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
|
Angeloni E, Germelli L, Costa B, Martini C, Da Pozzo E. Neurosteroids and Translocator Protein (TSPO) in neuroinflammation. Neurochem Int 2024; 182:105916. [PMID: 39681140 DOI: 10.1016/j.neuint.2024.105916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Neurosteroids have a crucial role in physiological intrinsic regulations of the Central Nervous System functions. They are derived from peripheral steroidogenic sources and from the de novo neurosteroidogenic capacity of brain cells. Significant alterations of neurosteroid levels have been frequently observed in neuroinflammation and neurodegenerative diseases. Such level fluctuations may be useful for both diagnosis and treatment of these pathological conditions. Beyond steroid administration, enhancing the endogenous production by Translocator Protein (TSPO) targeting has been proposed to restore these altered pathological levels. However, the neurosteroid quantification and the prediction of their final effects are often troublesome, sometimes controversial and context dependent, due to the complexity of neurosteroid biosynthetic pathway and to the low produced amounts. The aim of this review is to report recent advances, and technical limitations, in neurosteroid-related strategies against neuroinflammation.
Collapse
Affiliation(s)
- Elisa Angeloni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Lorenzo Germelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.
| |
Collapse
|
3
|
Bäckström T, Doverskog M, Blackburn TP, Scharschmidt BF, Felipo V. Allopregnanolone and its antagonist modulate neuroinflammation and neurological impairment. Neurosci Biobehav Rev 2024; 161:105668. [PMID: 38608826 DOI: 10.1016/j.neubiorev.2024.105668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
Neuroinflammation accompanies several brain disorders, either as a secondary consequence or as a primary cause and may contribute importantly to disease pathogenesis. Neurosteroids which act as Positive Steroid Allosteric GABA-A receptor Modulators (Steroid-PAM) appear to modulate neuroinflammation and their levels in the brain may vary because of increased or decreased local production or import from the systemic circulation. The increased synthesis of steroid-PAMs is possibly due to increased expression of the mitochondrial cholesterol transporting protein (TSPO) in neuroinflammatory tissue, and reduced production may be due to changes in the enzymatic activity. Microglia and astrocytes play an important role in neuroinflammation, and their production of inflammatory mediators can be both activated and inhibited by steroid-PAMs and GABA. What is surprising is the finding that both allopregnanolone, a steroid-PAM, and golexanolone, a novel GABA-A receptor modulating steroid antagonist (GAMSA), can inhibit microglia and astrocyte activation and normalize their function. This review focuses on the role of steroid-PAMs in neuroinflammation and their importance in new therapeutic approaches to CNS and liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
4
|
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
|
5
|
Pollock NM, Fernandes JP, Woodfield J, Moussa E, Hlavay B, Branton WG, Wuest M, Mohammadzadeh N, Schmitt L, Plemel JR, Julien O, Wuest F, Power C. Gasdermin D activation in oligodendrocytes and microglia drives inflammatory demyelination in progressive multiple sclerosis. Brain Behav Immun 2024; 115:374-393. [PMID: 37914099 DOI: 10.1016/j.bbi.2023.10.022] [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: 04/13/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023] Open
Abstract
Neuroinflammation coupled with demyelination and neuro-axonal damage in the central nervous system (CNS) contribute to disease advancement in progressive multiple sclerosis (P-MS). Inflammasome activation accompanied by proteolytic cleavage of gasdermin D (GSDMD) results in cellular hyperactivation and lytic death. Using multiple experimental platforms, we investigated the actions of GSDMD within the CNS and its contributions to P-MS. Brain tissues from persons with P-MS showed significantly increased expression of GSDMD, NINJ1, IL-1β, and -18 within chronic active demyelinating lesions compared to MS normal appearing white matter and nonMS (control) white matter. Conditioned media (CM) from stimulated GSDMD+/+ human macrophages caused significantly greater cytotoxicity of oligodendroglial and neuronal cells, compared to CM from GSDMD-/- macrophages. Oligodendrocytes and CNS macrophages displayed increased Gsdmd immunoreactivity in the central corpus callosum (CCC) of cuprizone (CPZ)-exposed Gsdmd+/+ mice, associated with greater demyelination and reduced oligodendrocyte precursor cell proliferation, compared to CPZ-exposed Gsdmd-/- animals. CPZ-exposed Gsdmd+/+ mice exhibited significantly increased G-ratios and reduced axonal densities in the CCC compared to CPZ-exposed Gsdmd-/- mice. Proteomic analyses revealed increased brain complement C1q proteins and hexokinases in CPZ-exposed Gsdmd-/- animals. [18F]FDG PET imaging showed increased glucose metabolism in the hippocampus and whole brain with intact neurobehavioral performance in Gsdmd-/- animals after CPZ exposure. GSDMD activation in CNS macrophages and oligodendrocytes contributes to inflammatory demyelination and neuroaxonal injury, offering mechanistic and potential therapeutic insights into P-MS pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Laura Schmitt
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton AB, Canada.
| | | | | | | | - Christopher Power
- Department of Medicine (Neurology), Canada; Department of Medical Microbiology & Immunology, Canada.
| |
Collapse
|
6
|
Roczkowsky A, Limonta D, Fernandes JP, Branton WG, Clarke M, Hlavay B, Noyce RS, Joseph JT, Ogando NS, Das SK, Elaish M, Arbour N, Evans DH, Langdon K, Hobman TC, Power C. COVID-19 Induces Neuroinflammation and Suppresses Peroxisomes in the Brain. Ann Neurol 2023; 94:531-546. [PMID: 37190821 DOI: 10.1002/ana.26679] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Peroxisome injury occurs in the central nervous system (CNS) during multiple virus infections that result in neurological disabilities. We investigated host neuroimmune responses and peroxisome biogenesis factors during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection using a multiplatform strategy. METHODS Brain tissues from coronavirus disease 2019 (COVID-19) (n = 12) and other disease control (ODC) (n = 12) patients, as well as primary human neural cells and Syrian hamsters, infected with a clinical variant of SARS-CoV-2, were investigated by droplet digital polymerase chain reaction (ddPCR), quantitative reverse transcriptase PCR (RT-qPCR), and immunodetection methods. RESULTS SARS-CoV-2 RNA was detected in the CNS of 4 patients with COVID-19 with viral protein (NSP3 and spike) immunodetection in the brainstem. Olfactory bulb, brainstem, and cerebrum from patients with COVID-19 showed induction of pro-inflammatory transcripts (IL8, IL18, CXCL10, NOD2) and cytokines (GM-CSF and IL-18) compared to CNS tissues from ODC patients (p < 0.05). Peroxisome biogenesis factor transcripts (PEX3, PEX5L, PEX11β, and PEX14) and proteins (PEX3, PEX14, PMP70) were suppressed in the CNS of COVID-19 compared to ODC patients (p < 0.05). SARS-CoV-2 infection of hamsters revealed viral RNA detection in the olfactory bulb at days 4 and 7 post-infection while inflammatory gene expression was upregulated in the cerebrum of infected animals by day 14 post-infection (p < 0.05). Pex3 transcript levels together with catalase and PMP70 immunoreactivity were suppressed in the cerebrum of SARS-CoV-2 infected animals (p < 0.05). INTERPRETATION COVID-19 induced sustained neuroinflammatory responses with peroxisome biogenesis factor suppression despite limited brainstem SARS-CoV-2 neurotropism in humans. These observations offer insights into developing biomarkers and therapies, while also implicating persistent peroxisome dysfunction as a contributor to the neurological post-acute sequelae of COVID-19. ANN NEUROL 2023;94:531-546.
