1
|
Demetriou A, Lindqvist B, Ali HG, Shamekh MM, Varshney M, Inzunza J, Maioli S, Nilsson P, Nalvarte I. ERβ mediates sex-specific protection in the App-NL-G-F mouse model of Alzheimer's disease. Biol Sex Differ 2025; 16:29. [PMID: 40302001 PMCID: PMC12039102 DOI: 10.1186/s13293-025-00711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Menopausal loss of neuroprotective estrogen is thought to contribute to the sex differences in Alzheimer's disease (AD). Activation of estrogen receptor beta (ERβ) can be clinically relevant since it avoids the adverse systemic effects of ERα activation. However, very few studies have explored ERβ-mediated neuroprotection in AD, and no information on its contribution to the sex differences in AD exists. In the present study, we specifically explored the role of ERβ in mediating sex-specific protection against AD pathology in the AppNL-G-F knock-in mouse model of amyloidosis, and if surgical menopause (ovariectomy) modulates pathology in this model. METHODS We treated male and female AppNL-G-F knock-in mice with the clinically relevant and selective ERβ agonist LY500307. A subset of the females was ovariectomized prior to treatment. Y-maze and contextual fear conditioning tests were used to assess memory performance, and biochemical assays such as qPCR, immunohistochemistry, Western blot, and multiplex immunoassays, were used to evaluate amyloid pathology. RESULTS We found that Female AppNL-G-F mice had higher soluble Aβ levels in cortex and hippocampus than males and more activated microglia. ERβ activation protected against amyloid pathology and cognitive decline in both male and female AppNL-G-F mice. Although ovariectomy increased soluble amyloid beta (Aβ) in cortex and insoluble Aβ in hippocampus, as well as sustained neuroinflammation after ERβ activation, it had otherwise limited effects on pathology. We further identified that ERβ did not alter APP processing, but rather exerted its protection at least partly via microglia activation in a sex-specific manner. CONCLUSION Combined, we provide new understanding to the sex differences in AD by demonstrating that ERβ protects against AD pathology differently in males and females, warranting reassessment of ERβ in combating AD.
Collapse
Affiliation(s)
- Aphrodite Demetriou
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
| | - Birgitta Lindqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Heba G Ali
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mohamed M Shamekh
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
| | - Ivan Nalvarte
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden.
| |
Collapse
|
2
|
Li L, Liu S, Wang M, Li M, Liu Y, Chen H, Chen J, Tao W, Huang L, Zhao S. Gen inhibiting the Wnt/Ca 2+ signaling pathway alleviates cerebral ischemia/reperfusion injury. Sci Rep 2025; 15:4661. [PMID: 39920331 PMCID: PMC11805899 DOI: 10.1038/s41598-025-88136-8] [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: 10/22/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
Cerebral ischemia/reperfusion injury (CIRI) is a major complication of acute ischemic stroke (AIS), characterized by calcium overload, oxidative stress, and cell apoptosis. In this study, we investigated the therapeutic potential of Genistein (Gen) in alleviating CIRI by focusing on its effects on the Wnt/Ca2+ signaling pathway. Using a rat model of cerebral ischemia/reperfusion and in vitro experiments on PC12 cells, we observed that Gen treatment reduced infarct size, improved neurological function, and mitigated calcium overload, oxidative stress, and apoptosis. Further analysis revealed that Gen regulates key proteins in the Wnt/Ca2+ signaling pathway, including Wnt5a and Frizzled-2, effectively preventing intracellular calcium accumulation and subsequent damage. The knockdown of Frizzled-2 confirmed the pathway's role in mediating calcium overload and subsequent damage. Our findings suggest that Gen alleviates CIRI by inhibiting the Wnt/Ca2+ signaling pathway, positioning it as a promising candidate for therapeutic intervention in stroke treatment.
Collapse
Affiliation(s)
- Li Li
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China
- Department of Pathology, Anqing 116 Hospital, Anqing, 246000, Anhui, China
| | - Saisai Liu
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China
- Department of Pathology, The First People's Hospital of Jiande, HangZhou, 310000, Zhejiang, China
| | - Mengzhe Wang
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Mengjia Li
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Yi Liu
- Clinical Medical School, Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Haili Chen
- Clinical Medical School, Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Jie Chen
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Weiting Tao
- Nanchang Health Vocational College, Nanchang, 330000, Jiangxi, China
| | - Li Huang
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China.
| | - Shidi Zhao
- Department of Pathophysiology, Bengbu Medical University, Bengbu, 233030, Anhui, China.
| |
Collapse
|
3
|
Ansari S, Khahpay R, Khakpai F, Heidarzadeh Z, Khojasteh SMB. Comparison of pain modulatory effect of the LPGi estragon receptor on inflammatory pain between pro-estrus and estrus phases and OVX rats. Psychopharmacology (Berl) 2025; 242:129-147. [PMID: 39180591 DOI: 10.1007/s00213-024-06653-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/12/2024] [Indexed: 08/26/2024]
Abstract
The present study has investigated whether circulating estrogen level variations in the pro-estrus and estrus phases of the intact rats and estrogen depletion in the ovariectomized animals (OVX) adjust the formalin-induced nociceptive behaviors. During the pro-estrus and estrus phases of rats' estrus cycle and in the OVX rats, 17β-estradiol and ICI 182,780 (estrogen receptor antagonist) were administered into the right paragigantocellularis lateralis (LPGi) nucleus. Then, the formalin-induced flexing and licking responses were recorded for 60 min. The findings of this study revealed that intra-LPGi administration of 17β-estradiol (0.8 μmol) reduced the formalin-induced flexing and licking duration in pro-estrus and estrus rats (P < 0.001), suggesting an analgesic effect. 17β-Estradiol injection into the LPGi nucleus of OVX rats increased the flexing duration (P < 0.05) while decreasing the licking duration (P < 0.05) of the formalin test. The pain modulatory effect of 17β-estradiol on the flexing response was reversed by ICI 182,780 (15 nmol) in the pro-estrus (P < 0.001) and estrus rats (P < 0.001) but not in the OVX rats. Also, pretreatment of LPGi nucleus with ICI 182,780 reversed the analgesic effect of 17β-estradiol on the licking response in the pro-estrus (P < 0.05), estrus (P < 0.001), and OVX rats (P < 0.001). These results suggest that the pain threshold in intact female rats is modulated independently of the estrus state. Still, the basal level of plasma estrogen and the activation of its receptors are necessary for pain modulation.
Collapse
Affiliation(s)
- Sanam Ansari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Roghaieh Khahpay
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Fatemeh Khakpai
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Heidarzadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
4
|
Agrimi J, Bernardele L, Sbaiti N, Brondi M, D’Angelo D, Canato M, Marchionni I, Oeing CU, Barbara G, Vignoli B, Canossa M, Kaludercic N, Spolverato G, Raffaello A, Lodovichi C, Maschio MD, Paolocci N. Reiterated male-to-female violence disrupts hippocampal estrogen receptor β expression, prompting anxiety-like behavior. iScience 2024; 27:110585. [PMID: 39228787 PMCID: PMC11369378 DOI: 10.1016/j.isci.2024.110585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/29/2024] [Accepted: 07/23/2024] [Indexed: 09/05/2024] Open
Abstract
Intimate partner violence (IPV) is a significant public health concern whose neurological/behavioral sequelae remain to be mechanistically explained. Using a mouse model recapitulating an IPV scenario, we evaluated the female brain neuroendocrine alterations produced by a reiterated male-to-female violent interaction (RMFVI). RMFVI prompted anxiety-like behavior in female mice whose hippocampus displayed a marked neuronal loss and hampered neurogenesis, namely reduced BrdU-DCX-positive nuclei and diminished dendritic arborization in the dentate gyrus (DG): effects paralleled by a substantial downregulation of the estrogen receptor β (ERβ). After RMFVI, the DG harbored reduced brain-derived neurotrophic factor (BDNF) pools and tyrosine kinase receptor B (TrkB) phosphorylation. Accordingly, ERβ knockout (KO) mice had heightened anxiety and curtailed BDNF levels at baseline while dying prematurely during the RMFVI procedure. Strikingly, injecting an ERβ antagonist or agonist into the wild-type (WT) female hippocampus enhanced or reduced anxiety, respectively. Thus, reiterated male-to-female violence jeopardizes hippocampal homeostasis, perturbing the ERβ/BDNF axis and ultimately instigating anxiety and chronic stress.
Collapse
Affiliation(s)
- Jacopo Agrimi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lucia Bernardele
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Naeem Sbaiti
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Donato D’Angelo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marta Canato
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ivan Marchionni
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Christian U. Oeing
- Department of Internal Medicine and Cardiology, Charité University Medicine, Berlin, Germany
| | - Giussy Barbara
- Service for Sexual and Domestic Violence, Fondazione IRCSS, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Beatrice Vignoli
- Department of Physics, University of Trento, 38123 Trento, Italy
| | - Marco Canossa
- Department of Cellular, Computational, and Integrative Biology, University of Trento, Trento, Italy
| | - Nina Kaludercic
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Gaya Spolverato
- Department of Surgical Oncological and Gastrointestinal Sciences, University of Padova, Padova, Italy
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Claudia Lodovichi
- Neuroscience Institute -CNR, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
- Padova Neuroscience Center, Padova, Italy
| | - Marco Dal Maschio
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Padova Neuroscience Center, Padova, Italy
| | - Nazareno Paolocci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Salinero AE, Abi-Ghanem C, Venkataganesh H, Sura A, Smith RM, Thrasher CA, Kelly RD, Hatcher KM, NyBlom V, Shamlian V, Kyaw NR, Belanger KM, Gannon OJ, Stephens SBZ, Zuloaga DG, Zuloaga KL. Treatment with brain specific estrogen prodrug ameliorates cognitive effects of surgical menopause in mice. Horm Behav 2024; 164:105594. [PMID: 38917776 PMCID: PMC11330726 DOI: 10.1016/j.yhbeh.2024.105594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Menopause is an endocrine shift leading to increased vulnerability for cognitive impairment and dementia risk factors, in part due to loss of neuroprotective circulating estrogens. Systemic replacement of estrogen post-menopause has limitations, including risk for estrogen-sensitive cancers. A promising therapeutic approach therefore might be to deliver estrogen only to the brain. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in ovariectomized mice (a surgical menopause model). We treated mice with the prodrug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17β-estradiol only in the brain. Young and middle-aged C57BL/6 J mice received ovariectomy and subcutaneous implant containing vehicle or DHED and underwent cognitive testing to assess memory after 1-3.5 months of treatment. Low and medium doses of DHED did not alter metabolic status in middle-aged mice. In both age groups, DHED treatment improved spatial memory in ovariectomized mice. Additional testing in middle-aged mice showed that DHED treatment improved working and recognition memory in ovariectomized mice. These results lay the foundation for future studies determining if this intervention is as efficacious in models of dementia with comorbid risk factors.
