1
|
Goenaga J, Nanclares C, Hall M, Kofuji P, Mermelstein PG, Araque A. Estradiol Mediates Astrocyte-Neuron Communication in the Hippocampus. Mol Neurobiol 2025:10.1007/s12035-025-04905-6. [PMID: 40208551 DOI: 10.1007/s12035-025-04905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Accumulating evidence has revealed the existence of functional astrocyte-neuron communication based on the ability of astrocytes to respond to neurotransmitters and release gliotransmitters. However, little is known about how other signaling molecules, such as hormones, impact astrocyte function. Estradiol (E2) is an important hormone known to regulate neuronal activity, synaptic transmission, plasticity, and animal behavior. However, whether E2 specifically signals to astrocytes in situ and the functional consequences on astrocyte-neuron communication remain unknown. Therefore, we investigated the impact of estradiol on astrocyte activity and astrocyte-neuron communication in the mouse hippocampus. Using an RNAscope approach, we determined that estrogen receptors (ERα and ERβ) are expressed in astrocytes in both female and male mice. In both sexes, confocal imaging of hippocampal slices determined that astrocytes respond to locally applied E2 with calcium elevations. In pyramidal neurons, slow inward currents (SICs) are mediated by the activation of extrasynaptic NMDA receptors and indicate gliotransmission. Electrophysiological recordings of hippocampal neurons determined that E2 increases the frequency, but not the amplitude, of SICs. We also recorded excitatory synaptic transmission evoked by Schaffer collateral stimulation. Here, only in females, did E2 produce a reduction in excitatory synaptic transmission. The E2-induced effects on the astrocyte calcium signal and gliotransmission were prevented by the broad estrogen receptor antagonist ICI 182,780. Taken together, these results demonstrate the existence of estradiol-mediated astrocyte-neuron communication in both female and male mice. They reveal that E2 can signal to astrocytes and, through this signaling, E2 may regulate neuronal activity and synaptic transmission.
Collapse
Grants
- NIMH R01MH119355 National Institutes of Health, United States
- NIMH R01MH119355 National Institutes of Health, United States
- NIDA R01HD100007 National Institutes of Health, United States
- NIMH R01MH119355 National Institutes of Health, United States
- NIDA R01HD100007 National Institutes of Health, United States
- NIMH R01MH119355 National Institutes of Health, United States
- W911NF2110328 U.S. Department of Defense, United States
- W911NF2110328 U.S. Department of Defense, United States
- W911NF2110328 U.S. Department of Defense, United States
- W911NF2110328 U.S. Department of Defense, United States
Collapse
Affiliation(s)
- Julianna Goenaga
- Department of Neuroscience, University of Minnesota, 6 - 145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Carmen Nanclares
- Department of Neuroscience, University of Minnesota, 6 - 145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Megan Hall
- Department of Neuroscience, University of Minnesota, 6 - 145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, 6 - 145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, 6 - 145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, 6 - 145 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
2
|
de Assis GG, de Sousa MBC, Murawska-Ciałowicz E. Sex Steroids and Brain-Derived Neurotrophic Factor Interactions in the Nervous System: A Comprehensive Review of Scientific Data. Int J Mol Sci 2025; 26:2532. [PMID: 40141172 PMCID: PMC11942429 DOI: 10.3390/ijms26062532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Sex steroids and the neurotrophin brain-derived neurotrophic factor (BDNF) participate in neural tissue formation, phenotypic differentiation, and neuroplasticity. These processes are essential for the health and maintenance of the central nervous system. AIM The aim of our review is to elucidate the interaction mechanisms between BDNF and sex steroids in neuronal function. METHOD A series of searches were performed using Mesh terms for androgen/receptors, estrogen/receptors, and BDNF/receptors, and a collection of the scientific data available on PubMed up to February 2025 about mechanical interactions between BDNF and sex steroids was included in this literature review. DISCUSSION This review discussed the influence of sex steroids on the formation and/or maintenance of neural circuits via different mechanisms, including the regulation of BDNF expression and signaling. Estrogens exert a time- and region-specific effect on BDNF synthesis. The nuclear estrogen receptor can directly regulate BDNF expression, independently of the presence of estrogen, in neuronal cells, whereas progesterone and testosterone upregulate BDNF expression via their specific nuclear receptors. In addition, testosterone has a positive effect on BDNF release by glial cells, which lack androgen receptors.
Collapse
Affiliation(s)
- Gilmara Gomes de Assis
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, 4900-347 Viana do Castelo, Portugal
- Sport Physical Activity and Health Research & Innovation Center, 4900-347 Viana do Castelo, Portugal
| | | | - Eugenia Murawska-Ciałowicz
- Department of Physiology and Biomechanics, Wroclaw University of Health and Sport Sciences, 51-612 Wrocław, Poland;
| |
Collapse
|
3
|
Garcia-Segura LM, Méndez P, Arevalo MA, Azcoitia I. Neuroestradiol and neuronal development: Not an exclusive male tale anymore. Front Neuroendocrinol 2023; 71:101102. [PMID: 37689249 DOI: 10.1016/j.yfrne.2023.101102] [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: 07/26/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
The brain synthesizes a variety of neurosteroids, including neuroestradiol. Inhibition of neuroestradiol synthesis results in alterations in basic neurodevelopmental processes, such as neurogenesis, neuroblast migration, neuritogenesis and synaptogenesis. Although the neurodevelopmental actions of neuroestradiol are exerted in both sexes, some of them are sex-specific, such as the well characterized effects of neuroestradiol derived from the metabolism of testicular testosterone during critical periods of male brain development. In addition, recent findings have shown sex-specific actions of neuroestradiol on neuroblast migration, neuritic growth and synaptogenesis in females. Among other factors, the epigenetic regulation exerted by X linked genes, such as Kdm6a/Utx, may determine sex-specific actions of neuroestradiol in the female brain. This review evidences the impact of neuroestradiol on brain formation in both sexes and highlights the interaction of neural steriodogenesis, hormones and sex chromosomes in sex-specific brain development.
Collapse
Affiliation(s)
- Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain.
| | - Pablo Méndez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - M Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain.
| | - Iñigo Azcoitia
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain; Department of Cell Biology, Universidad Complutense de Madrid, C José Antonio Nováis 12, 28040 Madrid, Spain
| |
Collapse
|
4
|
Fukui K, Sato K, Murakawa S, Minami M, Amano T. Estrogen signaling modulates behavioral selection toward pups and amygdalohippocampal area in the rhomboid nucleus of the bed nucleus of the stria terminalis circuit. Neuropharmacology 2022; 204:108879. [PMID: 34785164 DOI: 10.1016/j.neuropharm.2021.108879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023]
Abstract
Gonadal steroid hormone influences behavioral choice of adult animals toward pups, parental or aggressive. We previously reported that long-term administration of 17β-estradiol (E2) to male mice during sexual maturation induces aggressive behavior toward conspecific pups, which is called "infanticide," and significantly enhanced excitatory synaptic transmission in the rhomboid nucleus of bed nucleus of the stria terminalis (BSTrh), which is an important brain region for infanticide. However, it is unclear how estrogen receptor-dependent signaling after sexual maturity regulates neural circuits including the BSTrh. Here we revealed that E2 administration to gonadectomized mice in adulthood elicited infanticidal behavior and enhanced excitatory synaptic transmission in the BSTrh by increasing the probability of glutamate release from the presynaptic terminalis. Next, we performed whole-brain mapping of E2-sensitive brain regions projecting to the BSTrh and found that amygdalohippocampal area (AHi) neurons that project to the BSTrh densely express estrogen receptor 1 (Esr1). Moreover, E2 treatment enhanced synaptic connectivity in the AHi-BSTrh pathway. Together, these results suggest that reinforcement of excitatory inputs from AHi neurons into the BSTrh by estrogen receptor-dependent signaling may contribute to the expression of infanticide.
