive microglia in neurodegenerative disorders, such as multiple sclerosis (MS). The M1 microglia phenotype plays a pro-inflammatory role while M2 is involved in anti-inflammatory processes in the brain. In this study, CPZ-induced demyelination mouse model was used to investigate the effect of progesterone (PRO) therapy on microglia activation and neuro-inflammation. Results showed that progesterone therapy (CPZ+PRO) decreased neurological behavioral deficits, as demonstrated by significantly decreased escape latencies, in comparison to CPZ mice. In addition, CPZ+PRO caused a significant reduction in the mRNA expression levels of M1-markers (iNOS, CD86, MHC-II and TNF-α) in the corpus callosum region, whereas the expression of M2-markers (Trem-2, CD206, Arg-1 and TGF-β) was significantly increased, in comparison to CPZ mice. Moreover, CPZ+PRO resulted in a significant decrease in the number of iNOS+ and Iba-1+/iNOS+ cells (M1), whereas TREM-2+ and Iba-1+/TREM-2+ cells (M2) significantly increased, in comparison to CPZ group. Furthermore, CPZ+PRO caused a significant decrease in mRNA and protein expression levels of NLRP3 and IL-18 (~2-fold), in comparison to the CPZ group. Finally, CPZ+PRO therapy was accompanied with reduced levels of demyelination, compared to CPZ, as confirmed by immunofluorescence to myelin basic protein (MBP) and Luxol Fast Blue (LFB) staining, as well as transmission electron microscopy (TEM) analysis. In summary, we reported for the first time that PRO therapy causes polarization of M2 microglia, attenuation of M1 phenotype, and suppression of NLRP3 inflammasome in a CPZ-induced demyelination model of MS.
Collapse
Affiliation(s)
- Roya Aryanpour
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, DSST, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Zeinab Namjoo
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Fatemeh Beigi Boroujeni
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Shahbeigi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Adib Zendehdel
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany; Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
31
|
Garay L, Gonzalez Giqueaux P, Guennoun R, Schumacher M, Gonzalez Deniselle MC, De Nicola AF. Progesterone treatment modulates mRNA OF neurosteroidogenic enzymes in a murine model of multiple sclerosis. J Steroid Biochem Mol Biol 2017; 165:421-429. [PMID: 27597394 DOI: 10.1016/j.jsbmb.2016.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 11/20/2022]
Abstract
Previous studies of experimental autoimmune encephalomyelitis (EAE) have shown that progesterone decreases inflammatory cell infiltration and proinflammatory factors, increases myelination and attenuates clinical grade of EAE mice. To elucidate potential mediators of these effects, we analyzed the mRNA expression of neurosteroidogenic enzymes in the spinal cord, in view of the protective role of steroids in EAE. We also analyzed mitochondrial morphology and dynamics (fusion and fission proteins), considering the role of mitochondria in neurosteroidogenesis. EAE was induced in C57Bl6 mice using MOG40-54 and killed on day 16 after induction. Using qPCR, we found in steroid-untreated EAE mice decreased mRNAs for the steroidogenic acute regulatory protein (Star), voltage-dependent anion channel (VDAC), P450scc (cholesterol side-chain cleavage), 5α-reductase, 3α-hydroxysteroid dehydrogenase (3α-HSD) and aromatase, whereas levels of 3β-hydroxysteroid dehydrogenase (3β-HSD) showed a large intra-group variance. We also found increased mRNA expression of 18Kd translocator protein (TSPO), which likely resulted from the reactive microgliosis in this model. EAE mice also showed pathological mitochondrial morphology and reduced expression of fission and fusion protein mRNAs. Most importantly, pretreatment with progesterone a week before EAE induction increased Star,VDAC, P450scc, 5α-reductase type I, 3α-HSD and aromatase mRNAs and did not modify 3β-HSD. TSPO mRNA was decreased, consequent with the inhibition of microgliosis. Mitochondrial morphology was improved and fission/fusion protein mRNAs were enhanced by progesterone treatment. Furthermore, progesterone protective effects on mitochondrial and endoplasmic reticulum may allow the recovery of neurosteroidogenesis. In this way, endogenously synthesized neurosteroids may reinforce the beneficial effects of exogenous progesterone previously shown in MS mice.