Collapse
Affiliation(s)
- Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Rachel M Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Christina A Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Katherine M Hatcher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Vanessa NyBlom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA; Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY 12222, USA
| | - Victoria Shamlian
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Kasey M Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Shannon B Z Stephens
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY 12222, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA.
| |
Collapse
|
6
|
Dai P, He J, Wei Y, Xu M, Zhao J, Zhou X, Tang H. High Dose of Estrogen Protects the Lungs from Ischemia-Reperfusion Injury by Downregulating the Angiotensin II Signaling Pathway. Inflammation 2024; 47:1248-1261. [PMID: 38386131 DOI: 10.1007/s10753-024-01973-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
We explored the sex difference in lung ischemia-reperfusion injury (LIRI) and the role and mechanism of estrogen (E2) and angiotensin II (Ang II) in LIRI. We established a model of LIRI in mice. E2, Ang II, E2 inhibitor (fulvestrant), and angiotensin II receptor blocker (losartan) were grouped for treatment. The lung wet/dry weight ratio, natural killer (NK) cells (by flow cytometry), neutrophils (by flow cytometry), expression of key proteins (by Western blot, immunohistochemistry, ELISA, and immunofluorescence), and expression of related protein mRNA (by qPCR) were detected. The ultrastructure of the alveolar epithelial cells was observed by transmission electron microscopy. We found that E2 and Ang II played an important role in the progression of LIRI. The two signaling pathways showed obvious antagonism, and E2 regulates LIRI in the different sexes by downregulating Ang II, leading to a better prognosis. E2 and losartan reduced the inflammatory cell infiltration in lung tissue and key inflammatory factors in serum while fulvestrant and Ang II had the opposite effect. The protective effect of E2 was related with AKT, p38, COX2, and HIF-1α.
Collapse
Affiliation(s)
- Peng Dai
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jutong He
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinping Zhao
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Xuefeng Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Demetriou A, Lindqvist B, Ali HG, Shamekh MM, Maioli S, Inzunza J, Varshney M, Nilsson P, Nalvarte I. ERβ mediates sex-specific protection in the App-NL-G-F mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604543. [PMID: 39091856 PMCID: PMC11291054 DOI: 10.1101/2024.07.22.604543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Menopausal loss of neuroprotective estrogen is thought to contribute to the sex differences in Alzheimer's disease (AD). Activation of estrogen receptor beta (ERβ) can be clinically relevant since it avoids the negative systemic effects of ERα activation. However, very few studies have explored ERβ-mediated neuroprotection in AD, and no information on its contribution to the sex differences in AD exists. In the present study we specifically explored the role of ERβ in mediating sex-specific protection against AD pathology in the clinically relevant App NL-G-F knock-in mouse model of amyloidosis, and if surgical menopause (ovariectomy) modulates pathology in this model. We treated male and female App NL-G-F mice with the selective ERβ agonist LY500307 and subset of the females was ovariectomized prior to treatment. Memory performance was assessed and a battery of biochemical assays were used to evaluate amyloid pathology and neuroinflammation. Primary microglial cultures from male and female wild-type and ERβ-knockout mice were used to assess ERβ's effect on microglial activation and phagocytosis. We find that ERβ activation protects against amyloid pathology and cognitive decline in male and female App NL-G-F mice. Ovariectomy increased soluble amyloid beta (Aβ) in cortex and insoluble Aβ in hippocampus, but had otherwise limited effects on pathology. We further identify that ERβ does not alter APP processing, but rather exerts its protection through amyloid scavenging that at least in part is mediated via microglia in a sex-specific manner. Combined, we provide new understanding to the sex differences in AD by demonstrating that ERβ protects against AD pathology differently in males and females, warranting reassessment of ERβ in combating AD.
Collapse
Affiliation(s)
- Aphrodite Demetriou
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Birgitta Lindqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Heba G. Ali
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Mohamed M. Shamekh
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| |
Collapse
|
8
|
Rishabh, Rohilla M, Bansal S, Bansal N, Chauhan S, Sharma S, Goyal N, Gupta S. Estrogen signalling and Alzheimer's disease: Decoding molecular mechanisms for therapeutic breakthrough. Eur J Neurosci 2024; 60:3466-3490. [PMID: 38726764 DOI: 10.1111/ejn.16360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 07/06/2024]
Abstract
In females, Alzheimer's disease (AD) incidences increases as compared to males due to estrogen deficiency after menopause. Estrogen therapy is the mainstay therapy for menopause and associated complications. Estrogen, a hormone with multifaceted physiological functions, has been implicated in AD pathophysiology. Estrogen plays a crucial role in amyloid precursor protein (APP) processing and overall neuronal health by regulating various factors such as brain-derived neurotrophic factor (BDNF), intracellular calcium signalling, death domain-associated protein (Daxx) translocation, glutamatergic excitotoxicity, Voltage-Dependent Anion Channel, Insulin-Like Growth Factor 1 Receptor, estrogen-metabolising enzymes and apolipoprotein E (ApoE) protein polymorphisms. All these factors impact the physiology of postmenopausal women. Estrogen replacement therapies play an important treatment strategy to prevent AD after menopause. However, use of these therapies may lead to increased risks of breast cancer, venous thromboembolism and cardiovascular disease. Various therapeutic approaches have been used to mitigate the effects of estrogen on AD. These include hormone replacement therapy, Selective Estrogen Receptor Modulators (SERMs), Estrogen Receptor Beta (ERβ)-Selective Agonists, Transdermal Estrogen Delivery, Localised Estrogen Delivery, Combination Therapies, Estrogen Metabolism Modulation and Alternative Estrogenic Compounds like genistein from soy, a notable phytoestrogen from plant sources. However, mechanism via which these approaches modulate AD in postmenopausal women has not been explained earlier thoroughly. Present review will enlighten all the molecular mechanisms of estrogen and estrogen replacement therapies in AD. Along-with this, the association between estrogen, estrogen-metabolising enzymes and ApoE protein polymorphisms will also be discussed in postmenopausal AD.
Collapse
Affiliation(s)
- Rishabh
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Manni Rohilla
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Seema Bansal
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Nitin Bansal
- Department of Pharmacy, Chaudhary Bansilal University, Bhiwani, India
| | - Samrat Chauhan
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sheenam Sharma
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Navjyoti Goyal
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Sumeet Gupta
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| |
Collapse
|
9
|
Chen C, Khanthiyong B, Thaweetee-Sukjai B, Charoenlappanit S, Roytrakul S, Thanoi S, Reynolds GP, Nudmamud-Thanoi S. Proteomic association with age-dependent sex differences in Wisconsin Card Sorting Test performance in healthy Thai subjects. Sci Rep 2023; 13:20238. [PMID: 37981639 PMCID: PMC10658079 DOI: 10.1038/s41598-023-46750-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/04/2023] [Indexed: 11/21/2023] Open
Abstract
Sex differences in cognitive function exist, but they are not stable and undergo dynamic change during the lifespan. However, our understanding of how sex-related neural information transmission evolves with age is still in its infancy. This study utilized the Wisconsin Card Sorting Test (WCST) and the label-free proteomics method with bioinformatic analysis to investigate the molecular mechanisms underlying age-related sex differences in cognitive performance in 199 healthy Thai subjects (aged 20-70 years), as well as explore the sex-dependent protein complexes for predicting cognitive aging. The results showed that males outperformed females in two of the five WCST sub-scores: %Corrects and %Errors. Sex differences in these scores were related to aging, becoming noticeable in those over 60. At the molecular level, differently expressed individual proteins and protein complexes between both sexes are associated with the potential N-methyl-D-aspartate type glutamate receptor (NMDAR)-mediated excitotoxicity, with the NMDAR complex being enriched exclusively in elderly female samples. These findings provided a preliminary indication that healthy Thai females might be more susceptible to such neurotoxicity, as evidenced by their cognitive performance. NMDAR protein complex enrichment in serum could be proposed as a potential indication for predicting cognitive aging in healthy Thai females.
Collapse
Affiliation(s)
- Chen Chen
- Medical Science Graduate Program, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | | | | | - Sawanya Charoenlappanit
- National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sittiruk Roytrakul
- National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Samur Thanoi
- School of Medical Sciences, University of Phayao, Phayao, Thailand.
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Sutisa Nudmamud-Thanoi
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand.
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand.
| |
Collapse
|
10
|
Salinero AE, Abi-Ghanem C, Venkataganesh H, Sura A, Smith RM, Thrasher CA, Kelly RD, Hatcher KM, NyBlom V, Shamlian V, Kyaw NR, Belanger KM, Gannon OJ, Stephens SB, Zuloaga DG, Zuloaga KL. Brain Specific Estrogen Ameliorates Cognitive Effects of Surgical Menopause in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552687. [PMID: 37609180 PMCID: PMC10441397 DOI: 10.1101/2023.08.09.552687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Menopause is a major endocrinological shift that leads to an increased vulnerability to the risk factors for cognitive impairment and dementia. This is thought to be due to the loss of circulating estrogens, which exert many potent neuroprotective effects in the brain. Systemic replacement of estrogen post-menopause has many limitations, including increased risk for estrogen-sensitive cancers. A more promising therapeutic approach therefore might be to deliver estrogen only to the brain thus limiting adverse peripheral side effects. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in post-menopausal mice. We modeled surgical menopause via bilateral ovariectomy (OVX). We treated mice with the pro-drug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17β-estradiol only in the brain. Young (2.5-month) and middle-aged (11-month-old) female C57BL/6J mice received ovariectomy and a subcutaneous implant containing vehicle (cholesterol) or DHED. At 3.5 months old (young group) and 14.5 months old (middle-aged group), mice underwent behavior testing to assess memory. DHED did not significantly alter metabolic status in middle-aged, post-menopausal mice. In both young and middle-aged mice, the brain-specific estrogen DHED improved spatial memory. Additional testing in middle-aged mice also showed that DHED improved working and recognition memory. These promising results lay the foundation for future studies aimed at determining if this intervention is as efficacious in models of dementia that have comorbid risk factors.
Collapse
Affiliation(s)
- Abigail E. Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Rachel M. Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Christina A. Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Richard D. Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Katherine M. Hatcher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Vanessa NyBlom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - Victoria Shamlian
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Kasey M. Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Olivia J. Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Shannon B.Z. Stephens
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Damian G. Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - Kristen L. Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| |
Collapse
|
11
|
Chen C, Khanthiyong B, Charoenlappanit S, Roytrakul S, Reynolds GP, Thanoi S, Nudmamud-Thanoi S. Cholinergic-estrogen interaction is associated with the effect of education on attenuating cognitive sex differences in a Thai healthy population. PLoS One 2023; 18:e0278080. [PMID: 37471329 PMCID: PMC10358962 DOI: 10.1371/journal.pone.0278080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/30/2023] [Indexed: 07/22/2023] Open
Abstract
The development of human brain is shaped by both genetic and environmental factors. Sex differences in cognitive function have been found in humans as a result of sexual dimorphism in neural information transmission. Numerous studies have reported the positive effects of education on cognitive functions. However, little work has investigated the effect of education on attenuating cognitive sex differences and the neural mechanisms behind it based on healthy population. In this study, the Wisconsin Card Sorting Test (WCST) was employed to examine sex differences in cognitive function in 135 Thai healthy subjects, and label-free quantitative proteomic method and bioinformatic analysis were used to study sex-specific neurotransmission-related protein expression profiles. The results showed sex differences in two WCST sub-scores: percentage of Total corrects and Total errors in the primary education group (Bayes factor>100) with males performed better, while such differences eliminated in secondary and tertiary education levels. Moreover, 11 differentially expressed proteins (DEPs) between men and women (FDR<0.1) were presented in both education groups, with majority of them upregulated in females. Half of those DEPs interacted directly with nAChR3, whereas the other DEPs were indirectly connected to the cholinergic pathways through interaction with estrogen. These findings provided a preliminary indication that a cholinergic-estrogen interaction relates to, and might underpin, the effect of education on attenuating cognitive sex differences in a Thai healthy population.