Collapse
Affiliation(s)
- Kiyoshiro Fukui
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Keiichiro Sato
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Shunsaku Murakawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan.
| |
Collapse
|
5
|
X-linked histone H3K27 demethylase Kdm6a regulates sexually dimorphic differentiation of hypothalamic neurons. Cell Mol Life Sci 2021; 78:7043-7060. [PMID: 34633482 PMCID: PMC8558156 DOI: 10.1007/s00018-021-03945-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/26/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022]
Abstract
Several X-linked genes are involved in neuronal differentiation and may contribute to the generation of sex dimorphisms in the brain. Previous results showed that XX hypothalamic neurons grow faster, have longer axons, and exhibit higher expression of the neuritogenic gene neurogenin 3 (Ngn3) than XY before perinatal masculinization. Here we evaluated the participation of candidate X-linked genes in the development of these sex differences, focusing mainly on Kdm6a, a gene encoding for an H3K27 demethylase with functions controlling gene expression genome-wide. We established hypothalamic neuronal cultures from wild-type or transgenic Four Core Genotypes mice, a model that allows evaluating the effect of sex chromosomes independently of gonadal type. X-linked genes Kdm6a, Eif2s3x and Ddx3x showed higher expression in XX compared to XY neurons, regardless of gonadal sex. Moreover, Kdm6a expression pattern with higher mRNA levels in XX than XY did not change with age at E14, P0, and P60 in hypothalamus or under 17β-estradiol treatment in culture. Kdm6a pharmacological blockade by GSK-J4 reduced axonal length only in female neurons and decreased the expression of neuritogenic genes Neurod1, Neurod2 and Cdk5r1 in both sexes equally, while a sex-specific effect was observed in Ngn3. Finally, Kdm6a downregulation using siRNA reduced axonal length and Ngn3 expression only in female neurons, abolishing the sex differences observed in control conditions. Altogether, these results point to Kdm6a as a key mediator of the higher axogenesis and Ngn3 expression observed in XX neurons before the critical period of brain masculinization.
Collapse
|
6
|
Chow R, Wessels JM, Foster WG. Brain-derived neurotrophic factor (BDNF) expression and function in the mammalian reproductive Tract. Hum Reprod Update 2020; 26:545-564. [PMID: 32378708 DOI: 10.1093/humupd/dmaa008] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/13/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Neurotrophins of the nerve growth factor family are soluble polypeptides that are best known for their role in nerve growth, survival and differentiation in the central nervous system. A growing body of literature shows that neurotrophins and their receptors are also expressed throughout the reproductive tract. OBJECTIVE AND RATIONALE Neurotrophins are key regulatory proteins in reproductive physiology during development and throughout adult life. Of the neurotrophins, the literature describing the expression and function of brain-derived neurotrophic factor (BDNF) and its high-affinity receptor, neurotrophin receptor kinase-2 (NTRK2), has been expanding rapidly. We therefore conducted a systematic inductive qualitative review of the literature to better define the role of the BDNF in the reproductive tract. We postulate that BDNF and NTRK2 are central regulatory proteins throughout the reproductive system. SEARCH METHODS An electronic search of Medline (PubMed) and Web of Science for articles relating to BDNF and the reproductive system was carried out between January 2018 and February 2019. OUTCOMES In the ovary, BDNF expression and levels have been linked with follicle organisation during ovarian development, follicle recruitment and growth and oocyte maturation. In the endometrium, BDNF is involved in cell proliferation and neurogenesis. In contrast, literature describing the role of BDNF in other reproductive tissues is sparse and BDNF-NTRK2 signalling in the male reproductive tract has been largely overlooked. Whilst estradiol appears to be the primary regulator of BDNF expression, we also identified reports describing binding sites for glucocorticoid and myocyte enhancer factor-2, a calcium-response element through activation of an N-methyl-D-aspartate (NMDA) receptor, and aryl hydrocarbon receptor nuclear transporter protein-4 (ARNT) response elements in promoter regions of the BDNF gene. Expression is also regulated by multiple microRNAs and post-translational processing of precursor proteins and intracellular shuttling. BDNF-NTRK2 signalling is modulated through tissue specific receptor expression of either the full-length or truncated NTRK2 receptor; however, the functional importance remains to be elucidated. Dysregulation of BDNF expression and circulating concentrations have been implicated in several reproductive disorders including premature ovarian failure, endometriosis, pre-eclampsia, intra-uterine growth restriction (IUGR) and several reproductive cancers. WIDER IMPLICATIONS We conclude that BDNF and its receptors are key regulatory proteins central to gonadal development, ovarian regulation and uterine physiology, as well as embryo and placenta development. Furthermore, dysregulation of BDNF-NTRK2 in reproductive diseases suggests their potential role as candidate clinical markers of disease and potential therapeutic targets.
Collapse
Affiliation(s)
- R Chow
- Department of Obstetrics & Gynaecology, McMaster University, Hamilton, Ontario, Canada
| | - J M Wessels
- Department of Obstetrics & Gynaecology, McMaster University, Hamilton, Ontario, Canada
| | - W G Foster
- Department of Obstetrics & Gynaecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Tibolone regulates systemic metabolism and the expression of sex hormone receptors in the central nervous system of ovariectomised rats fed with high-fat and high-fructose diet. Brain Res 2020; 1748:147096. [DOI: 10.1016/j.brainres.2020.147096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/04/2023]
|
8
|
Cisternas CD, Cabrera Zapata LE, Mir FR, Scerbo MJ, Arevalo MA, Garcia-Segura LM, Cambiasso MJ. Estradiol-dependent axogenesis and Ngn3 expression are determined by XY sex chromosome complement in hypothalamic neurons. Sci Rep 2020; 10:8223. [PMID: 32427857 PMCID: PMC7237695 DOI: 10.1038/s41598-020-65183-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/14/2020] [Indexed: 01/15/2023] Open
Abstract
Hypothalamic neurons show sex differences in neuritogenesis, female neurons have longer axons and higher levels of the neuritogenic factor neurogenin 3 (Ngn3) than male neurons in vitro. Moreover, the effect of 17-β-estradiol (E2) on axonal growth and Ngn3 expression is only found in male-derived neurons. To investigate whether sex chromosomes regulate these early sex differences in neuritogenesis by regulating the E2 effect on Ngn3, we evaluated the growth and differentiation of hypothalamic neurons derived from the “four core genotypes” mouse model, in which the factors of “gonadal sex” and “sex chromosome complement” are dissociated. We showed that sex differences in neurite outgrowth are determined by sex chromosome complement (XX > XY). Moreover, E2 increased the mRNA expression of Ngn3 and axonal length only in XY neurons. ERα/β expressions are regulated by sex chromosome complement; however, E2-effect on Ngn3 expression in XY neurons was only fully reproduced by PPT, a specific ligand of ERα, and prevented by MPP, a specific antagonist of ERα. Together our data indicate that sex chromosomes regulate early development of hypothalamic neurons by orchestrating not only sex differences in neuritogenesis, but also regulating the effect of E2 on Ngn3 expression through activation of ERα in hypothalamic neurons.