Collapse
Affiliation(s)
- Laura Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry(,) Faculty of Medicine, University of Buenos Aires, Argentina
| | - Paula Gonzalez Giqueaux
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Physiology, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry(,) Faculty of Medicine, University of Buenos Aires, Argentina.
| |
Collapse
|
32
|
Chew LJ, DeBoy CA. Pharmacological approaches to intervention in hypomyelinating and demyelinating white matter pathology. Neuropharmacology 2016; 110:605-625. [PMID: 26116759 PMCID: PMC4690794 DOI: 10.1016/j.neuropharm.2015.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/10/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022]
Abstract
White matter disease afflicts both developing and mature central nervous systems. Both cell intrinsic and extrinsic dysregulation result in profound changes in cell survival, axonal metabolism and functional performance. Experimental models of developmental white matter (WM) injury and demyelination have not only delineated mechanisms of signaling and inflammation, but have also paved the way for the discovery of pharmacological approaches to intervention. These reagents have been shown to enhance protection of the mature oligodendrocyte cell, accelerate progenitor cell recruitment and/or differentiation, or attenuate pathological stimuli arising from the inflammatory response to injury. Here we highlight reports of studies in the CNS in which compounds, namely peptides, hormones, and small molecule agonists/antagonists, have been used in experimental animal models of demyelination and neonatal brain injury that affect aspects of excitotoxicity, oligodendrocyte development and survival, and progenitor cell function, and which have been demonstrated to attenuate damage and improve WM protection in experimental models of injury. The molecular targets of these agents include growth factor and neurotransmitter receptors, morphogens and their signaling components, nuclear receptors, as well as the processes of iron transport and actin binding. By surveying the current evidence in non-immune targets of both the immature and mature WM, we aim to better understand pharmacological approaches modulating endogenous oligodendroglia that show potential for success in the contexts of developmental and adult WM pathology. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| | - Cynthia A DeBoy
- Biology Department, Trinity Washington University, Washington, DC, USA
| |
Collapse
|
33
|
Vinuesa A, Pomilio C, Menafra M, Bonaventura MM, Garay L, Mercogliano MF, Schillaci R, Lux Lantos V, Brites F, Beauquis J, Saravia F. Juvenile exposure to a high fat diet promotes behavioral and limbic alterations in the absence of obesity. Psychoneuroendocrinology 2016; 72:22-33. [PMID: 27337091 DOI: 10.1016/j.psyneuen.2016.06.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 06/03/2016] [Accepted: 06/04/2016] [Indexed: 01/09/2023]
Abstract
The incidence of metabolic disorders including obesity, type 2 diabetes and metabolic syndrome have seriously increased in the last decades. These diseases - with growing impact in modern societies - constitute major risk factors for neurodegenerative disorders such as Alzheimer's disease (AD), sharing insulin resistance, inflammation and associated cognitive impairment. However, cerebral cellular and molecular pathways involved are not yet clearly understood. Thus, our aim was to study the impact of a non-severe high fat diet (HFD) that resembles western-like alimentary habits, particularly involving juvenile stages where the brain physiology and connectivity are in plain maturation. To this end, one-month-old C57BL/6J male mice were given either a control diet or HFD during 4 months. Exposure to HFD produced metabolic alterations along with changes in behavioral and central parameters, in the absence of obesity. Two-month-old HFD mice showed increased glycemia and plasmatic IL1β but these values normalized at the end of the HFD protocol at 5 months of age, probably representing an acute response that is compensated at later stages. After four months of HFD exposure, mice presented dyslipidemia, increased Lipoprotein-associated phospholipase A2 (Lp-PLA2) activity, hepatic insulin resistance and inflammation. Alterations in the behavioral profile of the HFD group were shown by the impediment in nest building behavior, deficiencies in short and mid-term spatial memories, anxious and depressive- like behavior. Regarding the latter disruptions in emotional processing, we found an increased neural activity in the amygdala, shown by a greater number of c-Fos+ nuclei. We found that hippocampal adult neurogenesis was decreased in HFD mice, showing diminished cell proliferation measured as Ki67+ cells and neuronal differentiation in SGZ by doublecortin labeling. These phenomena were accompanied by a neuroinflammatory and insulin-resistant state in the hippocampus, depicted by a reactive phenotype in Iba1+ microglia cells (increased in number and soma size) and an impaired response to insulin given by decreased phosphorylated Akt levels and increased levels of inhibitory phosphorylation of IRS1. Our data portray a set of alterations in behavioral and neural parameters as a consequence of an early-life exposure to a quite moderate high fat diet, many of which can resemble AD-related features. These results highly emphasize the need to study how metabolic and neurodegenerative disorders are interrelated in deep, thus allowing the finding of successful preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Angeles Vinuesa