Collapse
Affiliation(s)
- Chen Chen
- Medical Science Graduate Program, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | | | - Sawanya Charoenlappanit
- Functional Proteomics Technology Laboratory, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Gavin P. Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Samur Thanoi
- School of Medical Sciences, University of Phayao, Mae Ka, Phayao, Thailand
| | - Sutisa Nudmamud-Thanoi
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
12
|
Ehtezazi T, Rahman K, Davies R, Leach AG. The Pathological Effects of Circulating Hydrophobic Bile Acids in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:173-211. [PMID: 36994114 PMCID: PMC10041467 DOI: 10.3233/adr-220071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Recent clinical studies have revealed that the serum levels of toxic hydrophobic bile acids (deoxy cholic acid, lithocholic acid [LCA], and glycoursodeoxycholic acid) are significantly higher in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) when compared to control subjects. The elevated serum bile acids may be the result of hepatic peroxisomal dysfunction. Circulating hydrophobic bile acids are able to disrupt the blood-brain barrier and promote the formation of amyloid-β plaques through enhancing the oxidation of docosahexaenoic acid. Hydrophobic bile acid may find their ways into the neurons via the apical sodium-dependent bile acid transporter. It has been shown that hydrophobic bile acids impose their pathological effects by activating farnesoid X receptor and suppressing bile acid synthesis in the brain, blocking NMDA receptors, lowering brain oxysterol levels, and interfering with 17β-estradiol actions such as LCA by binding to E2 receptors (molecular modelling data exclusive to this paper). Hydrophobic bile acids may interfere with the sonic hedgehog signaling through alteration of cell membrane rafts and reducing brain 24(S)-hydroxycholesterol. This article will 1) analyze the pathological roles of circulating hydrophobic bile acids in the brain, 2) propose therapeutic approaches, and 3) conclude that consideration be given to reducing/monitoring toxic bile acid levels in patients with AD or aMCI, prior/in combination with other treatments.
Collapse
Affiliation(s)
- Touraj Ehtezazi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rhys Davies
- The Walton Centre, NHS Foundation Trust, Liverpool, UK
| | - Andrew G Leach
- School of Pharmacy, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Mitra S, Dash R, Sohel M, Chowdhury A, Munni YA, Ali C, Hannan MA, Islam T, Moon IS. Targeting Estrogen Signaling in the Radiation-induced Neurodegeneration: A Possible Role of Phytoestrogens. Curr Neuropharmacol 2023; 21:353-379. [PMID: 35272592 PMCID: PMC10190149 DOI: 10.2174/1570159x20666220310115004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/01/2022] [Accepted: 03/06/2022] [Indexed: 11/22/2022] Open
Abstract
Radiation for medical use is a well-established therapeutic method with an excellent prognosis rate for various cancer treatments. Unfortunately, a high dose of radiation therapy comes with its own share of side effects, causing radiation-induced non-specific cellular toxicity; consequently, a large percentage of treated patients suffer from chronic effects during the treatment and even after the post-treatment. Accumulating data evidenced that radiation exposure to the brain can alter the diverse cognitive-related signaling and cause progressive neurodegeneration in patients because of elevated oxidative stress, neuroinflammation, and loss of neurogenesis. Epidemiological studies suggested the beneficial effect of hormonal therapy using estrogen in slowing down the progression of various neuropathologies. Despite its primary function as a sex hormone, estrogen is also renowned for its neuroprotective activity and could manage radiation-induced side effects as it regulates many hallmarks of neurodegenerations. Thus, treatment with estrogen and estrogen-like molecules or modulators, including phytoestrogens, might be a potential approach capable of neuroprotection in radiation-induced brain degeneration. This review summarized the molecular mechanisms of radiation effects and estrogen signaling in the manifestation of neurodegeneration and highlighted the current evidence on the phytoestrogen mediated protective effect against radiationinduced brain injury. This existing knowledge points towards a new area to expand to identify the possible alternative therapy that can be taken with radiation therapy as adjuvants to improve patients' quality of life with compromised cognitive function.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju38066, Republic of Korea
| | - Md. Sohel
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Apusi Chowdhury
- Department of Pharmaceutical Science, North-South University, Dhaka-12 29, Bangladesh
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju38066, Republic of Korea
| | - Chayan Ali
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE-751 08, Sweden
| | - Md. Abdul Hannan
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh-2202, Bangladesh
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering (IBGE), Bangabandhu Sheikh Mujibur Rahman Agricultural University (BSMRAU), Gazipur, Bangladesh
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju38066, Republic of Korea
| |
Collapse
|
14
|
Queathem ED, Fitzgerald M, Welly R, Rowles CC, Schaller K, Bukhary S, Baines CP, Rector RS, Padilla J, Manrique-Acevedo C, Lubahn DB, Vieira-Potter VJ. Suppression of estrogen receptor beta classical genomic activity enhances systemic and adipose-specific response to chronic beta-3 adrenergic receptor (β3AR) stimulation. Front Physiol 2022; 13:920675. [PMID: 36213237 PMCID: PMC9534559 DOI: 10.3389/fphys.2022.920675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
White adipose tissue (WAT) dysfunction independently predicts cardiometabolic disease, yet there is a lack of effective adipocyte-targeting therapeutics. B3AR agonists enhance adipocyte mitochondrial function and hold potential in this regard. Based on enhanced sensitivity to B3AR-mediated browning in estrogen receptor (ER)alpha-null mice, we hypothesized that ERβ may enhance the WAT response to the B3AR ligand, CL316,243 (CL). Methods: Male and female wild-type (WT) and ERβ DNA binding domain knock-out (ERβDBDKO) mice fed high-fat diet (HFD) to induce obesity were administered CL (1 mg/kg) daily for 2 weeks. Systemic physiological assessments of body composition (EchoMRI), bioenergetics (metabolic chambers), adipocyte mitochondrial respiration (oroboros) and glucose tolerance were performed, alongside perigonadal (PGAT), subcutaneous (SQAT) and brown adipose tissue (BAT) protein expression assessment (Western blot). Mechanisms were tested in vitro using primary adipocytes isolated from WT mice, and from Esr2-floxed mice in which ERβ was knocked down. Statistical analyses were performed using 2 × 2 analysis of variance (ANOVA) for main effects of genotype (G) and treatment (T), as well as GxT interactions; t-tests were used to determine differences between in vitro treatment conditions (SPSS V24). Results: There were no genotype differences in HFD-induced obesity or systemic rescue effects of CL, yet ERβDBDKO females were more sensitive to CL-induced increases in energy expenditure and WAT UCP1 induction (GxT, p < 0.05), which coincided with greater WAT B3AR protein content among the KO (G, p < 0.05). Among males, who were more insulin resistant to begin with (no genotype differences before treatment), tended to be more sensitive to CL-mediated reduction in insulin resistance. With sexes combined, basal WAT mitochondrial respiration trended toward being lower in the ERβDBDKO mice, but this was completely rescued by CL (p < 0.05). Confirming prior work, CL increased adipose tissue ERβ protein (T, p < 0.05, all), an effect that was enhanced in WAT and BAT the female KO (GxT, p < 0.01). In vitro experiments indicated that an inhibitor of ERβ genomic function (PHTPP) synergized with CL to further increase UCP1 mRNA (p = 0.043), whereas full ERβ protein was required for UCP1 expression (p = 0.042). Conclusion: Full ERβ activity appears requisite and stimulatory for UCP1 expression via a mechanism involving non-classical ERβ signaling. This novel discovery about the role of ERβ in adipocyte metabolism may have important clinical applications.
Collapse
Affiliation(s)
- Eric D. Queathem
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Maggie Fitzgerald
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Rebecca Welly
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Candace C. Rowles
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Kylie Schaller
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Shahad Bukhary
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Christopher P. Baines
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO, United States
- Research Service, Truman VA Memorial Hospital, Columbia, MO, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Camila Manrique-Acevedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri Columbia School of Medicine, Columbia, MO, United States
| | - Dennis B. Lubahn
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Victoria J. Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
15
|
Marchant IC, Chabert S, Martínez-Pinto J, Sotomayor-Zárate R, Ramírez-Barrantes R, Acevedo L, Córdova C, Olivero P. Estrogen, Cognitive Performance, and Functional Imaging Studies: What Are We Missing About Neuroprotection? Front Cell Neurosci 2022; 16:866122. [PMID: 35634466 PMCID: PMC9133497 DOI: 10.3389/fncel.2022.866122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 01/20/2023] Open
Abstract
Menopause transition can be interpreted as a vulnerable state characterized by estrogen deficiency with detrimental systemic effects as the low-grade chronic inflammation that appears with aging and partly explains age-related disorders as cancer, diabetes mellitus and increased risk of cognitive impairment. Over the course of a lifetime, estrogen produces several beneficial effects in healthy neurological tissues as well as cardioprotective effects, and anti-inflammatory effects. However, clinical evidence on the efficacy of hormone treatment in menopausal women has failed to confirm the benefit reported in observational studies. Unambiguously, enhanced verbal memory is the most robust finding from longitudinal and cross-sectional studies, what merits consideration for future studies aiming to determine estrogen neuroprotective efficacy. Estrogen related brain activity and functional connectivity remain, however, unexplored. In this context, the resting state paradigm may provide valuable information about reproductive aging and hormonal treatment effects, and their relationship with brain imaging of functional connectivity may be key to understand and anticipate estrogen cognitive protective effects. To go in-depth into the molecular and cellular mechanisms underlying rapid-to-long lasting protective effects of estrogen, we will provide a comprehensive review of cognitive tasks used in animal studies to evaluate the effect of hormone treatment on cognitive performance and discuss about the tasks best suited to the demonstration of clinically significant differences in cognitive performance to be applied in human studies. Eventually, we will focus on studies evaluating the DMN activity and responsiveness to pharmacological stimulation in humans.
Collapse
Affiliation(s)
- Ivanny Carolina Marchant
- Laboratorio de Modelamiento en Medicina, Escuela de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
- Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
- *Correspondence: Ivanny Carolina Marchant
| | - Stéren Chabert
- Millennium Nucleus in Cardiovascular Magnetic Resonance, Santiago, Chile
- Escuela de Ingeniería Biomédica, Universidad de Valparaiso, Valparaíso, Chile
- Centro de Investigación y Desarrollo en Ingeniería en Salud, Universidad de Valparaíso, Valparaíso, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa, Valparaíso, Chile
- Laboratorio de Neuroquímica y Neurofarmacología, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa, Valparaíso, Chile
- Laboratorio de Neuroquímica y Neurofarmacología, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | - Lilian Acevedo
- Servicio de Neurología Hospital Carlos van Buren, Valparaíso, Chile
| | - Claudio Córdova
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Olivero
- Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
- Pablo Olivero
| |
Collapse
|
16
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [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: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
17
|
Taylor E, Heyland A. Evolution of non-genomic nuclear receptor function. Mol Cell Endocrinol 2022; 539:111468. [PMID: 34610359 DOI: 10.1016/j.mce.2021.111468] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/01/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022]
Abstract
Nuclear receptors (NRs) are responsible for the regulation of diverse developmental and physiological systems in metazoans. NR actions can be the result of genomic and non-genomic mechanisms depending on whether they act inside or outside of the nucleus respectively. While the actions of both mechanisms have been shown to be crucial to NR functions, non-genomic actions are considered less frequently than genomic actions. Furthermore, hypotheses on the origin and evolution of non-genomic NR signaling pathways are rarely discussed in the literature. Here we summarize non-genomic NR signaling mechanisms in the context of NR protein family evolution and animal phyla. We find that NRs across groups and phyla act via calcium flux as well as protein phosphorylation cascades (MAPK/PI3K/PKC). We hypothesize and discuss a possible synapomorphy of NRs in the NR1 and NR3 families, including the thyroid hormone receptor, vitamin D receptor, ecdysone receptor, retinoic acid receptor, steroid receptors, and others. In conclusion, we propose that the advent of non-genomic NR signaling may have been a driving force behind the expansion of NR diversity in Cnidarians, Placozoans, and Bilaterians.