Collapse
Affiliation(s)
- Carla Daniela Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Biología Bucal, Facultad de Odontología -Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lucas Ezequiel Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Franco Rafael Mir
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Julia Scerbo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Angeles Arevalo
- Instituto Cajal, CSIC, Madrid, Spain.,Ciber de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain.,Ciber de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina. .,Departamento de Biología Bucal, Facultad de Odontología -Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
9
|
Mir FR, Wilson C, Cabrera Zapata LE, Aguayo LG, Cambiasso MJ. Gonadal hormone-independent sex differences in GABA A receptor activation in rat embryonic hypothalamic neurons. Br J Pharmacol 2020; 177:3075-3090. [PMID: 32133616 DOI: 10.1111/bph.15037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE GABAA receptor functions are dependent on subunit composition, and, through their activation, GABA can exert trophic actions in immature neurons. Although several sex differences in GABA-mediated responses are known to be dependent on gonadal hormones, few studies have dealt with sex differences detected before the critical period of brain masculinisation. In this study, we assessed GABAA receptor functionality in sexually segregated neurons before brain hormonal masculinisation. EXPERIMENTAL APPROACH Ventromedial hypothalamic neurons were obtained from embryonic day 16 rat brains and grown in vitro for 2 days. Calcium imaging and electrophysiology recordings were carried out to assess GABAA receptor functional parameters. KEY RESULTS GABAA receptor activation elicited calcium entry in immature hypothalamic neurons mainly through L-type voltage-dependent calcium channels. Nifedipine blocked calcium entry more efficiently in male than in female neurons. There were more male than female neurons responding to GABA, and they needed more time to return to resting levels. Pharmacological characterisation revealed that propofol enhanced GABAA -mediated currents and blunted GABA-mediated calcium entry more efficiently in female neurons than in males. Testosterone treatment did not erase such sex differences. These data suggest sex differences in the expression of GABAA receptor subtypes. CONCLUSION AND IMPLICATIONS GABA-mediated responses are sexually dimorphic even in the absence of gonadal hormone influence, suggesting genetically biased differences. These results highlight the importance of GABAA receptors in hypothalamic neurons even before hormonal masculinisation of the brain.
Collapse
Affiliation(s)
- Franco R Mir
- Laboratorio de Neurofisiología, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Cátedra de Fisiología Animal, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina.,Cátedra de Fisiología Animal, Departamento de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de La Rioja, La Rioja, Argentina
| | - Carlos Wilson
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigación en Medicina Traslaciona, Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina
| | - Lucas E Cabrera Zapata
- Laboratorio de Neurofisiología, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Luis G Aguayo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - María Julia Cambiasso
- Laboratorio de Neurofisiología, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
10
|
Cabrera Zapata LE, Bollo M, Cambiasso MJ. Estradiol-Mediated Axogenesis of Hypothalamic Neurons Requires ERK1/2 and Ryanodine Receptors-Dependent Intracellular Ca 2+ Rise in Male Rats. Front Cell Neurosci 2019; 13:122. [PMID: 31001087 PMCID: PMC6454002 DOI: 10.3389/fncel.2019.00122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
17β-estradiol (E2) induces axonal growth through extracellular signal-regulated kinase 1 and 2 (ERK1/2)-MAPK cascade in hypothalamic neurons of male rat embryos in vitro, but the mechanism that initiates these events is poorly understood. This study reports the intracellular Ca2+ increase that participates in the activation of ERK1/2 and axogenesis induced by E2. Hypothalamic neuron cultures were established from 16-day-old male rat embryos and fed with astroglia-conditioned media for 48 h. E2-induced ERK phosphorylation was completely abolished by a ryanodine receptor (RyR) inhibitor (ryanodine) and partially attenuated by an L-type voltage-gated Ca2+ channel (L-VGCC) blocker (nifedipine), an inositol-1,4,5-trisphosphate receptor (IP3R) inhibitor (2-APB), and a phospholipase C (PLC) inhibitor (U-73122). We also conducted Ca2+ imaging recording using primary cultured neurons. The results show that E2 rapidly induces an increase in cytosolic Ca2+, which often occurs in repetitive Ca2+ oscillations. This response was not observed in the absence of extracellular Ca2+ or with inhibitory ryanodine and was markedly reduced by nifedipine. E2-induced axonal growth was completely inhibited by ryanodine. In summary, the results suggest that Ca2+ mobilization from extracellular space as well as from the endoplasmic reticulum is necessary for E2-induced ERK1/2 activation and axogenesis. Understanding the mechanisms of brain estrogenic actions might contribute to develop novel estrogen-based therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Lucas E Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Bollo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Cátedra de Biología Celular, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
11
|
Baud O, Berkane N. Hormonal Changes Associated With Intra-Uterine Growth Restriction: Impact on the Developing Brain and Future Neurodevelopment. Front Endocrinol (Lausanne) 2019; 10:179. [PMID: 30972026 PMCID: PMC6443724 DOI: 10.3389/fendo.2019.00179] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
The environment in which a fetus develops is not only important for its growth and maturation but also for its long-term postnatal health and neurodevelopment. Several hormones including glucocorticosteroids, estrogens and progesterone, insulin growth factor and thyroid hormones, carefully regulate the growth of the fetus and its metabolism during pregnancy by controlling the supply of nutrients crossing the placenta. In addition to fetal synthesis, hormones regulating fetal growth are also expressed and regulated in the placenta, and they play a key role in the vulnerability of the developing brain and its maturation. This review summarizes the current understanding and evidence regarding the involvement of hormonal dysregulation associated with intra-uterine growth restriction and its consequences on brain development.
Collapse
Affiliation(s)
- Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Department of Women-Children-Teenagers, University Hospitals Geneva, Geneva, Switzerland
- Inserm U1141, Sorbonne, Paris Diderot University, Paris, France
- *Correspondence: Olivier Baud
| | - Nadia Berkane
- Division of Obstetrics and Gynecology, Department of Women-Children-Teenagers, University Hospitals Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Mir FR, Carrer HF, Cambiasso MJ. Sex differences in depolarizing actions of GABA A receptor activation in rat embryonic hypothalamic neurons. Eur J Neurosci 2016; 45:521-527. [PMID: 27888546 DOI: 10.1111/ejn.13467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/27/2016] [Accepted: 11/02/2016] [Indexed: 11/28/2022]
Abstract
GABAA receptor activation exerts trophic actions in immature neurons through depolarization of resting membrane potential. The switch to its classical hyperpolarizing role is developmentally regulated. Previous results suggest that a hormonally biased sex difference exists at the onset of the switch in hypothalamic neurons. The aim of this work was to evaluate sex differences in GABAA receptor function of hypothalamic neurons before brain masculinization by gonadal hormones. Hypothalamic cells were obtained from embryonic day 16 male and female rat foetuses, 2 days before the peak of testosterone production by the foetal testis, and grown in vitro for 9 days. Whole-cell and perforated patch-clamp recordings were carried out in order to measure several electrophysiological parameters. Our results show that there are more male than female neurons responding with depolarization to muscimol. Additionally, among cells with depolarizing responses, males have higher and longer lasting responses than females. These results highlight the relevance of differences in neural cell sex irrespective of exposure to sex hormones.