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Carlos Pomilio
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Martin Menafra
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Laura Garay
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | | | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Victoria Lux Lantos
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Fernando Brites
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Beauquis
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
34
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
35
|
Rolf L, Damoiseaux J, Hupperts R, Huitinga I, Smolders J. Network of nuclear receptor ligands in multiple sclerosis: Common pathways and interactions of sex-steroids, corticosteroids and vitamin D3-derived molecules. Autoimmun Rev 2016; 15:900-10. [DOI: 10.1016/j.autrev.2016.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/08/2016] [Indexed: 01/12/2023]
|
36
|
Coronel MF, Labombarda F, González SL. Neuroactive steroids, nociception and neuropathic pain: A flashback to go forward. Steroids 2016; 110:77-87. [PMID: 27091763 DOI: 10.1016/j.steroids.2016.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/29/2016] [Accepted: 04/11/2016] [Indexed: 11/26/2022]
Abstract
The present review discusses the potential role of neurosteroids/neuroactive steroids in the regulation of nociceptive and neuropathic pain, and recapitulates the current knowledge on the main mechanisms involved in the reduction of pain, especially those occurring at the dorsal horn of the spinal cord, a crucial site for nociceptive processing. We will make special focus on progesterone and its derivative allopregnanolone, which have been shown to exert remarkable actions in order to prevent or reverse the maladaptive changes and pain behaviors that arise after nervous system damage in various experimental neuropathic conditions.
Collapse
Affiliation(s)
- María F Coronel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Facultad de Ciencias Biomédicas, Universidad Austral, Presidente Perón 1500, B1629AHJ, Pilar, Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratorio de Bioquímica Neuroendócrina, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG, Buenos Aires, Argentina
| | - Susana L González
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG, Buenos Aires, Argentina.
| |
Collapse
|
37
|
Ravikumar B, Crawford D, Dellovade T, Savinainen A, Graham D, Liere P, Oudinet JP, Webb M, Hering H. Differential efficacy of the TSPO ligands etifoxine and XBD-173 in two rodent models of Multiple Sclerosis. Neuropharmacology 2016; 108:229-37. [PMID: 27039042 DOI: 10.1016/j.neuropharm.2016.03.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/24/2016] [Accepted: 03/29/2016] [Indexed: 12/25/2022]
Abstract
Neurosteroids such as progesterone and allopregnanolone have been shown to exert neuroprotective effects under a variety of pathological or insult conditions, and there is evidence that the neurosteroid system is perturbed in Multiple Sclerosis (MS) patients. Neurosteroids are synthesized in the central nervous system (CNS) through a series of metabolic transformations, beginning with a rate-limiting step of cholesterol transport through the outer mitochondrial membrane via the transporter translocator protein (TSPO). We examined the effects of etifoxine and XBD-173, two different brain penetrant TSPO agonists, for their ability to ameliorate clinical signs in two different experimental autoimmune encephalitis (EAE) models. Etifoxine, as previously reported, was efficacious in EAE, while XBD-173 was not. Surprisingly, XBD-173, but not etifoxine elevated relevant neurosteroids in brain of female rats and differed in its ability to exert anti-inflammatory and direct neuroprotective effects in vitro as compared to etifoxine. We conclude that the neurosteroid elevations produced in brain by XBD-173 are not sufficient to ameliorate EAE and suggest that etifoxine may have additional mechanisms of action that provide therapeutic benefit in this model system.
Collapse
Affiliation(s)
- Brinda Ravikumar
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA
| | - Dan Crawford
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA
| | - Tammy Dellovade
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA
| | - Anneli Savinainen
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA
| | - Danielle Graham
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA
| | - Philippe Liere
- U1195 Inserm & University Paris-Sud, 80, rue du Général Leclerc, 94276 Kremlin-Bicetre, France
| | - Jean-Paul Oudinet
- U1195 Inserm & University Paris-Sud, 80, rue du Général Leclerc, 94276 Kremlin-Bicetre, France
| | - Mike Webb
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA
| | - Heike Hering
- EMD Serono Research and Development Institute, Billerica, MA 01821, USA.
