51
|
Abstract
Wnt signaling has emerged in recent years as a major player in both nervous system development and adult synaptic plasticity. Of particular relevance to researchers studying learning and memory, Wnt signaling is critical for normal functioning of the hippocampus, a brain region that is essential for many types of memory formation and whose dysfunction is implicated in numerous neurodegenerative and psychiatric conditions. Impaired hippocampal Wnt signaling is implicated in several of these conditions, however, little is known about how Wnt signaling mediates hippocampal memory formation. This review will provide a general overview of Wnt signaling and discuss evidence demonstrating a key role for Wnt signaling in hippocampal memory formation in both normal and disease states. The regulation of Wnt signaling by ovarian sex steroid hormones will also be highlighted, given that the neuroprotection afforded by Wnt-hormone interactions may have significant implications for cognitive function in aging, neurodegenerative disease, and ischemic injury.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, WI, USA
| |
Collapse
|
52
|
Zhang L, Xiong W, Xiong Y, Liu H, Liu Y. 17 β-Estradiol promotes vascular endothelial growth factor expression via the Wnt/β-catenin pathway during the pathogenesis of endometriosis. Mol Hum Reprod 2016; 22:526-35. [DOI: 10.1093/molehr/gaw025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
|
53
|
Sabogal-Guáqueta AM, Osorio E, Cardona-Gómez GP. Linalool reverses neuropathological and behavioral impairments in old triple transgenic Alzheimer's mice. Neuropharmacology 2016; 102:111-20. [PMID: 26549854 PMCID: PMC4698173 DOI: 10.1016/j.neuropharm.2015.11.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 10/16/2015] [Accepted: 11/02/2015] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is an age-related progressive neurodegenerative disorder. Several types of treatments have been tested to block or delay the onset of the disease, but none have been completely successful. Diet, lifestyle and natural products are currently the main scientific focuses. Here, we evaluate the effects of oral administration of the monoterpene linalool (25 mg/kg), every 48 h for 3 months, on aged (21-24 months old) mice with a triple transgenic model of AD (3xTg-AD) mice. Linalool-treated 3xTg-AD mice showed improved learning and spatial memory and greater risk assessment behavior during the elevated plus maze. Hippocampi and amygdalae from linalool-treated 3xTg-AD mice exhibited a significant reduction in extracellular β-amyloidosis, tauopathy, astrogliosis and microgliosis as well as a significant reduction in the levels of the pro-inflammatory markers p38 MAPK, NOS2, COX2 and IL-1β. Together, our findings suggest that linalool reverses the histopathological hallmarks of AD and restores cognitive and emotional functions via an anti-inflammatory effect. Thus, linalool may be an AD prevention candidate for preclinical studies.
Collapse
Affiliation(s)
- Angélica Maria Sabogal-Guáqueta
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area - School of Medicine, SIU, University of Antioquia UdeA, Calle 70 No, 52-21, Medellin, Colombia
| | - Edison Osorio
- Grupo de Investigación en Sustancias Bioactivas, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquía UdeA, Calle 70 No, 52-21, Medellin, Colombia
| | - Gloria Patricia Cardona-Gómez
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area - School of Medicine, SIU, University of Antioquia UdeA, Calle 70 No, 52-21, Medellin, Colombia.
| |
Collapse
|
54
|
Piedrahita D, Castro-Alvarez JF, Boudreau RL, Villegas-Lanau A, Kosik KS, Gallego-Gomez JC, Cardona-Gómez GP. β-Secretase 1's Targeting Reduces Hyperphosphorilated Tau, Implying Autophagy Actors in 3xTg-AD Mice. Front Cell Neurosci 2016; 9:498. [PMID: 26778963 PMCID: PMC4705306 DOI: 10.3389/fncel.2015.00498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/11/2015] [Indexed: 01/08/2023] Open
Abstract
β-site APP cleaving enzyme 1 (BACE1) initiates APP cleavage, which has been reported to be an inducer of tau pathology by altering proteasome functions in Alzheimer’s disease (AD). However, the exact relationship between BACE1 and PHF (Paired Helical Filaments) formation is not clear. In this study, we confirm that BACE1 and Hsc70 are upregulated in the brains of AD patients, and we demonstrate that both proteins show enhanced expression in lipid rafts from AD-affected triple transgenic mouse brains. BACE1 targeting increased Hsc70 levels in the membrane and cytoplasm fractions and downregulated Hsp90 and CHIP in the nucleus in the hippocampi of 3xTg-AD mice. However, these observations occurred in a proteasome-independent manner in vitro. The BACE1miR-induced reduction of soluble hyperphosphorylated tau was associated with a decrease in MAPK activity. However, the BACE1 RNAi-mediated reduction of hyperphosphorylated tau was only blocked by 3-MA (3-methyladenine) in vitro, and it resulted in the increase of Hsc70 and LAMP2 in lipid rafts from hippocampi of 3xTg-AD mice, and upregulation of survival and homeostasis signaling. In summary, our findings suggest that BACE1 silencing neuroprotects reducing soluble hyperphosphorylated tau, modulating certain autophagy-related proteins in aged 3xTg-AD mice.
Collapse
Affiliation(s)
- Diego Piedrahita
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| | - John Fredy Castro-Alvarez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| | | | - Andres Villegas-Lanau
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia Medellín, Colombia
| | - Kenneth S Kosik
- Department of Molecular Cellular Developmental Biology, Neuroscience Research Institute, University of California Santa Barbara Santa Barbara, CA, USA
| | - Juan Carlos Gallego-Gomez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| | - Gloria Patricia Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| |
Collapse
|
55
|
Proline-, glutamic acid-, and leucine-rich protein 1 mediates estrogen rapid signaling and neuroprotection in the brain. Proc Natl Acad Sci U S A 2015; 112:E6673-82. [PMID: 26627258 DOI: 10.1073/pnas.1516729112] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
17-β estradiol (E2) has been implicated as neuroprotective in a variety of neurodegenerative disorders. However, the underlying mechanism remains unknown. Here, we provide genetic evidence, using forebrain-specific knockout (FBKO) mice, that proline-, glutamic acid-, and leucine-rich protein 1 (PELP1), an estrogen receptor coregulator protein, is essential for the extranuclear signaling and neuroprotective actions of E2 in the hippocampal CA1 region after global cerebral ischemia (GCI). E2-mediated extranuclear signaling (including activation of extracellular signal-regulated kinase and Akt) and antiapoptotic effects [such as attenuation of JNK signaling and increase in phosphorylation of glycogen synthase kinase-3β (GSK3β)] after GCI were compromised in PELP1 FBKO mice. Mechanistic studies revealed that PELP1 interacts with GSK3β, E2 modulates interaction of PELP1 with GSK3β, and PELP1 is a novel substrate for GSK3β. RNA-seq analysis of control and PELP1 FBKO mice after ischemia demonstrated alterations in several genes related to inflammation, metabolism, and survival in PELP1 FBKO mice, as well as a significant reduction in the activation of the Wnt/β-catenin signaling pathway. In addition, PELP1 FBKO studies revealed that PELP1 is required for E2-mediated neuroprotection and for E2-mediated preservation of cognitive function after GCI. Collectively, our data provide the first direct in vivo evidence, to our knowledge, of an essential role for PELP1 in E2-mediated rapid extranuclear signaling, neuroprotection, and cognitive function in the brain.
Collapse
|
56
|
Xiong W, Zhang L, Yu L, Xie W, Man Y, Xiong Y, Liu H, Liu Y. Estradiol promotes cells invasion by activating β-catenin signaling pathway in endometriosis. Reproduction 2015; 150:507-16. [PMID: 26432349 PMCID: PMC4633770 DOI: 10.1530/rep-15-0371] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 10/02/2015] [Indexed: 12/29/2022]
Abstract
Endometriosis is an estrogen-dependent disease that involves the adhesion, invasion, and angiogenesis of endometrial tissues outside of the uterine cavity. We hypothesized that a link exists between estrogen and beta-catenin (β-catenin) signaling in the pathogenesis of endometriosis. Human endometrial stromal cells (HESCs) were separated from eutopic endometrial tissues that were obtained from patients with endometriosis. β-catenin expression and cells invasiveness ability were up-regulated by 17β-estradiol (E2) in an estrogen receptor (ESR)-dependent manner, whereas β-catenin siRNA abrogated this phenomenon. Moreover, co-immunoprecipitation and dual immunofluorescence studies confirmed ESR1, β-catenin, and lymphoid enhancer factor 1/T cell factor 3 co-localization in the nucleus in HESCs after E2 treatment. To determine the role of β-catenin signaling in the implantation of ectopic endometrium, we xenotransplanted eutopic endometrium from endometriosis patients into ovariectomized severe combined immunodeficiency mice. The implantation of the endometrium was suppressed by β-catenin siRNA. Collectively, studies regarding β-catenin signaling are critical for improving our understanding of the pathogenesis of estrogen-induced endometriosis, which can translate into the development of treatments and therapeutic strategies for endometriosis.