Collapse
Affiliation(s)
- A Roczkowsky
- Department of Medicine, University of Alberta, Edmonton, AB, USA
| | - D Limonta
- Department of Cell Biology, University of Alberta, Edmonton, AB, USA
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, USA
| | - J P Fernandes
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, USA
| | - W G Branton
- Department of Medicine, University of Alberta, Edmonton, AB, USA
| | - M Clarke
- Department of Medicine, University of Alberta, Edmonton, AB, USA
| | - B Hlavay
- Department of Medicine, University of Alberta, Edmonton, AB, USA
| | - R S Noyce
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, USA
| | - J T Joseph
- Department of Pathology, University of Calgary, Calgary, AB, USA
| | - N S Ogando
- Department of Medicine, University of Alberta, Edmonton, AB, USA
| | - S K Das
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, USA
| | - M Elaish
- Department of Cell Biology, University of Alberta, Edmonton, AB, USA
| | - N Arbour
- Department of Neuroscience, University of Montreal, and CHUM, Montreal, QC, Canada
| | - D H Evans
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, USA
| | - K Langdon
- Department of Pathology, University of Calgary, Calgary, AB, USA
| | - T C Hobman
- Department of Cell Biology, University of Alberta, Edmonton, AB, USA
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, USA
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, USA
| | - C Power
- Department of Medicine, University of Alberta, Edmonton, AB, USA
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, USA
| |
Collapse
|
7
|
Angeloni E, Germelli L, Marchetti L, Da Pozzo E, Tremolanti C, Wetzel CH, Baglini E, Taliani S, Da Settimo F, Martini C, Costa B. The human microglial surveillant phenotype is preserved by de novo neurosteroidogenesis through the control of cholesterol homeostasis: Crucial role of 18 kDa Translocator Protein. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166751. [PMID: 37169037 DOI: 10.1016/j.bbadis.2023.166751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
Neurodegenerative disease-associated microglia commonly exhibit harmful cholesterol accumulation that impairs their ability to resolve the neuroinflammatory response, contributing to disease onset and progression. Neurosteroids, whose levels have been often found significantly altered in brain diseases, are the most potent endogenous anti-inflammatory molecules exerting beneficial effects on activities of brain cells, including microglia. For the first time, the impact of neurosteroidogenesis on cholesterol homeostasis for the immune surveillance phenotype maintenance was investigated in a human microglia in vitro model. To enhance and inhibit neurosteroidogenesis, pharmacological stimulation and knock-down of 18 kDa Translocator Protein (TSPO), which is involved in the neurosteroidogenesis rate-limiting step, were used as experimental approaches, respectively. The obtained results point to an essential autocrine control of neurosteroidogenesis in orchestrating cholesterol trafficking in human microglia. TSPO pharmacological stimulation ensured cholesterol turnover by strengthening cholesterol efflux systems and preserving healthy immune surveillant phenotype. Conversely, TSPO knock-down induced an impairment of the controlled interplay among cholesterol synthesis, efflux, and metabolism mechanisms, leading to an excessive cholesterol accumulation and acquisition of a chronically activated dysfunctional phenotype. In this model, the exogenous neurosteroid administration restored proper the cholesterol clearance. The TSPO ability in promoting native neurosteroidogenesis opens the way to restore cholesterol homeostasis, and thus to maintain microglia proper functionality for the treatment of neuroinflammation-related brain diseases.
Collapse
Affiliation(s)
- Elisa Angeloni
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Lorenzo Germelli
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Laura Marchetti
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy; Center for Instrument Sharing University of Pisa (CISUP), Lungarno Pacinotti, 43/44, 56126 Pisa, Italy.
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy; Center for Instrument Sharing University of Pisa (CISUP), Lungarno Pacinotti, 43/44, 56126 Pisa, Italy.
| | - Chiara Tremolanti
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93059 Regensburg, Germany.
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy; Center for Instrument Sharing University of Pisa (CISUP), Lungarno Pacinotti, 43/44, 56126 Pisa, Italy.
| |
Collapse
|
8
|
Amanollahi M, Jameie M, Rezaei N. Neuroinflammation as a potential therapeutic target in neuroimmunological diseases. TRANSLATIONAL NEUROIMMUNOLOGY, VOLUME 7 2023:475-504. [DOI: 10.1016/b978-0-323-85841-0.00021-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Hatipoglu E, Hacioglu Y, Polat Y, Arslan HF, Oner S, Ekmekci OB, Niyazoglu M. Do neurosteroids have impact on depression and cognitive functions in cases with acromegaly? Growth Horm IGF Res 2022; 66:101496. [PMID: 35952406 DOI: 10.1016/j.ghir.2022.101496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/23/2022] [Accepted: 07/24/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Neurosteroids (NSs) are a distinct hormone group and, they are known for their contribution into the status of mood and cognitive functions. Whether they are also involved in the mood disturbances and cognition in acromegaly is not known. Herein we aimed to evaluate the relation of mood status and cognitive functions with the NS levels in cases with acromegaly. DESIGN A total of 33 cases with acromegaly composed the acromegaly group (AG) and, 30 age and gender-matched cases without acromegaly composed the control group (CG). The levels of Allopregnanolone (AP), pregnenolone (PRG), 24S-hydroxycholesterol (24OHC), dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), androsterone (ADT), GH and IGF-1 were measured in each group. Beck Depression Inventory (BDI) was used to assess depressive symptoms, whereas an extensive neuropsychological assessment with several neurocognitive tests were carried out for each subject by an experienced psychologist. RESULTS Cases with acromegaly had lower 24OHC and DHEA levels (p = 0.002 and p = 0.007, respectively) in comparison to CG. Of the cognitive functions time to complete 1 s Series was significantly higher and, the scores on Switching Verbal Fluency Test, Boston Naming Test (BNT)-semantic and BNT-phonological, the highest learning point of Oktem Verbal Memory Processes Test (VMPT) were significantly lower in cases with acromegaly in comparison to those in controls (p = 0.004, p = 0.01, p < 0.001, p = 0.02 and p = 0.05, respectively). KAS-perseveration errors were higher in CG (p = 0.03). In AG the levels of AP were negatively correlated with the scores on Months backward Test (MBT), Animal Naming Test, Construction, BNT-spontaneous and positively correlated with BNT-incorrect answers; PRG was positively correlated with VMPT-retention scores, ADT was negatively correlated with MBT and 3 s Series scores, DHEAS was positively correlated with VMPT-the highest learning point whereas it was negatively correlated with MBT scores. Additionally, the scores on BDI were positively correlated with DHEA levels in AG. CONCLUSION Cognitive changes may be encountered in acromegaly and, neurosteroids may contribute to the changes in certain cognitive functions.
Collapse
Affiliation(s)
- Esra Hatipoglu
- Division of Endocrinology, Department of Internal Medicine, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey.