| |
Collapse
|
38
|
Au A, Feher A, McPhee L, Jessa A, Oh S, Einstein G. Estrogens, inflammation and cognition. Front Neuroendocrinol 2016; 40:87-100. [PMID: 26774208 DOI: 10.1016/j.yfrne.2016.01.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/02/2016] [Accepted: 01/11/2016] [Indexed: 01/15/2023]
Abstract
The effects of estrogens are pleiotropic, affecting multiple bodily systems. Changes from the body's natural fluctuating levels of estrogens, through surgical removal of the ovaries, natural menopause, or the administration of exogenous estrogens to menopausal women have been independently linked to an altered immune profile, and changes to cognitive processes. Here, we propose that inflammation may mediate the relationship between low levels of estrogens and cognitive decline. In order to determine what is known about this connection, we review the literature on the cognitive effects of decreased estrogens due to oophorectomy or natural menopause, decreased estrogens' role on inflammation--both peripherally and in the brain--and the relationship between inflammation and cognition. While this review demonstrates that much is unknown about the intersection between estrogens, cognition, inflammation, we propose that there is an important interaction between these literatures.
Collapse
Affiliation(s)
- April Au
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Anita Feher
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Lucy McPhee
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Ailya Jessa
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Soojin Oh
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Gillian Einstein
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
39
|
Kashani IR, Hedayatpour A, Pasbakhsh P, Kafami L, Khallaghi B, Malek F. Progesterone Enhanced Remyelination in the Mouse Corpus Callosum after Cuprizone Induced Demyelination. IRANIAN JOURNAL OF MEDICAL SCIENCES 2015; 40:507-14. [PMID: 26538779 PMCID: PMC4628141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Progesterone as a sex steroid hormone is thought to affect and prevent demyelination, but its role in promoting myelin repair is far less investigated. In this study, remyelinating potential of progesterone in corpus callosum was evaluated on an experimental model of MS. METHODS In this experimental study, adult male C57BL/6 mice were fed with 0.2% (w/w) cuprizone in ground breeder chow ad libitum for 6 weeks. At day zero, after cuprizone removal, mice were divided randomly into two groups: (a) placebo group, which received saline pellet implant, (b) progesterone group, which received progesterone pellet implant. Some mice of the same age were fed with their normal diet to serve as the healthy control group. Two weeks after progesterone administration, Myelin content was assessed by Luxol-fast blue staining. The myelin basic protein (MBP) and proteolipid protein (PLP) expression were assessed using Western blot analysis and the changes in the number of oligodendrocytes and oligodendroglial progenitor cells were assessed by immunohistochemistry (IHC) and flow cytometry. RESULTS Luxol-fast blue staining revealed enhanced remyelination in the progesterone group when compared with the placebo group. Densitometry measurements of immunoblots demonstrated that MBP and PLP proteins contents were significantly increased in the progesterone group compared with the placebo group. Flow cytometry and IHC analysis showed increases in Olig2 and O4 cells in the progesterone group compared with the placebo group. CONCLUSION Overall, our results indicate that progesterone treatment can stimulate myelin production and that it may provide a feasible and practical way for remyelination in diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Iraj Ragerdi Kashani
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran,Correspondence: Iraj Ragerdi Kashani, PhD; Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, 16 Azar Street, Poursina Street, Tehran, Iran Tel: +98 21 64053410 Fax: +98 21 66419072
| | - Azim Hedayatpour
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Laya Kafami
- Department of Pathobiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran,Shefa Neurosciences Research Center, Tehran, Iran
| | | | - Fatemeh Malek
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Labombarda F, Jure I, Gonzalez S, Lima A, Roig P, Guennoun R, Schumacher M, De Nicola AF. A functional progesterone receptor is required for immunomodulation, reduction of reactive gliosis and survival of oligodendrocyte precursors in the injured spinal cord. J Steroid Biochem Mol Biol 2015; 154:274-84. [PMID: 26369614 DOI: 10.1016/j.jsbmb.2015.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/26/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022]
Abstract
The anti-inflammatory effects of progesterone have been increasingly recognized in several neuropathological models, including spinal cord inflammation. In the present investigation, we explored the regulation of proinflammatory factors and enzymes by progesterone at several time points after spinal cord injury (SCI) in male rats. We also demonstrated the role of the progesterone receptor (PR) in inhibiting inflammation and reactive gliosis, and in enhancing the survival of oligodendrocyte progenitors cells (OPC) in injured PR knockout (PRKO) mice receiving progesterone. First, after SCI in rats, progesterone greatly attenuated the injury-induced hyperexpression of the mRNAs of interleukin 1β (IL1β), IL6, tumor necrosis factor alpha (TNFα), inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2), all involved in oligodendrocyte damage. Second, the role of the PR was investigated in PRKO mice after SCI, in which progesterone failed to reduce the high expression of IL1β, IL6, TNFα and IκB-α mRNAs, the latter being considered an index of reduced NF-κB transactivation. These effects occurred in a time framework coincident with a reduction in the astrocyte and microglial responses. In contrast to wild-type mice, progesterone did not increase the density of OPC and did not prevent apoptotic death of these cells in PRKO mice. Our results support a role of PR in: (a) the anti-inflammatory effects of progesterone; (b) the modulation of astrocyte and microglial responses and (c) the prevention of OPC apoptosis, a mechanism that would enhance the commitment of progenitors to the remyelination pathway in the injured spinal cord.
