1
|
Shapira G, Karmon G, Hacohen-Kleiman G, Ganaiem M, Shazman S, Theotokis P, Grigoriadis N, Shomron N, Gozes I. ADNP is essential for sex-dependent hippocampal neurogenesis, through male unfolded protein response and female mitochondrial gene regulation. Mol Psychiatry 2024:10.1038/s41380-024-02879-w. [PMID: 39715923 DOI: 10.1038/s41380-024-02879-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/20/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Essential for brain formation and protective against tauopathy, activity-dependent neuroprotective protein (ADNP) is critical for neurogenesis and cognitive functions, while regulating steroid hormone biogenesis. As such, de novo mutations in ADNP lead to syndromic autism and somatic ADNP mutations parallel Alzheimer's disease progression. Furthermore, clinical trials with the ADNP fragment NAP (the investigational drug davunetide) showed efficacy in women suffering from the tauopathy progressive supranuclear palsy and differentially boosted memory in men (spatial) and women (verbal), exhibiting prodromal Alzheimer's disease. While autism is more prevalent in boys and Alzheimer's disease in women, both involve impaired neurogenesis. Here, we asked whether ADNP sex-dependently regulates neurogenesis. Using bromodeoxyuridine (BrdU) as a marker of neurogenesis, we identified two-fold higher labeling in the hippocampal sub-ventricular zone of ADNP-intact male versus female mice. Adnp haplo-insufficient (Adnp+/-) mice or mice CRSIPR/Cas9-edited to present the most prevalent neurodevelopmental ADNP syndrome mutation, p.Tyr718* (Tyr) showed dramatic reductions in male BrdU incorporation, resulting in mutated females presenting higher labeling than males. Treatment with NAP compensated for the male reduction of BrdU labeling. Mechanistically, hippocampal RNAseq revealed male-specific Tyr down-regulation of endoplasmic reticulum unfolded protein response genes critical for sex-dependent organogenesis. Newly discovered mitochondrial accessibility of ADNP was inhibited by the Tyr718* mutation further revealing female-specific Tyr downregulation of mitochondrial ATP6. NAP moderated much of the differential expression caused by p.Tyr718*, accompanied by the down-regulation of neurotoxic, pro-inflammatory and pro-apoptotic genes. Thus, ADNP is a key regulator of sex-dependent neurogenesis that acts by controlling canonical pathways, with NAP compensating for fundamental ADNP deficiencies, striding toward clinical development targeting the ADNP syndrome and related neurodevelopmental/neurodegenerative diseases.
Collapse
Affiliation(s)
- Guy Shapira
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Sagol School of Neuroscience, Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Gidon Karmon
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Gal Hacohen-Kleiman
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Maram Ganaiem
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Shula Shazman
- Department of Mathematics and Computer Science, The Open University of Israel, Ra'anana, 4353701, Israel
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Noam Shomron
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Sagol School of Neuroscience, Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
2
|
Thanh DD, Bich-Ngoc N, Paques C, Christian A, Herkenne S, Struman I, Muller M. The food dye Tartrazine disrupts vascular formation both in zebrafish larvae and in human primary endothelial cells. Sci Rep 2024; 14:30367. [PMID: 39639097 PMCID: PMC11621646 DOI: 10.1038/s41598-024-82076-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Tartrazine (E102) is a controversial coloring agent whose potential impacts on human health are not fully understood. Our study reveals the vascular disrupting effects of tartrazine (TTZ) on developing zebrafish embryos in vivo and on human umbilical vein endothelial cells in vitro. The dye was shown to cause dose-dependent hemorrhages in zebrafish embryos. Analyzing transgenic zebrafish harboring fluorescent endothelial cells revealed that TTZ treatment disrupted cell organization into vessels in both the sub-intestinal vein and the brain area. Assays on human umbilical vein endothelial cells demonstrated that TTZ inhibited endothelial proliferation, tube formation, and migration in a dose-dependent manner. Taken together, our results indicate for the first time that TTZ can affect endothelial cell properties, possibly by disrupting Rho family GTPase pathways which control the cytoskeleton. Our finding provides a credible explanation for many reported human health impacts and offers prospective applications for biomedicine.
Collapse
Affiliation(s)
- Dinh Duy Thanh
- Lab. for Organogenesis and Regeneration, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
- Department of Cell Biology, Faculty of Biology, VNU University of Science, Hanoi, 100000, Vietnam
| | - Nguyen Bich-Ngoc
- VNU School of Interdisciplinary Sciences and Arts, Vietnam National University, Hanoi, 100000, Vietnam
| | - Cécile Paques
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Aurélie Christian
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Stéphanie Herkenne
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Ingrid Struman
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Marc Muller
- Lab. for Organogenesis and Regeneration, GIGA-Institute, Université de Liège, Liège, 4000, Belgium.
| |
Collapse
|
3
|
Molinaro G, Bowles JE, Croom K, Gonzalez D, Mirjafary S, Birnbaum SG, Razak KA, Gibson JR, Huber KM. Female-specific dysfunction of sensory neocortical circuits in a mouse model of autism mediated by mGluR5 and estrogen receptor α. Cell Rep 2024; 43:114056. [PMID: 38581678 PMCID: PMC11112681 DOI: 10.1016/j.celrep.2024.114056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/26/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024] Open
Abstract
Little is known of the brain mechanisms that mediate sex-specific autism symptoms. Here, we demonstrate that deletion of the autism spectrum disorder (ASD)-risk gene, Pten, in neocortical pyramidal neurons (NSEPten knockout [KO]) results in robust cortical circuit hyperexcitability selectively in female mice observed as prolonged spontaneous persistent activity states. Circuit hyperexcitability in females is mediated by metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) signaling to mitogen-activated protein kinases (Erk1/2) and de novo protein synthesis. Pten KO layer 5 neurons have a female-specific increase in mGluR5 and mGluR5-dependent protein synthesis. Furthermore, mGluR5-ERα complexes are generally elevated in female cortices, and genetic reduction of ERα rescues enhanced circuit excitability, protein synthesis, and neuron size selectively in NSEPten KO females. Female NSEPten KO mice display deficits in sensory processing and social behaviors as well as mGluR5-dependent seizures. These results reveal mechanisms by which sex and a high-confidence ASD-risk gene interact to affect brain function and behavior.
Collapse
Affiliation(s)
- Gemma Molinaro
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jacob E Bowles
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Katilynne Croom
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, USA
| | - Darya Gonzalez
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Saba Mirjafary
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shari G Birnbaum
- Department of Psychiatry, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, USA; Department of Psychology, University of California, Riverside, Riverside, CA, USA
| | - Jay R Gibson
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kimberly M Huber
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Molinaro G, Bowles JE, Croom K, Gonzalez D, Mirjafary S, Birnbaum S, Razak KA, Gibson JR, Huber KM. Female specific dysfunction of sensory neocortical circuits in a mouse model of autism mediated by mGluR5 and Estrogen Receptor α. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.10.552857. [PMID: 37609208 PMCID: PMC10441407 DOI: 10.1101/2023.08.10.552857] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Autism manifests differently in males and females and the brain mechanisms that mediate these sex-dependent differences are unknown. Here, we demonstrate that deletion of the ASD-risk gene, Pten, in neocortical pyramidal neurons (NSE Pten KO) results in robust hyperexcitability of local neocortical circuits in female, but not male, mice, observed as prolonged, spontaneous persistent activity states (UP states). Circuit hyperexcitability in NSE Pten KO mice is mediated by enhanced and/or altered signaling of metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) to ERK and protein synthesis selectively in Pten deleted female neurons. In support of this idea, Pten deleted Layer 5 cortical neurons have female-specific increases in mGluR5 and mGluR5-driven protein synthesis. In addition, mGluR5-ERα complexes are elevated in female cortex and genetic reduction of ERα in Pten KO cortical neurons rescues circuit excitability, protein synthesis and enlarged neurons selectively in females. Abnormal timing and hyperexcitability of neocortical circuits in female NSE Pten KO mice are associated with deficits in temporal processing of sensory stimuli and social behaviors as well as mGluR5-dependent seizures. Female-specific cortical hyperexcitability and mGluR5-dependent seizures are also observed in a human disease relevant mouse model, germline Pten +/- mice. Our results reveal molecular mechanisms by which sex and a high impact ASD-risk gene interact to affect brain function and behavior.
Collapse
|
5
|
López-Merino E, Cuartero MI, Esteban JA, Briz V. Perinatal exposure to pesticides alters synaptic plasticity signaling and induces behavioral deficits associated with neurodevelopmental disorders. Cell Biol Toxicol 2023; 39:2089-2111. [PMID: 35137321 PMCID: PMC10547633 DOI: 10.1007/s10565-022-09697-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
Increasing evidence from animal and epidemiological studies indicates that perinatal exposure to pesticides cause developmental neurotoxicity and may increase the risk for psychiatric disorders such as autism and intellectual disability. However, the underlying pathogenic mechanisms remain largely elusive. This work was aimed at testing the hypothesis that developmental exposure to different classes of pesticides hijacks intracellular neuronal signaling contributing to synaptic and behavioral alterations associated with neurodevelopmental disorders (NDD). Low concentrations of organochlorine (dieldrin, endosulfan, and chlordane) and organophosphate (chlorpyrifos and its oxon metabolite) pesticides were chronically dosed ex vivo (organotypic rat hippocampal slices) or in vivo (perinatal exposure in rats), and then biochemical, electrophysiological, behavioral, and proteomic studies were performed. All the pesticides tested caused prolonged activation of MAPK/ERK pathway in a concentration-dependent manner. Additionally, some of them impaired metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD). In the case of the pesticide chlordane, the effect was attributed to chronic modulation of MAPK/ERK signaling. These synaptic alterations were reproduced following developmental in vivo exposure to chlordane and chlorpyrifos-oxon, and were also associated with prototypical behavioral phenotypes of NDD, including impaired motor development, increased anxiety, and social and memory deficits. Lastly, proteomic analysis revealed that these pesticides differentially regulate the expression of proteins in the hippocampus with pivotal roles in brain development and synaptic signaling, some of which are associated with NDD. Based on these results, we propose a novel mechanism of synaptic dysfunction, involving chronic overactivation of MAPK and impaired mGluR-LTD, shared by different pesticides which may have important implications for NDD.