Collapse
Affiliation(s)
- Wenqian Xiong
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Ling Zhang
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Lan Yu
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Wei Xie
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Yicun Man
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Yao Xiong
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Hengwei Liu
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| | - Yi Liu
- Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China Department of Obstetrics and GynecologyTongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, ChinaDepartment of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, 1277 JieFang Road, Wuhan, China
| |
Collapse
|
57
|
Arevalo MA, Azcoitia I, Gonzalez-Burgos I, Garcia-Segura LM. Signaling mechanisms mediating the regulation of synaptic plasticity and memory by estradiol. Horm Behav 2015; 74:19-27. [PMID: 25921586 DOI: 10.1016/j.yhbeh.2015.04.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/02/2015] [Accepted: 04/20/2015] [Indexed: 01/29/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". Estradiol participates in the regulation of the function and plasticity of synaptic circuits in key cognitive brain regions, such as the prefrontal cortex and the hippocampus. The mechanisms elicited by estradiol are mediated by the regulation of transcriptional activity by nuclear estrogen receptors and by intracellular signaling cascades activated by estrogen receptors associated with the plasma membrane. In addition, the mechanisms include the interaction of estradiol with the signaling of other factors involved in the regulation of cognition, such as brain derived neurotrophic factor, insulin-like growth factor-1 and Wnt. Modifications in these signaling pathways by aging or by a long-lasting ovarian hormone deprivation after menopause may impair the enhancing effects of estradiol on synaptic plasticity and cognition.
Collapse
Affiliation(s)
- Maria-Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avenida Doctor Arce 37, E-28002 Madrid, Spain
| | - Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense, Calle José Antonio Novais 12, Ciudad Universitaria, E-28040 Madrid, Spain
| | - Ignacio Gonzalez-Burgos
- Laboratorio de Psicobiología, División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jal. Mexico
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avenida Doctor Arce 37, E-28002 Madrid, Spain.
| |
Collapse
|
58
|
The Role of Wnt/β-Catenin Signaling Pathway in Disrupted Hippocampal Neurogenesis of Temporal Lobe Epilepsy: A Potential Therapeutic Target? Neurochem Res 2015; 40:1319-32. [PMID: 26012365 DOI: 10.1007/s11064-015-1614-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/06/2015] [Accepted: 05/12/2015] [Indexed: 02/05/2023]
Abstract
Temporal lobe epilepsy is one of the most common clinical neurological disorders. One of the major pathological findings in temporal lobe epilepsy is hippocampal sclerosis, characterized by massive neuronal loss and severe gliosis. The epileptogenesis process of temporal lobe epilepsy usually starts with initial precipitating insults, followed by neurodegeneration, abnormal hippocampus circuitry reorganization, and the formation of hypersynchronicity. Experimental and clinical evidence strongly suggests that dysfunctional neurogenesis is involved in the epileptogenesis. Recent data demonstrate that neurogenesis is induced by acute seizures or precipitating insults, whereas the capacity of neuronal recruitment and proliferation substantially decreases in the chronic phase of epilepsy. Participation of the Wnt/β-catenin signaling pathway in neurogenesis reveals its importance in epileptogenesis; its dysfunction contributes to the structural and functional abnormality of temporal lobe epilepsy, while rescuing this pathway exerts neuroprotective effects. Here, we summarize data supporting the involvement of Wnt/β-catenin signaling in the epileptogenesis of temporal lobe epilepsy. We also propose that the Wnt/β-catenin signaling pathway may serve as a promising therapeutic target for temporal lobe epilepsy treatment.
Collapse
|
59
|
Villamil-Ortiz JG, Cardona-Gomez GP. Comparative analysis of autophagy and tauopathy related markers in cerebral ischemia and Alzheimer's disease animal models. Front Aging Neurosci 2015; 7:84. [PMID: 26042033 PMCID: PMC4436888 DOI: 10.3389/fnagi.2015.00084] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/30/2015] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) and cerebral ischemia (CI) are neuropathologies that are characterized by aggregates of tau protein, a hallmark of cognitive disorder and dementia. Protein accumulation can be induced by autophagic failure. Autophagy is a metabolic pathway involved in the homeostatic recycling of cellular components. However, the role of autophagy in those tauopathies remains unclear. In this study, we performed a comparative analysis to identify autophagy related markers in tauopathy generated by AD and CI during short-term, intermediate, and long-term progression using the 3xTg-AD mouse model (aged 6,12, and 18 months) and the global CI 2-VO (2-Vessel Occlusion) rat model (1,15, and 30 days post-ischemia). Our findings confirmed neuronal loss and hyperphosphorylated tau aggregation in the somatosensory cortex (SS-Cx) of the 3xTg-AD mice in the late stage (aged 18 months), which was supported by a failure in autophagy. These results were in contrast to those obtained in the SS-Cx of the CI rats, in which we detected neuronal loss and tauopathy at 1 and 15 days post-ischemia, and this phenomenon was reversed at 30 days. We proposed that this phenomenon was associated with autophagy induction in the late stage, since the data showed a decrease in p-mTOR activity, an association of Beclin-1 and Vps34, a progressive reduction in PHF-1, an increase in LC3B puncta and autophago-lysosomes formation were observed. Furthermore, the survival pathways remained unaffected. Together, our comparative study suggest that autophagy could ameliorates tauopathy in CI but not in AD, suggesting a differential temporal approach to the induction of neuroprotection and the prevention of neurodegeneration.
Collapse
Affiliation(s)
| | - Gloria P. Cardona-Gomez
- *Correspondence: Gloria P. Cardona-Gomez, Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Calle 62 #52–59, Torre 1, Piso 4, Laboratorio 412, Antioquia, Medellín, Colombia
| |
Collapse
|
60
|
Castro-Alvarez JF, Uribe-Arias A, Cardona-Gómez GP. Cyclin-Dependent kinase 5 targeting prevents β-Amyloid aggregation involving glycogen synthase kinase 3β and phosphatases. J Neurosci Res 2015; 93:1258-66. [PMID: 25711385 DOI: 10.1002/jnr.23576] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 01/24/2023]
Abstract
Inappropriate activation of cyclin-dependent kinase 5 (CDK5) resulting from proteolytic release of the activator fragment p25 from the membrane contributes to the formation of neurofibrillary tangles, β-amyloid (βA) aggregation, and chronic neurodegeneration. At 18 months of age, 3× Tg-AD mice were sacrificed after either 3 weeks (short term) or 1 year (long term) of CDK5 knockdown. In short-term-treated animals, CDK5 knockdown reversed βA aggregation in the hippocampi via inhibitory phosphorylation of glycogen synthase kinase 3β Ser9 and activation of phosphatase PP2A. In long-term-treated animals, CDK5 knockdown induced a persistent reduction in CDK5 and prevented βA aggregation, but the effect on amyloid precursor protein processing was reduced, suggesting that yearly booster therapy would be required. These findings further validate CDK5 as a target for preventing or blocking amyloidosis in older transgenic mice.
Collapse
Affiliation(s)
- John Fredy Castro-Alvarez
- Cellular and Molecular Neurobiology Area, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellín, Colombia
| | - Alejandro Uribe-Arias
- Cellular and Molecular Neurobiology Area, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellín, Colombia
| | - Gloria Patricia Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellín, Colombia
| |
Collapse
|
61
|
Andrographolide activates the canonical Wnt signalling pathway by a mechanism that implicates the non-ATP competitive inhibition of GSK-3β: autoregulation of GSK-3β in vivo. Biochem J 2015; 466:415-30. [DOI: 10.1042/bj20140207] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Andrographolide activates the canonical Wnt pathway and induces the transcription of Wnt target genes through a mechanism independent of Wnt ligand binding to its receptor, by direct substrate-competitive inhibition of GSK-3.
Collapse
|
62
|
Sabogal-Guáqueta AM, Muñoz-Manco JI, Ramírez-Pineda JR, Lamprea-Rodriguez M, Osorio E, Cardona-Gómez GP. The flavonoid quercetin ameliorates Alzheimer's disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer's disease model mice. Neuropharmacology 2015; 93:134-45. [PMID: 25666032 DOI: 10.1016/j.neuropharm.2015.01.027] [Citation(s) in RCA: 299] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is the most common senile dementia in the world. Although important progress has been made in understanding the pathogenesis of AD, current therapeutic approaches provide only modest symptomatic relief. In this study, we evaluated the neuroprotective effect of quercetin (25 mg/kg) administration via i.p. injection every 48 h for 3 months on aged (21-24 months old) triple transgenic AD model (3xTg-AD) mice. Our data show that quercetin decreases extracellular β-amyloidosis, tauopathy, astrogliosis and microgliosis in the hippocampus and the amygdala. These results were supported by a significant reduction in the paired helical filament (PHF), β-amyloid (βA) 1-40 and βA 1-42 levels and a decrease in BACE1-mediated cleavage of APP (into CTFβ). Additionally, quercetin induced improved performance on learning and spatial memory tasks and greater risk assessment behavior based on the elevated plus maze test. Together, these findings suggest that quercetin reverses histological hallmarks of AD and protects cognitive and emotional function in aged 3xTg-AD mice.
Collapse
Affiliation(s)
- Angélica Maria Sabogal-Guáqueta
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, University of Antioquia, Calle 70 No. 52-21, Medellin, Colombia
| | - Juan Ignacio Muñoz-Manco
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, University of Antioquia, Calle 70 No. 52-21, Medellin, Colombia
| | - Jose R Ramírez-Pineda
- Grupo de Investigación en Sustancias Bioactivas, Facultad de Química Farmacéutica, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia; Group of Immunomodulation, Faculty of Medicine, University of Antioquia, Calle 70 No. 52-21, Medellin, Colombia
| | | | - Edison Osorio
- Grupo de Investigación en Sustancias Bioactivas, Facultad de Química Farmacéutica, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Gloria Patricia Cardona-Gómez
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, University of Antioquia, Calle 70 No. 52-21, Medellin, Colombia.
| |
Collapse
|
63
|
Abstract
Despite decades of research aimed at identifying the causes of postpartum depression (PPD), PPD remains common, and the causes are poorly understood. Many have attributed the onset of PPD to the rapid perinatal change in reproductive hormones. Although a number of human and nonhuman animal studies support the role of reproductive hormones in PPD, several studies have failed to detect an association between hormone concentrations and PPD. The purpose of this review is to examine the hypothesis that fluctuations in reproductive hormone levels during pregnancy and the postpartum period trigger PPD in susceptible women. We discuss and integrate the literature on animal models of PPD and human studies of reproductive hormones and PPD. We also discuss alternative biological models of PPD to demonstrate the potential for multiple PPD phenotypes and to describe the complex interplay of changing reproductive hormones and alterations in thyroid function, immune function, hypothalamic-pituitary-adrenal (HPA) axis function, lactogenic hormones, and genetic expression that may contribute to affective dysfunction. There are 3 primary lines of inquiry that have addressed the role of reproductive hormones in PPD: nonhuman animal studies, correlational studies of postpartum hormone levels and mood symptoms, and hormone manipulation studies. Reproductive hormones influence virtually every biological system implicated in PPD, and a subgroup of women seem to be particularly sensitive to the effects of perinatal changes in hormone levels. We propose that these women constitute a "hormone-sensitive" PPD phenotype, which should be studied independent of other PPD phenotypes to identify underlying pathophysiology and develop novel treatment targets.