Collapse
Affiliation(s)
- Florencia Labombarda
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Ignacio Jure
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Susana Gonzalez
- Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina; Laboratory of Nociception and Neuropathic Pain, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Rachida Guennoun
- U1195 Inserm and Université Paris-Sud, 94276 Le Kremlin-Bicêtre, France
| | | | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
41
|
The progesterone receptor agonist Nestorone holds back proinflammatory mediators and neuropathology in the wobbler mouse model of motoneuron degeneration. Neuroscience 2015; 308:51-63. [DOI: 10.1016/j.neuroscience.2015.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/20/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022]
|
42
|
Bernardes D, Brambilla R, Bracchi-Ricard V, Karmally S, Dellarole A, Carvalho-Tavares J, Bethea JR. Prior regular exercise improves clinical outcome and reduces demyelination and axonal injury in experimental autoimmune encephalomyelitis. J Neurochem 2015; 136 Suppl 1:63-73. [PMID: 26364732 DOI: 10.1111/jnc.13354] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 08/18/2015] [Accepted: 08/21/2015] [Indexed: 12/22/2022]
Abstract
Although previous studies have shown that forced exercise modulates inflammation and is therapeutic acutely for experimental autoimmune encephalomyelitis (EAE), the long-term benefits have not been evaluated. In this study, we investigated the effects of preconditioning exercise on the clinical and pathological progression of EAE. Female C57BL/6 mice were randomly assigned to either an exercised (Ex) or unexercised (UEx) group and all of them were induced for EAE. Mice in the Ex group had an attenuated clinical score relative to UEx mice throughout the study. At 42 dpi, flow cytometry analysis showed a significant reduction in B cells, CD4(+) T cells, and CD8(+) T cells infiltrating into the spinal cord in the Ex group compared to UEx. Ex mice also had a significant reduction in myelin damage with a corresponding increase in proteolipid protein expression. Finally, Ex mice had a significant reduction in axonal damage. Collectively, our study demonstrates for the first time that a prolonged and forced preconditioning protocol of exercise improves clinical outcome and attenuates pathological hallmarks of EAE at chronic disease. In this study, we show that a program of 6 weeks of preconditioning exercise promoted a significant reduction of cells infiltrating into the spinal cord, a significant reduction in myelin damage and a significant reduction in axonal damage in experimental autoimmune encephalomyelitis (EAE) mice at 42 dpi. Collectively, our study demonstrates for the first time that a preconditioning protocol of exercise improves clinical outcome and attenuates pathological hallmarks of EAE at chronic disease.
Collapse
Affiliation(s)
- Danielle Bernardes
- Departamento de Fisiologia e Biofísica, Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,CAPES Foundation, Ministry of Education of Brazil, Brasília, DF, Brazil.,The Miami Project To Cure Paralysis, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Roberta Brambilla
- The Miami Project To Cure Paralysis, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Valerie Bracchi-Ricard
- The Miami Project To Cure Paralysis, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Department of Biology, Drexel University, Philadelphia, Philadelphia, USA
| | - Shaffiat Karmally
- The Miami Project To Cure Paralysis, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Anna Dellarole
- The Miami Project To Cure Paralysis, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Juliana Carvalho-Tavares
- Departamento de Fisiologia e Biofísica, Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - John R Bethea
- Department of Biology, Drexel University, Philadelphia, Philadelphia, USA
| |
Collapse
|
43
|
Garcia-Ovejero D, González S, Paniagua-Torija B, Lima A, Molina-Holgado E, De Nicola AF, Labombarda F. Progesterone reduces secondary damage, preserves white matter, and improves locomotor outcome after spinal cord contusion. J Neurotrauma 2014; 31:857-71. [PMID: 24460450 DOI: 10.1089/neu.2013.3162] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Progesterone is an anti-inflammatory and promyelinating agent after spinal cord injury, but its effectiveness on functional recovery is still controversial. In the current study, we tested the effects of chronic progesterone administration on tissue preservation and functional recovery in a clinically relevant model of spinal cord lesion (thoracic contusion). Using magnetic resonance imaging, we observed that progesterone reduced both volume and rostrocaudal extension of the lesion at 60 days post-injury. In addition, progesterone increased the number of total mature oligodendrocytes, myelin basic protein immunoreactivity, and the number of axonal profiles at the epicenter of the lesion. Further, progesterone treatment significantly improved motor outcome as assessed using the Basso-Bresnahan-Beattie scale for locomotion and CatWalk gait analysis. These data suggest that progesterone could be considered a promising therapeutical candidate for spinal cord injury.