Collapse
Affiliation(s)
| | - María I Cuartero
- Neurovascular Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José A Esteban
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
| | - Víctor Briz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
6
|
Mohanty S, Kamolvit W, Scheffschick A, Björklund A, Tovi J, Espinosa A, Brismar K, Nyström T, Schröder JM, Östenson CG, Aspenström P, Brauner H, Brauner A. Diabetes downregulates the antimicrobial peptide psoriasin and increases E. coli burden in the urinary bladder. Nat Commun 2022; 13:4983. [PMID: 36127330 PMCID: PMC9489794 DOI: 10.1038/s41467-022-32636-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Diabetes is known to increase susceptibility to infections, partly due to impaired granulocyte function and changes in the innate immunity. Here, we investigate the effect of diabetes, and high glucose on the expression of the antimicrobial peptide, psoriasin and the putative consequences for E. coli urinary tract infection. Blood, urine, and urine exfoliated cells from patients are studied. The influence of glucose and insulin is examined during hyperglycemic clamps in individuals with prediabetes and in euglycemic hyperinsulinemic clamped patients with type 1 diabetes. Important findings are confirmed in vivo in type 2 diabetic mice and verified in human uroepithelial cell lines. High glucose concentrations induce lower psoriasin levels and impair epithelial barrier function together with altering cell membrane proteins and cytoskeletal elements, resulting in increasing bacterial burden. Estradiol treatment restores the cellular function with increasing psoriasin and bacterial killing in uroepithelial cells, confirming its importance during urinary tract infection in hyperglycemia. In conclusion, our findings present the effects and underlying mechanisms of high glucose compromising innate immunity.
Collapse
Affiliation(s)
- Soumitra Mohanty
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Division of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Witchuda Kamolvit
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Division of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Anneli Björklund
- Center for Diabetes, Academic Specialist Center, Stockholm County Council, Solna, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Division of Internal Medicine, Unit for Diabetes Research, Karolinska Institutet, South Hospital, Stockholm, Sweden
| | - Jens M Schröder
- Department of Dermatology, Venerology and Allergology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Claes-Göran Östenson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Pontus Aspenström
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Hanna Brauner
- Department of Medicine, Solna, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Annelie Brauner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Division of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
7
|
Brökling J, Brunne B, Rune GM. Sex-dependent responsiveness of hippocampal neurons to sex neurosteroids: A role of Arc/Arg3.1. J Neuroendocrinol 2022; 34:e13090. [PMID: 35081672 DOI: 10.1111/jne.13090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 11/28/2022]
Abstract
Sex steroids, such as estradiol (E2 ) and dihydrotestosterone (DHT), regulate hippocampal plasticity and memory in a sex-dependent manner. Because the activity-regulated cytoskeleton protein Arc/Arg3.1 is essential for long-term memory formation and synaptic plasticity, we investigated the expression of Arc/Arg3.1 with respect to its responsiveness to E2 and DHT in male and female hippocampal neurons. For the first time, we show that, in hippocampal neurons, Arc/Arg3.1 expression is sex-dependently regulated by sex steroids. No difference in the expression between sexes was observed under control conditions. Using a quantitative real-time polymerase chain reaction, western blot analysis and quantitative immunoreactivity, upregulation of Arc/Arg3.1 protein expression was observed in specifically female hippocampal neurons after application of E2 to the cultures. Conversely, upregulation of Arc/Arg3.1 was seen in specifically male neurons after application of DHT. A quantitative real-time PCR revealed that the sex-dependency was most pronounced on the mRNA level. Most importantly, the effects of E2 in cultures of female animals were abolished when neuron-derived E2 synthesis was inhibited. Our results point to a potentially important role of Arc/Arg3.1 regarding sex-dependency in sex steroid-induced synaptic plasticity in the hippocampus.
Collapse
Affiliation(s)
- Janina Brökling
- Institute of Neuroanatomy, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Bianka Brunne
- Institute of Neuroanatomy, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Gabriele M Rune
- Institute of Cell Biology and Neurobiology, Universitätsmedizin Charité Berlin, Berlin, Germany
| |
Collapse
|
8
|
Beamish SB, Frick KM. A Putative Role for Ubiquitin-Proteasome Signaling in Estrogenic Memory Regulation. Front Behav Neurosci 2022; 15:807215. [PMID: 35145382 PMCID: PMC8821141 DOI: 10.3389/fnbeh.2021.807215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/20/2021] [Indexed: 12/31/2022] Open
Abstract
Sex steroid hormones such as 17β-estradiol (E2) are critical neuromodulators of hippocampal synaptic plasticity and hippocampus-dependent memory in both males and females. However, the mechanisms through which E2 regulates memory formation in both sexes remain unclear. Research to date suggests that E2 regulates hippocampus-dependent memory by activating numerous cell-signaling cascades to promote the synthesis of proteins that support structural changes at hippocampal synapses. However, this work has largely overlooked the equally important contributions of protein degradation mediated by the ubiquitin proteasome system (UPS) in remodeling the synapse. Despite being critically implicated in synaptic plasticity and successful formation of long-term memories, it remains unclear whether protein degradation mediated by the UPS is necessary for E2 to exert its beneficial effects on hippocampal plasticity and memory formation. The present article provides an overview of the receptor and signaling mechanisms so far identified as critical for regulating hippocampal E2 and UPS function in males and females, with a particular emphasis on the ways in which these mechanisms overlap to support structural integrity and protein composition of hippocampal synapses. We argue that the high degree of correspondence between E2 and UPS activity warrants additional study to examine the contributions of ubiquitin-mediated protein degradation in regulating the effects of sex steroid hormones on cognition.
Collapse
|
9
|
Feng Y, Shi R, Hu J, Lou S. Effects of neural-derived estradiol on actin polymerization and synaptic plasticity-related proteins in prefrontal and hippocampal cells of mice. Steroids 2022; 177:108935. [PMID: 34715132 DOI: 10.1016/j.steroids.2021.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
Neural-derived 17β-estradiol (E2) plays an important role in the synaptic plasticity of the hippocampus and prefrontal cortex, but the mechanism is not well defined. This study was designed to explore the effect and mechanism of neural-derived E2 on synaptic plasticity of the hippocampus and prefrontal cortex. Primary cultured hippocampal and prefrontal cells in mice were randomly divided into the DMSO (D), aromatase (Rate-limiting enzymes for E2 synthesizes) inhibitor letrozole (L), and ERs antagonist (MPG) treated groups. After intervention for 48 h, the cell was collected, and then, the expressions of AMPA-receptor subunit GluR1 (GluR1), synaptophysin (SYN), p-21-Activated kinase (PAK) phosphorylation, Rho kinase (ROCK), p-Cofilin, F-actin, and G-actin proteins were detected. Letrozole or ER antagonists inhibited the expression of GluR1, F-actin/G-actin, p-PAK and p-Cofilin proteins in prefrontal cells significantly. And the expressions of GluR1 and F-actin/G-actin proteins were declined in hippocampal cells markedly after adding letrozole or ERs antagonists. In conclusion, neural-derived E2 and ERs regulated the synaptic plasticity, possibly due to promoting actin polymerization in prefrontal and hippocampal cells. The regional specificity in the effect of neural-derived E2 and ERs on the actin polymerization-related pathway may provide a theoretical basis for the functional differences between the hippocampus and prefrontal cortex.
Collapse
Affiliation(s)
- Yu Feng
- Shanghai University of Sport, Kinesiology, Shanghai, China
| | - Rengfei Shi
- Shanghai University of Sport, Kinesiology, Shanghai, China
| | - Jingyun Hu
- Shanghai University of Sport, Kinesiology, Shanghai, China
| | - Shujie Lou
- Shanghai University of Sport, Kinesiology, Shanghai, China.
| |
Collapse
|
10
|
From Menopause to Neurodegeneration-Molecular Basis and Potential Therapy. Int J Mol Sci 2021; 22:ijms22168654. [PMID: 34445359 PMCID: PMC8395405 DOI: 10.3390/ijms22168654] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
The impacts of menopause on neurodegenerative diseases, especially the changes in steroid hormones, have been well described in cell models, animal models, and humans. However, the therapeutic effects of hormone replacement therapy on postmenopausal women with neurodegenerative diseases remain controversial. The steroid hormones, steroid hormone receptors, and downstream signal pathways in the brain change with aging and contribute to disease progression. Estrogen and progesterone are two steroid hormones which decline in circulation and the brain during menopause. Insulin-like growth factor 1 (IGF-1), which plays an import role in neuroprotection, is rapidly decreased in serum after menopause. Here, we summarize the actions of estrogen, progesterone, and IGF-1 and their signaling pathways in the brain. Since the incidence of Alzheimer’s disease (AD) is higher in women than in men, the associations of steroid hormone changes and AD are emphasized. The signaling pathways and cellular mechanisms for how steroid hormones and IGF-1 provide neuroprotection are also addressed. Finally, the molecular mechanisms of potential estrogen modulation on N-methyl-d-aspartic acid receptors (NMDARs) are also addressed. We provide the viewpoint of why hormone therapy has inconclusive results based on signaling pathways considering their complex response to aging and hormone treatments. Nonetheless, while diagnosable AD may not be treatable by hormone therapy, its preceding stage of mild cognitive impairment may very well be treatable by hormone therapy.
Collapse
|
11
|
Li X, Johann S, Rune GM, Bender RA. Sex-specific Regulation of Spine Density and Synaptic Proteins by G-protein-coupled Estrogen Receptor (GPER)1 in Developing Hippocampus. Neuroscience 2021; 472:35-50. [PMID: 34364953 DOI: 10.1016/j.neuroscience.2021.07.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/19/2022]
Abstract
G-protein-coupled-estrogen-receptor 1 (GPER1) is a membrane-bound receptor that mediates estrogen signaling via intracellular signaling cascades. We recently showed that GPER1 promotes the distal dendritic enrichment of hyperpolarization activated and cyclic nucleotide-gated (HCN)1 channels in CA1 stratum lacunosum-moleculare (SLM), suggesting a role of GPER1-mediated signaling in neuronal plasticity. Here we studied whether this role involves processes of structural plasticity, such as the regulation of spine and synapse density in SLM. In organotypic entorhino-hippocampal cultures from mice expressing eGFP, we analyzed spine densities in SLM after treatment with GPER1 agonist G1 (20 nM). G1 significantly increased the density of "non-stubby" spines (maturing spines with a spine head and a neck), but did so only in cultures from female mice. In support of this finding, the expression of synaptic proteins was sex-specifically altered in the cultures: G1 increased the protein (but not mRNA) expression of PSD95 and reduced the p-/n-cofilin ratio only in cultures from females. Application of E2 (2 nM) reproduced the sex-specific effect on spine density in SLM, but only partially on the expression of synaptic proteins. Spine synapse density was, however, not altered after G1-treatment, suggesting that the increased spine density did not translate into an increased spine synapse density in the culture model. Taken together, our results support a role of GPER1 in mediating structural plasticity in CA1 SLM, but suggest that in developing hippocampus, this role is sex-specific.