| | - Yalcin Hacioglu
- Division of Family Medicine, Department of Internal Medicine, Ministry of Health's Istanbul Education and Research Hospital, Istanbul, Turkey
| | - Yeliz Polat
- Department of Psychology, Ministry of Health's Istanbul Education and Research Hospital, Istanbul, Turkey
| | - Hilmi Furkan Arslan
- Department of Biochemistry, Ministry of Health's Istanbul Education and Research Hospital, Istanbul, Turkey
| | - Sena Oner
- Department of Biochemistry, Cerrahpasa University, Cerrahpasa Medical Faculty, Istanbul, Turkey
| | - Ozlem Balci Ekmekci
- Department of Biochemistry, Cerrahpasa University, Cerrahpasa Medical Faculty, Istanbul, Turkey
| | - Mutlu Niyazoglu
- Division of Endocrinology, Department of Internal Medicine, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
10
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
11
|
Vitku J, Hill M, Kolatorova L, Kubala Havrdova E, Kancheva R. Steroid Sulfation in Neurodegenerative Diseases. Front Mol Biosci 2022; 9:839887. [PMID: 35281259 PMCID: PMC8904904 DOI: 10.3389/fmolb.2022.839887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Steroid sulfation and desulfation participates in the regulation of steroid bioactivity, metabolism and transport. The authors focused on sulfation and desulfation balance in three neurodegenerative diseases: Alzheimer´s disease (AD), Parkinson´s disease (PD), and multiple sclerosis (MS). Circulating steroid conjugates dominate their unconjugated counterparts, but unconjugated steroids outweigh their conjugated counterparts in the brain. Apart from the neurosteroid synthesis in the central nervous system (CNS), most brain steroids cross the blood-brain barrier (BBB) from the periphery and then may be further metabolized. Therefore, steroid levels in the periphery partly reflect the situation in the brain. The CNS steroids subsequently influence the neuronal excitability and have neuroprotective, neuroexcitatory, antidepressant and memory enhancing effects. They also exert anti-inflammatory and immunoprotective actions. Like the unconjugated steroids, the sulfated ones modulate various ligand-gated ion channels. Conjugation by sulfotransferases increases steroid water solubility and facilitates steroid transport. Steroid sulfates, having greater half-lives than their unconjugated counterparts, also serve as a steroid stock pool. Sulfotransferases are ubiquitous enzymes providing massive steroid sulfation in adrenal zona reticularis and zona fasciculata.. Steroid sulfatase hydrolyzing the steroid conjugates is exceedingly expressed in placenta but is ubiquitous in low amounts including brain capillaries of BBB which can rapidly hydrolyze the steroid sulfates coming across the BBB from the periphery. Lower dehydroepiandrosterone sulfate (DHEAS) plasma levels and reduced sulfotransferase activity are considered as risk factors in AD patients. The shifted balance towards unconjugated steroids can participate in the pathophysiology of PD and anti-inflammatory effects of DHEAS may counteract the MS.
Collapse
Affiliation(s)
- Jana Vitku
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
- *Correspondence: Jana Vitku,
| | - Martin Hill
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| | - Lucie Kolatorova
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| | - Eva Kubala Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Radmila Kancheva
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| |
Collapse
|
12
|
Lentiviral Infections Persist in Brain despite Effective Antiretroviral Therapy and Neuroimmune Activation. mBio 2021; 12:e0278421. [PMID: 34903055 PMCID: PMC8669467 DOI: 10.1128/mbio.02784-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV infection persists in different tissue reservoirs among people with HIV (PWH) despite effective antiretroviral therapy (ART). In the brain, lentiviruses replicate principally in microglia and trafficking macrophages. The impact of ART on this viral reservoir is unknown. We investigated the activity of contemporary ART in various models of lentivirus brain infection. HIV-1 RNA and total and integrated DNA were detected in cerebral cortex from all PWH (n = 15), regardless of ART duration or concurrent plasma viral quantity and, interestingly, integrated proviral DNA levels in brain were significantly higher in the aviremic ART-treated group (P < 0.005). Most ART drugs tested (dolutegravir, ritonavir, raltegravir, and emtricitabine) displayed significantly lower 50% effective concentration (EC50) values in lymphocytes than in microglia, except tenofovir, which showed 1.5-fold greater activity in microglia (P < 0.05). In SIV-infected Chinese rhesus macaques, despite receiving suppressive (n = 7) or interrupted (n = 8) ART, brain tissues had similar SIV-encoded RNA and total and integrated DNA levels compared to brains from infected animals without ART (n = 3). SIV and HIV-1 capsid antigens were immunodetected in brain, principally in microglia/macrophages, regardless of ART duration and outcome. Antiviral immune responses were comparable in the brains of ART-treated and untreated HIV- and SIV-infected hosts. Both HIV-1 and SIV persist in brain tissues despite contemporary ART, with undetectable virus in blood. ART interruption exerted minimal effect on the SIV brain reservoir and did not alter the neuroimmune response profile. These studies underscore the importance of augmenting ART potency in different tissue compartments. IMPORTANCE Antiretroviral therapy (ART) suppresses HIV-1 in plasma and CSF to undetectable levels. However, the impact of contemporary ART on HIV-1 brain reservoirs remains uncertain. An active viral reservoir in the brain during ART could lead to rebound systemic infection after cessation of therapy, development of drug resistance mutations, and neurological disease. ART's impact, including its interruption, on brain proviral DNA remains unclear. The present studies show that in different experimental platforms, contemporary ART did not suppress viral burden in the brain, regardless of ART component regimen, the duration of therapy, and its interruption. Thus, new strategies for effective HIV-1 suppression in the brain are imperative to achieve sustained HIV suppression.
Collapse
|
13
|
Yang F, Wu SC, Ling ZX, Chao S, Zhang LJ, Yan XM, He L, Yu LM, Zhao LY. Altered Plasma Metabolic Profiles in Chinese Patients With Multiple Sclerosis. Front Immunol 2021; 12:792711. [PMID: 34975894 PMCID: PMC8715987 DOI: 10.3389/fimmu.2021.792711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease that leads to the demyelination of nerve axons. An increasing number of studies suggest that patients with MS exhibit altered metabolic profiles, which might contribute to the course of MS. However, the alteration of metabolic profiles in Chinese patients with MS and their potential roles in regulating the immune system remain elusive. In this study, we performed a global untargeted metabolomics approach in plasma samples from 22 MS-affected Chinese patients and 21 healthy subjects. A total of 42 differentially abundant metabolites (DAMs) belonging to amino acids, lipids, and carbohydrates were identified in the plasma of MS patients and compared with those in healthy controls. We observed an evident reduction in the levels of amino acids, such as L-tyrosine, L-isoleucine, and L-tryptophan, whereas there was a great increase in the levels of L-glutamic acid and L-valine in MS-affected patients. The levels of lipid and carbohydrate metabolites, such as sphingosine 1-phosphate and myo-inositol, were also reduced in patients with MS. In addition, the concentrations of proinflammatory cytokines, such as IL-17 and TNF-α, were significantly increased, whereas those of several anti-inflammatory cytokines and chemokines, such as IL-1ra, IL-7, and MIP-1α, were distinctly reduced in the plasma of MS patients compared with those in healthy subjects. Interestingly, some DAMs, such as L-tryptophan and sphingosine 1-phosphate, showed an evident negative correlation with changes in the level of TNF-α and IL-17, while tightly positively correlating with altered concentrations of anti-inflammatory cytokines and chemokines, such as MIP-1α and RANTES. Our results revealed that altered metabolomic profiles might contribute to the pathogenesis and course of MS disease by modulating immuno-inflammatory responses in the peripheral system, which is essential for eliciting autoimmune responses in the central nervous system, thus resulting in the progression of MS. This study provides potential clues for developing therapeutic strategies for MS in the near future.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Institutes for Shanghai Pudong Decoding Life, Research Center for Lin He Academician New Medicine, Shanghai, China
| | - Shao-chang Wu
- Department of Geriatrics and Clinical Laboratory, Lishui Second People’s Hospital, Lishui, China
| | - Zong-xin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Microbe & Host Health, Linyi University, Linyi, China
| | - Shan Chao
- Institutes for Shanghai Pudong Decoding Life, Research Center for Lin He Academician New Medicine, Shanghai, China
| | - Li-juan Zhang
- Department of Geriatrics and Clinical Laboratory, Lishui Second People’s Hospital, Lishui, China
| | - Xiu-mei Yan
- Department of Geriatrics and Clinical Laboratory, Lishui Second People’s Hospital, Lishui, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Li-mei Yu
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Long-you Zhao, ; Li-mei Yu,
| | - Long-you Zhao
- Department of Geriatrics and Clinical Laboratory, Lishui Second People’s Hospital, Lishui, China
- *Correspondence: Long-you Zhao, ; Li-mei Yu,
| |
Collapse
|
14
|
Rustichelli C, Monari E, Avallone R, Bellei E, Bergamini S, Tomasi A, Ferrari A. Dehydroepiandrosterone sulfate, dehydroepiandrosterone, 5α-dihydroprogesterone and pregnenolone in women with migraine: Analysis of serum levels and correlation with age, migraine years and frequency. J Pharm Biomed Anal 2021; 206:114388. [PMID: 34597839 DOI: 10.1016/j.jpba.2021.114388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/30/2022]
Abstract
Migraine is a very painful, disabling and extremely common disorder among the world's adult population, especially women, and it is associated with a variety of comorbidities. Neuroactive steroids exhibit pleiotropic actions on the nervous system. Alterations in their peripheral and central levels could be involved in the pathogenesis, still not fully understood, of migraine and its comorbidities. The purpose of our exploratory study was to determine and compare the serum levels of dehydroepiandrosterone sulfate (DHEAS), dehydroepiandrosterone (DHEA), 5α-dihydroprogesterone (DHP) and pregnenolone (PREGNE) between women suffering from migraine without aura (MO group, n = 30) and age-matched non-headache women as controls (C group, n = 30). Correlations with age, migraine years and frequency were also analyzed. The patients were enrolled at a headache center; controls were patients' contacts. Calibrators and serum samples were spiked with the internal standards (ISs) solution and treated to deplete proteins and phospholipids. The obtained extracts were evaporated to dryness, derivatized and analyzed by LC-MS/MS in multiple reaction monitoring mode. Analytes' levels were determined by interpolation on the regression curves, generated from the analyte quantifier ion peak area to the corresponding IS. MO group presented significantly lower levels of DHEAS, DHEA and DHP compared to C group (P < 0.05, Student't-test) and the neurosteroid levels negatively correlated with years of migraine and migraine days/3 months (P < 0.05, linear regression analysis). These results parallel to previous studies showing reduced serum levels of allopregnanolone and pregnenolone sulfate in women with migraine. The low serum levels found for both excitatory and inhibitory neurosteroids suggested that women with migraine might suffer from inadequate neuroprotection, anti-inflammation activity and pain modulation. These deficits might underlie the migraine chronification process and represent the link between migraine and its various comorbidities.