Collapse
Affiliation(s)
- Franco R Mir
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Hugo F Carrer
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - María J Cambiasso
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.,Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
13
|
Rossetti MF, Cambiasso MJ, Holschbach MA, Cabrera R. Oestrogens and Progestagens: Synthesis and Action in the Brain. J Neuroendocrinol 2016; 28. [PMID: 27306650 DOI: 10.1111/jne.12402] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/14/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022]
Abstract
When steroids, such as pregnenolone, progesterone and oestrogen, are synthesised de novo in neural tissues, they are more specifically referred to as neurosteroids. These neurosteroids bind specific receptors to promote essential brain functions. Pregnenolone supports cognition and protects mouse hippocampal cells against glutamate and amyloid peptide-induced cell death. Progesterone promotes myelination, spinogenesis, synaptogenesis, neuronal survival and dendritic growth. Allopregnanolone increases hippocampal neurogenesis, neuronal survival and cognitive functions. Oestrogens, such as oestradiol, regulate synaptic plasticity, reproductive behaviour, aggressive behaviour and learning. In addition, neurosteroids are neuroprotective in animal models of Alzheimer's disease, Parkinson's disease, brain injury and ageing. Using in situ hybridisation and/or immunohistochemistry, steroidogenic enzymes, including cytochrome P450 side-chain cleavage, 3β-hydroxysteroid dehydrogenase/Δ5-Δ4 isomerase, cytochrome P450arom, steroid 5α-reductase and 3α-hydroxysteroid dehydrogenase, have been detected in numerous brain regions, including the hippocampus, hypothalamus and cerebral cortex. In the present review, we summarise some of the studies related to the synthesis and function of oestrogens and progestagens in the central nervous system.
Collapse
Affiliation(s)
- M F Rossetti
- Departamento de Bioquímica Clínica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Instituto de Salud y Ambiente del Litoral, CONICET-Universidad Nacional del Litoral, Santa Fe, Argentina
| | - M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M A Holschbach
- Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - R Cabrera
- Instituto de Investigaciones Biomédicas, INBIOMED-IMBECU-CONICET, Universidad de Mendoza, Mendoza, Argentina
| |
Collapse
|
14
|
Yang W, Zhang H. Effects of hindlimb unloading on neurotrophins in the rat spinal cord and soleus muscle. Brain Res 2015; 1630:1-9. [PMID: 26529644 DOI: 10.1016/j.brainres.2015.10.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/15/2015] [Accepted: 10/25/2015] [Indexed: 10/22/2022]
Abstract
The aim of the present study was to investigate the effects of hindlimb unloading (HU) on the expression of neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF), together with the expression of their high-affinity receptors tropomyosin receptor kinase C (TrkC) and tropomyosin receptor kinase B (TrkB), in lumbar (L4-6) segment of the spinal cord and in the soleus muscle. The mRNA and protein levels of the genes of interest were compared using quantitative PCR and western blot assays. Immunohistochemistry for NT-3 and BDNF was used to detect the levels of protein in the motoneurons in the lateral motor column. In this study, NT-3 and BDNF mRNA and protein expression were significantly increased in the spinal cord and soleus muscle after HU. NT-3 immunoreactivity, but not BDNF immunoreactivity, was significantly increased in the large motoneurons located in lateral motor column after 14 days of HU. The level of TrkC protein in the spinal cord and soleus muscle were significantly elevated after both 7 days and 14 days of HU. However, TrkC mRNA, TrkB mRNA and TrkB protein levels did not change significantly. Elevated BDNF, NT-3 and TrkC levels in the neuromuscular system indicate that neurotrophins are involved in HU-induced neuromuscular plasticity. NT-3 is a candidate to mediate the synaptic efficacy between alpha motoneurons and group Ia afferents.
Collapse
Affiliation(s)
- Wei Yang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China.
| | - Hao Zhang
- Key Laboratory of Ministry of Education, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
15
|
Kranz GS, Hahn A, Kaufmann U, Küblböck M, Hummer A, Ganger S, Seiger R, Winkler D, Swaab DF, Windischberger C, Kasper S, Lanzenberger R. White matter microstructure in transsexuals and controls investigated by diffusion tensor imaging. J Neurosci 2014; 34:15466-75. [PMID: 25392513 PMCID: PMC4699258 DOI: 10.1523/jneurosci.2488-14.2014] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/16/2014] [Accepted: 10/02/2014] [Indexed: 12/22/2022] Open
Abstract
Biological causes underpinning the well known gender dimorphisms in human behavior, cognition, and emotion have received increased attention in recent years. The advent of diffusion-weighted magnetic resonance imaging has permitted the investigation of the white matter microstructure in unprecedented detail. Here, we aimed to study the potential influences of biological sex, gender identity, sex hormones, and sexual orientation on white matter microstructure by investigating transsexuals and healthy controls using diffusion tensor imaging (DTI). Twenty-three female-to-male (FtM) and 21 male-to-female (MtF) transsexuals, as well as 23 female (FC) and 22 male (MC) controls underwent DTI at 3 tesla. Fractional anisotropy, axial, radial, and mean diffusivity were calculated using tract-based spatial statistics (TBSS) and fiber tractography. Results showed widespread significant differences in mean diffusivity between groups in almost all white matter tracts. FCs had highest mean diffusivities, followed by FtM transsexuals with lower values, MtF transsexuals with further reduced values, and MCs with lowest values. Investigating axial and radial diffusivities showed that a transition in axial diffusivity accounted for mean diffusivity results. No significant differences in fractional anisotropy maps were found between groups. Plasma testosterone levels were strongly correlated with mean, axial, and radial diffusivities. However, controlling for individual estradiol, testosterone, or progesterone plasma levels or for subjects' sexual orientation did not change group differences. Our data harmonize with the hypothesis that fiber tract development is influenced by the hormonal environment during late prenatal and early postnatal brain development.