Collapse
Affiliation(s)
- Elias Taylor
- University of Guelph, College of Biological Sciences, Integrative Biology, Guelph, ON N1G-2W1, Canada.
| | - Andreas Heyland
- University of Guelph, College of Biological Sciences, Integrative Biology, Guelph, ON N1G-2W1, Canada.
| |
Collapse
|
18
|
Maioli S, Leander K, Nilsson P, Nalvarte I. Estrogen receptors and the aging brain. Essays Biochem 2021; 65:913-925. [PMID: 34623401 PMCID: PMC8628183 DOI: 10.1042/ebc20200162] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022]
Abstract
The female sex hormone estrogen has been ascribed potent neuroprotective properties. It signals by binding and activating estrogen receptors that, depending on receptor subtype and upstream or downstream effectors, can mediate gene transcription and rapid non-genomic actions. In this way, estrogen receptors in the brain participate in modulating neural differentiation, proliferation, neuroinflammation, cholesterol metabolism, synaptic plasticity, and behavior. Circulating sex hormones decrease in the course of aging, more rapidly at menopause in women, and slower in men. This review will discuss what this drop entails in terms of modulating neuroprotection and resilience in the aging brain downstream of spatiotemporal estrogen receptor alpha (ERα) and beta (ERβ) signaling, as well as in terms of the sex differences observed in Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, controversies related to ER expression in the brain will be discussed. Understanding the spatiotemporal signaling of sex hormones in the brain can lead to more personalized prevention strategies or therapies combating neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Maioli
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Karin Leander
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| |
Collapse
|
19
|
Willems S, Zaienne D, Merk D. Targeting Nuclear Receptors in Neurodegeneration and Neuroinflammation. J Med Chem 2021; 64:9592-9638. [PMID: 34251209 DOI: 10.1021/acs.jmedchem.1c00186] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors, also known as ligand-activated transcription factors, regulate gene expression upon ligand signals and present as attractive therapeutic targets especially in chronic diseases. Despite the therapeutic relevance of some nuclear receptors in various pathologies, their potential in neurodegeneration and neuroinflammation is insufficiently established. This perspective gathers preclinical and clinical data for a potential role of individual nuclear receptors as future targets in Alzheimer's disease, Parkinson's disease, and multiple sclerosis, and concomitantly evaluates the level of medicinal chemistry targeting these proteins. Considerable evidence suggests the high promise of ligand-activated transcription factors to counteract neurodegenerative diseases with a particularly high potential of several orphan nuclear receptors. However, potent tools are lacking for orphan receptors, and limited central nervous system exposure or insufficient selectivity also compromises the suitability of well-studied nuclear receptor ligands for functional studies. Medicinal chemistry efforts are needed to develop dedicated high-quality tool compounds for the therapeutic validation of nuclear receptors in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| |
Collapse
|
20
|
Gu Y, Chen X, Fu S, Liu W, Wang Q, Liu KJ, Shen J. Astragali Radix Isoflavones Synergistically Alleviate Cerebral Ischemia and Reperfusion Injury Via Activating Estrogen Receptor-PI3K-Akt Signaling Pathway. Front Pharmacol 2021; 12:533028. [PMID: 33692686 PMCID: PMC7937971 DOI: 10.3389/fphar.2021.533028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 01/15/2021] [Indexed: 11/13/2022] Open
Abstract
Isoflavones are major neuroprotective components of a medicinal herb Astragali Radix, against cerebral ischemia-reperfusion injury but the mechanisms of neuroprotection remain unclear. Calycosin and formononetin are two major AR isoflavones while daidzein is the metabolite of formononetin after absorption. Herein, we aim to investigate the synergistic neuroprotective effects of those isoflavones of Astragali Radix against cerebral ischemia-reperfusion injury. Calycosin, formononetin and daidzein were organized with different combinations whose effects observed in both in vitro and in vivo experimental models. In the in vitro study, primary cultured neurons were subjected to oxygen-glucose deprivation plus reoxygenation (OGD/RO) or l-glutamate treatment. In the in vivo study, rats were subjected to middle cerebral artery occlusion to induce cerebral ischemia and reperfusion. All three isoflavones pre-treatment alone decreased brain infarct volume and improved neurological deficits in rats, and dose-dependently attenuated neural death induced by l-glutamate treatment and OGD/RO in cultured neurons. Interestingly, the combined formulas of those isoflavones revealed synergistically activated estrogen receptor (estrogen receptors)-PI3K-Akt signaling pathway. Using ER antagonist and phosphatidylinositol 3-kinase (PI3K) inhibitor blocked the neuroprotective effects of those isoflavones. In conclusion, isoflavones could synergistically alleviate cerebral ischemia-reperfusion injury via activating ER-PI3K-Akt pathway.
Collapse
Affiliation(s)
- Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Chinese Medicine, Haikou, China.,School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Xi Chen
- Department of Core Facility, The People's Hospital of Bao-an Shenzhen, Shenzhen, China.,School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Shuping Fu
- School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Wenlan Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ke-Jian Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States
| | - Jiangang Shen
- School of Chinese Medicine, University of Hong Kong, Hong Kong, China.,The University of Hong Kong-Shenzhen, Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
21
|
Fels JA, Casalena GA, Manfredi G. Sex and oestrogen receptor β have modest effects on gene expression in the mouse brain posterior cortex. Endocrinol Diabetes Metab 2021; 4:e00191. [PMID: 33532622 PMCID: PMC7831211 DOI: 10.1002/edm2.191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/12/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Sex differences in brain cortical function affect cognition, behaviour and susceptibility to neural diseases, but the molecular basis of sexual dimorphism in cortical function is still largely unknown. Oestrogen and oestrogen receptors (ERs), specifically ERβ, the most abundant ER in the cortex, may play a role in determining sex differences in gene expression, which could underlie functional sex differences. However, further investigation is needed to address brain region specificity of the effects of sex and ERβ on gene expression. The goal of this study was to investigate sex differences in gene expression in the mouse posterior cortex, where sex differences in transcription have never been examined, and to determine how genetic ablation of ERβ affects transcription. Methods In this study, we performed unbiased transcriptomics on RNA from the posterior cortex of adult wild-type and ERβ knockout mice (n = 4/sex/genotype). We used unbiased clustering to analyse whole-transcriptome changes between the groups. We also performed differential expression analysis on the data using DESeq2 to identify specific changes in gene expression. Results We found only 27 significantly differentially expressed genes (DEGs) in wild-type (WT) males vs females, of which 17 were autosomal genes. Interestingly, in ERβKO males vs females all the autosomal DEGs were lost. Gene Ontology analysis of the subset of DEGs with sex differences only in the WT cortex revealed a significant enrichment of genes annotated with the function 'cation channel activity'. Moreover, within each sex we found only a few DEGs in ERβKO vs WT mice (8 and 5 in males and females, respectively). Conclusions Overall, our results suggest that in the adult mouse posterior cortex there are surprisingly few sex differences in gene expression, and those that exist are mainly related to cation channel activity. Additionally, they indicate that brain region-specific functional effects of ERβ may be largely post-transcriptional.
Collapse
Affiliation(s)
- Jasmine A. Fels
- Feil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | | | - Giovanni Manfredi
- Feil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
22
|
Kövesdi E, Szabó-Meleg E, Abrahám IM. The Role of Estradiol in Traumatic Brain Injury: Mechanism and Treatment Potential. Int J Mol Sci 2020; 22:E11. [PMID: 33374952 PMCID: PMC7792596 DOI: 10.3390/ijms22010011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023] Open
Abstract
Patients surviving traumatic brain injury (TBI) face numerous neurological and neuropsychological problems significantly affecting their quality of life. Extensive studies over the past decades have investigated pharmacological treatment options in different animal models, targeting various pathological consequences of TBI. Sex and gender are known to influence the outcome of TBI in animal models and in patients, respectively. Apart from its well-known effects on reproduction, 17β-estradiol (E2) has a neuroprotective role in brain injury. Hence, in this review, we focus on the effect of E2 in TBI in humans and animals. First, we discuss the clinical classification and pathomechanism of TBI, the research in animal models, and the neuroprotective role of E2. Based on the results of animal studies and clinical trials, we discuss possible E2 targets from early to late events in the pathomechanism of TBI, including neuroinflammation and possible disturbances of the endocrine system. Finally, the potential relevance of selective estrogenic compounds in the treatment of TBI will be discussed.
Collapse
Affiliation(s)
- Erzsébet Kövesdi
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Center for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pecs, Hungary;
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pecs, Hungary;
| | - István M. Abrahám
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Center for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pecs, Hungary;
| |
Collapse
|
23
|
Xiong YC, Chen T, Yang XB, Deng CL, Ning QL, Quan R, Yu XR. 17β-Oestradiol Attenuates the Photoreceptor Apoptosis in Mice with Retinitis Pigmentosa by Regulating N-myc Downstream Regulated Gene 2 Expression. Neuroscience 2020; 452:280-294. [PMID: 33246060 DOI: 10.1016/j.neuroscience.2020.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/23/2020] [Accepted: 11/08/2020] [Indexed: 10/22/2022]
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of retinal degenerative diseases in which the final pathological feature is photoreceptor cell apoptosis. Currently, the pathogenesis of RP remains poorly understood and therapeutics are ineffective. 17β-Oestradiol (βE2) is universally acknowledged as a neuroprotective factor in neurodegenerative diseases and has manifested neuroprotective effects in a light-induced retinal degeneration model. Recently, we identified N-myc downstream regulated gene 2 (NDRG2) suppression as a molecular marker of mouse retinal photoreceptor-specific cell death. βE2 has also been reported to regulate NDRG2 in salivary acinar cells. Therefore, in this study, we investigated whether βE2 plays a protective role in RP and regulates NDRG2 in photoreceptor cells. To this end, we generated RP models and observed that βE2 not only reduced the apoptosis of photoreceptor cells, but also restored the level of NDRG2 expression in RP models. Then, we showed that siNDRG2 inhibits the anti-apoptotic effect of βE2 on photoreceptor cells in a cellular RP model. Subsequently, we used a classic oestrogen receptor (ER) antagonist to attenuate the effects of βE2, suggesting that βE2 exerted its effects on RP models via the classic ERs. In addition, we performed a bioinformatics analysis, and the results indicated that the reported oestrogen response element (ERE) sequence is present in the promoter region of the mouse NDRG2 gene. Overall, our results suggest that βE2 attenuated the apoptosis of photoreceptor cells in RP models by maintaining NDRG2 expression via a classic ER-mediated mechanism.
Collapse
Affiliation(s)
- Ye-Cheng Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Tao Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiao-Bei Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chun-Lei Deng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qi-Lan Ning
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Rui Quan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiao-Rui Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
24
|
Wang YX, Xia ZH, Jiang X, Li LX, Wang HG, An D, Liu YQ. Genistein inhibits amyloid peptide 25-35-induced neuronal death by modulating estrogen receptors, choline acetyltransferase and glutamate receptors. Arch Biochem Biophys 2020; 693:108561. [PMID: 32857999 DOI: 10.1016/j.abb.2020.108561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/31/2020] [Accepted: 08/22/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE To explore genistein, the most active component of soy isoflavones, on viability, expression of estrogen receptor (ER) subtypes, choline acetyltransferase (ChAT), and glutamate receptor subunits in amyloid peptide 25-35-induced hippocampal neurons, providing valuable data and basic information for neuroprotective effect of genistein in Aβ25-35-induced neuronal injury. METHODS We established an in vitro model of Alzheimer's disease by exposing primary hippocampal neurons of newborn rats to amyloid peptide 25-35 (20 μM) for 24 h and observing the effects of genistein (10 μM, 3 h) on viability, expression of ER subtypes, ChAT, NMDA receptor subunit NR2B and AMPA receptor subunit GluR2 in Aβ25-35-induced hippocampal neurons. RESULTS We found that amyloid peptide 25-35 exposure reduced the viability of hippocampal neurons. Meanwhile, amyloid peptide 25-35 exposure decreased the expression of ER subtypes, ChAT and GluR2, and increased the expression of NR2B. Genistein at least partially reversed the effects of amyloid peptide 25-35 in hippocampal neurons. CONCLUSION Genistein could increase the expression of ChAT as a consequence of activating estrogen receptor subtypes, modulating the expression of NR2B and GluR2, and thereby ameliorating the status of hippocampal neurons and exerting neuroprotective effects against amyloid peptide 25-35. Our data suggest that genistein might represent a potential cell-targeted therapy which could be a promising approach to treating AD.