Collapse
Affiliation(s)
- Xiaoyu Li
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Roland A Bender
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany.
| |
Collapse
|
12
|
Sheppard PAS, Asling HA, Walczyk-Mooradally A, Armstrong SE, Elad VM, Lalonde J, Choleris E. Protein synthesis and actin polymerization in the rapid effects of 17β-estradiol on short-term social memory and dendritic spine dynamics in female mice. Psychoneuroendocrinology 2021; 128:105232. [PMID: 33892375 DOI: 10.1016/j.psyneuen.2021.105232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/04/2021] [Accepted: 04/10/2021] [Indexed: 11/28/2022]
Abstract
Estrogens rapidly facilitate learning and memory, including social recognition - the ability of an animal to recognize another. In ovariectomized female mice, systemic or dorsal hippocampal administration of 17β-estradiol (E2) facilitates short-term social recognition memory within 40 min. Within the same timeframe, E2 increases dendritic spine density in CA1 dorsal hippocampal neurons of behavioural task-naïve mice and in hippocampal sections. Mechanisms underlying these effects remain unclear. Estrogens rapidly modulate actin cytoskeletal dynamics through actin polymerization and the translation of key synaptic proteins. We first determined doses of actin polymerization inhibitor latrunculin A (LAT) and protein synthesis inhibitor anisomycin (ANI) that would block short-term social recognition memory when infused into the dorsal hippocampus of ovariectomized female mice 15 min prior to testing. The highest doses that did not block social recognition prevented the facilitating effects of E2, whereas DNA transcription inhibitor, actinomycin D, could not block social recognition. As task performance may interfere with E2-facilitated increases in dendritic spine density, dendritic spine density and length were examined in task-performing and task-naïve mice. E2 increased dendritic spine density 15 but not 40 min following treatment, regardless of whether the animal had performed the social recognition task. This effect was blocked by LAT, but not ANI. Thus, both actin polymerization and protein synthesis are necessary for E2 to rapidly facilitate social recognition, whereas actin polymerization, but not protein synthesis, is required for the rapid increase in dendritic spine density brought on by E2.
Collapse
Affiliation(s)
- Paul A S Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Hayley A Asling
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | - Sabrina E Armstrong
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Vissy M Elad
- Department of Human Health and Nutrition Sciences, University of Guelph, Guelph, ON, Canada; Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
13
|
Rial D, Puighermanal E, Chazalon M, Valjent E, Schiffmann SN, de Kerchove d'Exaerde A. Mammalian Target of Rapamycin-RhoA Signaling Impairments in Direct Striatal Projection Neurons Induce Altered Behaviors and Striatal Physiology in Mice. Biol Psychiatry 2020; 88:945-954. [PMID: 32711953 DOI: 10.1016/j.biopsych.2020.05.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND As an integrator of molecular pathways, mTOR (mammalian target of rapamycin) has been associated with diseases including neurodevelopmental, psychiatric, and neurodegenerative disorders such as autism spectrum disorder, schizophrenia, and Huntington's disease. An important brain area involved in all these diseases is the striatum. However, the mechanisms behind how mTOR is involved in striatal physiology and its relative role in distinct neuronal populations in these striatal-related diseases still remain to be clarified. METHODS Using Drd1-Cre mTOR-conditional knockout male mice, we combined behavioral, biochemical, electrophysiological, and morphological analysis aiming to untangle the role of mTOR in direct pathway striatal projection neurons and how this would impact on striatal physiology. RESULTS Our results indicate deep behavioral changes in absence of mTOR in Drd1-expressing neurons such as decreased spontaneous locomotion, impaired social interaction, and decreased marble-burying behavior. These alterations were accompanied by a Kv1.1-induced increase in the fast phase of afterhyperpolarization and coincident decreased distal spine density in striatal direct pathway striatal projection neurons. The physiological changes were mechanistically independent of protein synthesis but sensitive to pharmacological blockade of transforming protein RhoA activity. CONCLUSIONS These results identify mTOR signaling as an important regulator of striatal functions through an intricate mechanism involving RhoA and culminating in Kv1.1 overfunction, which could be targeted to treat striatal-related monogenic disorders associated with the mTOR signaling pathway.
Collapse
Affiliation(s)
- Daniel Rial
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emma Puighermanal
- Institut de Génétique Foncionnelle (IGF), Centre National de la Recherche Scientifique (CNRS), (Institut National de la Santé et de la Recherche Médicale (INSERM), University of Montpellier, Montpellier, France
| | - Marine Chazalon
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emmanuel Valjent
- Institut de Génétique Foncionnelle (IGF), Centre National de la Recherche Scientifique (CNRS), (Institut National de la Santé et de la Recherche Médicale (INSERM), University of Montpellier, Montpellier, France
| | - Serge N Schiffmann
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alban de Kerchove d'Exaerde
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
14
|
Estradiol Induces Epithelial to Mesenchymal Transition of Human Glioblastoma Cells. Cells 2020; 9:cells9091930. [PMID: 32825553 PMCID: PMC7564468 DOI: 10.3390/cells9091930] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/02/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
The mesenchymal phenotype of glioblastoma multiforme (GBM), the most frequent and malignant brain tumor, is associated with the worst prognosis. The epithelial–mesenchymal transition (EMT) is a cell plasticity mechanism involved in GBM malignancy. In this study, we determined 17β-estradiol (E2)-induced EMT by changes in cell morphology, expression of EMT markers, and cell migration and invasion assays in human GBM-derived cell lines. E2 (10 nM) modified the shape and size of GBM cells due to a reorganization of actin filaments. We evaluated EMT markers expression by RT-qPCR, Western blot, and immunofluorescence.We found that E2 upregulated the expression of the mesenchymal markers, vimentin, and N-cadherin. Scratch and transwell assays showed that E2 increased migration and invasion of GBM cells. The estrogen receptor-α (ER-α)-selective agonist 4,4’,4’’-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT, 10 nM) affected similarly to E2 in terms of the expression of EMT markers and cell migration, and the treatment with the ER-α antagonist methyl-piperidino-pyrazole (MPP, 1 μM) blocked E2 and PPT effects. ER-β-selective agonist diarylpropionitrile (DNP, 10 nM) and antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazole[1,5-a]pyrimidin-3-yl]phenol (PHTPP, 1 μM) showed no effects on EMT marker expression. These data suggest that E2 induces EMT activation through ER-α in human GBM-derived cells.
Collapse
|
15
|
McLellan MA, Skelly DA, Dona MSI, Squiers GT, Farrugia GE, Gaynor TL, Cohen CD, Pandey R, Diep H, Vinh A, Rosenthal NA, Pinto AR. High-Resolution Transcriptomic Profiling of the Heart During Chronic Stress Reveals Cellular Drivers of Cardiac Fibrosis and Hypertrophy. Circulation 2020; 142:1448-1463. [PMID: 32795101 PMCID: PMC7547893 DOI: 10.1161/circulationaha.119.045115] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Cardiac fibrosis is a key antecedent to many types of cardiac dysfunction including heart failure. Physiological factors leading to cardiac fibrosis have been recognized for decades. However, the specific cellular and molecular mediators that drive cardiac fibrosis, and the relative effect of disparate cell populations on cardiac fibrosis, remain unclear. Methods: We developed a novel cardiac single-cell transcriptomic strategy to characterize the cardiac cellulome, the network of cells that forms the heart. This method was used to profile the cardiac cellular ecosystem in response to 2 weeks of continuous administration of angiotensin II, a profibrotic stimulus that drives pathological cardiac remodeling. Results: Our analysis provides a comprehensive map of the cardiac cellular landscape uncovering multiple cell populations that contribute to pathological remodeling of the extracellular matrix of the heart. Two phenotypically distinct fibroblast populations, Fibroblast-Cilp and Fibroblast-Thbs4, emerged after induction of tissue stress to promote fibrosis in the absence of smooth muscle actin–expressing myofibroblasts, a key profibrotic cell population. After angiotensin II treatment, Fibroblast-Cilp develops as the most abundant fibroblast subpopulation and the predominant fibrogenic cell type. Mapping intercellular communication networks within the heart, we identified key intercellular trophic relationships and shifts in cellular communication after angiotensin II treatment that promote the development of a profibrotic cellular microenvironment. Furthermore, the cellular responses to angiotensin II and the relative abundance of fibrogenic cells were sexually dimorphic. Conclusions: These results offer a valuable resource for exploring the cardiac cellular landscape in health and after chronic cardiovascular stress. These data provide insights into the cellular and molecular mechanisms that promote pathological remodeling of the mammalian heart, highlighting early transcriptional changes that precede chronic cardiac fibrosis.
Collapse
Affiliation(s)
- Micheal A McLellan
- The Jackson Laboratory, Bar Harbor, ME (M.A.M., D.A.S., G.T.S., R.P., N.A.R.).,Graduate School of Biomedical Sciences, Tufts University, Boston, MA (M.A.M.)
| | - Daniel A Skelly
- The Jackson Laboratory, Bar Harbor, ME (M.A.M., D.A.S., G.T.S., R.P., N.A.R.)
| | - Malathi S I Dona
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., G.E.F., T.L.G., C.D.C., A.R.P.)
| | - Galen T Squiers
- The Jackson Laboratory, Bar Harbor, ME (M.A.M., D.A.S., G.T.S., R.P., N.A.R.)
| | - Gabriella E Farrugia
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., G.E.F., T.L.G., C.D.C., A.R.P.)
| | - Taylah L Gaynor
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., G.E.F., T.L.G., C.D.C., A.R.P.)
| | - Charles D Cohen
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., G.E.F., T.L.G., C.D.C., A.R.P.)
| | - Raghav Pandey
- The Jackson Laboratory, Bar Harbor, ME (M.A.M., D.A.S., G.T.S., R.P., N.A.R.)
| | - Henry Diep
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia (T.L.G, C.D.C., H.D., A.V., A.R.P.)
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia (T.L.G, C.D.C., H.D., A.V., A.R.P.)
| | - Nadia A Rosenthal
- The Jackson Laboratory, Bar Harbor, ME (M.A.M., D.A.S., G.T.S., R.P., N.A.R.)
| | - Alexander R Pinto
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., G.E.F., T.L.G., C.D.C., A.R.P.).,Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia (T.L.G, C.D.C., H.D., A.V., A.R.P.)
| |
Collapse
|
16
|
Camacho-Arroyo I, Piña-Medina AG, Bello-Alvarez C, Zamora-Sánchez CJ. Sex hormones and proteins involved in brain plasticity. VITAMINS AND HORMONES 2020; 114:145-165. [PMID: 32723542 DOI: 10.1016/bs.vh.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
It is well known that peripheral sex steroid hormones cross the blood-brain barrier and control a broad spectrum of reproductive behaviors. However, their role in other essential brain functions was investigated since the 1980s, when the accumulation of pregnenolone and dehydroepiandrosterone in the brain of mammalian species was determined. Since then, numerous studies have demonstrated the participation of sex hormones in brain plasticity processes. Sex hormones through both genomic and non-genomic mechanisms of action are capable of inducing gene transcription or activating signaling cascades that result in the promotion of different physiological and pathological events of brain plasticity, such as remodeling or formation of dendritic spines, neurogenesis, synaptogenesis or myelination. In this chapter, we will present the effects of sex hormones and proteins involved in brain plasticity.