Collapse
|
64
|
Qu N, Zhou XY, Han L, Wang L, Xu JX, Zhang T, Chu J, Chen Q, Wang JZ, Zhang Q, Tian Q. Combination of PPT with LiCl Treatment Prevented Bilateral Ovariectomy-Induced Hippocampal-Dependent Cognition Deficit in Rats. Mol Neurobiol 2014; 53:894-904. [DOI: 10.1007/s12035-014-9050-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/02/2014] [Indexed: 12/17/2022]
|
65
|
Gao YRE, Walters KA, Desai R, Zhou H, Handelsman DJ, Simanainen U. Androgen receptor inactivation resulted in acceleration in pubertal mammary gland growth, upregulation of ERα expression, and Wnt/β-catenin signaling in female mice. Endocrinology 2014; 155:4951-63. [PMID: 25076121 DOI: 10.1210/en.2014-1226] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The androgen receptor (AR) is widely expressed in mammary cells of female mammals including humans and mice, indicating a possible role for AR-mediated androgen actions in breast development, function, and pathology, although the specific mechanisms remain unclear. To elucidate the mechanisms of androgen action in mammary gland physiology and development, we used AR-knockout (AR(Δex3)KO) female mice with a universally expressed, transcriptionally inactive AR protein harboring an in-frame deletion of its second zinc finger. Although in sexually mature wild-type (WT) and AR(ex3Δ)KO females, the mammary epithelial growth was fully extended to the edge of the fat pad, during puberty, AR(ex3Δ)KO females exhibit significantly accelerated mammary ductal growth and an increased number of terminal end buds compared with WT females. Accelerated AR(ex3Δ)KO female mammary growth was associated with significantly increased mammary epithelial ERα expression and activated Wnt/β-catenin signaling as shown by increased Wnt4 expression and accumulation of nuclear β-catenin. These findings are consistent with increased mammary estrogen exposure although ovarian estradiol content was unchanged compared with WT females. Furthermore, treatment with the potent pure androgen DHT markedly reduced ductal extension and terminal end bud numbers in WT but not in AR(Δex3)KO females, further supporting the concept that AR-mediated, androgen-induced suppression of murine mammary growth is a physiological characteristic of puberty. In summary, our findings reveal an inhibitory role of AR-mediated androgen actions in pubertal mammary gland development by reducing epithelial cell proliferation and could be mediated by regulation of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yan Ru Ellen Gao
- Andrology Laboratory (Y.R.G., K.A.W., R.D., D.J.H., U.S.) and Bone research Program (H.Z.), ANZAC Research Institute, University of Sydney, Sydney New South Wales 2139, Australia
| | | | | | | | | | | |
Collapse
|
66
|
Arevalo MA, Azcoitia I, Garcia-Segura LM. The neuroprotective actions of oestradiol and oestrogen receptors. Nat Rev Neurosci 2014; 16:17-29. [PMID: 25423896 DOI: 10.1038/nrn3856] [Citation(s) in RCA: 323] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hormones regulate homeostasis by communicating through the bloodstream to the body's organs, including the brain. As homeostatic regulators of brain function, some hormones exert neuroprotective actions. This is the case for the ovarian hormone 17β-oestradiol, which signals through oestrogen receptors (ERs) that are widely distributed in the male and female brain. Recent discoveries have shown that oestradiol is not only a reproductive hormone but also a brain-derived neuroprotective factor in males and females and that ERs coordinate multiple signalling mechanisms that protect the brain from neurodegenerative diseases, affective disorders and cognitive decline.
Collapse
Affiliation(s)
- Maria-Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain
| | - Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense, E-28040 Madrid, Spain
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain
| |
Collapse
|
67
|
Castro-Alvarez JF, Uribe-Arias SA, Kosik KS, Cardona-Gómez GP. Long- and short-term CDK5 knockdown prevents spatial memory dysfunction and tau pathology of triple transgenic Alzheimer's mice. Front Aging Neurosci 2014; 6:243. [PMID: 25309427 PMCID: PMC4159979 DOI: 10.3389/fnagi.2014.00243] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 08/26/2014] [Indexed: 11/13/2022] Open
Abstract
CDK5 is a member of the cyclin-dependent kinase family with diverse functions in both the developing and mature nervous system. The inappropriate activation of CDK5 due to the proteolytic release of the activator fragment p25 from the membrane contributes to the formation of neurofibrillary tangles and chronic neurodegeneration. At 18 months of age 3xTg-AD mice were sacrificed after 1 year (long term) or 3 weeks (short term) of CDK5 knockdown. In long-term animals CDK5 knockdown prevented insoluble Tau formation in the hippocampi and prevented spatial memory impairment. In short-term animals, CDK5 knockdown showed reduction of CDK5, reversed Tau aggregation, and improved spatial memory compared to scrambled treated old 3xTg-AD mice. Neither long-term nor short-term CDK5 knock-down had an effect on old littermates. These findings further validate CDK5 as a target for Alzheimer’s disease both as a preventive measure and after the onset of symptoms.
Collapse
Affiliation(s)
- John F Castro-Alvarez
- Cellular and Molecular Neurobiology Area, Neuroscience Group of Antioquia, Faculty of Medicine, University Research Headquarters, University of Antioquia Medellín Colombia
| | - S Alejandro Uribe-Arias
- Cellular and Molecular Neurobiology Area, Neuroscience Group of Antioquia, Faculty of Medicine, University Research Headquarters, University of Antioquia Medellín Colombia
| | - Kenneth S Kosik
- Department of Molecular Cellular Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara Santa Barbara, CA USA
| | - Gloria P Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Neuroscience Group of Antioquia, Faculty of Medicine, University Research Headquarters, University of Antioquia Medellín Colombia
| |
Collapse
|
68
|
Allopregnanolone as a mediator of affective switching in reproductive mood disorders. Psychopharmacology (Berl) 2014; 231:3557-67. [PMID: 24846476 PMCID: PMC4135022 DOI: 10.1007/s00213-014-3599-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/14/2014] [Indexed: 01/15/2023]
Abstract
RATIONALE Reproductive mood disorders, including premenstrual dysphoria (PMD) and postpartum depression (PPD), are characterized by affective dysregulation that occurs during specific reproductive states. The occurrence of illness onset during changes in reproductive endocrine function has generated interest in the role of gonadal steroids in the pathophysiology of reproductive mood disorders, yet the mechanisms by which the changing hormone milieu triggers depression in susceptible women remain poorly understood. OBJECTIVES This review focuses on one of the neurosteroid metabolites of progesterone - allopregnanolone (ALLO) - that acutely regulates neuronal function and may mediate affective dysregulation that occurs concomitant with changes in reproductive endocrine function. We describe the role of the "neuroactive" steroids estradiol and progesterone in reproductive endocrine-related mood disorders to highlight the potential mechanisms by which ALLO might contribute to their pathophysiology. Finally, using existing data, we test the hypothesis that changes in ALLO levels may trigger affective dysregulation in susceptible women. RESULTS Although there is no reliable evidence that basal ALLO levels distinguish those with PMD or PPD from those without, existing animal models suggest potential mechanisms by which specific reproductive states may unmask susceptibility to affective dysregulation. Consistent with these models, initially euthymic women with PMD and those with a history of PPD show a negative association between depressive symptoms and circulating ALLO levels following progesterone administration. CONCLUSIONS Existing animal models and our own preliminary data suggest that ALLO may play an important role in the pathophysiology of reproductive mood disorders by triggering affective dysregulation in susceptible women.
Collapse
|
69
|
Epigenetics and autism. AUTISM RESEARCH AND TREATMENT 2013; 2013:826156. [PMID: 24151554 PMCID: PMC3787640 DOI: 10.1155/2013/826156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/17/2013] [Accepted: 08/01/2013] [Indexed: 12/19/2022]
Abstract
This review identifies mechanisms for altering DNA-histone interactions of cell chromatin to upregulate or downregulate gene expression that could serve as epigenetic targets for therapeutic interventions in autism. DNA methyltransferases (DNMTs) can phosphorylate histone H3 at T6. Aided by protein kinase C β 1, the DNMT lysine-specific demethylase-1 prevents demethylation of H3 at K4. During androgen-receptor-(AR-) dependent gene activation, this sequence may produce AR-dependent gene overactivation which may partly explain the male predominance of autism. AR-dependent gene overactivation in conjunction with a DNMT mechanism for methylating oxytocin receptors could produce high arousal inputs to the amygdala resulting in aberrant socialization, a prime characteristic of autism. Dysregulation of histone methyltransferases and histone deacetylases (HDACs) associated with low activity of methyl CpG binding protein-2 at cytosine-guanine sites in genes may reduce the capacity for condensing chromatin and silencing genes in frontal cortex, a site characterized by decreased cortical interconnectivity in autistic subjects. HDAC1 inhibition can overactivate mRNA transcription, a putative mechanism for the increased number of cerebral cortical columns and local frontal cortex hyperactivity in autistic individuals. These epigenetic mechanisms underlying male predominance, aberrant social interaction, and low functioning frontal cortex may be novel targets for autism prevention and treatment strategies.