Collapse
Affiliation(s)
- Yu-Xiang Wang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China; Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Zhen-Hong Xia
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xue Jiang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Li-Xia Li
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hong-Gang Wang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Di An
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan-Qiang Liu
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
25
|
Ramírez-Barrantes R, Carvajal-Zamorano K, Rodriguez B, Cordova C, Lozano C, Simon F, Díaz P, Muñoz P, Marchant I, Latorre R, Castillo K, Olivero P. TRPV1-Estradiol Stereospecific Relationship Underlies Cell Survival in Oxidative Cell Death. Front Physiol 2020; 11:444. [PMID: 32528302 PMCID: PMC7265966 DOI: 10.3389/fphys.2020.00444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/09/2020] [Indexed: 12/31/2022] Open
Abstract
17β-estradiol is a neuronal survival factor against oxidative stress that triggers its protective effect even in the absence of classical estrogen receptors. The polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channel has been proposed as a steroid receptor implied in tissue protection against oxidative damage. We show here that TRPV1 is sufficient condition for 17β-estradiol to enhance metabolic performance in injured cells. Specifically, in TRPV1 expressing cells, the application of 17β-estradiol within the first 3 h avoided H2O2-dependent mitochondrial depolarization and the activation of caspase 3/7 protecting against the irreversible damage triggered by H2O2. Furthermore, 17β-estradiol potentiates TRPV1 single channel activity associated with an increased open probability. This effect was not observed after the application of 17α-estradiol. We explored the TRPV1-Estrogen relationship also in primary culture of hippocampal-derived neurons and observed that 17β-estradiol cell protection against H2O2-induced damage was independent of estrogen receptors pathway activation, membrane started and stereospecific. These results support the role of TRPV1 as a 17β-estradiol-activated ionotropic membrane receptor coupling with mitochondrial function and cell survival.
Collapse
Affiliation(s)
- Ricardo Ramírez-Barrantes
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Tecnología Médica, Universidad Andrés Bello, Viña del Mar, Chile
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Belen Rodriguez
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Cordova
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlo Lozano
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Simon
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
| | - Paula Díaz
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Muñoz
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Ivanny Marchant
- Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Olivero
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
26
|
Uddin MS, Rahman MM, Jakaria M, Rahman MS, Hossain MS, Islam A, Ahmed M, Mathew B, Omar UM, Barreto GE, Ashraf GM. Estrogen Signaling in Alzheimer's Disease: Molecular Insights and Therapeutic Targets for Alzheimer's Dementia. Mol Neurobiol 2020; 57:2654-2670. [PMID: 32297302 DOI: 10.1007/s12035-020-01911-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/23/2020] [Indexed: 01/04/2023]
Abstract
Estrogens play a crucial physiological function in the brain; however, debates exist concerning the role of estrogens in Alzheimer's disease (AD). Women during pre-, peri-, or menopause periods are more susceptible for developing AD, suggesting the connection of sex factors and a decreased estrogen signaling in AD pathogenesis. Yet, the underlying mechanism of estrogen-mediated neuroprotection is unclarified and is complicated by the existence of estrogen-related factors. Consequently, a deeper analysis of estrogen receptor (ER) expression and estrogen-metabolizing enzymes could interpret the importance of estrogen in age-linked cognitive alterations. Previous studies propose that hormone replacement therapy may attenuate AD onset in postmenopausal women, demonstrating that estrogen signaling is important for the development and progression of AD. For example, ERα exerts neuroprotection against AD by maintaining intracellular signaling cascades and study reported reduced expression of ERα in hippocampal neurons of AD patients. Similarly, reduced expression of ERβ in female AD patients has been associated with abnormal function in mitochondria and improved markers of oxidative stress. In this review, we discuss the critical interaction between estrogen signaling and AD. Moreover, we highlight the potential of targeting estrogen-related signaling for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | - Md Motiar Rahman
- Graduate School of Innovative Life Science, University of Toyama, Gofuku 3190, Toyama, 930-8555, Japan
| | - Md Jakaria
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Md Sohanur Rahman
- Graduate School of Innovative Life Science, University of Toyama, Gofuku 3190, Toyama, 930-8555, Japan
| | - Md Sarwar Hossain
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Ariful Islam
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Muniruddin Ahmed
- Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Ulfat Mohammed Omar
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
- Health Research Institute, University of Limerick, Limerick, Ireland.
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
27
|
Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer's Disease. Cell Mol Neurobiol 2020; 40:283-299. [PMID: 31502112 PMCID: PMC11448899 DOI: 10.1007/s10571-019-00733-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is described as cognitive and memory impairments with a sex-related epidemiological profile, affecting two times more women than men. There is emerging evidence that alternations in the hippocampal neurogenesis occur at the early stage of AD. Therapies that may effectively slow, stop, or regenerate the dying neurons in AD are being extensively investigated in the last few decades, but none has yet been found to be effective. The regulation of endogenous neurogenesis is one of the main therapeutic targets for AD. Mounting evidence indicates that the neurosteroid estradiol (17β-estradiol) plays a supporting role in neurogenesis, neuronal activity, and synaptic plasticity of AD. This effect may provide preventive and/or therapeutic approaches for AD. In this article, we discuss the molecular mechanism of potential estradiol modulatory action on endogenous neurogenesis, synaptic plasticity, and cognitive function in AD.
Collapse
Affiliation(s)
- Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Vahid Hajali
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Moradi
- Histology and Embryology Group, Basic Science Department, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, Münster, Germany.
| |
Collapse
|
28
|
Zhang W, Wu H, Xu Q, Chen S, Sun L, Jiao C, Wang L, Fu F, Feng Y, Qian X, Chen X. Estrogen modulation of pain perception with a novel 17β-estradiol pretreatment regime in ovariectomized rats. Biol Sex Differ 2020; 11:2. [PMID: 31918752 PMCID: PMC6953313 DOI: 10.1186/s13293-019-0271-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/21/2019] [Indexed: 11/10/2022] Open
Abstract
Estrogen plays substantial roles in pain modulation; however, studies concerning sex hormones and nociception often yield confusing results. The discrepancy could be a result of lack of consensus to regard estrogen as a variable when working with animal models; thus, the influence of hormones’ fluctuations on nociception has continually been neglected. In the present study, we designed a novel hormone substitution model to aid us to evaluate the effects of estrogen’s long-term alterations on ovariectomy (OVX)-induced mechanical hyperalgesia and the expression of estrogen receptors(ERs). OVX rats were implanted with slow-release estrogen pellets at differently arranged time points and doses, such that a gradual elevation or decrease of serum estrogen levels following a relatively stable period of estrogen replacement was achieved in rats. Our results demonstrated that gradual estrogen depletion rather than elevation following the stable period of estrogen substitution in OVX rats alleviated OVX-induced mechanical hyperalgesia in a dose-independent manner, and the opposite estrogen increase or decrease paradigms differently regulate the expression of spinal ERs. Specifically, in rats rendered to continuously increased serum estrogen, the early phase estrogen-induced anti-nociception effect in OVX rats was eliminated, which was accompanied by an over-activation of ERα and a strong depression of ERβ, while in the OVX rats subject to gradual decrease of estrogen replacement, both ERα and ERβ increased modestly compared with the OVX group. Thus, the present study demonstrated that estrogen increase or decrease modulate nociception differently through change of spinal ERs.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Qi Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Sheng Chen
- Zhejiang University School of Medicine, 866th Yuhangtang Road, Hangzhou, 310058, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Luyang Wang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Feng Fu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Ying Feng
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Xiaowei Qian
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China.
| |
Collapse
|
29
|
Finney CA, Shvetcov A, Westbrook RF, Jones NM, Morris MJ. The role of hippocampal estradiol in synaptic plasticity and memory: A systematic review. Front Neuroendocrinol 2020; 56:100818. [PMID: 31843506 DOI: 10.1016/j.yfrne.2019.100818] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/29/2019] [Accepted: 12/11/2019] [Indexed: 12/31/2022]
Abstract
The consolidation of long-term memory is influenced by various neuromodulators. One of these is estradiol, a steroid hormone that is synthesized both in peripheral endocrine tissue and in the brain, including the hippocampus. Here, we examine the evidence regarding the role of estradiol in the hippocampus, specifically, in memory formation and its effects on the molecular mechanisms underlying synaptic plasticity. We conclude that estradiol improves memory consolidation and, thereby, long-term memory. Previous studies have shown that it does this in three, interconnected ways: (1) via functional changes in excitatory activity, (2) signaling changes in calcium dynamics, protein phosphorylation and protein expression, and (3) structural changes to synaptic morphology. Through a functional network analysis of proteins affected by estradiol, we identify potential protein-protein interactions that further support a role for estradiol in modulating synaptic plasticity as well as highlight signaling pathways that may be involved in these changes within the hippocampus.
Collapse
Affiliation(s)
- C A Finney
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - A Shvetcov
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - R F Westbrook
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - N M Jones
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - M J Morris
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
30
|
Wang Y, Hernandez G, Mack WJ, Schneider LS, Yin F, Brinton RD. Retrospective analysis of phytoSERM for management of menopause-associated vasomotor symptoms and cognitive decline: a pilot study on pharmacogenomic effects of mitochondrial haplogroup and APOE genotype on therapeutic efficacy. Menopause 2020; 27:57-65. [PMID: 31567873 PMCID: PMC7100617 DOI: 10.1097/gme.0000000000001418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE PhytoSERM is a selective estrogen receptor beta (ERβ) modulator comprised of three phytoestrogens: genistein, daidzein, and S-equol. The PhytoSERM formulation promotes estrogenic action in the brain while largely inactive or inhibitory in reproductive tissue. A phase Ib/IIa clinical trial (ClinicalTrial.gov ID: NCT01723917) of PhytoSERM demonstrated safety and pharmacokinetics profile of PhytoSERM. While this study was not powered for efficacy analysis, we conducted a pilot, retrospective analysis to identify potential responders to PhytoSERM treatment, and to determine the optimal populations to pursue in a phase II clinical trial of efficacy of the PhytoSERM formulation. METHODS In this retrospective analysis involving 46 participants (n = 16, placebo; n = 18, 50 mg/d PhytoSERM; and n = 12, 100 mg/d PhytoSERM), the therapeutic effect of PhytoSERM was stratified by 2 genetic risk modulators for Alzheimer's disease: mitochondrial haplogroup and APOE genotype. RESULTS Our retrospective responder analysis indicated that participants on 50 mg of daily PhytoSERM (PS50) for 12 weeks significantly reduced hot flash frequency compared with their baseline (mean [95% CI])-1.61, [-2.79, -0.42], P = 0.007). Participants on 50 mg of PhytoSERM also had significantly greater reduction in hot flash frequency at 12 weeks compared with the placebo group (-1.38, -0.17 [median PS50, median placebo], P = 0.04). Fifty milligrams of daily PhytoSERM also preserved cognitive function in certain aspects of verbal learning and executive function. Our analysis further suggests that mitochondrial haplogroup and APOE genotype can modify PhytoSERM response. CONCLUSION Our data support a precision medicine approach for further development of PhytoSERM as a safe and effective alternative to hormone therapy for menopause-associated hot flash and cognitive decline. While definitive determination of PhytoSERM efficacy is limited by the small sample size, these data provide a reasonable rationale to extend analyses to a larger study set powered to address statistical significance.