Collapse
Affiliation(s)
- Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| | - Ana Gabriela Piña-Medina
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Carmen J Zamora-Sánchez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
17
|
Abstract
17β-Estradiol (E2) is a potent steroid hormone of both gonadal and neuronal origin that exerts profound effects on neuroplasticity in several brain regions. Dendritic spine and synapse formation and rearrangements are modulated and mediated by estrogens. In this chapter, we highlighted the essential background concerning the effects of E2 on synaptic rearrangements accompanied by synaptic plasticity in E2-sensitive brain regions that mediate learning and memory, i.e., cortex and hippocampus. We also address details of the molecular mechanisms underlying E2 regulation of spine dynamics. The proposed models of action of E2 overlaps with that for well-established synaptic modulators, such as adenosine. Thus, the possible synergistic effects of those two molecules in respect to synaptic rearrangement and plasticity were presented.
Collapse
|
18
|
Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer's Disease. Cell Mol Neurobiol 2020; 40:283-299. [PMID: 31502112 DOI: 10.1007/s10571-019-00733-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is described as cognitive and memory impairments with a sex-related epidemiological profile, affecting two times more women than men. There is emerging evidence that alternations in the hippocampal neurogenesis occur at the early stage of AD. Therapies that may effectively slow, stop, or regenerate the dying neurons in AD are being extensively investigated in the last few decades, but none has yet been found to be effective. The regulation of endogenous neurogenesis is one of the main therapeutic targets for AD. Mounting evidence indicates that the neurosteroid estradiol (17β-estradiol) plays a supporting role in neurogenesis, neuronal activity, and synaptic plasticity of AD. This effect may provide preventive and/or therapeutic approaches for AD. In this article, we discuss the molecular mechanism of potential estradiol modulatory action on endogenous neurogenesis, synaptic plasticity, and cognitive function in AD.
Collapse
Affiliation(s)
- Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Vahid Hajali
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Moradi
- Histology and Embryology Group, Basic Science Department, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, Münster, Germany.
| |
Collapse
|
19
|
Negrón AL, Yu G, Boehm U, Acosta-Martínez M. Targeted Deletion of PTEN in Kisspeptin Cells Results in Brain Region- and Sex-Specific Effects on Kisspeptin Expression and Gonadotropin Release. Int J Mol Sci 2020; 21:ijms21062107. [PMID: 32204355 PMCID: PMC7139936 DOI: 10.3390/ijms21062107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Kisspeptin-expressing neurons in the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC) of the hypothalamus relay hormonal and metabolic information to gonadotropin-releasing hormone neurons, which in turn regulate pituitary and gonadal function. Phosphatase and tensin homolog (PTEN) blocks phosphatidylinositol 3-kinase (PI3K), a signaling pathway utilized by peripheral factors to transmit their signals. However, whether PTEN signaling in kisspeptin neurons helps to integrate peripheral hormonal cues to regulate gonadotropin release is unknown. To address this question, we generated mice with a kisspeptin cell-specific deletion of Pten (Kiss-PTEN KO), and first assessed kisspeptin protein expression and gonadotropin release in these animals. Kiss-PTEN KO mice displayed a profound sex and region-specific kisspeptin neuron hyperthrophy. We detected both kisspeptin neuron hyperthrophy as well as increased kisspeptin fiber densities in the AVPV and ARC of Kiss-PTEN KO females and in the ARC of Kiss-PTEN KO males. Moreover, Kiss-PTEN KO mice showed a reduced gonadotropin release in response to gonadectomy. We also found a hyperactivation of mTOR, a downstream PI3K target and central regulator of cell metabolism, in the AVPV and ARC of Kiss-PTEN KO females but not males. Fasting, known to inhibit hypothalamic kisspeptin expression and luteinizing hormone levels, failed to induce these changes in Kiss-PTEN KO females. We conclude that PTEN signaling regulates kisspeptin protein synthesis in both sexes and that its role as a metabolic signaling molecule in kisspeptin neurons is sex-specific.
Collapse
Affiliation(s)
- Ariel L. Negrón
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY 11794, USA;
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Guiqin Yu
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421 Homburg, Germany;
| | - Maricedes Acosta-Martínez
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA;
- Correspondence: ; Tel.: +1-631-444-6075; Fax: +1-631-444-3432
| |
Collapse
|
20
|
Baudry M. Calpain-1 and Calpain-2 in the Brain: Dr. Jekill and Mr Hyde? Curr Neuropharmacol 2020; 17:823-829. [PMID: 30819083 PMCID: PMC7052842 DOI: 10.2174/1570159x17666190228112451] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/24/2019] [Accepted: 02/08/2019] [Indexed: 11/22/2022] Open
Abstract
While the calpain system has now been discovered for over 50 years, there is still a paucity of information regard-ing the organization and functions of the signaling pathways regulated by these proteases, although calpains play critical roles in many cell functions. Moreover, calpain overactivation has been shown to be involved in numerous diseases. Among the 15 calpain isoforms identified, calpain-1 (aka µ-calpain) and calpain-2 (aka m-calpain) are ubiquitously distributed in most tissues and organs, including the brain. We have recently proposed that calpain-1 and calpain-2 play opposite functions in the brain, with calpain-1 activation being required for triggering synaptic plasticity and neuroprotection (Dr. Jekill), and calpain-2 limiting the extent of plasticity and being neurodegenerative (Mr. Hyde). Calpain-mediated cleavage has been ob-served in cytoskeleton proteins, membrane-associated proteins, receptors/channels, scaffolding/anchoring proteins, and pro-tein kinases and phosphatases. This review will focus on the signaling pathways related to local protein synthesis, cytoskele-ton regulation and neuronal survival/death regulated by calpain-1 and calpain-2, in an attempt to explain the origin of the op-posite functions of these 2 calpain isoforms. This will be followed by a discussion of the potential therapeutic applications of selective regulators of these 2 calpain isoforms.
Collapse
Affiliation(s)
- Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| |
Collapse
|
21
|
Kumar A, Foster TC. G Protein-Coupled Estrogen Receptor: Rapid Effects on Hippocampal-Dependent Spatial Memory and Synaptic Plasticity. Front Endocrinol (Lausanne) 2020; 11:385. [PMID: 32587576 PMCID: PMC7298106 DOI: 10.3389/fendo.2020.00385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
In the hippocampus, estrogen regulates gene transcription linked to neuronal growth, neuroprotection, and the maintenance of memory function (1-3). The mechanism is likely to involve genomic regulation through classic estrogen receptor (ER) signaling cascades that influence transcription. Estrogens binding to classic nuclear ERs, alpha (ERα) and beta (ERβ), and have pleotropic effects on development, behavior, and neurophysiological functions, including synaptic plasticity and memory consolidation (4-7). In addition to ERα and ERβ, estrogen can also initiate activation of classical second messenger signaling cascades to influence the activity of G-proteins and a host of kinases, resulting in rapid changes in physiology (8-14). These rapid effects of estrogen are commonly mediated by membrane receptors. In the late 90s, multiple laboratories cloned cDNA/gene for an orphan G-protein-coupled receptor with very low homology with other G-protein-coupled receptors and named it G-protein-coupled receptor 30 (GPR30) (15-20). Later in 2007, the International Union of Basic and Clinical Pharmacology designated GPR30 as G protein-coupled estrogen receptor (GPER) (21); GPER is a seven-transmembrane G-protein-coupled receptor, predominantly expressed on the cell membrane (22). Interestingly, GPER is reported to mediate many of the rapid responses of estradiol in the adult brain, and is widely distributed in the mammalian brain including the plasma membrane of hippocampal neurons (23-31). GPER modulates second messenger signaling cascades involving Gαs- and Gαi/o-associated increase in cyclic adenosine monophosphate and phosphoinositide 3-kinase or Src protein kinase respectively (32, 33). Activation of GPER is also associated with phospholipase C, and the inositol receptor and ryanodine receptor-mediated increase in intracellular calcium (24, 34). This commentary is concentrated specifically on the possible rapid effects of GPER in hippocampal-dependent spatial memory function and synaptic plasticity.
Collapse
|
22
|
Cheboub A, Regouat N, Djidjik R, Slimani A, Hadj-Bekkouche F. Short-term aromatase inhibition induces prostatic alterations in adult wistar rat: A biochemical, histopathological and immunohistochemical study. Acta Histochem 2019; 121:151441. [PMID: 31522738 DOI: 10.1016/j.acthis.2019.151441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 08/08/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE This study aimed to evaluate the effects of estrogen reduction on amyloid deposition, some lipid metabolism and oxidative stress markers, PSA-like production and p63 expression in the prostate of the adult rat. METHODS Aromatase inhibitor: Formestane (4-OHA), was administrated to male rats, at a dose of 0.1 mg/kg b.w./day, for 10 days. The control group (CONT) received the same volume of placebo injection (NaCl 0.9%). RESULTS 4-OHA treatment induced a significant accumulation of intraprostatic cholesterol (138.90 ± 17.64 vs 85.12 ± 2.87, p = 0.01); against an insignificant diminution of malondialdehyde (412.6 ± 54.35 vs 842.70 ± 336.50, p > 0.05) and glutathione (2.40 ± 0.23 vs 3.65 ± 0.88, p > 0.05). This was associated with a significant decrease of nitric oxide (31.76 ± 7.07 vs 179.40 ± 58.35, p = 0.024). Additionally, 4-OHA significantly increased the intraprostatic production of PSA-like (11.12 ± 2.78 vs 3.91 ± 0.43, p = 0.043). The prostatic histology revealed an amyloid deposition, in all prostatic lobes and a smooth muscle layer growth (p < 0.05); especially significant in the dorsal and lateral lobes. Theses lobes manifested a basal cells proliferation, with a 3-fold increase of p63 expression (p < 0.001). The ventral lobe presented epithelial atrophy (37.80 ± 16.20 vs 167.60 ± 5.16, p < 0.05); with occasional and significant proliferative foci (247.00 ± 9.573 vs 167.60 ± 5.16 p < 0.05). DISCUSSION AND CONCLUSION Aromatase inhibition, in the adult male rat, alters the prostatic function by reducing nitric oxide availability and inducing amyloid deposition along with limiting the differentiation of basal cells, through a lobe-specific p63-overexpression.