Collapse
Affiliation(s)
- Cecilia Rustichelli
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 41125 Modena, Italy
| | - Emanuela Monari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, Proteomic Lab, University of Modena and Reggio Emilia, via del Pozzo, 41124 Modena, Italy
| | - Rossella Avallone
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 41125 Modena, Italy
| | - Elisa Bellei
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, Proteomic Lab, University of Modena and Reggio Emilia, via del Pozzo, 41124 Modena, Italy
| | - Stefania Bergamini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, Proteomic Lab, University of Modena and Reggio Emilia, via del Pozzo, 41124 Modena, Italy
| | - Aldo Tomasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, Proteomic Lab, University of Modena and Reggio Emilia, via del Pozzo, 41124 Modena, Italy
| | - Anna Ferrari
- Department of Biomedical, Metabolic and Neural Sciences, Unit of Medical Toxicology, Headache Centre and Drug Abuse, University of Modena and Reggio Emilia, via del Pozzo, 41124 Modena, Italy
| |
Collapse
|
15
|
Amato A, Ragonese P, Ingoglia S, Schiera G, Schirò G, Di Liegro CM, Salemi G, Di Liegro I, Proia P. Lactate Threshold Training Program on Patients with Multiple Sclerosis: A Multidisciplinary Approach. Nutrients 2021; 13:nu13124284. [PMID: 34959834 PMCID: PMC8704660 DOI: 10.3390/nu13124284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/29/2022] Open
Abstract
Physical activity could play a key role in improving the quality of life, particularly in patients with nervous system diseases such as multiple sclerosis (MS). Through lactacid anaerobic training, this study aims to investigate the effects at a bio-psycho-physical level to counteract the chronic fatigue associated with the pathology, and to improve mental health at a psychological and neurotrophic level. Eight subjects (age: 34.88 ± 4.45 years) affected by multiple sclerosis were involved. A lactate threshold training program was administered biweekly for 12 weeks at the beginning of the study (T0), at the end of the study (T1) and at 9 months after the end of the study (T2), with physical, psychological and hematochemicals parameters, and dietary habits being tested. The results obtained confirmed that lactacid exercise can influence brain-derived neurotrophic factor (BDNF) levels as well as dehydroepiandrosterone sulfate (DHEAS) levels. In addition, levels of baseline lactate, which could be best used as an energy substrate, showed a decrease after the protocol training. Self-efficacy regarding worries and concerns management significantly increased from T0 to T1. The eating attitudes test (EAT-26) did not highlight any eating disease in the patients with a normal diet enrolled in our study. Physical exercise also greatly influenced the patients psychologically and emotionally, increasing their self-esteem. Lactate threshold training, together with dietary habits, appears to exert synergic positive effects on inflammation, neural plasticity and neuroprotection, producing preventive effects on MS symptoms and progression.
Collapse
Affiliation(s)
- Alessandra Amato
- Department of Psychology, Educational Science and Human Movement, Research Unit, University of Palermo, 90100 Palermo, Italy; (A.A.); (S.I.)
| | - Paolo Ragonese
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90100 Palermo, Italy; (P.R.); (G.S.); (G.S.); (I.D.L.)
| | - Sonia Ingoglia
- Department of Psychology, Educational Science and Human Movement, Research Unit, University of Palermo, 90100 Palermo, Italy; (A.A.); (S.I.)
| | - Gabriella Schiera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90100 Palermo, Italy; (G.S.); (C.M.D.L.)
| | - Giuseppe Schirò
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90100 Palermo, Italy; (P.R.); (G.S.); (G.S.); (I.D.L.)
| | - Carlo Maria Di Liegro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90100 Palermo, Italy; (G.S.); (C.M.D.L.)
| | - Giuseppe Salemi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90100 Palermo, Italy; (P.R.); (G.S.); (G.S.); (I.D.L.)
| | - Italia Di Liegro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90100 Palermo, Italy; (P.R.); (G.S.); (G.S.); (I.D.L.)
| | - Patrizia Proia
- Department of Psychology, Educational Science and Human Movement, Research Unit, University of Palermo, 90100 Palermo, Italy; (A.A.); (S.I.)
- Correspondence:
| |
Collapse
|
16
|
Ysrraelit MC, Correale J. Impact of Andropause on Multiple Sclerosis. Front Neurol 2021; 12:766308. [PMID: 34803897 PMCID: PMC8602357 DOI: 10.3389/fneur.2021.766308] [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: 08/28/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Andropause results from the natural decrease in testosterone levels that occurs with age. In contrast to menopause, which is a universal, well-characterized process associated with absolute gonadal failure, andropause ensues after gradual decline of both hypothalamic-pituitary-gonadal axis activity, as well as of testicular function, a process which usually develops over a period of many years. Increasing evidence on greater risk of Multiple sclerosis (MS) associated with lower testosterone levels is being reported. Likewise, epidemiological studies have shown a later age of onset of MS in men, relative to women, which could perhaps respond to the decline in protective testosterone levels. In this review, we will discuss the role of androgens in the development and function of the innate and adaptive immune response, as well as in neuroprotective mechanisms relevant to MS. Testosterone effects observed in different animal models and in epidemiological studies in humans will be discussed, as well as their correlation with physical disability and cognitive function levels. Finally, published and ongoing clinical trials exploring the role of androgens, particularly at key stages of sexual maturation, will be reviewed.