Collapse
Affiliation(s)
- Daniel Garcia-Ovejero
- 1 Laboratorio de Neuroinflamación, Hospital Nacional de Parapléjicos , Toledo, Spain
| | | | | | | | | | | | | |
Collapse
|
44
|
Efficacy of the selective progesterone receptor agonist Nestorone for chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 276:89-97. [DOI: 10.1016/j.jneuroim.2014.08.619] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 11/17/2022]
|
45
|
Irwin RW, Solinsky CM, Brinton RD. Frontiers in therapeutic development of allopregnanolone for Alzheimer's disease and other neurological disorders. Front Cell Neurosci 2014; 8:203. [PMID: 25126056 PMCID: PMC4115668 DOI: 10.3389/fncel.2014.00203] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/04/2014] [Indexed: 01/08/2023] Open
Abstract
Allopregnanolone (Allo), a neurosteroid, has emerged as a promising promoter of endogenous regeneration in brain. In a mouse model of Alzheimer’s disease, Allo induced neurogenesis, oligodendrogenesis, white matter generation and cholesterol homeostasis while simultaneously reducing β-amyloid and neuroinflammatory burden. Allo activates signaling pathways and gene expression required for regeneration of neural stem cells and their differentiation into neurons. In parallel, Allo activates systems to sustain cholesterol homeostasis and reduce β-amyloid generation. To advance Allo into studies for chronic human neurological conditions, we examined translational and clinical parameters: dose, regimen, route, formulation, outcome measures, and safety regulations. A treatment regimen of once per week at sub-sedative doses of Allo was optimal for regeneration and reduction in Alzheimer’s pathology. This regimen had a high safety profile following chronic exposure in aged normal and Alzheimer’s mice. Formulation of Allo for multiple routes of administration has been developed for both preclinical and clinical testing. Preclinical evidence for therapeutic efficacy of Allo spans multiple neurological diseases including Alzheimer’s, Parkinson’s, multiple sclerosis, Niemann-Pick, diabetic neuropathy, status epilepticus, and traumatic brain injury. To successfully translate Allo as a therapeutic for multiple neurological disorders, it will be necessary to tailor dose and regimen to the targeted therapeutic mechanisms and disease etiology. Treatment paradigms conducted in accelerated disease models in young animals have a low probability of successful translation to chronic diseases in adult and aged humans. Gender, genetic risks, stage and burden of disease are critical determinants of efficacy. This review focuses on recent advances in development of Allo for Alzheimer’s disease (AD) that have the potential to accelerate therapeutic translation for multiple unmet neurological needs.