Collapse
Affiliation(s)
- Amina Cheboub
- Faculty of Biology Sciences, University of Sciences and Technology Houari Boumediene, Algeria.
| | - Nadia Regouat
- Faculty of Biology Sciences, University of Sciences and Technology Houari Boumediene, Algeria
| | - Reda Djidjik
- Immunology Service of Isaad Hassani-Beni Messous Hospital, Algiers, Algeria
| | - Assia Slimani
- Pathological Anatomy Service of Isaad Hassani-Beni Messous Hospital, Algiers, Algeria
| | - Fatima Hadj-Bekkouche
- Faculty of Biology Sciences, University of Sciences and Technology Houari Boumediene, Algeria
| |
Collapse
|
23
|
Dorsal Hippocampal Actin Polymerization Is Necessary for Activation of G-Protein-Coupled Estrogen Receptor (GPER) to Increase CA1 Dendritic Spine Density and Enhance Memory Consolidation. J Neurosci 2019; 39:9598-9610. [PMID: 31628182 DOI: 10.1523/jneurosci.2687-18.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 09/03/2019] [Accepted: 10/13/2019] [Indexed: 11/21/2022] Open
Abstract
Activation of the membrane estrogen receptor G-protein-coupled estrogen receptor (GPER) in ovariectomized mice via the GPER agonist G-1 mimics the beneficial effects of 17β-estradiol (E2) on hippocampal CA1 spine density and memory consolidation, yet the cell-signaling mechanisms mediating these effects remain unclear. The present study examined the role of actin polymerization and c-Jun N-terminal kinase (JNK) phosphorylation in mediating effects of dorsal hippocampally infused G-1 on CA1 dendritic spine density and consolidation of object recognition and spatial memories in ovariectomized mice. We first showed that object learning increased apical CA1 spine density in the dorsal hippocampus (DH) within 40 min. We then found that DH infusion of G-1 increased both CA1 spine density and phosphorylation of the actin polymerization regulator cofilin, suggesting that activation of GPER may increase spine morphogenesis through actin polymerization. As with memory consolidation in our previous work (Kim et al., 2016), effects of G-1 on CA1 spine density and cofilin phosphorylation depended on JNK phosphorylation in the DH. Also consistent with our previous findings, E2-induced cofilin phosphorylation was not dependent on GPER activation. Finally, we found that infusion of the actin polymerization inhibitor, latrunculin A, into the DH prevented G-1 from increasing apical CA1 spine density and enhancing both object recognition and spatial memory consolidation. Collectively, these data demonstrate that GPER-mediated hippocampal spinogenesis and memory consolidation depend on JNK and cofilin signaling, supporting a critical role for actin polymerization in the GPER-induced regulation of hippocampal function in female mice.SIGNIFICANCE STATEMENT Emerging evidence suggests that G-protein-coupled estrogen receptor (GPER) activation mimics effects of 17β-estradiol on hippocampal memory consolidation. Unlike canonical estrogen receptors, GPER activation is associated with reduced cancer cell proliferation; thus, understanding the molecular mechanisms through which GPER regulates hippocampal function may provide new avenues for the development of drugs that provide the cognitive benefits of estrogens without harmful side effects. Here, we demonstrate that GPER increases CA1 dendritic spine density and hippocampal memory consolidation in a manner dependent on actin polymerization and c-Jun N-terminal kinase phosphorylation. These findings provide novel insights into the role of GPER in mediating hippocampal morphology and memory consolidation, and may suggest first steps toward new therapeutics that more safely and effectively reduce memory decline in menopausal women.
Collapse
|
24
|
Hippocampus-specific Rictor knockdown inhibited 17β-estradiol induced neuronal plasticity and spatial memory improvement in ovariectomized mice. Behav Brain Res 2019; 364:50-61. [DOI: 10.1016/j.bbr.2019.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/08/2019] [Accepted: 02/09/2019] [Indexed: 11/19/2022]
|
25
|
Sheppard PAS, Choleris E, Galea LAM. Structural plasticity of the hippocampus in response to estrogens in female rodents. Mol Brain 2019; 12:22. [PMID: 30885239 PMCID: PMC6423800 DOI: 10.1186/s13041-019-0442-7] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 01/05/2023] Open
Abstract
It is well established that estrogens affect neuroplasticity in a number of brain regions. In particular, estrogens modulate and mediate spine and synapse formation as well as neurogenesis in the hippocampal formation. In this review, we discuss current research exploring the effects of estrogens on dendritic spine plasticity and neurogenesis with a focus on the modulating factors of sex, age, and pregnancy. Hormone levels, including those of estrogens, fluctuate widely across the lifespan from early life to puberty, through adulthood and into old age, as well as with pregnancy and parturition. Dendritic spine formation and modulation are altered both by rapid (likely non-genomic) and classical (genomic) actions of estrogens and have been suggested to play a role in the effects of estrogens on learning and memory. Neurogenesis in the hippocampus is influenced by age, the estrous cycle, pregnancy, and parity in female rodents. Furthermore, sex differences exist in hippocampal cellular and molecular responses to estrogens and are briefly discussed throughout. Understanding how structural plasticity in the hippocampus is affected by estrogens and how these effects can influence function and be influenced by other factors, such as experience and sex, is critical and can inform future treatments in conditions involving the hippocampus.
Collapse
Affiliation(s)
- Paul A. S. Sheppard
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Elena Choleris
- Department of Psychology & Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1 Canada
| | - Liisa A. M. Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
26
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
27
|
Kirshner ZZ, Gibbs RB. Use of the REVERT ® total protein stain as a loading control demonstrates significant benefits over the use of housekeeping proteins when analyzing brain homogenates by Western blot: An analysis of samples representing different gonadal hormone states. Mol Cell Endocrinol 2018; 473:156-165. [PMID: 29396126 PMCID: PMC6045444 DOI: 10.1016/j.mce.2018.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/21/2018] [Accepted: 01/22/2018] [Indexed: 01/01/2023]
Abstract
Western blot is routinely used to quantify differences in the levels of target proteins in tissues. Standard methods typically use measurements of housekeeping proteins to control for variations in loading and protein transfer. This is problematic, however, when housekeeping proteins also are affected by experimental conditions such as injury, disease, and/or gonadal hormone manipulations. Our goal was to evaluate an alternative and perhaps superior method for conducting Western blot analysis of brain tissue homogenates from rats with distinct physiologically relevant gonadal hormone states. Tissues were collected from the hippocampus, frontal cortex, and striatum of young adult female rats that either were ovariectomized to model surgical menopause, or were treated with the ovatotoxin 4-vinylcyclohexene diepoxide (VCD) to model transitional menopause. Tissues also were collected from rats with a normal estrous cycle killed at proestrus when estradiol levels are high, and at diestrus when estradiol levels are low. Western blot detection of α-tubulin, β-actin, and GAPDH was performed and were compared for sensitivity and reliability with a fluorescent total protein stain (REVERT®). Results show that the total protein stain was much less variable across samples and had a greater linear range than α-tubulin, β-actin, or GAPDH. The stain was stable and easy to use, and did not interfere with the immunodetection or multiplexed detection of the housekeeping proteins. In addition, we show that normalization of our data to total protein, but not to GAPDH, revealed significant differences in α-tubulin expression in the hippocampus as a function of treatment, and that gel-to-gel consistency in measuring differences between paired samples run on multiple gels was significantly better when data were normalized to total protein than when normalized to GAPDH. These results demonstrate that the REVERT® total protein stain can be used in Western blot analysis of brain tissue homogenates to control for variations in loading and protein transfer, and provides significant advantages over the use of housekeeping proteins for quantifying changes in the levels of multiple target proteins.
Collapse
Affiliation(s)
- Z Z Kirshner
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - R B Gibbs
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
28
|
Paletta P, Sheppard PAS, Matta R, Ervin KSJ, Choleris E. Rapid effects of estrogens on short-term memory: Possible mechanisms. Horm Behav 2018; 104:88-99. [PMID: 29847771 DOI: 10.1016/j.yhbeh.2018.05.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/24/2018] [Accepted: 05/26/2018] [Indexed: 01/11/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Estrogens affect learning and memory through rapid and delayed mechanisms. Here we review studies on rapid effects on short-term memory. Estradiol rapidly improves social and object recognition memory, spatial memory, and social learning when administered systemically. The dorsal hippocampus mediates estrogen rapid facilitation of object, social and spatial short-term memory. The medial amygdala mediates rapid facilitation of social recognition. The three estrogen receptors, α (ERα), β (ERβ) and the G-protein coupled estrogen receptor (GPER) appear to play different roles depending on the task and brain region. Both ERα and GPER agonists rapidly facilitate short-term social and object recognition and spatial memory when administered systemically or into the dorsal hippocampus and facilitate social recognition in the medial amygdala. Conversely, only GPER can facilitate social learning after systemic treatment and an ERβ agonist only rapidly improved short-term spatial memory when given systemically or into the hippocampus, but also facilitates social recognition in the medial amygdala. Investigations into the mechanisms behind estrogens' rapid effects on short term memory showed an involvement of the extracellular signal-regulated kinase (ERK) and the phosphoinositide 3-kinase (PI3K) kinase pathways. Recent evidence also showed that estrogens interact with the neuropeptide oxytocin in rapidly facilitating social recognition. Estrogens can increase the production and/or release of oxytocin and other neurotransmitters, such as dopamine and acetylcholine. Therefore, it is possible that estrogens' rapid effects on short-term memory may occur through the regulation of various neurotransmitters, although more research is need on these interactions as well as the mechanisms of estrogens' actions on short-term memory.
Collapse
Affiliation(s)
- Pietro Paletta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Paul A S Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Richard Matta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Kelsy S J Ervin
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
29
|
Dossat AM, Wright KN, Strong CE, Kabbaj M. Behavioral and biochemical sensitivity to low doses of ketamine: Influence of estrous cycle in C57BL/6 mice. Neuropharmacology 2018; 130:30-41. [PMID: 29175352 PMCID: PMC5749639 DOI: 10.1016/j.neuropharm.2017.11.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/31/2017] [Accepted: 11/12/2017] [Indexed: 12/26/2022]
Abstract
RATIONALE Low-dose ketamine is a rapid-acting antidepressant, to which female rodents are more sensitive as compared to males. However, the mechanism mediating this sex difference in ketamine sensitivity remains elusive. OBJECTIVES We sought to determine whether male and female mice differ in their behavioral sensitivity to low doses of ketamine, and uncover how ovarian hormones influence females' ketamine sensitivity. We also aimed to uncover some of the molecular mechanism(s) in mood-related brain regions that mediate sex differences in ketamine antidepressant effects. METHODS Male and female mice (freely-cycling, diestrus 1 [D1], proestrus [Pro], or D1 treated with an estrogen receptor (ER) α, ERβ, or progesterone receptor (PR) agonist) received ketamine (0, 1.5, or 3 mg/kg, intraperitoneally) and were tested in the forced swim test (FST) 30 min later. Ketamine's influence over synaptic plasticity markers in the prefrontal cortex (PFC) and hippocampus (HPC) of males, D1, and Pro females was quantified by Western blot 1 h post-treatment. RESULTS Males, freely cycling females, D1 and Pro females exhibited antidepressant-like responses to 3 mg/kg ketamine. Pro females were the only group where ketamine exhibited an antidepressant effect at 1.5 mg/kg. D1 females treated with an agonist for ERα or ERβ exhibited an antidepressant-like response to 1.5 mg/kg ketamine. Ketamine (3 mg/kg) increased synaptic plasticity-related proteins in the PFC and HPC of males, D1, and Pro females. Yet, Pro females exhibited an increase in p-Akt and p-CaMKIIα in response to 1.5 and 3 mg/kg ketamine. CONCLUSION Our results indicate that females' enhanced sensitivity to ketamine during Pro is likely mediated through estradiol acting on ERα and ERβ, leading to greater activation of synaptic plasticity-related kinases within the PFC and HPC.