Collapse
Affiliation(s)
| | | | | | - Martin Küblböck
- MR Centre of Excellence, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria, and
| | - Allan Hummer
- MR Centre of Excellence, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria, and
| | | | - Rene Seiger
- Departments of Psychiatry and Psychotherapy and
| | | | - Dick F Swaab
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| | - Christian Windischberger
- MR Centre of Excellence, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria, and
| | | | | |
Collapse
|
16
|
Tsukahara S, Kanaya M, Yamanouchi K. Neuroanatomy and sex differences of the lordosis-inhibiting system in the lateral septum. Front Neurosci 2014; 8:299. [PMID: 25278832 PMCID: PMC4166118 DOI: 10.3389/fnins.2014.00299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 09/02/2014] [Indexed: 12/21/2022] Open
Abstract
Female sexual behavior in rodents, termed lordosis, is controlled by facilitatory and inhibitory systems in the brain. It has been well demonstrated that a neural pathway from the ventromedial hypothalamic nucleus (VMN) to the midbrain central gray (MCG) is essential for facilitatory regulation of lordosis. The neural pathway from the arcuate nucleus to the VMN, via the medial preoptic nucleus, in female rats mediates transient suppression of lordosis, until female sexual receptivity is induced. In addition to this pathway, other regions are involved in inhibitory regulation of lordosis in female rats. The lordosis-inhibiting systems exist not only in the female brain but also in the male brain. The systems contribute to suppression of heterotypical sexual behavior in male rats, although they have the potential ability to display lordosis. The lateral septum (LS) exerts an inhibitory influence on lordosis in both female and male rats. This review focuses on the neuroanatomy and sex differences of the lordosis-inhibiting system in the LS. The LS functionally and anatomically links to the MCG to exert suppression of lordosis. Neurons of the intermediate part of the LS (LSi) serve as lordosis-inhibiting neurons and project axons to the MCG. The LSi-MCG neural connection is sexually dimorphic, and formation of the male-like LSi-MCG neural connection is affected by aromatized testosterone originating from the testes in the postnatal period. The sexually dimorphic LSi-MCG neural connection may reflect the morphological basis of sex differences in the inhibitory regulation of lordosis in rats.
Collapse
Affiliation(s)
- Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Moeko Kanaya
- Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Korehito Yamanouchi
- Department of Human Behavior and Environment Sciences, Faculty of Human Sciences, Waseda University Saitama, Japan
| |
Collapse
|
17
|
Wang C, Jie C, Dai X. Possible roles of astrocytes in estrogen neuroprotection during cerebral ischemia. Rev Neurosci 2014; 25:255-68. [PMID: 24566361 DOI: 10.1515/revneuro-2013-0055] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/29/2014] [Indexed: 01/08/2023]
Abstract
17β-Estradiol (E2), one of female sex hormones, has well-documented neuroprotective effects in a variety of clinical and experimental disorders of the central cerebral ischemia, including stroke and neurodegenerative diseases. The cellular mechanisms that underlie these protective effects of E2 are uncertain because a number of different cell types express estrogen receptors in the central nervous system. Astrocytes are the most abundant cells in the central nervous system and provide structural and nutritive support of neurons. They interact with neurons by cross-talk, both physiologically and pathologically. Proper astrocyte function is particularly important for neuronal survival under ischemic conditions. Dysfunction of astrocytes resulting from ischemia significantly influences the responses of other brain cells to injury. Recent studies demonstrate that estrogen receptors are expressed in astrocytes, indicating that E2 may exert multiple regulatory actions on astrocytes. Cerebral ischemia induced changes in the expression of estrogen receptors in astrocytes. In the present review, we summarize the data in support of possible roles for astrocytes in the mediation of neuroprotection by E2 against cerebral ischemia.
Collapse
|
18
|
Scerbo MJ, Freire-Regatillo A, Cisternas CD, Brunotto M, Arevalo MA, Garcia-Segura LM, Cambiasso MJ. Neurogenin 3 mediates sex chromosome effects on the generation of sex differences in hypothalamic neuronal development. Front Cell Neurosci 2014; 8:188. [PMID: 25071448 PMCID: PMC4086225 DOI: 10.3389/fncel.2014.00188] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/19/2014] [Indexed: 01/01/2023] Open
Abstract
The organizational action of testosterone during critical periods of development is the cause of numerous sex differences in the brain. However, sex differences in neuritogenesis have been detected in primary neuronal hypothalamic cultures prepared before the peak of testosterone production by fetal testis. In the present study we assessed the hypothesis of that cell-autonomous action of sex chromosomes can differentially regulate the expression of the neuritogenic gene neurogenin 3 (Ngn3) in male and female hypothalamic neurons, generating sex differences in neuronal development. Neuronal cultures were prepared from male and female E14 mouse hypothalami, before the fetal peak of testosterone. Female neurons showed enhanced neuritogenesis and higher expression of Ngn3 than male neurons. The silencing of Ngn3 abolished sex differences in neuritogenesis, decreasing the differentiation of female neurons. The sex difference in Ngn3 expression was determined by sex chromosomes, as demonstrated using the four core genotypes mouse model, in which a spontaneous deletion of the testis-determining gene Sry from the Y chromosome was combined with the insertion of the Sry gene onto an autosome. In addition, the expression of Ngn3, which is also known to mediate the neuritogenic actions of estradiol, was increased in the cultures treated with the hormone, but only in those from male embryos. Furthermore, the hormone reversed the sex differences in neuritogenesis promoting the differentiation of male neurons. These findings indicate that Ngn3 mediates both cell-autonomous actions of sex chromosomes and hormonal effects on neuritogenesis.
Collapse
Affiliation(s)
- María J Scerbo
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET - Universidad Nacional de Córdoba Córdoba, Argentina
| | | | - Carla D Cisternas
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET - Universidad Nacional de Córdoba Córdoba, Argentina ; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba Córdoba, Argentina
| | - Mabel Brunotto
- Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba Córdoba, Argentina
| | - Maria A Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas Madrid, Spain
| | | | - María J Cambiasso
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET - Universidad Nacional de Córdoba Córdoba, Argentina ; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba Córdoba, Argentina
| |
Collapse
|
19
|
Gonzalez B, Ratner LD, Scerbo MJ, Di Giorgio NP, Poutanen M, Huhtaniemi IT, Calandra RS, Lux-Lantos VAR, Cambiasso MJ, Rulli SB. Elevated hypothalamic aromatization at the onset of precocious puberty in transgenic female mice hypersecreting human chorionic gonadotropin: effect of androgens. Mol Cell Endocrinol 2014; 390:102-11. [PMID: 24755422 DOI: 10.1016/j.mce.2014.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/03/2014] [Accepted: 04/10/2014] [Indexed: 01/26/2023]
Abstract
Transgenic female mice overexpressing the α- and β- subunits of human chorionic gonadotropin (hCGαβ+) exhibited precocious puberty, as evidenced by early vaginal opening. Chronically elevated hCG in 21-day-old hCGαβ+ females stimulated gonadal androgen production, which exerted negative feedback over the endogenous gonadotropin synthesis, and activated the hypothalamic GnRH pulsatility and gene expression. Transgenic females also exhibited elevated hypothalamic aromatization in the preoptic area (POA), which is the sexually-differentiated area that controls the LH surge in adulthood. Ovariectomy at 14 days of age was unable to rescue this phenotype. However, the blockade of androgen action by flutamide from postnatal day 6 onwards reduced the aromatase levels in the POA of hCGαβ+ females. Our results suggest that early exposure of females to androgen action during a critical period between postnatal days 6-14 induces sex-specific organizational changes of the brain, which affect the aromatase expression in the POA at the onset of precocious puberty.