Collapse
Affiliation(s)
- Ronald W Irwin
- Department of Pharmacology and Pharmaceutical Sciences, Pharmaceutical Sciences Center, School of Pharmacy, University of Southern California Los Angeles, CA, USA
| | - Christine M Solinsky
- Clinical and Experimental Therapeutics Program, School of Pharmacy, University of Southern California Los Angeles, CA, USA
| | - Roberta Diaz Brinton
- Department of Pharmacology and Pharmaceutical Sciences, Pharmaceutical Sciences Center, School of Pharmacy, University of Southern California Los Angeles, CA, USA ; Department of Neurology, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
46
|
Mouihate A. TLR4-mediated brain inflammation halts neurogenesis: impact of hormonal replacement therapy. Front Cell Neurosci 2014; 8:146. [PMID: 24904290 PMCID: PMC4034512 DOI: 10.3389/fncel.2014.00146] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 05/07/2014] [Indexed: 12/27/2022] Open
Abstract
Experimental and epidemiological data show that the severity and the duration of brain inflammation are attenuated in females compared to males. This attenuated brain inflammation is ascribed to 17β-estradiol. However, several studies suggest that 17β-estradiol is also endowed with proinflammatory properties. The aim of the present study is to assess the effect of hormonal replacement therapies on lipopolysaccharide (LPS)-induced brain inflammation and its consequent effect on newly born neurons. Bilaterally ovariectomized rats received intrastriatal injection of LPS (250 ng/μl) and were subsequently given daily subcutaneous injections of either vehicle, 17β-estradiol (25 μg/kg) or 17β-estradiol and progesterone (5 mg/kg). Microglial activation and newly born neurons in the rostral migratory stream were monitored using double immunofluorescence. Nuclear factor κB (NFκB) signaling pathway and its target inflammatory proteins were assessed by either western blot [cyclooxygenase-2 (COX-2) and interleukin-6 (IL-6)] or enzyme-linked immunosorbent assay [tumor necrosis factor-α (TNF-α)]. LPS-induced activation of microglia, promoted NFκB signaling pathway and enhanced the production of proinflammatory proteins (TNF-α and COX-2). These proinflammatory responses were not attenuated by 17β-estradiol injection. Supplementation of 17β-estradiol with progesterone significantly dampened these proinflammatory processes. Interestingly, LPS-induced brain inflammation dampened the number of newly born neurons in the rostral migratory stream. Administration of combined 17β-estradiol and progesterone resulted in a significantly higher number of newly born neurons when compared to those seen in rats given either vehicle or 17β-estradiol alone. These data strongly suggest that combined 17β-estradiol and progesterone, and not 17β-estradiol alone, rescues neurogenesis from the deleterious effect of brain inflammation likely via the inhibition of the signaling pathways leading to the activation of proinflammatory genes.
Collapse
Affiliation(s)
- Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University Safat, Kuwait
| |
Collapse
|
47
|
Rahn EJ, Iannitti T, Donahue RR, Taylor BK. Sex differences in a mouse model of multiple sclerosis: neuropathic pain behavior in females but not males and protection from neurological deficits during proestrus. Biol Sex Differ 2014; 5:4. [PMID: 24581045 PMCID: PMC3974112 DOI: 10.1186/2042-6410-5-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/31/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS), a demyelinating disease of the central nervous system, is one of the most prevalent neurological disorders in the industrialized world. This disease afflicts more than two million people worldwide, over two thirds of which are women. MS is typically diagnosed between the ages of 20-40 and can produce debilitating neurological impairments including muscle spasticity, muscle paralysis, and chronic pain. Despite the large sex disparity in MS prevalence, clinical and basic research investigations of how sex and estrous cycle impact development, duration, and severity of neurological impairments and pain symptoms are limited. To help address these questions, we evaluated behavioral signs of sensory and motor functions in one of the most widely characterized animal models of MS, the experimental autoimmune encephalomyelitis (EAE) model. METHODS C57BL/6 male and female mice received flank injection of complete Freund's adjuvant (CFA) or CFA plus myelin oligodendrocyte glycoprotein 35-55 (MOG35-55) to induce EAE. Experiment 1 evaluated sex differences of EAE-induced neurological motor deficits and neuropathic pain-like behavior over 3 weeks, while experiment 2 evaluated the effect of estrous phase in female mice on the same behavioral measures for 3 months. EAE-induced neurological motor deficits including gait analysis and forelimb grip strength were assessed. Neuropathic pain-like behaviors evaluated included sensitivity to mechanical, cold, and heat stimulations. Estrous cycle was determined daily via vaginal lavage. RESULTS MOG35-55-induced EAE produced neurological impairments (i.e., motor dysfunction) including mild paralysis and decreases in grip strength in both females and males. MOG35-55 produced behavioral signs of neuropathic pain-mechanical and cold hypersensitivity-in females, but not males. MOG35-55 did not change cutaneous heat sensitivity in either sex. Administration of CFA or CFA + MOG35-55 prolonged the time spent in diestrus for 2 weeks, after which normal cycling returned. MOG35-55 produced fewer neurological motor deficits when mice were in proestrus relative to non-proestrus phases. CONCLUSIONS We conclude that female mice are superior to males for the study of neuropathic pain-like behaviors associated with MOG35-55-induced EAE. Further, proestrus may be protective against EAE-induced neurological deficits, thus necessitating further investigation into the impact that estrous cycle exerts on MS symptoms.