Collapse
Affiliation(s)
- Yiwei Wang
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Gerson Hernandez
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Wendy J Mack
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Lon S Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Fei Yin
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Roberta D Brinton
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| |
Collapse
|
31
|
Azcoitia I, Barreto GE, Garcia-Segura LM. Molecular mechanisms and cellular events involved in the neuroprotective actions of estradiol. Analysis of sex differences. Front Neuroendocrinol 2019; 55:100787. [PMID: 31513774 DOI: 10.1016/j.yfrne.2019.100787] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/27/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022]
Abstract
Estradiol, either from peripheral or central origin, activates multiple molecular neuroprotective and neuroreparative responses that, being mediated by estrogen receptors or by estrogen receptor independent mechanisms, are initiated at the membrane, the cytoplasm or the cell nucleus of neural cells. Estrogen-dependent signaling regulates a variety of cellular events, such as intracellular Ca2+ levels, mitochondrial respiratory capacity, ATP production, mitochondrial membrane potential, autophagy and apoptosis. In turn, these molecular and cellular actions of estradiol are integrated by neurons and non-neuronal cells to generate different tissue protective responses, decreasing blood-brain barrier permeability, oxidative stress, neuroinflammation and excitotoxicity and promoting synaptic plasticity, axonal growth, neurogenesis, remyelination and neuroregeneration. Recent findings indicate that the neuroprotective and neuroreparative actions of estradiol are different in males and females and further research is necessary to fully elucidate the causes for this sex difference.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - George E Barreto
- Department of Biological Sciences, School of Natural Sciences, University of Limerick, Limerick, Ireland.
| | - Luis M Garcia-Segura
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain; Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain.
| |
Collapse
|
32
|
Zhang P, Fu Y, Ju J, Wan D, Su H, Wang Z, Rui H, Jin Q, Le Y, Hou R. Estradiol inhibits fMLP-induced neutrophil migration and superoxide production by upregulating MKP-2 and dephosphorylating ERK. Int Immunopharmacol 2019; 75:105787. [PMID: 31401382 DOI: 10.1016/j.intimp.2019.105787] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/20/2019] [Accepted: 07/25/2019] [Indexed: 01/03/2023]
Abstract
Estrogen has been reported to inhibit neutrophil infiltration related inflammation and suppress neutrophils migration in vitro, but the underlying mechanism is not fully understood. By using HL-60 differentiated neutrophil-like cells (dHL-60) and human neutrophils, we examined the effect of 17-β estradiol (E2) on cell migration and superoxide production in response to chemotactic peptide N-formyl-methionyl-leucyl-phenylalanine (fMLP) and explored the mechanisms involved. We found that fMLP significantly induced dHL-60 cell and neutrophil migration and superoxide production, which was inhibited by ERK inhibitor PD98059. E2 significantly inhibited fMLP-induced dHL-60 cell and neutrophil migration and superoxide production at both physiological and pharmacological concentrations. Mechanistic studies showed that pretreatment of these cells with E2 rapidly elevated the protein level of mitogen-activated protein kinase phosphatase 2 (MKP-2) and inhibited fMLP-induced ERK phosphorylation. Pretreatment of these cells with estrogen receptor (ER) antagonist ICI 182780 reversed the inhibition of fMP-induced cell migration and superoxide production, and the induction of MKP-2 expression and the suppression of fMP-induced ERK phosphorylation by E2. However, pretreatment of cells with G-protein coupled ER antagonist G15 had no such effect. Collectively, these results demonstrate that fMLP stimulates neutrophil chemotaxis and superoxide production through activating ERK, and indicate that ER-mediated upregulation of MKP-2 may dephosphorylate ERK and contribute to the inhibitory effect of E2 on neutrophil activation by fMLP. Our study reveals new mechanisms involved in the anti-inflammatory activity of estrogen.
Collapse
Affiliation(s)
- Ping Zhang
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Yi Fu
- Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215007, China
| | - Jihui Ju
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Dapeng Wan
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Hao Su
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Zhaodong Wang
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Huajuan Rui
- Department of Clinical Laboratory, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Qianheng Jin
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruixing Hou
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China; Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215100, China.
| |
Collapse
|
33
|
Kim KH, Cho Y, Lee J, Jeong H, Lee Y, Kim SI, Kim CH, Lee HW, Nam KT. Sexually dimorphic leanness and hypermobility in p16 Ink4a/CDKN2A-deficient mice coincides with phenotypic changes in the cerebellum. Sci Rep 2019; 9:11167. [PMID: 31371816 PMCID: PMC6671985 DOI: 10.1038/s41598-019-47676-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 07/22/2019] [Indexed: 12/31/2022] Open
Abstract
p16Ink4a/CDKN2A is a tumor suppressor that critically regulates the cell cycle. Indeed, p16Ink4a deficiency promotes tumor formation in various tissues. We now report that p16Ink4a deficiency in female mice, but not male mice, induces leanness especially in old age, as indicated by lower body weight and smaller white adipose tissue, although other major organs are unaffected. Unexpectedly, the integrity, number, and sizes of adipocytes in white adipose tissue were unaffected, as was macrophage infiltration. Hence, hypermobility appeared to be accountable for the phenotype, since food consumption was not altered. Histological analysis of the cerebellum and deep cerebellar nuclei, a vital sensorimotor control center, revealed increased proliferation of neuronal cells and improved cerebellum integrity. Expression of estrogen receptor β (ERβ) and PCNA also increased in deep cerebellar nuclei, implying crosstalk between p16Ink4a and ERβ. Furthermore, p16Ink4a deficiency expands LC3B+ cells and GFAP+ astrocytes in response to estrogen. Collectively, the data suggest that loss of p16INK4a induces sexually dimorphic leanness in female mice, which appears to be due to protection against cerebellar senescence by promoting neuronal proliferation and homeostasis via ERβ.
Collapse
Affiliation(s)
- Kwang H Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yejin Cho
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jaehoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center, Yonsei University, Seoul, 03722, Republic of Korea
| | - Haengdueng Jeong
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yura Lee
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Soo In Kim
- Department of Otorhinolaryngology, Korea Mouse Sensory Phenotyping Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chang-Hoon Kim
- Department of Otorhinolaryngology, Korea Mouse Sensory Phenotyping Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
34
|
Pharmacokinetics and safety profile of single-dose administration of an estrogen receptor β-selective phytoestrogenic (phytoSERM) formulation in perimenopausal and postmenopausal women. Menopause 2019; 25:191-196. [PMID: 28926513 DOI: 10.1097/gme.0000000000000984] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Selected estrogen receptor β-selective phytoestrogen (phytoSERM), a preparation of genistein, daidzein, and S-equol, has an 83-fold selective affinity for estrogen receptor (ER) β, and may promote neuronal survival and estrogenic mechanisms in the brain without exerting feminizing activity in the periphery. The aim of this study was to assess the safety, tolerability, and single-dose pharmacokinetics of the phytoSERM formulation in perimenopausal and postmenopausal women. METHODS Eighteen women aged 45 to 60 years from a 12-week clinical trial evaluating cognitive performance and vasomotor symptoms were randomly assigned to placebo, 50 mg, or 100 mg phytoSERM treatment groups. Plasma levels of the three parent phytoestrogens and their metabolites were measured before and at 2, 4, 6, 8, and 24 hours after ingestion by isotope dilution high-performance liquid chromatography (HPLC) electrospray ionization tandem mass spectrometry. RESULTS Plasma concentrations of genistein, daidzein, and S-equol peaked at 9, 6, and 4 hours, respectively, for the 50-mg dose, and at 6, 6, and 5 hours, respectively, for the 100-mg dose. The maximum concentration (Cmax) and area under the curve (AUC) for the three parent compounds were greater in the 100-mg dose group, indicating a dose-dependent change in concentration with the phytoSERM treatment. No adverse events were elicited. CONCLUSIONS A single-dose oral administration of the phytoSERM formulation was well-tolerated and did not elicit any adverse events. It was rapidly absorbed, reached high plasma concentrations, and showed a linear dose-concentration response in its pharmacokinetics. These findings are consistent with previously reported parameters for each parent compound (Clinicaltrials.gov NCT01723917).
Collapse
|
35
|
Mohajeri M, Martín-Jiménez C, Barreto GE, Sahebkar A. Effects of estrogens and androgens on mitochondria under normal and pathological conditions. Prog Neurobiol 2019; 176:54-72. [DOI: 10.1016/j.pneurobio.2019.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/23/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
|
36
|
Fels JA, Manfredi G. Sex Differences in Ischemia/Reperfusion Injury: The Role of Mitochondrial Permeability Transition. Neurochem Res 2019; 44:2336-2345. [PMID: 30863968 DOI: 10.1007/s11064-019-02769-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/23/2022]
Abstract
Brain and heart ischemia are among the leading causes of death and disability in both men and women, but there are significant sex differences in the incidence and severity of these diseases. Ca2+ dysregulation in response to ischemia/reperfusion injury (I/RI) is a well-recognized pathogenic mechanism leading to the death of affected cells. Excess intracellular Ca2+ causes mitochondrial matrix Ca2+ overload that can result in mitochondrial permeability transition (MPT), which can have severe consequences for mitochondrial function and trigger cell death. Recent findings indicate that estrogens and their related receptors are involved in the regulation of MPT, suggesting that sex differences in I/RI could be linked to estrogen-dependent modulation of mitochondrial Ca2+. Here, we review the evidence supporting sex differences in I/RI and the role of estrogen and estrogen receptors in producing these differences, the involvement of mitochondrial Ca2+ overload in disease pathogenesis, and the estrogen-dependent modulation of MPT that may contribute to sex differences.
Collapse
Affiliation(s)
- Jasmine A Fels
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st St., RR506, New York, NY, 10065, USA.,Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st St., RR506, New York, NY, 10065, USA.
| |
Collapse
|
37
|
Latent Sex Differences in Molecular Signaling That Underlies Excitatory Synaptic Potentiation in the Hippocampus. J Neurosci 2018; 39:1552-1565. [PMID: 30578341 DOI: 10.1523/jneurosci.1897-18.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/27/2018] [Accepted: 12/13/2018] [Indexed: 01/25/2023] Open
Abstract
Excitatory synapses can be potentiated by chemical neuromodulators, including 17β-estradiol (E2), or patterns of synaptic activation, as in long-term potentiation (LTP). Here, we investigated kinases and calcium sources required for acute E2-induced synaptic potentiation in the hippocampus of each sex and tested whether sex differences in kinase signaling extend to LTP. We recorded EPSCs from CA1 pyramidal cells in hippocampal slices from adult rats and used specific inhibitors of kinases and calcium sources. This revealed that, although E2 potentiates synapses to the same degree in each sex, cAMP-activated protein kinase (PKA) is required to initiate potentiation only in females. In contrast, mitogen-activated protein kinase, Src tyrosine kinase, and rho-associated kinase are required for initiation in both sexes; similarly, Ca2+/calmodulin-activated kinase II is required for expression/maintenance of E2-induced potentiation in both sexes. Calcium source experiments showed that L-type calcium channels and calcium release from internal stores are both required for E2-induced potentiation in females, whereas in males, either L-type calcium channel activation or calcium release from stores is sufficient to permit potentiation. To investigate the generalizability of a sex difference in the requirement for PKA in synaptic potentiation, we tested how PKA inhibition affects LTP. This showed that, although the magnitude of both high-frequency stimulation-induced and pairing-induced LTP is the same between sexes, PKA is required for LTP in females but not males. These results demonstrate latent sex differences in mechanisms of synaptic potentiation in which distinct molecular signaling converges to common functional endpoints in males and females.SIGNIFICANCE STATEMENT Chemical- and activity-dependent neuromodulation alters synaptic strength in both male and female brains, yet few studies have compared mechanisms of neuromodulation between the sexes. Here, we studied molecular signaling that underlies estrogen-induced and activity-dependent potentiation of excitatory synapses in the hippocampus. We found that, despite similar magnitude increases in synaptic strength in males and females, the roles of cAMP-regulated protein kinase, internal calcium stores, and L-type calcium channels differ between the sexes. Therefore, latent sex differences in which the same outcome is achieved through distinct underlying mechanisms in males and females include kinase and calcium signaling involved in synaptic potentiation, demonstrating that sex is an important factor in identification of molecular targets for therapeutic development based on mechanisms of neuromodulation.