Collapse
Affiliation(s)
- Maria C Ysrraelit
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Jorge Correale
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
17
|
Infection of Glia by Human Pegivirus Suppresses Peroxisomal and Antiviral Signaling Pathways. J Virol 2021; 95:e0107421. [PMID: 34524914 DOI: 10.1128/jvi.01074-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human pegivirus (HPgV) infects peripheral leukocytes but was recently shown to be a neurotropic virus associated with leukoencephalitis in humans. In the present study, we investigated the neural cell tropism of HPgV as well as its effects on host immune responses. HPgV wild type (WT) and a mutant virus with a deletion in the HPgV NS2 gene (ΔNS2) were able to productively infect human astrocytes and microglia but not neurons or an oligodendrocyte-derived cell line. Of note, the ΔNS2 virus replicated better than WT pegivirus in astrocytes, with both viruses being able to subsequently infect and spread in fresh human astrocyte cultures. Infection of human glia by HPgV WT and ΔNS2 viruses resulted in suppression of peroxisome-associated genes, including PEX11B, ABCD1, PEX7, ABCD3, PEX3, and PEX5L, during peak viral production, which was accompanied by reduced expression of IFNB, IRF3, IRF1, and MAVS, particularly in ΔNS2-infected cells. These data were consistent with analyses of brain tissue from patients infected with HPgV in which we observed suppression of peroxisome and type I interferon gene transcripts, including PEX11B, ABCD3, IRF1, and IRF3, with concurrent loss of PMP70 immunoreactivity in glia. Our data indicate that human astrocytes and microglia are permissive to HPgV infection, resulting in peroxisome injury and suppressed antiviral signaling that is influenced by viral diversity. IMPORTANCE Human pegiviruses are detected in 1 to 5% of the general population, principally infecting leukocytes, although their effects on human health remain uncertain. Here, we show that human pegivirus infects specific neural cell types in culture and human brain and, like other neurotropic flaviviruses, causes suppression of peroxisome and antiviral signaling pathways, which could favor ongoing viral infection and perhaps confer susceptibility to the development of neurological disease.
Collapse
|
18
|
Li Z, Liu Y, Jia A, Cui Y, Feng J. Cerebrospinal fluid cells immune landscape in multiple sclerosis. J Transl Med 2021; 19:125. [PMID: 33766068 PMCID: PMC7995713 DOI: 10.1186/s12967-021-02804-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Background Multiple Sclerosis (MS) is a potentially devastating autoimmune neurological disorder, which characteristically induces demyelination of white matter in the brain and spinal cord. Methods In this study, three characteristics of the central nervous system (CNS) immune microenvironment occurring during MS onset were explored; immune cell proportion alteration, differential gene expression profile, and related pathways. The raw data of two independent datasets were obtained from the ArrayExpress database; E-MTAB-69, which was used as a derivation cohort, and E-MTAB-2374 which was used as a validation cohort. Differentially expressed genes (DEGs) were identified by the false discovery rate (FDR) value of < 0.05 and |log2 (Fold Change)|> 1, for further analysis. Then, functional enrichment analyses were performed to explore the pathways associated with MS onset. The gene expression profiles were analyzed using CIBERSORT to identify the immune type alterations involved in MS disease. Results After verification, the proportion of five types of immune cells (plasma cells, monocytes, macrophage M2, neutrophils and eosinophils) in cerebrospinal fluid (CSF) were revealed to be significantly altered in MS cases compared to the control group. Thus, the complement and coagulation cascades and the systemic lupus erythematosus (SLE) pathways may play critical roles in MS. We identified NLRP3, LILRB2, C1QB, CD86, C1QA, CSF1R, IL1B and TLR2 as eight core genes correlated with MS. Conclusions Our study identified the change in the CNS immune microenvironment of MS cases by analysis of the in silico data using CIBERSORT. Our data may assist in providing directions for further research as to the molecular mechanisms of MS and provide future potential therapeutic targets in treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02804-7.
Collapse
Affiliation(s)
- Zijian Li
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yongchao Liu
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Aili Jia
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yueran Cui
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
19
|
Jolivel V, Brun S, Binamé F, Benyounes J, Taleb O, Bagnard D, De Sèze J, Patte-Mensah C, Mensah-Nyagan AG. Microglial Cell Morphology and Phagocytic Activity Are Critically Regulated by the Neurosteroid Allopregnanolone: A Possible Role in Neuroprotection. Cells 2021; 10:698. [PMID: 33801063 PMCID: PMC8004004 DOI: 10.3390/cells10030698] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Microglial cells are key players in neural pathogenesis and microglial function regulation appears to be pivotal in controlling neuroinflammatory/neurological diseases. Here, we investigated the effects and mechanism of action of neurosteroid allopregnanolone (ALLO) on murine microglial BV-2 cells and primary microglia in order to determine ALLO-induced immunomodulatory potential and to provide new insights for the development of both natural and safe neuroprotective strategies targeting microglia. Indeed, ALLO-treatment is increasingly suggested as beneficial in various models of neurological disorders but the underlying mechanisms have not been elucidated. Therefore, the microglial cells were cultured with various serum concentrations to mimic the blood-brain-barrier rupture and to induce their activation. Proliferation, viability, RT-qPCR, phagocytosis, and morphology analyzes, as well as migration with time-lapse imaging and quantitative morphodynamic methods, were combined to investigate ALLO actions on microglia. BV-2 cells express subunits of GABA-A receptor that mediates ALLO activity. ALLO (10µM) induced microglial cell process extension and decreased migratory capacity. Interestingly, ALLO modulated the phagocytic activity of BV-2 cells and primary microglia. Our results, which show a direct effect of ALLO on microglial morphology and phagocytic function, suggest that the natural neurosteroid-based approach may contribute to developing effective strategies against neurological disorders that are evoked by microglia-related abnormalities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 1 rue Eugène Boeckel, 67000 Strasbourg, France; (V.J.); (S.B.); (F.B.); (J.B.); (O.T.); (D.B.); (J.D.S.); (C.P.-M.)
| |
Collapse
|
20
|
Biosynthesis and signalling functions of central and peripheral nervous system neurosteroids in health and disease. Essays Biochem 2021; 64:591-606. [PMID: 32756865 PMCID: PMC7517341 DOI: 10.1042/ebc20200043] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Neurosteroids are steroid hormones synthesised de novo in the brain and peripheral nervous tissues. In contrast to adrenal steroid hormones that act on intracellular nuclear receptors, neurosteroids directly modulate plasma membrane ion channels and regulate intracellular signalling. This review provides an overview of the work that led to the discovery of neurosteroids, our current understanding of their intracellular biosynthetic machinery, and their roles in regulating the development and function of nervous tissue. Neurosteroids mediate signalling in the brain via multiple mechanisms. Here, we describe in detail their effects on GABA (inhibitory) and NMDA (excitatory) receptors, two signalling pathways of opposing function. Furthermore, emerging evidence points to altered neurosteroid function and signalling in neurological disease. This review focuses on neurodegenerative diseases associated with altered neurosteroid metabolism, mainly Niemann-Pick type C, multiple sclerosis and Alzheimer disease. Finally, we summarise the use of natural and synthetic neurosteroids as current and emerging therapeutics alongside their potential use as disease biomarkers.
Collapse
|
21
|
Saito LB, Fernandes JP, Smith MJ, Doan MAL, Branton WG, Schmitt LM, Wuest M, Monaco MC, Major EO, Wuest F, Power C. Intranasal anti-caspase-1 therapy preserves myelin and glucose metabolism in a model of progressive multiple sclerosis. Glia 2020; 69:216-229. [PMID: 32882086 DOI: 10.1002/glia.23896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory demyelination and axonal injury in the central nervous system (CNS) are cardinal features of progressive multiple sclerosis (MS), and linked to activated brain macrophage-like cells (BMCs) including resident microglia and trafficking macrophages. Caspase-1 is a pivotal mediator of inflammation and cell death in the CNS. We investigated the effects of caspase-1 activation and its regulation in models of MS. Brains from progressive MS and non-MS patients, as well as cultured human oligodendrocytes were examined by transcriptomic and morphological methods. Next generation transcriptional sequencing of progressive MS compared to non-MS patients' normal appearing white matter (NAWM) showed induction of caspase-1 as well as other inflammasome-associated genes with concurrent suppression of neuron-specific genes. Oligodendrocytes exposed to TNFα exhibited upregulation of caspase-1 with myelin gene suppression in a cell differentiation state-dependent manner. Brains from cuprizone-exposed mice treated by intranasal delivery of the caspase-1 inhibitor, VX-765 or its vehicle, were investigated in morphological and molecular studies, as well as by fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging. Cuprizone exposure resulted in BMC and caspase-1 activation accompanied by demyelination and axonal injury, which was abrogated by intranasal VX-765 treatment. FDG-PET imaging revealed suppressed glucose metabolism in the thalamus, hippocampus and cortex of cuprizone-exposed mice that was restored with VX-765 treatment. These studies highlight the caspase-1 dependent interactions between inflammation, demyelination, and glucose metabolism in progressive MS and associated models. Intranasal delivery of an anti-caspase-1 therapy represents a promising therapeutic approach for progressive MS and other neuro-inflammatory diseases.