Collapse
|
70
|
Younesi E, Hofmann-Apitius M. A network model of genomic hormone interactions underlying dementia and its translational validation through serendipitous off-target effect. J Transl Med 2013; 11:177. [PMID: 23885764 PMCID: PMC3733613 DOI: 10.1186/1479-5876-11-177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/03/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND While the majority of studies have focused on the association between sex hormones and dementia, emerging evidence supports the role of other hormone signals in increasing dementia risk. However, due to the lack of an integrated view on mechanistic interactions of hormone signaling pathways associated with dementia, molecular mechanisms through which hormones contribute to the increased risk of dementia has remained unclear and capacity of translating hormone signals to potential therapeutic and diagnostic applications in relation to dementia has been undervalued. METHODS Using an integrative knowledge- and data-driven approach, a global hormone interaction network in the context of dementia was constructed, which was further filtered down to a model of convergent hormone signaling pathways. This model was evaluated for its biological and clinical relevance through pathway recovery test, evidence-based analysis, and biomarker-guided analysis. Translational validation of the model was performed using the proposed novel mechanism discovery approach based on 'serendipitous off-target effects'. RESULTS Our results reveal the existence of a well-connected hormone interaction network underlying dementia. Seven hormone signaling pathways converge at the core of the hormone interaction network, which are shown to be mechanistically linked to the risk of dementia. Amongst these pathways, estrogen signaling pathway takes the major part in the model and insulin signaling pathway is analyzed for its association to learning and memory functions. Validation of the model through serendipitous off-target effects suggests that hormone signaling pathways substantially contribute to the pathogenesis of dementia. CONCLUSIONS The integrated network model of hormone interactions underlying dementia may serve as an initial translational platform for identifying potential therapeutic targets and candidate biomarkers for dementia-spectrum disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Erfan Younesi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany
- Bonn-Aachen International Center for Information Technology, University of Bonn, Bonn, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany
- Bonn-Aachen International Center for Information Technology, University of Bonn, Bonn, Germany
| |
Collapse
|
71
|
Ruiz-Palmero I, Hernando M, Garcia-Segura LM, Arevalo MA. G protein-coupled estrogen receptor is required for the neuritogenic mechanism of 17β-estradiol in developing hippocampal neurons. Mol Cell Endocrinol 2013; 372:105-15. [PMID: 23545157 DOI: 10.1016/j.mce.2013.03.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 02/28/2013] [Accepted: 03/21/2013] [Indexed: 01/29/2023]
Abstract
Estradiol promotes neuritogenesis in developing hippocampal neurons by a mechanism involving the upregulation of neurogenin 3, a Notch-regulated transcription factor. In this study we have explored whether G-protein coupled estrogen receptor 1 (GPER) participates in this hormonal action. GPER agonists (17β-estradiol, G1, ICI 182,780) increased neurogenin 3 expression and neuritogenesis in mouse primary hippocampal neurons and this effect was blocked by the GPER antagonist G15 and by a siRNA for GPER. In addition, GPER agonists increased Akt phosphorylation in ser473, which is indicative of the activation of phosphoinositide-3-kinase (PI3K). G15 or GPER silencing prevented the estrogenic induction of Akt phosphorylation. Furthermore, the PI3K inhibitor wortmannin prevented the effect of G1 and estradiol on neurogenin 3 expression and the effect of estradiol on neuritogenesis. These findings suggest that GPER participates in the control of hippocampal neuritogenesis by a mechanism involving the activation of PI3K signaling.
Collapse
|
72
|
Varea O, Escoll M, Diez H, Garrido J, Wandosell F. Oestradiol signalling through the Akt–mTORC1–S6K1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1052-64. [DOI: 10.1016/j.bbamcr.2012.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/11/2012] [Accepted: 12/26/2012] [Indexed: 12/12/2022]
|
73
|
Cardona-Rossinyol A, Mir M, Caraballo-Miralles V, Lladó J, Olmos G. Neuroprotective effects of estradiol on motoneurons in a model of rat spinal cord embryonic explants. Cell Mol Neurobiol 2013; 33:421-32. [PMID: 23322321 PMCID: PMC11498032 DOI: 10.1007/s10571-013-9908-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/05/2013] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset degenerative disorder characterized by motoneuron death. Clinical and experimental studies in animal models of ALS have found gender differences in the incidence and onset of disease, suggesting that female hormones may play a beneficial role. Cumulative evidence indicates that 17β-estradiol (17βE2) has a neuroprotective role in the central nervous system. We have previously developed a new culture system by using rat spinal cord embryonic explants in which motoneurons have the singularity of migrating outside the spinal cord, growing as a monolayer in the presence of glial cells. In this study, we have validated this new culture system as a useful model for studying neuroprotection by estrogens on spinal cord motoneurons. We show for the first time that spinal cord motoneurons express classical estrogen receptors and that 17βE2 activates, specifically in these cells, the Akt anti-apoptotic signaling pathway and two of their downstream effectors: GSK-3β and Bcl-2. To further validate our system, we demonstrated neuroprotective effects of 17βE2 on spinal cord motoneurons when exposed to the proinflammatory cytokines TNF-α and IFN-γ. These effects of 17βE2 were fully reverted in the presence of the estrogen receptor antagonist ICI 182,780. Our new culture model and the results presented here may provide the basis for further studies on the effects of estrogens, and selective estrogen receptor modulators, on spinal cord motoneurons in the context of ALS or other motoneuron diseases.
Collapse
Affiliation(s)
- Andrea Cardona-Rossinyol
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Margalida Mir
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Víctor Caraballo-Miralles
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Jerònia Lladó
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Gabriel Olmos
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| |
Collapse
|
74
|
Witty CF, Gardella LP, Perez MC, Daniel JM. Short-term estradiol administration in aging ovariectomized rats provides lasting benefits for memory and the hippocampus: a role for insulin-like growth factor-I. Endocrinology 2013; 154:842-52. [PMID: 23264616 DOI: 10.1210/en.2012-1698] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We previously demonstrated that aged ovariectomized rats that had received prior estradiol treatment in middle age exhibited enhanced spatial memory and increased levels of estrogen receptor (ER)-α in the hippocampus long after estradiol treatment was terminated. The implication for cognition of increased levels of ERα resulting from prior estradiol exposure is unknown. In the absence of estrogens, growth factors, including IGF-I, can induce ERα-mediated transcription through ligand-independent mechanisms. Our current goal was to determine whether IGF-I mediates the ability of short-term exposure to estradiol to exert long-term effects on cognition and the hippocampus of aging females. Ovariectomized middle-aged rats were implanted with estradiol or cholesterol vehicle capsules. After 40 days, all capsules were removed and drug treatments were initiated. Half of each hormone treatment group received chronic intracerebroventricular delivery of the IGF-I receptor antagonist JB1, and the other half received artificial cerebrospinal fluid vehicle. Rats were tested on a spatial memory radial-arm maze task and hippocampi were immunostained for proteins of interest by Western blotting. As expected, previous treatment with estradiol enhanced spatial memory and increased levels of ERα in the hippocampus. JB1 reversed these effects. Previous treatment with estradiol resulted in lasting increases in levels of IGF-I receptors and phosphorylation of ERK/MAPK, a downstream signaling molecule of both ERα and IGF-I receptors, and increased levels of the ERα-regulated protein, choline acetyltransferase. JB1 blocked effects on ERK/MAPK and choline acetyltransferase. Results indicate that activation of IGF-I receptors is necessary for prior estradiol exposure to exert lasting impact on the hippocampus and memory.
Collapse
|
75
|
Chen CS, Tseng YT, Hsu YY, Lo YC. Nrf2-Keap1 antioxidant defense and cell survival signaling are upregulated by 17β-estradiol in homocysteine-treated dopaminergic SH-SY5Y cells. Neuroendocrinology 2013; 97:232-41. [PMID: 22948038 DOI: 10.1159/000342692] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 08/09/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS A recent neuroimaging study discovered the neurotoxicity effects of homocysteine (Hcy), which is only seen in elderly women. Estrogens exert a variety of actions on brain function that influence cognitive function, mood, and neuroprotection. The Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant defense pathway has been well-known to afford neuroprotection. Here, we first demonstrate the roles of Nrf2-Keap1 in 17β-estradiol (E2) cytoprotection and Hcy toxicity and the protective mechanisms of E2 on Hcy cytotoxicity in human dopaminergic SH-SY5Y cells. METHODS Cell viability was determined by trypan blue method. Protein expression was determined by Western blot analysis. Superoxide dismutase (SOD) activity was determined by ELISA. Reactive oxygen species (ROS) production was determined by flow cytometry. RESULTS In Hcy-treated SH-SY5Y cells, E2 increased cell viability, attenuated ROS production, activated Akt signaling and inhibited glycogen synthase kinase-3β (GSK-3β), a kinase known to participate in neurodegeneration. Moreover, E2 treatment led to Nrf2 dissociation from Keap1, the main negative regulator of Nrf2 activity in the cytoplasm, and increased the protein level of Nrf2 in the nucleus, with a significant increase in HO-1 expression and SOD activity in Hcy-treated cells. E2-induced Nrf2 activation was attenuated by the PI3K inhibitor LY294002 and the estrogen receptor antagonist ICI 182,780. Further, E2 decreased Hcy-induced apoptotic death by upregulating the antiapoptotic protein Bcl-2, decreasing cytochrome c release from mitochondria, and attenuating apoptotic cascade activation (Bax, caspase-9, and caspase-3). CONCLUSION E2 activates cell survival signaling and Nrf2-Keap1 antioxidant defense pathway and attenuates Hcy cytotoxicity.