Collapse
|
38
|
Speth RC, D'Ambra M, Ji H, Sandberg K. A heartfelt message, estrogen replacement therapy: use it or lose it. Am J Physiol Heart Circ Physiol 2018; 315:H1765-H1778. [PMID: 30216118 PMCID: PMC6336974 DOI: 10.1152/ajpheart.00041.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/24/2022]
Abstract
The issue of cardiovascular and cognitive health in women is complex. During the premenopausal phase of life, women have healthy blood pressure levels that are lower than those of age-matched men, and they have less cardiovascular disease. However, in the postmenopausal stage of life, blood pressure in women increases, and they are increasingly susceptible to cardiovascular disease, cognitive impairments, and dementia, exceeding the incidence in men. The major difference between pre- and postmenopausal women is the loss of estrogen. Thus, it seemed logical that postmenopausal estrogen replacement therapy, with or without progestin, generally referred to as menopausal hormone treatment (MHT), would prevent these adverse sequelae. However, despite initially promising results, a major randomized clinical trial refuted the benefits of MHT, leading to its falling from favor. However, reappraisal of this study in the framework of a "critical window," or "timing hypothesis," has changed our perspective on the benefit-to-risk ratio of MHT, and this review discusses the historical, current, and future approaches to MHT.
Collapse
Affiliation(s)
- Robert C Speth
- College of Pharmacy, Nova Southeastern University , Fort Lauderdale, Florida
- Department of Pharmacology and Physiology, College of Medicine, Georgetown University , Washington, District of Columbia
| | | | - Hong Ji
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University , Washington, District of Columbia
| | | |
Collapse
|
39
|
Mendell AL, MacLusky NJ. Neurosteroid Metabolites of Gonadal Steroid Hormones in Neuroprotection: Implications for Sex Differences in Neurodegenerative Disease. Front Mol Neurosci 2018; 11:359. [PMID: 30344476 PMCID: PMC6182082 DOI: 10.3389/fnmol.2018.00359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022] Open
Abstract
Gonadal steroid hormones are neurotrophic and neuroprotective. These effects are modulated by local metabolism of the hormones within the brain. Such control is necessary to maintain normal function, as several signaling pathways that are activated by gonadal steroid hormones in the brain can also become dysregulated in disease. Metabolites of the gonadal steroid hormones—particularly 3α-hydroxy, 5α-reduced neurosteroids—are synthesized in the brain and can act through different mechanisms from their parent steroids. These metabolites may provide a mechanism for modulating the responses to their precursor hormones, thereby providing a regulatory influence on cellular responses. In addition, there is evidence that the 3α-hydroxy, 5α-reduced neurosteroids are neuroprotective in their own right, and therefore may contribute to the overall protection conferred by their precursors. In this review article, the rapidly growing body of evidence supporting a neuroprotective role for this class of neurosteroids will be considered, including a discussion of potential mechanisms that may be involved. In addition, we explore the hypothesis that differences between males and females in local neurosteroid production may contribute to sex differences in the development of neurodegenerative disease.
Collapse
Affiliation(s)
- Ari Loren Mendell
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Neil James MacLusky
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
40
|
Min Z, Tang Y, Hu XT, Zhu BL, Ma YL, Zha JS, Deng XJ, Yan Z, Chen GJ. Cosmosiin Increases ADAM10 Expression via Mechanisms Involving 5'UTR and PI3K Signaling. Front Mol Neurosci 2018; 11:198. [PMID: 29942252 PMCID: PMC6004422 DOI: 10.3389/fnmol.2018.00198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
The α-secretase “a disintegrin and metalloproteinase domain-containing protein” (ADAM10) is involved in the processing of amyloid precursor protein (APP). Upregulation of ADAM10 precludes the generation of neurotoxic β-amyloid protein (Aβ) and represents a plausible therapeutic strategy for Alzheimer’s disease (AD). In this study, we explored compounds that can potentially promote the expression of ADAM10. Therefore, we performed high-throughput small-molecule screening in SH-SY5Y (human neuroblastoma) cells that stably express a luciferase reporter gene driven by the ADAM10 promoter, including a portion of its 5’-untranslated region (5’UTR). This has led to the discovery of cosmosiin (apigenin 7-O-β-glucoside). Here, we report that in human cell lines (SH-SY5Y and HEK293), cosmosiin proportionally increased the levels of the immature and mature forms of the ADAM10 protein without altering its mRNA level. This effect was attenuated by translation inhibitors or by deleting the 5’UTR of ADAM10, suggesting that a translational mechanism was responsible for the increased levels of ADAM10. Luciferase deletion assays revealed that the first 144 nucleotides of the 5’UTR were necessary for mediating the cosmosiin-induced enhancement of ADAM10 expression in SH-SY5Y cells. Cosmosiin failed to increase the levels of the ADAM10 protein in murine cells, which lack native expression of the ADAM10 transcript containing the identified 5’UTR element. The potential signaling pathway may involve phosphatidylinositide 3-kinase (PI3K) because pharmacological inhibition of PI3K attenuated the effect of cosmosiin on the expression of the ADAM10 protein. Finally, cosmosiin attenuated Aβ generation because the levels of Aβ40/42 in HEK-APP cells were significantly reduced after cosmosiin treatment. Collectively, we found that the first 144 nucleotides of the ADAM10 5’UTR, and PI3K signaling, are involved in cosmosiin-induced enhancement of the expression of ADAM10 protein. These results suggest that cosmosiin may be a potential therapeutic agent in the treatment of AD.
Collapse
Affiliation(s)
- Zhuo Min
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Ying Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Tong Hu
- Department of Neurology, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Bing-Lin Zhu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuan-Lin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Jing-Si Zha
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Juan Deng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, United States
| | - Guo-Jun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
41
|
Burstein SR, Kim HJ, Fels JA, Qian L, Zhang S, Zhou P, Starkov AA, Iadecola C, Manfredi G. Estrogen receptor beta modulates permeability transition in brain mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2018; 1859:423-433. [PMID: 29550215 PMCID: PMC5912174 DOI: 10.1016/j.bbabio.2018.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/08/2018] [Accepted: 03/12/2018] [Indexed: 12/24/2022]
Abstract
Recent evidence highlights a role for sex and hormonal status in regulating cellular responses to ischemic brain injury and neurodegeneration. A key pathological event in ischemic brain injury is the opening of a mitochondrial permeability transition pore (MPT) induced by excitotoxic calcium levels, which can trigger irreversible damage to mitochondria accompanied by the release of pro-apoptotic factors. However, sex differences in brain MPT modulation have not yet been explored. Here, we show that mitochondria isolated from female mouse forebrain have a lower calcium threshold for MPT than male mitochondria, and that this sex difference depends on the MPT regulator cyclophilin D (CypD). We also demonstrate that an estrogen receptor beta (ERβ) antagonist inhibits MPT and knockout of ERβ decreases the sensitivity of mitochondria to the CypD inhibitor, cyclosporine A. These results suggest a functional relationship between ERβ and CypD in modulating brain MPT. Moreover, co-immunoprecipitation studies identify several ERβ binding partners in mitochondria. Among these, we investigate the mitochondrial ATPase as a putative site of MPT regulation by ERβ. We find that previously described interaction between the oligomycin sensitivity-conferring subunit of ATPase (OSCP) and CypD is decreased by ERβ knockout, suggesting that ERβ modulates MPT by regulating CypD interaction with OSCP. Functionally, in primary neurons and hippocampal slice cultures, modulation of ERβ has protective effects against glutamate toxicity and oxygen glucose deprivation, respectively. Taken together, these results reveal a novel pathway of brain MPT regulation by ERβ that could contribute to sex differences in ischemic brain injury and neurodegeneration.
Collapse
Affiliation(s)
- Suzanne R Burstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Hyun Jeong Kim
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Jasmine A Fels
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Liping Qian
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Sheng Zhang
- Proteomics and Mass Spectrometry Facility, 139 Biotechnology Building, Cornell University, 526 Campus Road, Ithaca, NY 14853, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Anatoly A Starkov
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA.
| |
Collapse
|
42
|
Estrogen receptor β controls proliferation of enteric glia and differentiation of neurons in the myenteric plexus after damage. Proc Natl Acad Sci U S A 2018; 115:5798-5803. [PMID: 29760072 DOI: 10.1073/pnas.1720267115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Injury to the enteric nervous system (ENS) can cause several gastrointestinal (GI) disorders including achalasia, irritable bowel syndrome, and gastroparesis. Recently, a subpopulation of enteric glial cells with neuronal stem/progenitor properties (ENSCs) has been identified in the adult ENS. ENSCs have the ability of reconstituting the enteric neuronal pool after damage of the myenteric plexus. Since the estrogen receptor β (ERβ) is expressed in enteric glial cells and neurons, we investigated whether a selective ERβ agonist, LY3201, can influence neuronal and glial cell differentiation. Myenteric ganglia from the murine muscularis externa were isolated and cultured in either glial cell medium or neuronal medium. In glial cell medium, the number of glial progenitor cells (Sox10+) was increased by fourfold in the presence of LY3201. In the neuronal medium supplemented with an antimitotic agent to block glial cell proliferation, LY3201 elicited a 2.7-fold increase in the number of neurons (neurofilament+ or HuC/D+). In addition, the effect of LY3201 was evaluated in vivo in two murine models of enteric neuronal damage and loss, namely, high-fat diet and topical application of the cationic detergent benzalkonium chloride (BAC) on the intestinal serosa, respectively. In both models, treatment with LY3201 significantly increased the recovery of neurons after damage. Thus, LY3201 was able to stimulate glial-to-neuron cell differentiation in vitro and promoted neurogenesis in the damaged myenteric plexus in vivo. Overall, our study suggests that selective ERβ agonists may represent a therapeutic tool to treat patients suffering from GI disorders, caused by excessive neuronal/glial cell damage.
Collapse
|
43
|
Mendell AL, Chung BY, Creighton CE, Kalisch BE, Bailey CD, MacLusky NJ. Neurosteroid metabolites of testosterone and progesterone differentially inhibit ERK phosphorylation induced by amyloid β in SH-SY5Y cells and primary cortical neurons. Brain Res 2018; 1686:83-93. [DOI: 10.1016/j.brainres.2018.02.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/12/2017] [Accepted: 02/16/2018] [Indexed: 12/31/2022]
|
44
|
Wilkenfeld SR, Lin C, Frigo DE. Communication between genomic and non-genomic signaling events coordinate steroid hormone actions. Steroids 2018; 133:2-7. [PMID: 29155216 PMCID: PMC5864526 DOI: 10.1016/j.steroids.2017.11.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/07/2017] [Accepted: 11/10/2017] [Indexed: 01/30/2023]
Abstract
Steroid hormones are lipophilic molecules produced in one cell that can travel great distances within the body to elicit biological effects in another cell. In the canonical pathway, steroid hormone binding to a nuclear receptor (NR), often in the cytoplasm, causes the receptor to undergo a conformational change and translocate to the nucleus, where it interacts with specific sequences of DNA to regulate transcription. In addition to the classical genomic mechanism of action, alternate mechanisms of steroid activity have emerged that involve rapid, non-genomic signaling. The distinction between these two major mechanisms of action lies in the subcellular location of the initiating steroid hormone action. Importantly, the mechanisms of action are not exclusive, in that each can affect the activity of the other. Here, we describe the different types of genomic and non-genomic steroid hormone signaling mechanisms and how they can influence one another to ultimately regulate biology. Further, we discuss the approaches being used to study the non-genomic signaling events and address important caveats to be considered when designing new experiments. Thus, this minireview can serve as an introduction to the diverse signaling mechanisms of steroid hormones and offers initial, experimental guidance to those entering the field.