Collapse
Affiliation(s)
- Amanda M Dossat
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306, United States
| | - Katherine N Wright
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306, United States
| | - Caroline E Strong
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306, United States
| | - Mohamed Kabbaj
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306, United States.
| |
Collapse
|
30
|
Rodiño-Janeiro BK, Martínez C, Fortea M, Lobo B, Pigrau M, Nieto A, González-Castro AM, Salvo-Romero E, Guagnozzi D, Pardo-Camacho C, Iribarren C, Azpiroz F, Alonso-Cotoner C, Santos J, Vicario M. Decreased TESK1-mediated cofilin 1 phosphorylation in the jejunum of IBS-D patients may explain increased female predisposition to epithelial dysfunction. Sci Rep 2018; 8:2255. [PMID: 29396473 PMCID: PMC5797119 DOI: 10.1038/s41598-018-20540-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/17/2018] [Indexed: 02/08/2023] Open
Abstract
Disturbed intestinal epithelial barrier and mucosal micro-inflammation characterize irritable bowel syndrome (IBS). Despite intensive research demonstrating ovarian hormones modulation of IBS severity, there is still limited knowledge on the mechanisms underlying female predominance in this disorder. Our aim was to identify molecular pathways involved in epithelial barrier dysfunction and female predominance in diarrhea-predominant IBS (IBS-D) patients. Total RNA and protein were obtained from jejunal mucosal biopsies from healthy controls and IBS-D patients meeting the Rome III criteria. IBS severity was recorded based on validated questionnaires. Gene and protein expression profiles were obtained and data integrated to explore biological and molecular functions. Results were validated by western blot. Tight junction signaling, mitochondrial dysfunction, regulation of actin-based motility by Rho, and cytoskeleton signaling were differentially expressed in IBS-D. Decreased TESK1-dependent cofilin 1 phosphorylation (pCFL1) was confirmed in IBS-D, which negatively correlated with bowel movements only in female participants. In conclusion, deregulation of cytoskeleton dynamics through TESK1/CFL1 pathway underlies epithelial intestinal dysfunction in the small bowel mucosa of IBS-D, particularly in female patients. Further understanding of the mechanisms involving sex-mediated regulation of mucosal epithelial integrity may have significant preventive, diagnostic, and therapeutic implications for IBS.
Collapse
Affiliation(s)
- Bruno K Rodiño-Janeiro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.
| | - Cristina Martínez
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Marina Fortea
- Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Beatriz Lobo
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Marc Pigrau
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Adoración Nieto
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Ana María González-Castro
- Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Eloísa Salvo-Romero
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.,Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Danila Guagnozzi
- Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Cristina Pardo-Camacho
- Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Cristina Iribarren
- Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
| | - Fernando Azpiroz
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain
| | - Carmen Alonso-Cotoner
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain
| | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain.
| | - Maria Vicario
- Translational Mucosal Immunology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca; Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain
| |
Collapse
|
31
|
Xing FZ, Zhao YG, Zhang YY, He L, Zhao JK, Liu MY, Liu Y, Zhang JQ. Nuclear and membrane estrogen receptor antagonists induce similar mTORC2 activation-reversible changes in synaptic protein expression and actin polymerization in the mouse hippocampus. CNS Neurosci Ther 2018; 24:495-507. [PMID: 29352507 DOI: 10.1111/cns.12806] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/23/2017] [Accepted: 12/24/2017] [Indexed: 11/28/2022] Open
Abstract
AIMS Estrogens play pivotal roles in hippocampal synaptic plasticity through nuclear receptors (nERs; including ERα and ERβ) and the membrane receptor (mER; also called GPR30), but the underlying mechanism and the contributions of nERs and mER remain unclear. Mammalian target of rapamycin complex 2 (mTORC2) is involved in actin cytoskeleton polymerization and long-term memory, but whether mTORC2 is involved in the regulation of hippocampal synaptic plasticity by ERs is unclear. METHODS We treated animals with nER antagonists (MPP/PHTPP) or the mER antagonist (G15) alone or in combination with A-443654, an activator of mTORC2. Then, we examined the changes in hippocampal SRC-1 expression, mTORC2 signaling (rictor and phospho-AKTSer473), actin polymerization (phospho-cofilin and profilin-1), synaptic protein expression (GluR1, PSD95, spinophilin, and synaptophysin), CA1 spine density, and synapse density. RESULTS All of the examined parameters except synaptophysin expression were significantly decreased by MPP/PHTPP and G15 treatment. MPP/PHTPP and G15 induced a similar decrease in most parameters except p-cofilin, GluR1, and spinophilin expression. The ER antagonist-induced decreases in these parameters were significantly reversed by mTORC2 activation, except for the change in SRC-1, rictor, and synaptophysin expression. CONCLUSIONS nERs and mER contribute similarly to the changes in proteins and structures associated with synaptic plasticity, and mTORC2 may be a novel target of hippocampal-dependent dementia such as Alzheimer's disease as proposed by previous studies.
Collapse
Affiliation(s)
- Fang-Zhou Xing
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Yan-Gang Zhao
- Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Yuan-Yuan Zhang
- Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing, China
| | - Ji-Kai Zhao
- Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Meng-Ying Liu
- Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Yan Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Ji-Qiang Zhang
- Department of Neurobiology, Third Military Medical University, Chongqing, China
| |
Collapse
|
32
|
McCarthny CR, Du X, Wu YC, Hill RA. Investigating the Interactive Effects of Sex Steroid Hormones and Brain-Derived Neurotrophic Factor during Adolescence on Hippocampal NMDA Receptor Expression. Int J Endocrinol 2018; 2018:7231915. [PMID: 29666640 PMCID: PMC5831834 DOI: 10.1155/2018/7231915] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/05/2017] [Accepted: 10/19/2017] [Indexed: 12/18/2022] Open
Abstract
Sex steroid hormones have neuroprotective properties which may be mediated by brain-derived neurotrophic factor (BDNF). This study sought to determine the interactive effects of preadolescent hormone manipulation and BDNF heterozygosity (+/-) on hippocampal NMDA-R expression. Wild-type and BDNF+/- mice were gonadectomised, and females received either 17β-estradiol or progesterone treatment, while males received either testosterone or dihydrotestosterone (DHT) treatment. Dorsal (DHP) and ventral hippocampus (VHP) were dissected, and protein expression of GluN1, GluN2A, GluN2B, and PSD-95 was assessed by Western blot analysis. Significant genotype × OVX interactions were found for GluN1 and GluN2 expression within the DHP of female mice, suggesting modulation of select NMDA-R levels by female sex hormones is mediated by BDNF. Furthermore, within the DHP BDNF+/- mice show a hypersensitive response to hormone treatment on GluN2 expression which may result from upstream alterations in TrkB phosphorylation. In contrast to the DHP, the VHP showed no effects of hormone manipulation but significant effects of genotype on NMDA-R expression. Castration had no effect on NMDA-R expression; however, androgen treatment had selective effects on GluN2B. These data show case distinct, interactive roles for sex steroid hormones and BDNF in the regulation of NMDA-R expression that are dependent on dorsal versus ventral hippocampal region.
Collapse
Affiliation(s)
- Cushla R. McCarthny
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Xin Du
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - YeeWen Candace Wu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachel A. Hill
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
33
|
Zhao Y, He L, Zhang Y, Zhao J, Liu Z, Xing F, Liu M, Feng Z, Li W, Zhang J. Estrogen receptor alpha and beta regulate actin polymerization and spatial memory through an SRC-1/mTORC2-dependent pathway in the hippocampus of female mice. J Steroid Biochem Mol Biol 2017; 174:96-113. [PMID: 28789972 DOI: 10.1016/j.jsbmb.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 07/29/2017] [Accepted: 08/03/2017] [Indexed: 12/18/2022]
Abstract
Aging-related decline of estrogens, especially 17β-estradiol (E2), has been shown to play an important role in the impairment of learning and memory in dementias, such as Alzheimer's disease (AD), but the underlying molecular mechanisms are poorly understood. In this study, we first demonstrated decreases in E2 signaling (aromatase, classic estrogen receptor ERα and ERβ and their coactivator SRC-1), mTORC2 signaling (Rictor and phospho-AKTser473) and actin polymerization (phospho-Cofilin, Profilin-1 and the F-actin/G-actin ratio) in the hippocampus of old female mice compared with those levels detected in the adult hippocampus. We then showed that ERα and ERβ antagonists induced a significant decrease in SRC-1, mTORC2 signaling, actin polymerization, and CA1 spine density, as well as impairments of learning and memory; however, ovariectomy-induced changes of these parameters could be significantly reversed by treatment with ER agonists. We further showed that expression of SRC-1, mTORC2 signaling and actin polymerization could be upregulated by E2 treatment, and the effects of E2 were blocked by the ER antagonists but mimicked by the agonists. We also showed that the lentivirus-mediated SRC-1 knockdown significantly inhibited the agonist-activated mTORC2 signaling and actin polymerization, and the lentivirus-mediated Rictor knockdown also significantly inhibited the agonist-activated actin polymerization. Finally, we demonstrated that the ERα and ERβ antagonists induced a disruption in actin polymerization and an impairment of spatial memory, which were rescued by activation of mTORC2. Taken together, the above results clearly demonstrated an mTORC2-dependent regulation of actin polymerization that contributed to the effects of ERα and ERβ on spatial learning, which may provide a novel target for the prevention and treatment of E2-related dementia in the aged population.
Collapse
Affiliation(s)
- Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jikai Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Zhi Liu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Fangzhou Xing
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Mengying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Ziqi Feng
- Cadet Brigade, Third Military Medical University, Chongqing 400038, China
| | - Wei Li
- School of Nursing, Third Military Medical University, Chongqing 400038, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
34
|
The non-coding RNA BC1 regulates experience-dependent structural plasticity and learning. Nat Commun 2017; 8:293. [PMID: 28819097 PMCID: PMC5561022 DOI: 10.1038/s41467-017-00311-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/19/2017] [Indexed: 11/09/2022] Open
Abstract
The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders. Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.