Collapse
Affiliation(s)
- Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | | | | | | |
Collapse
|
76
|
Estrogen regulation of Dkk1 and Wnt/β-Catenin signaling in neurodegenerative disease. Brain Res 2012; 1514:63-74. [PMID: 23261660 DOI: 10.1016/j.brainres.2012.12.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/11/2012] [Indexed: 11/22/2022]
Abstract
17β-estradiol (E2 or estrogen) is an endogenous steroid hormone that is well known to exert neuroprotection. Along these lines, one mechanism through which E2 protects the hippocampus from cerebral ischemia is by preventing the post-ischemic elevation of Dkk1, a neurodegenerative factor that serves as an antagonist of the canonical Wnt signaling pathway, and simultaneously inducing pro-survival Wnt/β-Catenin signaling in hippocampal neurons. Intriguingly, while expression of Dkk1 is required for proper neural development, overexpression of Dkk1 is characteristic of many neurodegenerative diseases, such as stroke, Alzheimer's disease, Parkinson's disease, and temporal lobe epilepsy. In this review, we will briefly summarize the canonical Wnt signaling pathway, highlight the current literature linking alterations of Dkk1 and Wnt/β-Catenin signaling with neurological disease, and discuss E2's role in maintaining the delicate balance of Dkk1 and Wnt/β-Catenin signaling in the adult brain. Finally, we will consider the implications of long-term E2 deprivation and hormone therapy on this crucial neural pathway. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
|
77
|
Ooishi Y, Kawato S, Hojo Y, Hatanaka Y, Higo S, Murakami G, Komatsuzaki Y, Ogiue-Ikeda M, Kimoto T, Mukai H. Modulation of synaptic plasticity in the hippocampus by hippocampus-derived estrogen and androgen. J Steroid Biochem Mol Biol 2012; 131:37-51. [PMID: 22075082 DOI: 10.1016/j.jsbmb.2011.10.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/27/2011] [Accepted: 10/12/2011] [Indexed: 12/29/2022]
Abstract
The hippocampus synthesizes estrogen and androgen in addition to the circulating sex steroids. Synaptic modulation by hippocampus-derived estrogen or androgen is essential to maintain healthy memory processes. Rapid actions (1-2h) of 17β-estradiol (17β-E2) occur via synapse-localized receptors (ERα or ERβ), while slow genomic E2 actions (6-48h) occur via classical nuclear receptors (ERα or ERβ). The long-term potentiation (LTP), induced by strong tetanus or theta-burst stimulation, is not further enhanced by E2 perfusion in adult rats. Interestingly, E2 perfusion can rescue corticosterone (stress hormone)-induced suppression of LTP. The long-term depression is modulated rapidly by E2 perfusion. Elevation of the E2 concentration changes rapidly the density and head structure of spines in neurons. ERα, but not ERβ, drives this enhancement of spinogenesis. Kinase networks are involved downstream of ERα. Testosterone (T) or dihydrotestosterone (DHT) also rapidly modulates spinogenesis. Newly developed Spiso-3D mathematical analysis is used to distinguish these complex effects by sex steroids and kinases. It has been doubted that the level of hippocampus-derived estrogen and androgen may not be high enough to modulate synaptic plasticity. Determination of the accurate concentration of E2, T or DHT in the hippocampus is enabled by mass-spectrometric analysis in combination with new steroid-derivatization methods. The E2 level in the hippocampus is approximately 8nM for the male and 0.5-2nM for the female, which is much higher than that in circulation. The level of T and DHT is also higher than that in circulation. Taken together, hippocampus-derived E2, T, and DHT play a major role in modulation of synaptic plasticity.
Collapse
Affiliation(s)
- Yuuki Ooishi
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Arevalo MA, Ruiz-Palmero I, Scerbo MJ, Acaz-Fonseca E, Cambiasso MJ, Garcia-Segura LM. Molecular mechanisms involved in the regulation of neuritogenesis by estradiol: Recent advances. J Steroid Biochem Mol Biol 2012; 131:52-6. [PMID: 21971420 DOI: 10.1016/j.jsbmb.2011.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 09/19/2011] [Accepted: 09/21/2011] [Indexed: 12/13/2022]
Abstract
This review analyzes the signaling mechanisms activated by estradiol to regulate neuritogenesis in several neuronal populations. Estradiol regulates axogenesis by the activation of the mitogen activated protein kinase (MAPK) cascade through estrogen receptor α located in the plasma membrane. In addition, estradiol regulates MAPK signaling via the activation of protein kinase C and by increasing the expression of brain derived neurotrophic factor and tyrosine kinase receptor B. Estradiol also interacts with the signaling of insulin-like growth factor-I receptor through estrogen receptor α, modulating the phosphoinositide-3 kinase signaling pathway, which contributes to the stabilization of microtubules. Finally, estradiol modulates dendritogenesis by the inhibition of Notch signaling, by a mechanism that, at least in hippocampal neurons, is mediated by G-protein coupled receptor 30. This article is part of a Special Issue entitled 'Neurosteroids'.
Collapse
|
79
|
Pérez-Álvarez MJ, Maza MDC, Anton M, Ordoñez L, Wandosell F. Post-ischemic estradiol treatment reduced glial response and triggers distinct cortical and hippocampal signaling in a rat model of cerebral ischemia. J Neuroinflammation 2012; 9:157. [PMID: 22747981 PMCID: PMC3414748 DOI: 10.1186/1742-2094-9-157] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 05/29/2012] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Estradiol has been shown to exert neuroprotective effects in several neurodegenerative conditions, including cerebral ischemia. The presence of this hormone prior to ischemia attenuates the damage associated with such events in a rodent model (middle cerebral artery occlusion (MCAO)), although its therapeutic value when administered post-ischemia has not been assessed. Hence, we evaluated the effects of estradiol treatment after permanent MCAO (pMCAO) was induced in rats, studying the PI3K/AKT/GSK3/β-catenin survival pathway and the activation of SAPK-JNK in two brain areas differently affected by pMCAO: the cortex and hippocampus. In addition, we analyzed the effect of estradiol on the glial response to injury. METHODS Male rats were subjected to pMCAO and estradiol (0.04 mg/kg) was administered 6, 24, and 48 h after surgery. The animals were sacrificed 6 h after the last treatment, and brain damage was evaluated by immunohistochemical quantification of 'reactive gliosis' using antibodies against GFAP and Iba1. In addition, Akt, phospho-Akt(Ser473), phospho-Akt(Thr308), GSK3, phospho-GSK3(Ser21/9), β-catenin, SAPK-JNK, and pSAPK-JNK(Thr183/Tyr185) levels were determined in western blots of the ipsilateral cerebral cortex and hippocampus, and regional differences in neuronal phospho-Akt expression were determined by immunohistochemistry. RESULTS The increases in the percentage of GFAP- (5.25-fold) and Iba1- (1.8-fold) labeled cells in the cortex and hippocampus indicate that pMCAO induced 'reactive gliosis'. This effect was prevented by post-ischemic estradiol treatment; diminished the number of these cells to those comparable with control animals. pMCAO down-regulated the PI3K/AkT/GSK3/β-catenin survival pathway to different extents in the cortex and hippocampus, the activity of which was restored by estradiol treatment more efficiently in the cerebral cortex (the most affected region) than in the hippocampus. No changes in the phosphorylation of SAPK-JNK were observed 54 h after inducing pMCAO, whereas pMCAO did significantly decrease the phospho-Akt(Ser473) in neurons, an effect that was reversed by estradiol. CONCLUSION The present study demonstrates that post-pMCAO estradiol treatment attenuates ischemic injury in both neurons and glia, events in which the PI3K/AKT/GSK3/β-catenin pathway is at least partly involved. These findings indicate that estradiol is a potentially useful treatment to enhance recovery after human ischemic stroke.
Collapse
Affiliation(s)
- Maria Jose Pérez-Álvarez
- Departamento de Biología (Unidad docente Fisiología Animal), Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Maria del Carmen Maza
- Departamento de Biología (Unidad docente Fisiología Animal), Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marta Anton
- Departamento de Biología (Unidad docente Fisiología Animal), Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Lara Ordoñez
- Departamento de Biología (Unidad docente Fisiología Animal), Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, Univ. Autónoma de Madrid, Madrid, 28049, Spain
- Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa", CIBERNED-CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, C/Nicolás Cabrera n° 1, Madrid, 28049, Spain
| |
Collapse
|
80
|
Dellinger TH, Planutis K, Tewari KS, Holcombe RF. Role of canonical Wnt signaling in endometrial carcinogenesis. Expert Rev Anticancer Ther 2012; 12:51-62. [PMID: 22149432 DOI: 10.1586/era.11.194] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
While the role of Wnt signaling is well established in colorectal carcinogenesis, its function in gynecologic cancers has not been elucidated. Here, we describe the current state of knowledge of canonical Wnt signaling in endometrial cancer (EC), and its implications for future therapeutic targets. Deregulation of the Wnt/β-catenin signaling pathway in EC occurs by inactivating β-catenin mutations in approximately 10-45% of ECs, and via downregulation of Wnt antagonists by epigenetic silencing. The Wnt pathway is intimately involved with estrogen and progesterone, and emerging data implicate it in other important signaling pathways, such as mTOR and Hedgehog. While no therapeutic agents targeting the Wnt signaling pathway are currently in clinical trials, the preclinical data presented suggest a role for Wnt signaling in uterine carcinogenesis, with further research warranted to elucidate the mechanism of action and to proceed towards targeted cancer drug development.