Collapse
Affiliation(s)
- Betina Gonzalez
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina
| | - Laura D Ratner
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina
| | - María J Scerbo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Casilla de Correo 389, 5000 Córdoba, Argentina
| | - Noelia P Di Giorgio
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina
| | - Matti Poutanen
- Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland; Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| | - Ilpo T Huhtaniemi
- Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland; Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina
| | - Victoria A R Lux-Lantos
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina
| | - María J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Casilla de Correo 389, 5000 Córdoba, Argentina
| | - Susana B Rulli
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428 Buenos Aires, Argentina.
| |
Collapse
|
20
|
Yamaura K, Bi Y, Ishiwatari M, Oishi N, Fukata H, Ueno K. Sex differences in stress reactivity of hippocampal BDNF in mice are associated with the female preponderance of decreased locomotor activity in response to restraint stress. Zoolog Sci 2014; 30:1019-24. [PMID: 24320179 DOI: 10.2108/zsj.30.1019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The incidence and prevalence of depression is higher in women than in men, but the cause of this sex discrepancy remains unknown. Brain-derived neurotrophic factor (BDNF) is a key protein for maintaining neuronal integrity. The purpose of this study was to investigate the female preponderance in behavioral responsivity to restraint stress focusing on the stress reactivity of BDNF in the hippocampus. Male and female ICR mice were exposed to a 3-h session of restraint stress. Plasma corticosterone was measured by high-performance liquid chromatography. BDNF mRNA expression in the whole hippocampus was measured by quantitative real-time reverse transcription-polymerase chain reaction. Wheel-running activity was monitored during the dark period. In response to restraint stress, the increase in levels of serum corticosterone was higher in female than in male mice. Restraint stress resulted in decreased voluntary wheel-running behavior that was greater in female than male animals. In addition to these sex differences in stress reactivity, we found a significant sex difference in BDNF levels in the hippocampus of restraint-stressed mice; total BDNF levels significantly decreased in female mice, but not in male mice in response to the stress. Furthermore, BDNF exon I and IV mRNA expression also showed the same tendency. These data indicate that the reduction in levels of voluntary wheel-running activity in response to stress can be significantly influenced by sex. Moreover, our findings suggest a link between the sex differences in this behavioral response to stress and differential stress reactivity in the production of BDNF in the hippocampus.
Collapse
Affiliation(s)
- Katsunori Yamaura
- 1 Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Pfister A, Johnson A, Ellers O, Horch HW. Quantification of dendritic and axonal growth after injury to the auditory system of the adult cricket Gryllus bimaculatus. Front Physiol 2013; 3:367. [PMID: 23986706 PMCID: PMC3750946 DOI: 10.3389/fphys.2012.00367] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 08/27/2012] [Indexed: 12/13/2022] Open
Abstract
Dendrite and axon growth and branching during development are regulated by a complex set of intracellular and external signals. However, the cues that maintain or influence adult neuronal morphology are less well understood. Injury and deafferentation tend to have negative effects on adult nervous systems. An interesting example of injury-induced compensatory growth is seen in the cricket, Gryllus bimaculatus. After unilateral loss of an ear in the adult cricket, auditory neurons within the central nervous system (CNS) sprout to compensate for the injury. Specifically, after being deafferented, ascending neurons (AN-1 and AN-2) send dendrites across the midline of the prothoracic ganglion where they receive input from auditory afferents that project through the contralateral auditory nerve (N5). Deafferentation also triggers contralateral N5 axonal growth. In this study, we quantified AN dendritic and N5 axonal growth at 30 h, as well as at 3, 5, 7, 14, and 20 days after deafferentation in adult crickets. Significant differences in the rates of dendritic growth between males and females were noted. In females, dendritic growth rates were non-linear; a rapid burst of dendritic extension in the first few days was followed by a plateau reached at 3 days after deafferentation. In males, however, dendritic growth rates were linear, with dendrites growing steadily over time and reaching lengths, on average, twice as long as in females. On the other hand, rates of N5 axonal growth showed no significant sexual dimorphism and were linear. Within each animal, the growth rates of dendrites and axons were not correlated, indicating that independent factors likely influence dendritic and axonal growth in response to injury in this system. Our findings provide a basis for future study of the cellular features that allow differing dendrite and axon growth patterns as well as sexually dimorphic dendritic growth in response to deafferentation.
Collapse
Affiliation(s)
- Alexandra Pfister
- Department of Invertebrate Zoology, American Museum of Natural History New York, NY, USA
| | | | | | | |
Collapse
|
22
|
Sex and stress hormone influences on the expression and activity of brain-derived neurotrophic factor. Neuroscience 2012; 239:295-303. [PMID: 23211562 DOI: 10.1016/j.neuroscience.2012.10.073] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 01/02/2023]
Abstract
The neurotrophin, brain-derived neurotrophic factor (BDNF), is recognized as a key component in the regulation of CNS ontogeny, homeostasis and adult neuroplasticity. The importance of BDNF in CNS development and function is well documented by numerous reports from animal studies linking abnormal BDNF signaling to metabolic disturbances and anxiety or depressive-like behavior. Despite the diverse roles for BDNF in nearly all aspects of CNS physiology, the regulation of BDNF expression, as well as our understanding of the signaling mechanisms associated with this neurotrophin, remains incomplete. However, links between sex hormones such as estradiol and testosterone, as well as endogenous and synthetic glucocorticoids (GCs), have emerged as important mediators of BDNF expression and function. Examples of such regulation include brain region-specific induction of Bdnf mRNA in response to estradiol. Additional studies have also documented regulation of the expression of the high-affinity BDNF receptor Tropomyosin-Related Kinase B by estradiol, thus implicating sex steroids not only in the regulation of BDNF expression, but also in mechanisms of signaling associated with it. In addition to gonadal steroids, further evidence also suggests functional interaction between BDNF and GCs, such as in the regulation of corticotrophin-releasing hormone and other important neuropeptides. In this review, we provide an overview of the roles played by selected sex or stress hormones in the regulation of BDNF expression and signaling in the CNS.
Collapse
|
23
|
Hill RA. Interaction of sex steroid hormones and brain-derived neurotrophic factor-tyrosine kinase B signalling: relevance to schizophrenia and depression. J Neuroendocrinol 2012; 24:1553-61. [PMID: 22845879 DOI: 10.1111/j.1365-2826.2012.02365.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 07/10/2012] [Accepted: 07/21/2012] [Indexed: 01/25/2023]
Abstract
Sex steroid hormones and neurotrophic factors are involved in pruning and shaping the developing brain and have been implicated in the pathogenesis of neurodevelopmental disorders. Sex steroid hormones are also involved in the regulation of brain-derived neurotrophic factor expression. A review of the literature is provided on the relationship between brain-derived neurotrophic factor and sex steroid hormones, as well as the mechanisms behind this interaction, in the context of how this relationship may be involved in the development of neurodevelopmental psychiatric illnesses, such as schizophrenia and depression.