Collapse
Affiliation(s)
- Leina B Saito
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jason P Fernandes
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Mackenzie J Smith
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew A L Doan
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - William G Branton
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Laura M Schmitt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Melinda Wuest
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Frank Wuest
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Christopher Power
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
22
|
McKenzie BA, Fernandes JP, Doan MAL, Schmitt LM, Branton WG, Power C. Activation of the executioner caspases-3 and -7 promotes microglial pyroptosis in models of multiple sclerosis. J Neuroinflammation 2020; 17:253. [PMID: 32861242 PMCID: PMC7456507 DOI: 10.1186/s12974-020-01902-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pyroptosis is a type of proinflammatory regulated cell death (RCD) in which caspase-1 proteolytically cleaves gasdermin D (GSDMD) to yield a cytotoxic pore-forming protein. Recent studies have suggested that additional cell death pathways may interact with GSDMD under certain circumstances to execute pyroptosis. Microglia/macrophages in the central nervous system (CNS) undergo GSDMD-associated pyroptosis in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) but the contribution of other cell death pathways to this phenomenon is unknown. Herein, we tested the hypothesis that multiple RCD pathways underlie microglial pyroptosis in the context of neuroinflammation. METHODS A siRNA screen of genes with known RCD functions was performed in primary human microglia to evaluate their role in nigericin-induced pyroptosis using supernatant lactate dehydrogenase activity as a read-out of cell lysis. Activation of apoptotic executioner proteins and their contribution to pyroptosis was assessed using semi-quantitative confocal microscopy, high-sensitivity ELISA, immunoblot, cell lysis assays, and activity-based fluorescent probes. Quantification of pyroptosis-related protein expression was performed in CNS lesions from patients with progressive MS and mice with MOG35-55-induced EAE, and in matched controls. RESULTS Among progressive MS patients, activated caspase-3 was detected in GSDMD immunopositive pyroptotic microglia/macrophages within demyelinating lesions. In the siRNA screen, suppression of caspase-3/7, caspase-1, or GSDMD expression prevented plasma membrane rupture during pyroptosis. Upon exposure to pyroptotic stimuli (ATP or nigericin), human microglia displayed caspase-3/7 activation and cleavage of caspase-3/7-specific substrates (e.g., DFF45, ROCK1, and PARP), with accompanying features of pyroptosis including GSDMD immunopositive pyroptotic bodies, IL-1β release, and membrane rupture. Pyroptosis-associated nuclear condensation and pyroptotic body formation were suppressed by caspase-3/7 inhibition. Pharmacological and siRNA-mediated inhibition of caspase-1 diminished caspase-3/7 activation during pyroptosis. In mice with EAE-associated neurological deficits, activated caspase-3 colocalized with GSDMD immunopositivity in lesion-associated macrophages/microglia. CONCLUSIONS Activation of executioner caspases-3/7, widely considered key mediators of apoptosis, contributed to GSDMD-associated microglial pyroptosis under neuroinflammatory conditions. Collectively, these observations highlight the convergence of different cell death pathways during neuroinflammation and offer new therapeutic opportunities in neuroinflammatory disease.
Collapse
Affiliation(s)
- Brienne A McKenzie
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Jason P Fernandes
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Matthew A L Doan
- Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Laura M Schmitt
- Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada.,Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | | | - Christopher Power
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada. .,Department of Medicine, University of Alberta, Edmonton, AB, Canada. .,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
23
|
Roles of Progesterone, Testosterone and Their Nuclear Receptors in Central Nervous System Myelination and Remyelination. Int J Mol Sci 2020; 21:ijms21093163. [PMID: 32365806 PMCID: PMC7246940 DOI: 10.3390/ijms21093163] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Progesterone and testosterone, beyond their roles as sex hormones, are neuroactive steroids, playing crucial regulatory functions within the nervous system. Among these, neuroprotection and myelin regeneration are important ones. The present review aims to discuss the stimulatory effects of progesterone and testosterone on the process of myelination and remyelination. These effects have been demonstrated in vitro (i.e., organotypic cultures) and in vivo (cuprizone- or lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE)). Both steroids stimulate myelin formation and regeneration by acting through their respective intracellular receptors: progesterone receptors (PR) and androgen receptors (AR). Activation of these receptors results in multiple events involving direct transcription and translation, regulating general homeostasis, cell proliferation, differentiation, growth and myelination. It also ameliorates immune response as seen in the EAE model, resulting in a significant decrease in inflammation leading to a fast recovery. Although natural progesterone and testosterone have a therapeutic potential, their synthetic derivatives—the 19-norprogesterone (nestorone) and 7α-methyl-nortestosterone (MENT), already used as hormonal contraception or in postmenopausal hormone replacement therapies, may offer enhanced benefits for myelin repair. We summarize here a recent advancement in the field of myelin biology, to treat demyelinating disorders using the natural as well as synthetic analogs of progesterone and testosterone.
Collapse
|
24
|
Extra-adrenal glucocorticoid biosynthesis: implications for autoimmune and inflammatory disorders. Genes Immun 2020; 21:150-168. [PMID: 32203088 PMCID: PMC7276297 DOI: 10.1038/s41435-020-0096-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Glucocorticoid synthesis is a complex, multistep process that starts with cholesterol being delivered to the inner membrane of mitochondria by StAR and StAR-related proteins. Here its side chain is cleaved by CYP11A1 producing pregnenolone. Pregnenolone is converted to cortisol by the enzymes 3-βHSD, CYP17A1, CYP21A2 and CYP11B1. Glucocorticoids play a critical role in the regulation of the immune system and exert their action through the glucocorticoid receptor (GR). Although corticosteroids are primarily produced in the adrenal gland, they can also be produced in a number of extra-adrenal tissue including the immune system, skin, brain, and intestine. Glucocorticoid production is regulated by ACTH, CRH, and cytokines such as IL-1, IL-6 and TNFα. The bioavailability of cortisol is also dependent on its interconversion to cortisone which is inactive, by 11βHSD1/2. Local and systemic glucocorticoid biosynthesis can be stimulated by ultraviolet B, explaining its immunosuppressive activity. In this review, we want to emphasize that dysregulation of extra-adrenal glucocorticoid production can play a key role in a variety of autoimmune diseases including multiple sclerosis (MS), lupus erythematosus (LE), rheumatoid arthritis (RA), and skin inflammatory disorders such as psoriasis and atopic dermatitis (AD). Further research on local glucocorticoid production and its bioavailability may open doors into new therapies for autoimmune diseases.