Collapse
Affiliation(s)
- Thanh H Dellinger
- Divison of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of California, Irvine, Medical Center, 101 The City Drive, Building 56, Room 260, Orange, CA 92868, USA.
| | | | | | | |
Collapse
|
81
|
Barrera-Ocampo A, Gutierrez-Vargas J, Garcia-Segura LM, Cardona-Gómez GP. Glycogen synthase kinase-3β/β-catenin signaling in the rat hypothalamus during the estrous cycle. J Neurosci Res 2012; 90:1078-84. [PMID: 22331547 DOI: 10.1002/jnr.22816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/06/2011] [Accepted: 09/15/2011] [Indexed: 11/11/2022]
Abstract
During the estrous cycle, a remodeling of synapses on somas and dendritic spines occurs in the rat hypothalamic arcuate nucleus. The synaptic remodeling is known to be induced by estradiol, but the molecular mechanisms involved still have not been fully clarified. β-catenin is known to regulate synaptic plasticity, so we have assessed possible modifications of β-catenin in the rat mediobasal hypothalamus during the estrous cycle. Our findings indicate that β-catenin expression is increased during proestrus and estrus in comparison with diestrus day. This increase was accompanied by an enhanced phosphorylation of Akt in Ser473 and of glycogen synthase kinase-3β (GSK3β) in Ser9. Also, the association of β-catenin with the synaptic protein PSD95 was increased during these same stages of the estrous cycle, whereas the levels of synapsin I were significantly decreased in proestrus. These findings suggest that Akt/GSK3β/β-catenin signaling is involved in the synaptic modifications that occur in the basal hypothalamus during the estrous cycle.
Collapse
Affiliation(s)
- Alvaro Barrera-Ocampo
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellín, Colombia
| | | | | | | |
Collapse
|
82
|
Effect of a chronic treatment with 17β-estradiol on striatal dopamine neurotransmission and the Akt/GSK3 signaling pathway in the brain of ovariectomized monkeys. Psychoneuroendocrinology 2012; 37:280-91. [PMID: 21763075 DOI: 10.1016/j.psyneuen.2011.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/03/2011] [Accepted: 06/16/2011] [Indexed: 02/07/2023]
Abstract
The present experiments sought the effect of chronic treatment with 17β-estradiol on striatal dopaminergic activity and the Akt/GSK3 signaling pathway in the brain of monkeys. Eight female monkeys (Macacca fascicularis) were ovariectomized (OVX) and a month later, half received a month treatment with 17β-estradiol and the other with vehicle. The DA transporter (DAT) was measured by autoradiography with [(125)I]RTI-121 and the vesicular DA transporter (VMAT(2)) with [(3)H]TBZ-OH at three rostro-caudal levels (anterior, middle and posterior) of the caudate nucleus and putamen subdivided in their lateral/medial, ventral/dorsal sub-regions. Specific binding to DAT was increased in all sub-regions of the caudate nucleus and the putamen of 17β-estradiol-treated compared to vehicle-treated monkeys whereas specific binding to VMAT(2) remained unchanged. We measured by Western blot the phosphorylated forms of Akt at serine 473 and threonine 308, GSK3β at serine 9 and tyrosine 216 and GSK3α at serine 21 in anterior, middle and posterior caudate nucleus and putamen. 17β-Estradiol treatment increased in all the caudate nucleus and putamen pAkt (Ser473)/βIII-tubulin, pGSK3β (Ser9)/βIII-tubulin and in putamen Akt/βIII-tubulin compared to vehicle-treated monkeys. In anterior and middle putamen, pAkt (Thr308)/βIII-tubulin was also increased in monkeys treated with 17β-estradiol. pGSK3β (Tyr216)/βIII-tubulin and pGSK3α (Ser21)/βIII-tubulin remained unchanged by the 17β-estradiol treatment. These results suggest that 17β-estradiol activates striatal DA neurotransmission in primates as reflected with increased DAT specific binding and downstream activation of Akt/GSK3 signaling. This supports a beneficial role of a chronic treatment with 17β-estradiol by increasing the activity of signaling pathways implicated in cell survival.
Collapse
|
83
|
Wandosell F, Varea O, Arevalo MA, Garcia-Segura LM. Oestradiol regulates β-catenin-mediated transcription in neurones. J Neuroendocrinol 2012; 24:191-4. [PMID: 21722217 DOI: 10.1111/j.1365-2826.2011.02186.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Oestradiol acts in the brain by multiple mechanisms, including the regulation of transcriptional activity through classical oestrogen receptors, α and β, and by the activation of membrane/cytoplasm-initiated signalling cascades. In neuroblastoma cells, primary neurones in culture and in the brain in vivo, oestradiol activates the phosphoinositide 3-kinase/Akt/glycogen synthase kinase 3 signalling pathway by a mechanism involving oestrogen receptor α. Through this pathway, oestradiol regulates the stability of β-catenin, induces the translocation of β-catenin to the cell nucleus and regulates β-catenin-mediated transcription through the T cell factor/DNA complex. Genomic analyses in neuroblastoma cells have revealed that the set of genes regulated by oestradiol through β-catenin is not identical to that regulated by the Wnt signalling pathway, revealing a new mechanism for oestradiol signalling in neurones.
Collapse
Affiliation(s)
- F Wandosell
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | | | | | | |
Collapse
|
84
|
Pinto-Almazán R, Calzada-Mendoza CC, Campos-Lara MG, Guerra-Araiza C. Effect of chronic administration of estradiol, progesterone, and tibolone on the expression and phosphorylation of glycogen synthase kinase-3β and the microtubule-associated protein tau in the hippocampus and cerebellum of female rat. J Neurosci Res 2011; 90:878-86. [DOI: 10.1002/jnr.22808] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 08/25/2011] [Accepted: 09/01/2011] [Indexed: 01/26/2023]
|
85
|
Sharma A, Hu XT, Napier TC, Al-Harthi L. Methamphetamine and HIV-1 Tat down regulate β-catenin signaling: implications for methampetamine abuse and HIV-1 co-morbidity. J Neuroimmune Pharmacol 2011; 6:597-607. [PMID: 21744004 PMCID: PMC3714216 DOI: 10.1007/s11481-011-9295-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 06/30/2011] [Indexed: 12/13/2022]
Abstract
Methamphetamine (Meth) abuse exacerbates HIV-1-associated neurocognitive disorders (HAND). The underlying mechanism for this effect is not entirely clear but likely involves cooperation between Meth and HIV-1 virotoxins, such as the transactivator of transcription, Tat. HIV-1 Tat mediates damage in the CNS by inducing inflammatory processes including astrogliosis. Wnt/β-catenin signaling regulates survival processes for both neurons and astrocytes. Here, we evaluated the impact of Meth on the Wnt/β-catenin pathway in astrocytes transfected with Tat. Meth and Tat downregulated Wnt/β-catenin signaling by >50%, as measured by TOPflash reporter activity in both an astrocytoma cell line and primary human fetal astrocytes. Meth and Tat also downregulated LEF-1 transcript by >30%. LEF-1 is a key partner of β-catenin to regulate cognate gene expression. Interestingly, estrogen, which induces β-catenin signaling in a cell-type specific manner, at physiological concentrations of 1.5 and 3 nM normalized individual Meth and Tat effects on β-catenin signaling but not their combined effects. These findings suggest that Meth and Tat likely exert different mechanisms to mediate down regulation of β-catenin signaling. The consequences of which may contribute to the pathophysiologic effects of HIV-1 and Meth co-morbidity in the CNS.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Immunology/Microbiology, Rush University Medical Center, 1735 W. Harrison Street, 614 Cohn, Chicago, IL 60612, USA
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA
| | - Xiu-Ti Hu
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center for AIDS Research, Rush University Medical Center, Chicago, IL, USA
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA
| | - T. Celeste Napier
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center for AIDS Research, Rush University Medical Center, Chicago, IL, USA
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA
| | - Lena Al-Harthi
- Department of Immunology/Microbiology, Rush University Medical Center, 1735 W. Harrison Street, 614 Cohn, Chicago, IL 60612, USA
- Chicago Center for AIDS Research, Rush University Medical Center, Chicago, IL, USA
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
86
|
Medina M, Garrido JJ, Wandosell FG. Modulation of GSK-3 as a Therapeutic Strategy on Tau Pathologies. Front Mol Neurosci 2011; 4:24. [PMID: 22007157 PMCID: PMC3186940 DOI: 10.3389/fnmol.2011.00024] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 08/30/2011] [Indexed: 12/29/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is ubiquitously expressed and unusually active in resting, non-stimulated cells. In mammals, at least three proteins (α, β1, and β2), generated from two different genes, gsk-3α and gsk-3β, are widely expressed at both the RNA and protein levels although some tissues show preferential expression of some of the three proteins. Control of GSK-3 activity occurs by complex mechanisms that depend on specific signaling pathways, often controlling the inhibition of the kinase activity. GSK-3 appears to integrate different signaling pathways from a wide selection of cellular stimuli. The unique position of GSK-3 in modulating the function of a diverse series of proteins and its association with a wide variety of human disorders has attracted significant attention as a therapeutic target and as a means to understand the molecular basis of brain disorders. Different neurodegenerative diseases including frontotemporal dementia, progressive supranuclear palsy, and Alzheimer's disease, present prominent tau pathology such as tau hyperphosphorylation and aggregation and are collectively referred to as tauopathies. GSK-3 has also been associated to different neuropsychiatric disorders, like schizophrenia and bipolar disorder. GSK-3β is the major kinase to phosphorylate tau both in vitro and in vivo and has been proposed as a target for therapeutic intervention. The first therapeutic strategy to modulate GSK-3 activity was the direct inhibition of its kinase activity. This review will focus on the signaling pathways involved in the control of GSK-3 activity and its pathological deregulation. We will highlight different alternatives of GSK-3 modulation including the direct pharmacological inhibition as compared to the modulation by upstream regulators.