Collapse
Affiliation(s)
| | | | | | - Bradley K Taylor
- Department of Physiology, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|
48
|
Elkabes S, Nicot AB. Sex steroids and neuroprotection in spinal cord injury: a review of preclinical investigations. Exp Neurol 2014; 259:28-37. [PMID: 24440641 DOI: 10.1016/j.expneurol.2014.01.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/25/2013] [Accepted: 01/04/2014] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a debilitating condition that affects motor, sensory and autonomic functions. Subsequent to the first mechanical trauma, secondary events, which include inflammation and glial activation, exacerbate tissue damage and worsen functional deficits. Although these secondary injury mechanisms are amenable to therapeutic interventions, the efficacy of current approaches is inadequate. Further investigations are necessary to implement new therapies that can protect neural cells and attenuate some of the detrimental effects of inflammation while promoting regeneration. Studies on different animal models of SCI indicated that sex steroids, especially 17β-estradiol and progesterone, exert neuroprotective, anti-apoptotic and anti-inflammatory effects, ameliorate tissue sparing and improve functional deficits in SCI. As sex steroid receptors are expressed in a variety of cells including neurons, glia and immune system-related cells which infiltrate the injury epicenter, sex steroids could impact multiple processes simultaneously and in doing so, influence the outcomes of SCI. However, the translation of these pre-clinical findings into the clinical setting presents challenges such as the narrow therapeutic time window of sex steroid administration, the diversity of treatment regimens that have been employed in animal studies and the lack of sufficient information regarding the persistence of the effects in chronic SCI. The current review will summarize some of the major findings in this field and will discuss the challenges associated with the implementation of sex steroids as a promising treatment in human SCI.
Collapse
Affiliation(s)
- Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA.
| | - Arnaud B Nicot
- UMR 1064, INSERM, Nantes, France; Faculté de Médecine, Université de Nantes, France; ITUN, CHU de Nantes, France
| |
Collapse
|
49
|
De Nicola AF, Gonzalez Deniselle MC, Garay L, Meyer M, Gargiulo-Monachelli G, Guennoun R, Schumacher M, Carreras MC, Poderoso JJ. Progesterone protective effects in neurodegeneration and neuroinflammation. J Neuroendocrinol 2013; 25:1095-103. [PMID: 23639063 DOI: 10.1111/jne.12043] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 04/12/2013] [Accepted: 04/26/2013] [Indexed: 01/03/2023]
Abstract
Progesterone is a neuroprotective, promyelinating and anti-inflammatory factor for the nervous system. Here, we review the effects of progesterone in models of motoneurone degeneration and neuroinflammation. In neurodegeneration of the Wobbler mouse, a subset of spinal cord motoneurones showed increased activity of nitric oxide synthase (NOS), increased intramitochondrial NOS, decreased activity of respiratory chain complexes, and decreased activity and protein expression of Mn-superoxide dismutase type 2 (MnSOD2). Clinically, Wobblers suffered several degrees of motor impairment. Progesterone treatment restored the expression of neuronal markers, decreased the activity of NOS and enhanced complex I respiratory activity and MnSOD2. Long-term treatment with progesterone increased muscle strength, biceps weight and survival. Collectively, these data suggest that progesterone prevented neurodegeneration. To study the effects of progesterone in neuroinflammation, we employed mice with experimental autoimmune encephalomyelitis (EAE). EAE mice spinal cord showed increased mRNA levels of the inflammatory mediators tumour necrosis factor (TNF)α and its receptor TNFR1, the microglial marker CD11b, inducible NOS and the toll-like receptor 4. Progesterone pretreatment of EAE mice blocked the proinflammatory mediators, decreased Iba1+ microglial cells and attenuated clinical signs of EAE. Therefore, reactive glial cells became targets of progesterone anti-inflammatory effects. These results represent a starting point for testing the usefulness of neuroactive steroids in neurological disorders.
Collapse
Affiliation(s)
- A F De Nicola
- Department of Human Biochemistry, Faculty of Medicine, Instituto de Biologia y Medicina Experimental, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Si D, Wang H, Wang Q, Zhang C, Sun J, Wang Z, Zhang Z, Zhang Y. Progesterone treatment improves cognitive outcome following experimental traumatic brain injury in rats. Neurosci Lett 2013; 553:18-23. [DOI: 10.1016/j.neulet.2013.07.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 07/10/2013] [Accepted: 07/30/2013] [Indexed: 01/16/2023]
|