Collapse
|
35
|
Ding Y, Lu L, Xuan C, Han J, Ye S, Cao T, Chen W, Li A, Zhang X. Di- n -butyl phthalate exposure negatively influences structural and functional neuroplasticity via Rho-GTPase signaling pathways. Food Chem Toxicol 2017; 105:34-43. [DOI: 10.1016/j.fct.2017.03.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 01/15/2023]
|
36
|
Zhao Y, Yu Y, Zhang Y, He L, Qiu L, Zhao J, Liu M, Zhang J. Letrozole regulates actin cytoskeleton polymerization dynamics in a SRC-1 dependent manner in the hippocampus of mice. J Steroid Biochem Mol Biol 2017; 167:86-97. [PMID: 27866972 DOI: 10.1016/j.jsbmb.2016.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/24/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
In the hippocampus, local estrogens (E2) derived from testosterone that is catalyzed by aromatase play important roles in the regulation of hippocampal neural plasticity, but the underlying mechanisms remain unclear. The actin cytoskeleton contributes greatly to hippocampal synaptic plasticity; however, whether it is regulated by local E2 and the related mechanisms remain to be elucidated. In this study, we first examined the postnatal developmental profiles of hippocampal aromatase and specific proteins responsible for actin cytoskeleton dynamics. Then we used aromatase inhibitor letrozole (LET) to block local E2 synthesis and examined the changes of these proteins and steroid receptor coactivator-1 (SRC-1), the predominant coactivator for steroid nuclear receptors. Finally, SRC-1 specific RNA interference was used to examine the effects of SRC-1 on the expression of these actin remodeling proteins. The results showed a V-type profile for aromatase and increased profiles for actin cytoskeleton proteins in both male and female hippocampus without obvious sex differences. LET treatment dramatically decreased the F-actin/G-actin ratio, the expression of Rictor, phospho-AKT (ser473), Profilin-1, phospho-Cofilin (Ser3), and SRC-1 in a dose-dependent manner. In vitro studies demonstrated that LET induced downregulation of these proteins could be reversed by E2, and E2 induced increase of these proteins were significantly suppressed by SRC-1 shRNA interference. These results for the first time clearly demonstrated that local E2 inhibition could induce aberrant actin polymerization; they also showed an important role of SRC-1 in the mediation of local E2 action on hippocampal synaptic plasticity by regulation of actin cytoskeleton dynamics.
Collapse
Affiliation(s)
- Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Yanlan Yu
- Student Brigade, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Linli Qiu
- School of Nursing, Third Military Medical University, Chongqing 400038, China; Department of Nursing, Sichuan Nursing Vocational College, Chengdu 610100, China
| | - Jikai Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Mengying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
37
|
Zhu G, Briz V, Seinfeld J, Liu Y, Bi X, Baudry M. Calpain-1 deletion impairs mGluR-dependent LTD and fear memory extinction. Sci Rep 2017; 7:42788. [PMID: 28202907 PMCID: PMC5311935 DOI: 10.1038/srep42788] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/17/2017] [Indexed: 01/02/2023] Open
Abstract
Recent studies indicate that calpain-1 is required for the induction of long-term potentiation (LTP) elicited by theta-burst stimulation in field CA1 of hippocampus. Here we determined the contribution of calpain-1 in another type of synaptic plasticity, the long-term depression (LTD) elicited by activation of type-I metabotropic glutamate receptors (mGluR-LTD). mGluR-LTD was associated with calpain-1 activation following T-type calcium channel opening, and resulted in the truncation of a regulatory subunit of PP2A, B56α. This signaling pathway was required for both the early and late phase of Arc translation during mGluR-LTD, through a mechanism involving mTOR and ribosomal protein S6 activation. In contrast, in hippocampal slices from calpain-1 knock-out (KO) mice, application of the mGluR agonist, DHPG, did not result in B56α truncation, increased Arc synthesis and reduced levels of membrane GluA1-containing AMPA receptors. Consistently, mGluR-LTD was impaired in calpain-1 KO mice, and the impairment could be rescued by phosphatase inhibitors, which also restored Arc translation in response to DHPG. Furthermore, calpain-1 KO mice exhibited impairment in fear memory extinction to tone presentation. These results indicate that calpain-1 plays a critical role in mGluR-LTD and is involved in many forms of synaptic plasticity and learning and memory.
Collapse
Affiliation(s)
- Guoqi Zhu
- Graduate College of Biomedical Sciences, Pomona, CA 91766, USA
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Victor Briz
- Graduate College of Biomedical Sciences, Pomona, CA 91766, USA
- VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium
| | - Jeff Seinfeld
- Graduate College of Biomedical Sciences, Pomona, CA 91766, USA
| | - Yan Liu
- Graduate College of Biomedical Sciences, Pomona, CA 91766, USA
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona, CA 91766, CA 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona, CA 91766, CA 91766, USA
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Pomona, CA 91766, USA
| |
Collapse
|
38
|
Alexander A, Irving AJ, Harvey J. Emerging roles for the novel estrogen-sensing receptor GPER1 in the CNS. Neuropharmacology 2017; 113:652-660. [DOI: 10.1016/j.neuropharm.2016.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 02/06/2023]
|
39
|
Estrogen Modulates ubc9 Expression and Synaptic Redistribution in the Brain of APP/PS1 Mice and Cortical Neurons. J Mol Neurosci 2017; 61:436-448. [DOI: 10.1007/s12031-017-0884-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 01/09/2017] [Indexed: 12/26/2022]
|
40
|
Mitrović N, Zarić M, Drakulić D, Martinović J, Sévigny J, Stanojlović M, Nedeljković N, Grković I. 17β-Estradiol-Induced Synaptic Rearrangements Are Accompanied by Altered Ectonucleotidase Activities in Male Rat Hippocampal Synaptosomes. J Mol Neurosci 2016; 61:412-422. [PMID: 27981418 DOI: 10.1007/s12031-016-0877-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022]
Abstract
17β-Estradiol (E2) rapidly, by binding to membrane estrogen receptors, activates cell signaling cascades which induce formation of new dendritic spines in the hippocampus of males as in females, but the interaction with other metabolic processes, such as extracellular adenine nucleotides metabolism, are currently unknown. Extracellular adenine nucleotides play significant roles, controlling excitatory glutamatergic synapses and development of neural circuits and synaptic plasticity. Their precise regulation in the synaptic cleft is tightly controlled by ecto-nucleoside triphosphate diphosphohydrolase (NTPDase)/ecto-5'-nucleotidase (eN) enzyme chain. Therefore, we sought to clarify whether a single systemic injection of E2 in male rats is accompanied by changes in the expression of the pre- and postsynaptic proteins and downstream kinases linked to E2-induced synaptic rearrangement as well as alterations in NTPDase/eN pathway in the hippocampal synaptosomes. Obtained data showed activation of mammalian target of rapamycin and upregulation of key synaptic proteins necessary for spine formation, 24 h after systemic E2 administration. In E2-mediated conditions, we found downregulation of NTPDase1 and NTPDase2 and attenuation of adenine nucleotide hydrolysis by NTPDase/eN enzyme chain, without changes in NTPDase3 properties and augmentation of synaptic tissue-nonspecific alkaline phosphatase (TNAP) activity. Despite reduced NTPDase activities, increased TNAP activity probably prevents toxic accumulation of ATP in the extracellular milieu and also hydrolyzes accumulated ADP due to unchanged NTPDase3 activity. Thus, our initial evaluation supports idea of specific roles of different ectonucleotidases and their coordinated actions in E2-mediated spine remodeling and maintenance.
Collapse
Affiliation(s)
- Nataša Mitrović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Marina Zarić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Dunja Drakulić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Jelena Martinović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC, G1V 0A6, Canada.,Centre de recherche du CHU de Québec, Université Laval, Québec, QC, G1V 4G2, Canada
| | - Miloš Stanojlović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Nadežda Nedeljković
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Studentski trg 3, Belgrade, 11000, Serbia
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia.
| |
Collapse
|
41
|
Osmanovic-Barilar J, Salkovic-Petrisi M. Evaluating the Role of Hormone Therapy in Postmenopausal Women with Alzheimer’s Disease. Drugs Aging 2016; 33:787-808. [DOI: 10.1007/s40266-016-0407-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
42
|
Estradiol-Mediated Spine Changes in the Dorsal Hippocampus and Medial Prefrontal Cortex of Ovariectomized Female Mice Depend on ERK and mTOR Activation in the Dorsal Hippocampus. J Neurosci 2016; 36:1483-9. [PMID: 26843632 DOI: 10.1523/jneurosci.3135-15.2016] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Dendritic spine plasticity underlies the formation and maintenance of memories. Both natural fluctuations and systemic administration of 17β-estradiol (E2) alter spine density in the dorsal hippocampus (DH) of rodents. DH E2 infusion enhances hippocampal-dependent memory by rapidly activating extracellular signal-regulated kinase (ERK)-dependent signaling of mammalian target of rapamycin (mTOR), a key protein synthesis pathway involved in spine remodeling. Here, we investigated whether infusion of E2 directly into the DH drives spine changes in the DH and other brain regions, and identified cell-signaling pathways that mediate these effects. E2 significantly increased basal and apical spine density on CA1 pyramidal neurons 30 min and 2 h after infusion. DH E2 infusion also significantly increased basal spine density on pyramidal neurons in the medial prefrontal cortex (mPFC) 2 h later, suggesting that E2-mediated activity in the DH drives mPFC spinogenesis. The increase in CA1 and mPFC spine density observed 2 h after intracerebroventricular infusion of E2 was blocked by DH infusion of an ERK or mTOR inhibitor. DH E2 infusion did not affect spine density in the dentate gyrus or ventromedial hypothalamus, suggesting specific effects of E2 on the DH and mPFC. Collectively, these data demonstrate that DH E2 treatment elicits ERK- and mTOR-dependent spinogenesis on CA1 and mPFC pyramidal neurons, effects that may support the memory-enhancing effects of E2. SIGNIFICANCE STATEMENT Although systemically injected 17β-estradiol (E2) increases CA1 dendritic spine density, the molecular mechanisms regulating E2-induced spinogenesis in vivo are largely unknown. We found that E2 infused directly into the dorsal hippocampus (DH) increased CA1 spine density 30 min and 2 h later. Surprisingly, DH E2 infusion also increased spine density in the medial prefrontal cortex (mPFC), suggesting that estrogenic regulation of the DH influences mPFC spinogenesis. Moreover, inhibition of ERK and mTOR activation in the DH prevented E2 from increasing DH and mPFC spines, demonstrating that DH ERK and mTOR activation is necessary for E2-induced spinogenesis in the DH and mPFC. These findings provide novel insights into the molecular mechanisms through which E2 mediates dendritic spine density in CA1 and mPFC.