Collapse
Affiliation(s)
- Miguel Medina
- Research Department, Noscira S.A., Tres Cantos Madrid, Spain
| | | | | |
Collapse
|
87
|
Marin R, Marrero-Alonso J, Fernández C, Cury D, Díaz M. Membrane-initiated signaling of estrogen related to neuroprotection. "Social networks" are required. Horm Mol Biol Clin Investig 2011; 7:393-401. [PMID: 25961340 DOI: 10.1515/hmbci.2011.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 06/06/2011] [Indexed: 11/15/2022]
Abstract
Numerous studies indicate that estrogens are crucial in normal brain functioning and preservation against different injuries. At the neuronal membrane, estrogens, binding to estrogen receptors (ERs) or other surface targets, exert rapid actions involving a plethora of signaling pathways that may converge in neuronal survival. Emerging work reveals that at least part of these actions may require the compartmentalization of ERs in signaling platforms, composed of macromolecular signaling proteins and particular lipid composition integrated in lipid rafts. These particular microstructures may provide the optimal microenvironment to trigger multiple ER interactions that may be crucial for neuroprotection against different brain impairments, such as Alzheimer's disease (AD). In this order of ideas, recent evidence has demonstrated that a membrane ER (mER) physically interacts with a voltage-dependent anion channel (VDAC) in lipid rafts from septal, hippocampal and cortical neurons, and these interactions may have important consequences in the alternative mechanisms developed by estrogens to achieve neuroprotection against amyloid beta (Aβ)-induced toxicity. This review includes a survey of some of the rapid mechanisms developed by estrogen to prevent neuronal death, and the ER interactions that are involved in the structural maintenance and signal transduction mechanisms important for neuronal survival against AD neuro-pathology. A special emphasis is put on the biological relevance of neuronal membrane VDAC in Aβ-related neurotoxicity, and the potential modulation of this channel as a part of a signaling complex with mER, which may be modified in AD brains.
Collapse
|
88
|
Etgen AM, Jover-Mengual T, Suzanne Zukin R. Neuroprotective actions of estradiol and novel estrogen analogs in ischemia: translational implications. Front Neuroendocrinol 2011; 32:336-52. [PMID: 21163293 PMCID: PMC3080451 DOI: 10.1016/j.yfrne.2010.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/15/2022]
Abstract
This review highlights our investigations into the neuroprotective efficacy of estradiol and other estrogenic agents in a clinically relevant animal model of transient global ischemia, which causes selective, delayed death of hippocampal CA1 neurons and associated cognitive deficits. We find that estradiol rescues a significant number of CA1 pyramidal neurons that would otherwise die in response to global ischemia, and this is true when hormone is provided as a long-term pretreatment at physiological doses or as an acute treatment at the time of reperfusion. In addition to enhancing neuronal survival, both forms of estradiol treatment induce measurable cognitive benefit in young animals. Moreover, estradiol and estrogen analogs that do not bind classical nuclear estrogen receptors retain their neuroprotective efficacy in middle-aged females deprived of ovarian hormones for a prolonged duration (8weeks). Thus, non-feminizing estrogens may represent a new therapeutic approach for treating the neuronal damage associated with global ischemia.
Collapse
Affiliation(s)
- Anne M. Etgen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
- Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Teresa Jover-Mengual
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Valencia, Campus de Burjassot, Avda. Vicent Andrés Estellés, s/n, 46100 Burjassot, Valencia SPAIN
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| |
Collapse
|
89
|
β-Catenin Is a Positive Regulator of Estrogen Receptor-α Function in Breast Cancer Cells. Cancers (Basel) 2011; 3:2990-3001. [PMID: 24212942 PMCID: PMC3759182 DOI: 10.3390/cancers3032990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/18/2011] [Accepted: 07/19/2011] [Indexed: 11/21/2022] Open
Abstract
Estrogen receptor-alpha (ERα) is a key factor in the development of breast cancer in humans. The expression and activity of ERα is regulated by a multitude of intracellular and extracellular signals. Here we show a cross-talk between β-catenin and ERα in human breast cancer cells. Knockdown of β-catenin by RNAi resulted in significant reduction of ERα mRNA and/or protein levels in MCF-7, T-47D, and BT-474 breast cancer cells and in significant reduction of estradiol-induced expression of the ERα target genes pS2 and GREB1. In addition β-catenin silencing resulted in significant decrease of growth of MCF-7 cells both in the absence and presence of estradiol. β-catenin and ERα could not be co-immunoprecipitated by ERα antibodies from lysates of E2-treated or untreated cells suggesting lack of direct physical interaction. It is concluded that β-catenin is a positive regulator of ERα mRNA and protein expression.
Collapse
|
90
|
Decrease of Tau hyperphosphorylation by 17β estradiol requires sphingosine kinase in a glutamate toxicity model. Neurochem Res 2011; 34:2206-14. [PMID: 19543831 DOI: 10.1007/s11064-009-0017-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2009] [Indexed: 01/08/2023]
Abstract
Several studies have linked estrogens with sphingosine kinase (SphK) activity, enzyme responsible of sphingosine-1-phosphate synthesis (S-1P), however their possible interaction in the nervous system is not documented yet. In the present study, we developed a glutamate toxicity model in SH-SY5Y cells to evaluate the possible effect of the inhibition of SphK activity on the protective capability of 17β-estradiol (E2). Glutamate induced cytoskeletal actin changes associated to cytotoxic stress, significant increase of apoptotic-like nuclear fragmentation, Tau hyperphosphorylation and increase of p25/p35 cleavage. These effects were prevented by E2 pre-treatment during 24 h. Although the inhibition of SphK did not block this protective effect, significantly increased Tau hyperphosphorylation by glutamate, in a way that was not reverted by E2. Our results suggest that the decrease of glutamate-induced Tau hyperphosphorylation by 17β-estradiol requires SphK.
Collapse
|
91
|
Wnt/Β-catenin and sex hormone signaling in endometrial homeostasis and cancer. Oncotarget 2011; 1:674-84. [PMID: 21317462 DOI: 10.18632/oncotarget.101007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
A delicate balance between estrogen and progestagen signaling underlies proper functioning of the female reproductive tract and, in particular, the monthly re- and degenerative phases characteristic of the menstrual cycle. Here, we propose that the canonical Wnt/β-catenin signaling pathway may underlie this finely tuned hormonal equilibrium in endometrial homeostasis and, upon its constitutive activation, lead to neoplastic transformation of the endometrium. During the menstrual cycle, estradiol will enhance Wnt/β-catenin signaling in the proliferative phase, while progesterone inhibits Wnt/β-catenin signaling, thus restraining estrogens' proliferative actions, during the secretory phase. In case of enhanced or unopposed estrogen signaling, constitutive activation of Wnt/β-catenin signaling will trigger endometrial hyperplasia, which may develop further into endometrial cancer.
Collapse
|
92
|
Abstract
While a distinct minicolumnar phenotype seems to be an underlying factor in a significant portion of cases of autism, great attention is being paid not only to genetics but to epigenetic factors which may lead to development of the conditions. Here we discuss the indivisible role the molecular environment plays in cellular function, particularly the pivotal position which the transcription factor and adhesion molecule, β-catenin, occupies in cellular growth. In addition, the learning environment is not only integral to postnatal plasticity, but the prenatal environment plays a vital role during corticogenesis, neuritogenesis, and synaptogenesis as well. To illustrate these points in the case of autism, we review important findings in genetics studies (e.g., PTEN, TSC1/2, FMRP, MeCP2, Neurexin-Neuroligin) and known epigenetic factors (e.g., valproic acid, estrogen, immune system, ultrasound) which may predispose towards the minicolumnar and connectivity patterns seen in the conditions, showing how one-gene mutational syndromes and exposure to certain CNS teratogens may ultimately lead to comparable phenotypes. This in turn may shed greater light on how environment and complex genetics combinatorially give rise to a heterogenetic group of conditions such as autism.
Collapse
Affiliation(s)
- Emily L. Williams
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY 40202, USA
| | - Manuel F. Casanova
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
93
|
Medina M, Wandosell F. Deconstructing GSK-3: The Fine Regulation of Its Activity. Int J Alzheimers Dis 2011; 2011:479249. [PMID: 21629747 PMCID: PMC3100567 DOI: 10.4061/2011/479249] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 02/28/2011] [Indexed: 01/12/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) unique position in modulating the function of a diverse series of proteins in combination with its association with a wide variety of human disorders has attracted significant attention to the protein both as a therapeutic target and as a means to understand the molecular basis of these disorders. GSK-3 is ubiquitously expressed and, unusually, constitutively active in resting, unstimulated cells. In mammals, GSK-3α and β are each expressed widely at both the RNA and protein levels although some tissues show preferential levels of some of the two proteins. Neither gene appears to be acutely regulated at the transcriptional level, whereas the proteins are controlled posttranslationally, largely through protein-protein interactions or by posttranslational regulation. Control of GSK-3 activity thus occurs by complex mechanisms that are each dependent upon specific signalling pathways. Furthermore, GSK-3 appears to be a cellular nexus, integrating several signalling systems, including several second messengers and a wide selection of cellular stimulants. This paper will focus on the different ways to control GSK-3 activity (phosphorylation, protein complex formation, truncation, subcellular localization, etc.), the main signalling pathways involved in its control, and its pathological deregulation.