Collapse
Affiliation(s)
- R A Hill
- Behavioural Neuroscience Laboratory, Mental Health Research Institute, Melbourne, Australia.
| |
Collapse
|
24
|
Juncos LI, Martín FL, Baigorria ST, Pasqualini ME, Fiore MC, Eynard AR, Juncos LA, García NH. Atorvastatin improves sodium handling and decreases blood pressure in salt-loaded rats with chronic renal insufficiency. Nutrition 2012; 28:e23-8. [DOI: 10.1016/j.nut.2012.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Revised: 02/07/2012] [Accepted: 02/07/2012] [Indexed: 11/25/2022]
|
25
|
17β-estradiol regulates the sexually dimorphic expression of BDNF and TrkB proteins in the song system of juvenile zebra finches. PLoS One 2012; 7:e43687. [PMID: 22952738 PMCID: PMC3432032 DOI: 10.1371/journal.pone.0043687] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/23/2012] [Indexed: 11/19/2022] Open
Abstract
Mature brain derived neurotrophic factor (BDNF) plays critical roles in development of brain structure and function, including neurogenesis, axon growth, cell survival and processes associated with learning. Expression of this peptide is regulated by estradiol (E2). The zebra finch song system is sexually dimorphic - only males sing and the brain regions controlling song are larger and have more cells in males compared to females. Masculinization of this system is partially mediated by E2, and earlier work suggests that BDNF with its high affinity receptor TrkB may also influence this development. The present study evaluated expression of multiple forms of both BDNF and TrkB in the developing song system in juvenile males and females treated with E2 or a vehicle control. Using immunohistochemistry and Western blot analysis, BDNF was detected across the song nuclei of 25-day-old birds. Westerns allowed the pro- and mature forms of BDNF to be individually identified, and proBDNF to be quantified. Several statistically significant effects of sex existed in both the estimated total number of BDNF+ cells and relative concentration of proBDNF, varying across the regions and methodologies. E2 modulated BDNF expression, although the specific nature of the regulation depended on brain region, sex and the technique used. Similarly, TrkB (both truncated and full-length isoforms) was detected by Western blot in the song system of juveniles of both sexes, and expression was regulated by E2. In the context of earlier research on these molecules in the developing song system, this work provides a critical step in describing specific forms of BDNF and TrkB, and how they can be mediated by sex and E2. As individual isoforms of each can have opposing effects on mechanisms, such as cell survival, it will now be important to investigate in depth their specific functions in song system maturation.
Collapse
|
26
|
Arevalo MA, Ruiz-Palmero I, Scerbo MJ, Acaz-Fonseca E, Cambiasso MJ, Garcia-Segura LM. Molecular mechanisms involved in the regulation of neuritogenesis by estradiol: Recent advances. J Steroid Biochem Mol Biol 2012; 131:52-6. [PMID: 21971420 DOI: 10.1016/j.jsbmb.2011.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 09/19/2011] [Accepted: 09/21/2011] [Indexed: 12/13/2022]
Abstract
This review analyzes the signaling mechanisms activated by estradiol to regulate neuritogenesis in several neuronal populations. Estradiol regulates axogenesis by the activation of the mitogen activated protein kinase (MAPK) cascade through estrogen receptor α located in the plasma membrane. In addition, estradiol regulates MAPK signaling via the activation of protein kinase C and by increasing the expression of brain derived neurotrophic factor and tyrosine kinase receptor B. Estradiol also interacts with the signaling of insulin-like growth factor-I receptor through estrogen receptor α, modulating the phosphoinositide-3 kinase signaling pathway, which contributes to the stabilization of microtubules. Finally, estradiol modulates dendritogenesis by the inhibition of Notch signaling, by a mechanism that, at least in hippocampal neurons, is mediated by G-protein coupled receptor 30. This article is part of a Special Issue entitled 'Neurosteroids'.
Collapse
|
27
|
Azcoitia I, Santos-Galindo M, Arevalo MA, Garcia-Segura LM. Role of astroglia in the neuroplastic and neuroprotective actions of estradiol. Eur J Neurosci 2010; 32:1995-2002. [DOI: 10.1111/j.1460-9568.2010.07516.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
28
|
Actions of estrogens on glial cells: Implications for neuroprotection. Biochim Biophys Acta Gen Subj 2010; 1800:1106-12. [DOI: 10.1016/j.bbagen.2009.10.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 09/29/2009] [Accepted: 10/01/2009] [Indexed: 01/21/2023]
|
29
|
Interactions of estradiol and insulin-like growth factor-I signalling in the nervous system: new advances. PROGRESS IN BRAIN RESEARCH 2010; 181:251-72. [PMID: 20478442 DOI: 10.1016/s0079-6123(08)81014-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Estradiol and insulin-like growth factor-I (IGF-I) interact in the brain to regulate a variety of developmental and neuroplastic events. Some of these interactions are involved in the control of hormonal homeostasis and reproduction. However, the interactions may also potentially impact on affection and cognition by the regulation of adult neurogenesis in the hippocampus and by promoting neuroprotection under neurodegenerative conditions. Recent studies suggest that the interaction of estradiol and IGF-I is also relevant for the control of cholesterol homeostasis in neural cells. The molecular mechanisms involved in the interaction of estradiol and IGF-I include the cross-regulation of the expression of estrogen and IGF-I receptors, the regulation of estrogen receptor-mediated transcription by IGF-I and the regulation of IGF-I receptor signalling by estradiol. Current investigations are evidencing the role exerted by key signalling molecules, such as glycogen synthase kinase 3 and beta-catenin, in the cross-talk of estrogen receptors and IGF-I receptors in neural cells.
Collapse
|
30
|
Estrogen receptor α is expressed on the cell-surface of embryonic hypothalamic neurons. Neuroscience 2008; 154:1173-7. [DOI: 10.1016/j.neuroscience.2008.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Revised: 04/11/2008] [Accepted: 05/02/2008] [Indexed: 11/19/2022]
|
31
|
Gorosito SV, Cambiasso MJ. Axogenic effect of estrogen in male rat hypothalamic neurons involves Ca(2+), protein kinase C, and extracellular signal-regulated kinase signaling. J Neurosci Res 2008; 86:145-57. [PMID: 17722067 DOI: 10.1002/jnr.21466] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
17-beta-Estradiol (E2) stimulates the growth of axons in male-derived hypothalamic neurons in vitro. This effect is not exerted through the classical intracellular estrogen receptor (ER) but depends on a membrane mechanism involving TrkB. In the present study, we investigate the intracellular signaling cascade that mediates the axogenic effect of E2. Treatment with an intracellular Ca(2+) chelator, a Ca(2+)-dependent protein kinase C (PKC) inhibitor, or two specific inhibitors of extracellular signal-regulated kinases (ERK) mitogen-activated protein kinases (MAPK) completely inhibited the E2-induced axogenesis. E2 and the membrane-impermeant construct E2BSA rapidly induced phosphorylation of ERK, which was blocked by the specific inhibitor of the ERK pathway UO126 but not by the ER antagonist ICI 182,780. Decrease of intracellular free Ca(2+) or disruption of PKC activation by Ro 32-0432 attenuated ERK activation, indicating the confluence of signals in the MAPK pathway. Subcellular analysis of ERK demonstrated that the phospho-ERK signal is augmented in the nucleus after 15 min of E2 stimulation. We have also shown that E2 increased phosphorylation of CREB via ERK signaling. In summary, this study demonstrates that E2, probably via a membrane-associated receptor, induces axonal growth by activating CREB phosphorylation through ERK signaling by a mechanism involving Ca(2+) and PKC activation.