Collapse
|
25
|
Biliktu M, Senol SP, Temiz-Resitoglu M, Guden DS, Horat MF, Sahan-Firat S, Sevim S, Tunctan B. Pharmacological inhibition of soluble epoxide hydrolase attenuates chronic experimental autoimmune encephalomyelitis by modulating inflammatory and anti-inflammatory pathways in an inflammasome-dependent and -independent manner. Inflammopharmacology 2020; 28:1509-1524. [PMID: 32128702 DOI: 10.1007/s10787-020-00691-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
We aimed to determine the effect of soluble epoxide hydrolase (sEH) inhibition on chronic experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS), associated with changes in inflammasome-dependent and -independent inflammatory and anti-inflammatory pathways in the CNS of mice. C57BL/6 mice were used to induce chronic EAE by using an injection of MOG35-55 peptide/PT. Animals were observed daily and scored for EAE signs for 25 days after immunization. Following the induction of EAE, the scores were increased after 9 days and reached peak value as determined by ≥ 2 or ≤ 3 with 8% mortality rate on day 17. On day 17, mice were administered daily PBS, DMSO, or TPPU (a potent sEH inhibitor) (1, 3, or 10 mg/kg) until the end of the study. TPPU only at 3 mg/kg dose decreased the AUC values calculated from EAE scores obtained during the disease compared to EAE and vehicle control groups. On day 25, TPPU also caused an increase in the PPARα/β/γ and NLRC3 proteins and a decrease in the proteins of TLR4, MyD88, NF-κB p65, p-NF-κB p65, iNOS/nNOS, COX-2, NLRC4, ASC, caspase-1 p20, IL-1β, caspase-11 p20, NOX subunits (gp91phox and p47phox), and nitrotyrosine in addition to 14,15-DHET and IL-1β levels compared to EAE and vehicle control groups. Our findings suggest that pharmacological inhibition of sEH attenuates chronic EAE likely because of enhanced levels of anti-inflammatory EETs in addition to PPARα/β/γ and NLRC3 expression associated with suppressed inflammatory TLR4/MyD88/NF-κB signalling pathway, NLRC4/ASC/pro-caspase-1 inflammasome, caspase-11 inflammasome, and NOX activity that are responsible for inflammatory mediator formation in the CNS of mice.
Collapse
Affiliation(s)
- Merve Biliktu
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Sefika Pinar Senol
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Meryem Temiz-Resitoglu
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Mehmet Furkan Horat
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Serhan Sevim
- Department of Neurology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey.
| |
Collapse
|
26
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
27
|
Yilmaz C, Karali K, Fodelianaki G, Gravanis A, Chavakis T, Charalampopoulos I, Alexaki VI. Neurosteroids as regulators of neuroinflammation. Front Neuroendocrinol 2019; 55:100788. [PMID: 31513776 DOI: 10.1016/j.yfrne.2019.100788] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/12/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is a physiological protective response in the context of infection and injury. However, neuroinflammation, especially if chronic, may also drive neurodegeneration. Neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and traumatic brain injury (TBI), display inflammatory activation of microglia and astrocytes. Intriguingly, the central nervous system (CNS) is a highly steroidogenic environment synthesizing steroids de novo, as well as metabolizing steroids deriving from the circulation. Neurosteroid synthesis can be substantially affected by neuroinflammation, while, in turn, several steroids, such as 17β-estradiol, dehydroepiandrosterone (DHEA) and allopregnanolone, can regulate neuroinflammatory responses. Here, we review the role of neurosteroids in neuroinflammation in the context of MS, AD, PD and TBI and describe underlying molecular mechanisms. Moreover, we introduce the concept that synthetic neurosteroid analogues could be potentially utilized for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Canelif Yilmaz
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Kanelina Karali
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
28
|
De Kleijn KMA, Zuure WA, Peijnenborg J, Heuvelmans JM, Martens GJM. Reappraisal of Human HOG and MO3.13 Cell Lines as a Model to Study Oligodendrocyte Functioning. Cells 2019; 8:cells8091096. [PMID: 31533280 PMCID: PMC6769895 DOI: 10.3390/cells8091096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Myelination of neuronal axons is essential for proper brain functioning and requires mature myelinating oligodendrocytes (myOLs). The human OL cell lines HOG and MO3.13 have been widely used as in vitro models to study OL (dys) functioning. Here we applied a number of protocols aimed at differentiating HOG and MO3.13 cells into myOLs. However, none of the differentiation protocols led to increased expression of terminal OL differentiation or myelin-sheath formation markers. Surprisingly, the applied protocols did cause changes in the expression of markers for early OLs, neurons, astrocytes and Schwann cells. Furthermore, we noticed that mRNA expression levels in HOG and MO3.13 cells may be affected by the density of the cultured cells. Finally, HOG and MO3.13 co-cultured with human neuronal SH-SY5Y cells did not show myelin formation under several pro-OL-differentiation and pro-myelinating conditions. Together, our results illustrate the difficulty of inducing maturation of HOG and MO3.13 cells into myOLs, implying that these oligodendrocytic cell lines may not represent an appropriate model to study the (dys)functioning of human (my)OLs and OL-linked disease mechanisms.
Collapse
Affiliation(s)
- Kim M A De Kleijn
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, 6525AJ Nijmegen, The Netherlands.
- NeuroDrug Research, 6525 HP Nijmegen, The Netherlands.
| | - Wieteke A Zuure
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, 6525AJ Nijmegen, The Netherlands.
| | - Jolien Peijnenborg
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, 6525AJ Nijmegen, The Netherlands.
| | - Josje M Heuvelmans
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, 6525AJ Nijmegen, The Netherlands.
| | - Gerard J M Martens
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, 6525AJ Nijmegen, The Netherlands.
- NeuroDrug Research, 6525 HP Nijmegen, The Netherlands.
| |
Collapse
|
29
|
Absent in melanoma 2 regulates tumor cell proliferation in glioblastoma multiforme. J Neurooncol 2019; 144:265-273. [DOI: 10.1007/s11060-019-03230-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022]
|
30
|
Balcom EF, Doan MAL, Branton WG, Jovel J, Blevins G, Edguer B, Hobman TC, Yacyshyn E, Emery D, Box A, van Landeghem FKH, Power C. Human pegivirus-1 associated leukoencephalitis: Clinical and molecular features. Ann Neurol 2018; 84:781-787. [PMID: 30246885 DOI: 10.1002/ana.25343] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/14/2018] [Accepted: 09/10/2018] [Indexed: 01/23/2023]
Abstract
Etiologic diagnosis is uncertain in 35% to 50% of patients with encephalitis, despite its substantial global prevalence and disease burden. We report on 2 adult female patients with fatal leukoencephalitis associated with human pegivirus-1 (HPgV-1) brain infection. Neuroimaging showed inflammatory changes in cerebral white matter. Brain-derived HPgV-1 RNA sequences clustered phylogenetically with other pegiviruses despite an 87-nucleotide deletion in the viral nonstructural (NS)2 gene. Neuropathology disclosed lymphocyte infiltration and gliosis predominantly in brain white matter. HPgV-1 NS5A antigen was detected in lymphocytes as well as in astrocytes and oligodendrocytes. HPgV-1 neuroadaptation should be considered in the differential diagnosis of progressive leukoencephalitis in humans. Ann Neurol 2018;84:789-795.
Collapse
Affiliation(s)
- Erin F Balcom
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Matthew A L Doan
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | | | - Juan Jovel
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Gregg Blevins
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Beste Edguer
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Tom C Hobman
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Elaine Yacyshyn
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Derek Emery
- Department of Radiology & Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Adrian Box
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | - Frank K H van Landeghem
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada.,Department of Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Christopher Power
- Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
31
|
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
|
32
|
Powrie YSL, Smith C. Central intracrine DHEA synthesis in ageing-related neuroinflammation and neurodegeneration: therapeutic potential? J Neuroinflammation 2018; 15:289. [PMID: 30326923 PMCID: PMC6192186 DOI: 10.1186/s12974-018-1324-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023] Open
Abstract
It is a well-known fact that DHEA declines on ageing and that it is linked to ageing-related neurodegeneration, which is characterised by gradual cognitive decline. Although DHEA is also associated with inflammation in the periphery, the link between DHEA and neuroinflammation in this context is less clear. This review drew from different bodies of literature to provide a more comprehensive picture of peripheral vs central endocrine shifts with advanced age—specifically in terms of DHEA. From this, we have formulated the hypothesis that DHEA decline is also linked to neuroinflammation and that increased localised availability of DHEA may have both therapeutic and preventative benefit to limit neurodegeneration. We provide a comprehensive discussion of literature on the potential for extragonadal DHEA synthesis by neuroglial cells and reflect on the feasibility of therapeutic manipulation of localised, central DHEA synthesis.