Collapse
|
94
|
Zhang Z, Zhao R, Qi J, Wen S, Tang Y, Wang D. Inhibition of glycogen synthase kinase-3β by Angelica sinensis extract decreases β-amyloid-induced neurotoxicity and tau phosphorylation in cultured cortical neurons. J Neurosci Res 2010; 89:437-47. [DOI: 10.1002/jnr.22563] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 10/08/2010] [Accepted: 11/01/2010] [Indexed: 12/28/2022]
|
95
|
Wang Y, van der Zee M, Fodde R, Blok LJ. Wnt/Β-catenin and sex hormone signaling in endometrial homeostasis and cancer. Oncotarget 2010; 1:674-684. [PMID: 21317462 PMCID: PMC3248134 DOI: 10.18632/oncotarget.201] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 10/11/2010] [Indexed: 11/25/2022] Open
Abstract
A delicate balance between estrogen and progestagen signaling underlies proper functioning of the female reproductive tract and, in particular, the monthly re- and degenerative phases characteristic of the menstrual cycle. Here, we propose that the canonical Wnt/β-catenin signaling pathway may underlie this finely tuned hormonal equilibrium in endometrial homeostasis and, upon its constitutive activation, lead to neoplastic transformation of the endometrium. During the menstrual cycle, estradiol will enhance Wnt/β-catenin signaling in the proliferative phase, while progesterone inhibits Wnt/β-catenin signaling, thus restraining estrogens' proliferative actions, during the secretory phase. In case of enhanced or unopposed estrogen signaling, constitutive activation of Wnt/β-catenin signaling will trigger endometrial hyperplasia, which may develop further into endometrial cancer.
Collapse
Affiliation(s)
- Yongyi Wang
- Department of Obstetrics & Gynaecology, Josephine Nefkens Institute, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Departments of Pathology, Josephine Nefkens Institute, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Marten van der Zee
- Department of Obstetrics & Gynaecology, Josephine Nefkens Institute, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Departments of Pathology, Josephine Nefkens Institute, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Riccardo Fodde
- Departments of Pathology, Josephine Nefkens Institute, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Leen J Blok
- Department of Obstetrics & Gynaecology, Josephine Nefkens Institute, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
96
|
Piedrahita D, Hernández I, López-Tobón A, Fedorov D, Obara B, Manjunath BS, Boudreau RL, Davidson B, LaFerla F, Gallego-Gómez JC, Kosik KS, Cardona-Gómez GP. Silencing of CDK5 reduces neurofibrillary tangles in transgenic alzheimer's mice. J Neurosci 2010; 30:13966-76. [PMID: 20962218 PMCID: PMC3003593 DOI: 10.1523/jneurosci.3637-10.2010] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/24/2010] [Accepted: 08/26/2010] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease is a major cause of dementia for which treatments remain unsatisfactory. Cyclin-dependent kinase 5 (CDK5) is a relevant kinase that has been hypothesized to contribute to the tau pathology. Several classes of chemical inhibitors for CDK5 have been developed, but they generally lack the specificity to distinguish among various ATP-dependent kinases. Therefore, the efficacy of these compounds when tested in animal models cannot definitively be attributed to an effect on CDK5. However, RNA interference (RNAi) targeting of CDK5 is specific and can be used to validate CDK5 as a possible treatment target. We delivered a CDK5 RNAi by lentiviral or adenoassociated viral vectors and analyzed the results in vitro and in vivo. Silencing of CDK5 reduces the phosphorylation of tau in primary neuronal cultures and in the brain of wild-type C57BL/6 mice. Furthermore, the knockdown of CDK5 strongly decreased the number of neurofibrillary tangles in the hippocampi of triple-transgenic mice (3×Tg-AD mice). Our data suggest that this downregulation may be attributable to the reduction of the CDK5 availability in the tissue, without affecting the CDK5 kinase activity. In summary, our findings validate CDK5 as a reasonable therapeutic target for ameliorating tau pathology.
Collapse
Affiliation(s)
- Diego Piedrahita
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, AA 1226 Medellin, Colombia
| | - Israel Hernández
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, AA 1226 Medellin, Colombia
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California 93106
| | - Alejandro López-Tobón
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, AA 1226 Medellin, Colombia
| | | | - Boguslaw Obara
- Oxford e-Research Centre and Oxford Centre for Integrative Systems Biology, University of Oxford, Oxford OX1 3QG, United Kingdom
| | | | - Ryan L. Boudreau
- Viral Vector Core and Davidson Laboratory, University of Iowa, Iowa City, Iowa 52242, and
| | - Beverly Davidson
- Viral Vector Core and Davidson Laboratory, University of Iowa, Iowa City, Iowa 52242, and
| | - Frank LaFerla
- Institute for Brain Aging and Dementia, University of California, Irvine, Irvine, California 92697
| | - Juan Carlos Gallego-Gómez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, AA 1226 Medellin, Colombia
| | - Kenneth S. Kosik
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California 93106
| | - Gloria Patricia Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, AA 1226 Medellin, Colombia
| |
Collapse
|
97
|
Mukai H, Kimoto T, Hojo Y, Kawato S, Murakami G, Higo S, Hatanaka Y, Ogiue-Ikeda M. Modulation of synaptic plasticity by brain estrogen in the hippocampus. Biochim Biophys Acta Gen Subj 2010; 1800:1030-44. [DOI: 10.1016/j.bbagen.2009.11.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 10/15/2009] [Accepted: 11/02/2009] [Indexed: 12/31/2022]
|
98
|
Varea O, Arevalo MA, Garrido JJ, Garcia-Segura LM, Wandosell F, Mendez P. Interaction of estrogen receptors with insulin-like growth factor-I and Wnt signaling in the nervous system. Steroids 2010; 75:565-9. [PMID: 19778547 DOI: 10.1016/j.steroids.2009.09.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
Abstract
Estradiol signaling through estrogen receptors in the nervous system involves a variety of rapid membrane/cytoplasm-initiated events that are integrated with different mechanisms of transcriptional regulation. Here we review the role of glycogen synthase kinase 3 (GSK3) and beta-catenin in the coordination of membrane/cytoplasm-initiated and nuclear-initiated estrogen receptor signaling. Estradiol activates in vitro and in vivo the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway in neural cells. By activating this pathway through estrogen receptors, estradiol increases the levels of inactive GSK3beta (phosphorylated in serine 9). In turn, the inhibition of GSK3beta increases the stability of beta-catenin and its nuclear translocation. Then, beta-catenin exerts two different transcriptional effects: (i) regulates beta-catenin/T cell factor (TCF) mediated transcription in a similar but not identical way as Wnt ligands and (ii) regulates estrogen receptor mediated transcription after its association with estrogen receptor alpha. In addition, by the regulation of PI3K/Akt/GSK3/beta-catenin pathway, other factors such as insulin-like growth factor-I (IGF-I) regulate estrogen receptor mediated transcription. Therefore, GSK3 and beta-catenin allow the interaction of membrane/cytoplasm-initiated estrogen receptor signaling, IGF-I signaling, Wnt signaling and nuclear-initiated estrogen receptor signaling in the nervous system.
Collapse
Affiliation(s)
- Olga Varea
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) and Centro de Biología Molecular Severo Ochoa, CSIC-UAM, E-28049 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
99
|
Protective effect of isoflavones against homocysteine-mediated neuronal degeneration in SH-SY5Y cells. Amino Acids 2010; 39:785-94. [DOI: 10.1007/s00726-010-0523-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 02/10/2010] [Indexed: 01/02/2023]
|
100
|
Abstract
The ERalpha signaling pathway is one of the most important and most studied pathways in human breast cancer, yet numerous questions still exist such as how hormonally responsive cancers progress to a more aggressive and hormonally independent phenotype. We have noted that human breast cancers exhibit a strong direct correlation between ERalpha and E-cadherin expression by immunohistochemistry, suggesting that ERalpha signaling might regulate E-cadherin and implying that this regulation might influence epithelial-mesenchymal transition (EMT) and tumor progression. To investigate this hypothesis and the mechanisms behind it, we studied the effects of ERalpha signaling in ERalpha-transfected ERalpha-negative breast carcinoma cell lines, the MDA-MB-468 and the MDA-MB-231 and the effects of ERalpha knockdown in naturally expressing ERalpha-positive lines, MCF-7 and T47D. When ERalpha was overexpressed in the ERalpha-negative lines, 17beta-estradiol (E2) decreased slug and increased E-cadherin. Clones maximally exhibiting these changes grew more in clumps and became less invasive in Matrigel. When ERalpha was knocked down in the ERalpha-positive lines, slug increased, E-cadherin decreased, cells became spindly and exhibited increased Matrigel invasion. ERalpha signaling decreased slug expression by two different mechanisms: directly, by repression of slug transcription by the formation of a corepressor complex of ligand-activated ERalpha, HDAC inhibitor (HDAC1), and nuclear receptor corepressor (N-CoR) that bound the slug promoter in three half-site estrogen response elements (EREs); indirectly by phosphorylation and inactivation of GSK-3beta through phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt). The GSK-3beta inactivation, in turn, repressed slug expression and increased E-cadherin. In human breast cancer cases, there was a strong inverse correlation between slug and ERalpha and E-cadherin immunoreactivity. Our findings indicate that ERalpha signaling through slug regulates E-cadherin and EMT.
Collapse
|