Collapse
|
43
|
Garza-Lombó C, Gonsebatt ME. Mammalian Target of Rapamycin: Its Role in Early Neural Development and in Adult and Aged Brain Function. Front Cell Neurosci 2016; 10:157. [PMID: 27378854 PMCID: PMC4910040 DOI: 10.3389/fncel.2016.00157] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/30/2016] [Indexed: 01/14/2023] Open
Abstract
The kinase mammalian target of rapamycin (mTOR) integrates signals triggered by energy, stress, oxygen levels, and growth factors. It regulates ribosome biogenesis, mRNA translation, nutrient metabolism, and autophagy. mTOR participates in various functions of the brain, such as synaptic plasticity, adult neurogenesis, memory, and learning. mTOR is present during early neural development and participates in axon and dendrite development, neuron differentiation, and gliogenesis, among other processes. Furthermore, mTOR has been shown to modulate lifespan in multiple organisms. This protein is an important energy sensor that is present throughout our lifetime its role must be precisely described in order to develop therapeutic strategies and prevent diseases of the central nervous system. The aim of this review is to present our current understanding of the functions of mTOR in neural development, the adult brain and aging.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México México
| | - María E Gonsebatt
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México México
| |
Collapse
|
44
|
Abstract
Although calpain was proposed to participate in synaptic plasticity and learning and memory more than 30 years ago, the mechanisms underlying its activation and the roles of different substrates have remained elusive. Recent findings have provided evidence that the two major calpain isoforms in the brain, calpain-1 and calpain-2, play opposite functions in synaptic plasticity. In particular, while calpain-1 activation is the initial trigger for certain forms of synaptic plasticity, that is, long-term potentiation, calpain-2 activation restricts the extent of plasticity. Moreover, while calpain-1 rapidly cleaves regulatory and cytoskeletal proteins, calpain-2-mediated stimulation of local protein synthesis reestablishes protein homeostasis. These findings have important implications for our understanding of learning and memory and disorders associated with impairment in these processes.
Collapse
Affiliation(s)
- Victor Briz
- 1 KU Leuven, Center for Human Genetics and Leuven Institute for Neuroscience and Disease, Leuven, Belgium
- 2 VIB Center for the Biology of Disease, Leuven, Belgium
| | - Michel Baudry
- 3 Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
45
|
Briz V, Liu Y, Zhu G, Bi X, Baudry M. A novel form of synaptic plasticity in field CA3 of hippocampus requires GPER1 activation and BDNF release. J Cell Biol 2015; 210:1225-37. [PMID: 26391661 PMCID: PMC4586750 DOI: 10.1083/jcb.201504092] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/19/2015] [Indexed: 01/11/2023] Open
Abstract
Estrogen gates metabotropic glutamate receptor–dependent long-term depression at mossy fiber–CA3 synapses through a mechanism involving GPER1-mediated BDNF release, mTOR-dependent protein synthesis, and proteasome activity. Estrogen is an important modulator of hippocampal synaptic plasticity and memory consolidation through its rapid action on membrane-associated receptors. Here, we found that both estradiol and the G-protein–coupled estrogen receptor 1 (GPER1) specific agonist G1 rapidly induce brain-derived neurotrophic factor (BDNF) release, leading to transient stimulation of activity-regulated cytoskeleton-associated (Arc) protein translation and GluA1-containing AMPA receptor internalization in field CA3 of hippocampus. We also show that type-I metabotropic glutamate receptor (mGluR) activation does not induce Arc translation nor long-term depression (LTD) at the mossy fiber pathway, as opposed to its effects in CA1, and it only triggers LTD after GPER1 stimulation. Furthermore, this form of mGluR-dependent LTD is associated with ubiquitination and proteasome-mediated degradation of GluA1, and is prevented by proteasome inhibition. Overall, our study identifies a novel mechanism by which estrogen and BDNF regulate hippocampal synaptic plasticity in the adult brain.
Collapse
Affiliation(s)
- Victor Briz
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766 VIB Center for the Biology of Disease, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Yan Liu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766 College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Guoqi Zhu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766 Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei 230038, China
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
46
|
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
|
47
|
Abstract
Dendritic protein synthesis and actin cytoskeleton reorganization are important events required for the consolidation of hippocampal LTP and memory. However, the temporal and spatial relationships between these two processes remain unclear. Here, we report that treatment of adult rat hippocampal slices with BDNF or with tetraethylammonium (TEA), which induces a chemical form of LTP, produces a rapid and transient increase in RhoA protein levels. Changes in RhoA were restricted to dendritic spines of CA3 and CA1 and require de novo protein synthesis regulated by mammalian target of rapamycin (mTOR). BDNF-mediated stimulation of RhoA activity, cofilin phosphorylation, and actin polymerization were completely suppressed by protein synthesis inhibitors. Furthermore, intrahippocampal injections of RhoA antisense oligodeoxynucleotides inhibited theta burst stimulation (TBS)-induced RhoA upregulation in dendritic spines and prevented LTP consolidation. Addition of calpain inhibitors after BDNF or TEA treatment maintained RhoA levels elevated and prolonged the effects of BDNF and TEA on actin polymerization. Finally, the use of isoform-selective calpain inhibitors revealed that calpain-2 was involved in RhoA synthesis, whereas calpain-1 mediated RhoA degradation. Overall, this mechanism provides a novel link between dendritic protein synthesis and reorganization of the actin cytoskeleton in hippocampal dendritic spines during LTP consolidation.
Collapse
|
48
|
Sellers KJ, Erli F, Raval P, Watson IA, Chen D, Srivastava DP. Rapid modulation of synaptogenesis and spinogenesis by 17β-estradiol in primary cortical neurons. Front Cell Neurosci 2015; 9:137. [PMID: 25926772 PMCID: PMC4396386 DOI: 10.3389/fncel.2015.00137] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/23/2015] [Indexed: 01/06/2023] Open
Abstract
In the mammalian forebrain, the majority of excitatory synapses occur on dendritic spines. Changes in the number of these structures is important for brain development, plasticity and the refinement of neuronal circuits. The formation of excitatory synapses involves the coordinated formation of dendritic spines and targeting of multi-protein complexes to nascent connections. Recent studies have demonstrated that the estrogen 17β-estradiol (E2) can rapidly increase the number of dendritic spines, an effect consistent with the ability of E2 to rapidly influence cognitive function. However, the molecular composition of E2-induced spines and whether these protrusions form synaptic connections has not been fully elucidated. Moreover, which estrogen receptor(s) (ER) mediate these spine-morphogenic responses are not clear. Here, we report that acute E2 treatment results in the recruitment of postsynaptic density protein 95 (PSD-95) to novel dendritic spines. In addition neuroligin 1 (Nlg-1) and the NMDA receptor subunit GluN1 are recruited to nascent synapses in cortical neurons. The presence of these synaptic proteins at nascent synapses suggests that the machinery to allow pre- and post-synapses to form connections are present in E2-induced spines. We further demonstrate that E2 treatment results in the rapid and transient activation of extracellular signal-regulated kinase 1/2 (ERK1/2), Akt and the mammalian target of rapamycin (mTOR) signaling pathways. However, only ERK1/2 and Akt are required for E2-mediated spinogenesis. Using synthetic receptor modulators, we further demonstrate that activation of the estrogen receptor beta (ERβ) but not alpha (ERα) mimics rapid E2-induced spinogenesis and synaptogenesis. Taken together these findings suggest that in primary cortical neurons, E2 signaling via ERβ, but not through ERα, is capable of remodeling neuronal circuits by increasing the number of excitatory synapses.
Collapse
Affiliation(s)
- Katherine J Sellers
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Filippo Erli
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK ; Department of Biotechnology and Biosciences, Univeristy of Milano-Bicocca Milano, Italy
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Iain A Watson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Ding Chen
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| |
Collapse
|
49
|
Speranza L, Giuliano T, Volpicelli F, De Stefano ME, Lombardi L, Chambery A, Lacivita E, Leopoldo M, Bellenchi GC, di Porzio U, Crispino M, Perrone-Capano C. Activation of 5-HT7 receptor stimulates neurite elongation through mTOR, Cdc42 and actin filaments dynamics. Front Behav Neurosci 2015; 9:62. [PMID: 25814944 PMCID: PMC4356071 DOI: 10.3389/fnbeh.2015.00062] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 02/23/2015] [Indexed: 12/02/2022] Open
Abstract
Recent studies have indicated that the serotonin receptor subtype 7 (5-HT7R) plays a crucial role in shaping neuronal morphology during embryonic and early postnatal life. Here we show that pharmacological stimulation of 5-HT7R using a highly selective agonist, LP-211, enhances neurite outgrowth in neuronal primary cultures from the cortex, hippocampus and striatal complex of embryonic mouse brain, through multiple signal transduction pathways. All these signaling systems, involving mTOR, the Rho GTPase Cdc42, Cdk5, and ERK, are known to converge on the reorganization of cytoskeletal proteins that subserve neurite outgrowth. Indeed, our data indicate that neurite elongation stimulated by 5-HT7R is modulated by drugs affecting actin polymerization. In addition, we show, by 2D Western blot analyses, that treatment of neuronal cultures with LP-211 alters the expression profile of cofilin, an actin binding protein involved in microfilaments dynamics. Furthermore, by using microfluidic chambers that physically separate axons from the soma and dendrites, we demonstrate that agonist-dependent activation of 5-HT7R stimulates axonal elongation. Our results identify for the first time several signal transduction pathways, activated by stimulation of 5-HT7R, that converge to promote cytoskeleton reorganization and consequent modulation of axonal elongation. Therefore, the activation of 5-HT7R might represent one of the key elements regulating CNS connectivity and plasticity during development.
Collapse
Affiliation(s)
- Luisa Speranza
- Department of Biology, University of Naples Federico II Naples, Italy ; Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR Naples, Italy
| | - Teresa Giuliano
- Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR Naples, Italy
| | - Floriana Volpicelli
- Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR Naples, Italy ; Department of Pharmacy, University of Naples Federico II Naples, Italy
| | - M Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Istituto Pasteur Fondazione Cenci Bolognetti, University of Rome La Sapienza Rome, Italy
| | - Loredana Lombardi
- Department of Biology and Biotechnology "Charles Darwin", Istituto Pasteur Fondazione Cenci Bolognetti, University of Rome La Sapienza Rome, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples Naples, Italy ; IRCCS, Multimedica Milano, Italy
| | - Enza Lacivita
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari Bari, Italy
| | - Marcello Leopoldo
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari Bari, Italy
| | - Gian C Bellenchi
- Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR Naples, Italy
| | - Umberto di Porzio
- Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Carla Perrone-Capano
- Department of Biology, University of Naples Federico II Naples, Italy ; Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR Naples, Italy
| |
Collapse
|
50
|
Sellers K, Raval P, Srivastava DP. Molecular signature of rapid estrogen regulation of synaptic connectivity and cognition. Front Neuroendocrinol 2015; 36:72-89. [PMID: 25159586 DOI: 10.1016/j.yfrne.2014.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022]
Abstract
There is now a growing appreciation that estrogens are capable of rapidly activating a number of signaling cascades within the central nervous system. In addition, there are an increasing number of studies reporting that 17β-estradiol, the major biologically active estrogen, can modulate cognition within a rapid time frame. Here we review recent studies that have begun to uncover the molecular and cellular framework which contributes to estrogens ability to rapidly modulate cognition. We first describe the mechanisms by which estrogen receptors (ERs) can couple to intracellular signaling cascades, either directly, or via the transactivation of other receptors. Subsequently, we review the evidence that estrogen can rapidly modulate both neuronal function and structure in the hippocampus and the cortex. Finally, we will discuss how estrogens may influence cognitive function through the modulation of neuronal structure, and the implications this may have on the treatment of a range of brain disorders.
Collapse
Affiliation(s)
- Katherine Sellers
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK.
| |
Collapse
|