Collapse
Affiliation(s)
- Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Chenchu Lin
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Department of Biology and Biochemistry, University of Houston, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Molecular Medicine Program, The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
45
|
Céspedes Rubio ÁE, Pérez-Alvarez MJ, Lapuente Chala C, Wandosell F. Sex steroid hormones as neuroprotective elements in ischemia models. J Endocrinol 2018; 237:R65-R81. [PMID: 29654072 DOI: 10.1530/joe-18-0129] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022]
Abstract
Among sex steroid hormones, progesterone and estradiol have a wide diversity of physiological activities that target the nervous system. Not only are they carried by the blood stream, but also they are locally synthesized in the brain and for this reason, estradiol and progesterone are considered 'neurosteroids'. The physiological actions of both hormones range from brain development and neurotransmission to aging, illustrating the importance of a deep understanding of their mechanisms of action. In this review, we summarize key roles that estradiol and progesterone play in the brain. As numerous reports have confirmed a substantial neuroprotective role for estradiol in models of neurodegenerative disease, we focus this review on traumatic brain injury and stroke models. We describe updated data from receptor and signaling events triggered by both hormones, with an emphasis on the mechanisms that have been reported as 'rapid' or 'cytoplasmic actions'. Data showing the therapeutic effects of the hormones, used alone or in combination, are also summarized, with a focus on rodent models of middle cerebral artery occlusion (MCAO). Finally, we draw attention to evidence that neuroprotection by both hormones might be due to a combination of 'cytoplasmic' and 'nuclear' signaling.
Collapse
Affiliation(s)
- Ángel Enrique Céspedes Rubio
- Departamento de Sanidad AnimalGrupo de Investigación en Enfermedades Neurodegenerativas, Universidad del Tolima, Ibagué, Colombia
| | - Maria José Pérez-Alvarez
- Departamento de Biología (Fisiología Animal)Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Biología Molecular 'Severo Ochoa'Departamento de Neuropatología Molecular CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)Madrid, Spain
| | - Catalina Lapuente Chala
- Grupo de Investigación en Enfermedades NeurodegenerativasInvestigador Asociado Universidad del Tolima, Ibagué, Colombia
| | - Francisco Wandosell
- Centro de Biología Molecular 'Severo Ochoa'Departamento de Neuropatología Molecular CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)Madrid, Spain
| |
Collapse
|
46
|
Thakkar R, Sareddy GR, Zhang Q, Wang R, Vadlamudi RK, Brann D. PELP1: a key mediator of oestrogen signalling and actions in the brain. J Neuroendocrinol 2018; 30:10.1111/jne.12484. [PMID: 28485080 PMCID: PMC5785553 DOI: 10.1111/jne.12484] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 02/06/2023]
Abstract
Proline-, glutamic acid- and leucine-rich protein 1 (PELP1) is an oestrogen receptor (ER) coregulator protein identified by our collaborative group. Work from our laboratory and others has shown that PELP1 is a scaffold protein that interacts with ERs and kinase signalling factors, as well as proteins involved in chromatin remodelling and DNA repair. Its role in mediating 17β-oestradiol (E2 ) signalling and actions has been studied in detail in cancer cells, although only recently has attention turned to its role in the brain. In this review, we discuss the tissue, cellular and subcellular localisation of PELP1 in the brain. We also discuss recent evidence from PELP1 forebrain-specific knockout mice demonstrating a critical role of PELP1 in mediating both extranuclear and nuclear ER signalling in the brain, as well as E2 -induced neuroprotection, anti-inflammatory effects and regulation of cognitive function. Finally, the PELP1 interactome and unique gene network regulated by PELP1 in the brain is discussed, especially because it provides new insights into PELP1 biology, protein interactions and mechanisms of action in the brain. As a whole, the findings discussed in the present review indicate that PELP1 functions as a critical ER coregulator in the brain to mediate E2 signalling and actions.
Collapse
Affiliation(s)
- Roshni Thakkar
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Gangadhara Reddy Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Ruimin Wang
- Department of Neurobiology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX
| | - Darrell Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
- Corresponding Author: Dr. Darrell Brann, Regents’ Professor and Vice Chair, Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15 Street, CA-4004, Augusta, GA 30912, USA. Phone: 1-706-721-7779
| |
Collapse
|
47
|
Colón JM, González PA, Cajigas Á, Maldonado WI, Torrado AI, Santiago JM, Salgado IK, Miranda JD. Continuous tamoxifen delivery improves locomotor recovery 6h after spinal cord injury by neuronal and glial mechanisms in male rats. Exp Neurol 2018; 299:109-121. [PMID: 29037533 PMCID: PMC5723542 DOI: 10.1016/j.expneurol.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
No treatment is available for patients with spinal cord injury (SCI). Patients often arrive to the hospital hours after SCI suggesting the need of a therapy that can be used on a clinically relevant window. Previous studies showed that Tamoxifen (TAM) treatment 24h after SCI benefits locomotor recovery in female rats. Tamoxifen exerts beneficial effects in male and female rodents but a gap of knowledge exists on: the therapeutic window of TAM, the spatio-temporal mechanisms activated and if this response is sexually dimorphic. We hypothesized that TAM will favor locomotor recovery when administered up-to 24h after SCI in male Sprague-Dawley rats. Rats received a thoracic (T10) contusion using the MACSIS impactor followed by placebo or TAM (15mg/21days) pellets in a therapeutic window of 0, 6, 12, or 24h. Animals were sacrificed at 2, 7, 14, 28 or 35days post injury (DPI) to study the molecular and cellular changes in the acute and chronic stages. Immediate or delayed therapy (t=6h) improved locomotor function, increased white matter spared tissue, and neuronal survival. TAM reduced reactive gliosis during chronic stages and increased the expression of Olig-2. A significant difference was observed in estrogen receptor alpha between male and female rodents from 2 to 28 DPI suggesting a sexually dimorphic characteristic that could be related to the behavioral differences observed in the therapeutic window of TAM. This study supports the use of TAM in the SCI setting due to its neuroprotective effects but with a significant sexually dimorphic therapeutic window.
Collapse
Affiliation(s)
- Jennifer M Colón
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Pablo A González
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Ámbar Cajigas
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Wanda I Maldonado
- University of Puerto Rico Carolina Campus, Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, Carolina, PR 00984, USA.
| | - Aranza I Torrado
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - José M Santiago
- University of Puerto Rico Carolina Campus, Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, Carolina, PR 00984, USA.
| | - Iris K Salgado
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Jorge D Miranda
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| |
Collapse
|
48
|
Estrogen Receptor β Mediated Neuroprotective Efficacy of Cicer microphyllum Seed Extract in Global Hypoxia. Neurochem Res 2017; 42:3474-3489. [DOI: 10.1007/s11064-017-2395-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/17/2017] [Accepted: 08/23/2017] [Indexed: 10/19/2022]
|
49
|
Prakapenka AV, Bimonte-Nelson HA, Sirianni RW. Engineering poly(lactic-co-glycolic acid) (PLGA) micro- and nano-carriers for Controlled Delivery of 17β-Estradiol. Ann Biomed Eng 2017; 45:1697-1709. [PMID: 28634732 PMCID: PMC5599155 DOI: 10.1007/s10439-017-1859-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/23/2017] [Indexed: 12/29/2022]
Abstract
With menopause, circulating levels of 17β-estradiol (E2) markedly decrease. E2-based hormone therapy is prescribed to alleviate symptoms associated with menopause. E2 is also recognized for its beneficial effects in the central nervous system (CNS), such as enhanced cognitive function following abrupt hormonal loss associated with ovariectomy. For women with an intact uterus, an opposing progestogen component is required to decrease the risk of developing endometrial hyperplasia. While adding an opposing progestogen attenuates these detrimental effects on the uterus, it can attenuate the beneficial effects of E2 in the CNS. Poly(lactic-co-glycolic acid) (PLGA) micro- and nano- carriers (MNCs) have been heavily investigated for their ability to enhance the therapeutic activity of hydrophobic agents following exogenous administration, including E2. Multiple PLGA MNC formulation parameters, such as composition, molecular weight, and type of solvent used, can be altered to systematically manipulate the pharmacokinetic and pharmacodynamic profiles of encapsulated agents. Thus, there is an opportunity to enhance the therapeutic activity of E2 in the CNS through controlled delivery from PLGA MNCs. The aim of this review is to consider the fate of exogenously administered E2 and discuss how PLGA MNCs and route of administration can be used as strategies for controlled E2 delivery.
Collapse
Affiliation(s)
- Alesia V Prakapenka
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Rd., Phoenix, AZ, 85013, USA
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Rachael W Sirianni
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Rd., Phoenix, AZ, 85013, USA.
| |
Collapse
|
50
|
Jean A, Trouillet AC, Andrianarivelo NA, Mhaouty-Kodja S, Hardin-Pouzet H. Phospho-ERK and sex steroids in the mPOA: involvement in male mouse sexual behaviour. J Endocrinol 2017; 233:257-267. [PMID: 28356400 DOI: 10.1530/joe-17-0025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 12/20/2022]
Abstract
This paper aimed to investigate the mechanisms triggering ERK phosphorylation and its functional role in male sexual behaviour. ERK1/2-phosphorylated form was detected in the medial preoptic area of the hypothalamus (mPOA) during the sexual stimulation of naive and sexually experienced males who were killed 5 min after the first intromission. This mating-induced ERK phosphorylation was increased in sexually experienced males compared to that in naive mice. The functional role of the ERK1/2 pathway activation during sexual behaviour was explored with the administration of a MEK inhibitor, SL-327 (30 mg/kg, i.p.), 45 min before the contact with a receptive female. Inhibition of ERK phosphorylation was found to decrease sexual motivation in both naive and experienced males without altering their copulatory ability. The mechanisms potentially involved in this rapid ERK1/2 pathway activation were specified ex vivo on hypothalamic slices. A thirty-minute incubation with 100 nM of testosterone (T), dihydrotestosterone (DHT) or oestradiol (E2) led to ERK phosphorylation. No changes were observed after incubation with testosterone 3-(O-carboxymethyl)oxime-BSA (T-BSA), an impermeable to the plasma membrane form of testosterone. All these results indicate that ERK phosphorylation within the mPOA could be a key player in the motivational signalling pathway and considered as an index of sexual motivation. They also demonstrate the involvement of oestrogen receptor (ER) and androgen receptor (AR) transduction pathways in steroid-dependent ERK activation.
Collapse
Affiliation(s)
- Arnaud Jean
- Sorbonne UniversitésUPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris - Seine; Institut de Biologie Paris Seine, Paris, France
| | - Anne-Charlotte Trouillet
- Sorbonne UniversitésUPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris - Seine; Institut de Biologie Paris Seine, Paris, France
| | - Njiva Andry Andrianarivelo
- Sorbonne UniversitésUPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris - Seine; Institut de Biologie Paris Seine, Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne UniversitésUPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris - Seine; Institut de Biologie Paris Seine, Paris, France
| | - Hélène Hardin-Pouzet
- Sorbonne UniversitésUPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris - Seine; Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|