Collapse
Affiliation(s)
- Y S L Powrie
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - C Smith
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
33
|
D'yakonov VA, Tuktarova RA, Dzhemileva LU, Ishmukhametova SR, Yunusbaeva MM, Dzhemilev UM. Catalytic cyclometallation in steroid chemistry V: Synthesis of hybrid molecules based on steroid oximes and (5Z,9Z)-tetradeca-5,9-dienedioic acid as potential anticancer agents. Steroids 2018; 138:14-20. [PMID: 29894723 DOI: 10.1016/j.steroids.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/31/2018] [Accepted: 06/02/2018] [Indexed: 12/17/2022]
Abstract
Synthetic analogues of natural 5Z,9Z-dienoic acids - hybrid molecules based on the oximes of cholesterol, pregnenolone, and androsterone with 1,14-tetradeca-5Z,9Z-dienedicarboxylic acid - were synthesized for the first time and studied for antitumor activity in vitro. The acid was prepared using catalytic cyclomagnesiation of O-containing 1,2-dienes with Grignard reagent in the presence of Cp2TiCl2 as the key step. Using flow cytometry, it was shown for the first time that the new molecules are efficient apoptosis inducers in the HeLa, Hek293, U937, Jurkat, and K562.
Collapse
Affiliation(s)
- Vladimir A D'yakonov
- Institute of Petrochemistry and Catalysis of RAS, 141 Prospekt Oktyabrya, 450075 Ufa, Russian Federation.
| | - Regina A Tuktarova
- Institute of Petrochemistry and Catalysis of RAS, 141 Prospekt Oktyabrya, 450075 Ufa, Russian Federation
| | - Lilya U Dzhemileva
- Institute of Petrochemistry and Catalysis of RAS, 141 Prospekt Oktyabrya, 450075 Ufa, Russian Federation.
| | - Svetlana R Ishmukhametova
- Institute of Petrochemistry and Catalysis of RAS, 141 Prospekt Oktyabrya, 450075 Ufa, Russian Federation
| | - Milyausha M Yunusbaeva
- Institute of Petrochemistry and Catalysis of RAS, 141 Prospekt Oktyabrya, 450075 Ufa, Russian Federation
| | - Usein M Dzhemilev
- Institute of Petrochemistry and Catalysis of RAS, 141 Prospekt Oktyabrya, 450075 Ufa, Russian Federation
| |
Collapse
|
34
|
Abstract
Glial cell types were classified less than 100 years ago by del Rio-Hortega. For instance, he correctly surmised that microglia in pathologic central nervous system (CNS) were "voracious monsters" that helped clean the tissue. Although these historical predictions were remarkably accurate, innovative technologies have revealed novel molecular, cellular, and dynamic physiologic aspects of CNS glia. In this review, we integrate recent findings regarding the roles of glia and glial interactions in healthy and injured spinal cord. The three major glial cell types are considered in healthy CNS and after spinal cord injury (SCI). Astrocytes, which in the healthy CNS regulate neurotransmitter and neurovascular dynamics, respond to SCI by becoming reactive and forming a glial scar that limits pathology and plasticity. Microglia, which in the healthy CNS scan for infection/damage, respond to SCI by promoting axon growth and remyelination-but also with hyperactivation and cytotoxic effects. Oligodendrocytes and their precursors, which in healthy tissue speed axon conduction and support axonal function, respond to SCI by differentiating and producing myelin, but are susceptible to death. Thus, post-SCI responses of each glial cell can simultaneously stimulate and stifle repair. Interestingly, potential therapies could also target interactions between these cells. Astrocyte-microglia cross-talk creates a feed-forward loop, so shifting the response of either cell could amplify repair. Astrocytes, microglia, and oligodendrocytes/precursors also influence post-SCI cell survival, differentiation, and remyelination, as well as axon sparing. Therefore, optimizing post-SCI responses of glial cells-and interactions between these CNS cells-could benefit neuroprotection, axon plasticity, and functional recovery.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO, 80309, USA.
- Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO, 80309, USA.
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
35
|
Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc Natl Acad Sci U S A 2018; 115:E6065-E6074. [PMID: 29895691 DOI: 10.1073/pnas.1722041115] [Citation(s) in RCA: 352] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS of unknown cause that remains incurable. Inflammasome-associated caspases mediate the maturation and release of the proinflammatory cytokines IL-1β and IL-18 and activate the pore-forming protein gasdermin D (GSDMD). Inflammatory programmed cell death, pyroptosis, was recently shown to be mediated by GSDMD. Here, we report molecular evidence for GSDMD-mediated inflammasome activation and pyroptosis in both myeloid cells (macrophages/microglia) and, unexpectedly, in myelin-forming oligodendrocytes (ODCs) in the CNS of patients with MS and in the MS animal model, experimental autoimmune encephalomyelitis (EAE). We observed inflammasome activation and pyroptosis in human microglia and ODCs in vitro after exposure to inflammatory stimuli and demonstrate caspase-1 inhibition by the small-molecule inhibitor VX-765 in both cell types. GSDMD inhibition by siRNA transduction suppressed pyroptosis in human microglia. VX-765 treatment of EAE animals reduced the expression of inflammasome- and pyroptosis-associated proteins in the CNS, prevented axonal injury, and improved neurobehavioral performance. Thus, GSDMD-mediated pyroptosis in select glia cells is a previously unrecognized mechanism of inflammatory demyelination and represents a unique therapeutic opportunity for mitigating the disease process in MS and other CNS inflammatory diseases.
Collapse
|
36
|
Lou ZY, Cheng J, Wang XR, Zhao YF, Gan J, Zhou GY, Liu ZG, Xiao BG. The inhibition of CB 1 receptor accelerates the onset and development of EAE possibly by regulating microglia/macrophages polarization. J Neuroimmunol 2018; 317:37-44. [PMID: 29501084 DOI: 10.1016/j.jneuroim.2018.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022]
Abstract
Cannabinoid 1 receptor (CB1R) regulates the neuro-inflammatory and neurodegenerative damages of experimental autoimmune encephalomyelitis (EAE) and of multiple sclerosis (MS). The mechanism by which CB1R inhibition exerts inflammatory effects is still unclear. Here, we explored the cellular and molecular mechanisms of CB1R in the treatment of EAE by using a specific and selective CB1R antagonist SR141716A. Our study demonstrated that SR141716A accelerated the clinical onset and development of EAE, accompanied by body weight loss. SR141716A significantly up-regulated the expression of toll like receptor-4 (TLR-4) and nuclear factor-kappaB/p65 (NF-κB/p65) on microglia/macrophages of EAE mice as well as levels of inflammatory factors (TNF-α, IL-1β, IL-6) and chemokines (MCP-1, CX3CL1), accompanied by the shifts of cytokines from Th2 (IL-4, IL-10) to Th1 (IFN-γ)/Th17 (IL-17) in the spinal cords of EAE mice. Similar changes happened on splenic mononuclear cells (MNCs) except chemokine CX3CL1. Consistently, SR141716A promoted BV-2 microglia to release inflammatory factors (TNF-α, IL-1β, IL-6) while inhibited the production of IL-10 and chemokines (MCP-1, CX3CL1). Furthermore, when splenic CD4+ T cells co-cultured with SR141716A-administered BV-2 microglia, the levels of IL-4 and IL-10 were decreased while production of IL-17 and IFN-γ increased significantly. Our research indicated that inhibition of CB1R induced M1 phenotype-Th17 axis changed of microglia/macrophages through TLR-4 and NF-κB/p65 which accelerated the onset and development of EAE. Therefore, CB1R may be a promising target for the treatment of MS/EAE, but its complexity remains to be carefully considered and studied in further clinical application.
Collapse
Affiliation(s)
- Zhi-Yin Lou
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jie Cheng
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao-Rong Wang
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yong-Fei Zhao
- Department of Neurology, JinShan Hospital, Fudan Univeristy, Shanghai, China
| | - Jing Gan
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guo-Yu Zhou
- Department of Geriatric, Qilu Hospital, Shandong University, Jinan, China
| | - Zhen-Guo Liu
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|