Collapse
Affiliation(s)
- S V Gorosito
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
| | | |
Collapse
|
32
|
Abstract
Estradiol is the most potent and ubiquitous member of a class of steroid hormones called estrogens. Fetuses and newborns are exposed to estradiol derived from their mother, their own gonads, and synthesized locally in their brains. Receptors for estradiol are nuclear transcription factors that regulate gene expression but also have actions at the membrane, including activation of signal transduction pathways. The developing brain expresses high levels of receptors for estradiol. The actions of estradiol on developing brain are generally permanent and range from establishment of sex differences to pervasive trophic and neuroprotective effects. Cellular end points mediated by estradiol include the following: 1) apoptosis, with estradiol preventing it in some regions but promoting it in others; 2) synaptogenesis, again estradiol promotes in some regions and inhibits in others; and 3) morphometry of neurons and astrocytes. Estradiol also impacts cellular physiology by modulating calcium handling, immediate-early-gene expression, and kinase activity. The specific mechanisms of estradiol action permanently impacting the brain are regionally specific and often involve neuronal/glial cross-talk. The introduction of endocrine disrupting compounds into the environment that mimic or alter the actions of estradiol has generated considerable concern, and the developing brain is a particularly sensitive target. Prostaglandins, glutamate, GABA, granulin, and focal adhesion kinase are among the signaling molecules co-opted by estradiol to differentiate male from female brains, but much remains to be learned. Only by understanding completely the mechanisms and impact of estradiol action on the developing brain can we also understand when these processes go awry.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Physiology, University of Maryland Baltimore School of Medicine, Baltimore, Maryland 21201, USA.
| |
Collapse
|
33
|
Anger DL, Zhang B, Boutross-Tadross O, Foster WG. Tyrosine receptor kinase B (TrkB) protein expression in the human endometrium. Endocrine 2007; 31:167-73. [PMID: 17873329 DOI: 10.1007/s12020-007-0025-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 05/07/2007] [Accepted: 05/15/2007] [Indexed: 11/27/2022]
Abstract
Tyrosine kinase receptor B (TrkB) gene expression, a neurotrophic factor receptor expressed in the brain and ovary, has recently been identified in deep infiltrating endometriosis by gene array. TrkB is thought to be important in resistance to anchorage independent apoptosis (ANOIKIS) and thus could be important in the pathogenesis of endometriosis. However, TrkB protein expression in the eutopic endometrium of women with and without endometriosis is unknown. Therefore, we examined TrKB protein expression in the endometrium by Western blot (n = 50) and immunohistochemistry (n = 17). Immunoblots of endometrial biopsies were prepared from women with endometriosis (n = 21) vs. healthy controls (n = 29) undergoing benign gynecological surgery at McMaster University Medical Centre. TrkB protein expression was significantly higher in immunoblots from women with endometriosis compared to women without endometriosis. In samples of archived paraffin-embedded endometrial tissue TrkB was localized to the cytoplasm of epithelial cells of the eutopic endometrium from women with endometriosis (n = 7) and without endometriosis (n = 10). We conclude that TrkB protein is expressed in human endometrium. Our results also suggest that TrkB expression may be greater in women with endometriosis compared to women without endometriosis.
Collapse
Affiliation(s)
- Dana L Anger
- Reproductive Biology Division, Department of Obstetrics & Gynecology, HSC-3N52D, McMaster University, 1200 Main Street W, Hamilton, ON, Canada
| | | | | | | |
Collapse
|
34
|
Mendez P, Wandosell F, Garcia-Segura LM. Cross-talk between estrogen receptors and insulin-like growth factor-I receptor in the brain: cellular and molecular mechanisms. Front Neuroendocrinol 2006; 27:391-403. [PMID: 17049974 DOI: 10.1016/j.yfrne.2006.09.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Revised: 08/11/2006] [Accepted: 09/01/2006] [Indexed: 01/02/2023]
Abstract
Accumulating evidence suggests that insulin-like growth factor-I (IGF-I) and estradiol interact to regulate neural function. In this review, we focus on the cellular and molecular mechanisms involved in this interaction. The expression of estrogen receptors (ERs) and IGF-I receptor is cross-regulated in the central nervous system and many neurons and astrocytes coexpress both receptors. Furthermore, estradiol activates IGF-I receptor and its intracellular signaling. This effect may involve classical ERs since recent findings suggest that ERalpha may affect IGF-I actions in the brain by a direct interaction with some of the components of IGF-I signaling. In turn, IGF-I may regulate ER transcriptional activity in neuronal cells. In conclusion, ERs appear to be part of the signaling mechanism of IGF-I, and IGF-I receptor part of the mechanism of estradiol signaling in the nervous system.
Collapse
Affiliation(s)
- Pablo Mendez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), E-28002 Madrid, Spain
| | | | | |
Collapse
|
35
|
Etgen AM, González-Flores O, Todd BJ. The role of insulin-like growth factor-I and growth factor-associated signal transduction pathways in estradiol and progesterone facilitation of female reproductive behaviors. Front Neuroendocrinol 2006; 27:363-75. [PMID: 16904171 DOI: 10.1016/j.yfrne.2006.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Accepted: 06/12/2006] [Indexed: 12/24/2022]
Abstract
We are examining the role of insulin-like growth factor-I (IGF-I) and downstream signal transduction pathways associated with growth factors (e.g., mitogen-activated protein kinase, MAPK) in estradiol and progesterone facilitation of female reproductive behavior in rats. Brain IGF-I receptor activity is required for the long-term, priming actions of estradiol on the female reproductive axis. Infusions of an IGF-I receptor antagonist during estradiol priming blocks induction of hypothalamic alpha(1B)-adrenergic receptors and luteinizing hormone surges, and attenuates lordosis behavior. Infusion of MAPK and phosphatidylinositol-3-kinase inhibitors inhibitors during estradiol priming completely blocks hormone-facilitated lordosis. Because progestin receptors (PRs) can be phosphorylated and activated by MAPKs, growth factor signaling pathways may also participate in progesterone facilitation of reproductive behaviors. Infusion of a MAPK inhibitor in estradiol-primed rats blocks progestin facilitation and sequential inhibition of lordosis and proceptive behaviors. Interference with MAPK signaling also inhibits behavioral responses to cGMP and a delta-opioid agonist, both of which can activate MAPK in some cells. Thus MAPK is involved in the facilitation of lordosis and proceptive behaviors, perhaps by phosphorylation of hypothalamic PRs.
Collapse
Affiliation(s)
- Anne M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | | | |
Collapse
|
36
|
Abstract
Hormonal and locally produced steroids act in the nervous system as neuroendocrine regulators, as trophic factors and as neuromodulators and have a major impact on neural development and function. Glial cells play a prominent role in the local production of steroids and in the mediation of steroid effects on neurons and other glial cells. In this review, we examine the role of glia in the synthesis and metabolism of steroids and the functional implications of glial steroidogenesis. We analyze the mechanisms of steroid signaling on glia, including the role of nuclear receptors and the mechanisms of membrane and cytoplasmic signaling mediated by changes in intracellular calcium levels and activation of signaling kinases. Effects of steroids on functional parameters of glia, such as proliferation, myelin formation, metabolism, cytoskeletal reorganization, and gliosis are also reviewed, as well as the implications of steroid actions on glia for the regulation of synaptic function and connectivity, the regulation of neuroendocrine events, and the response of neural tissue to injury.
Collapse
|
37
|
Chen M, Lucas KG, Akum BF, Balasingam G, Stawicki TM, Provost JM, Riefler GM, Jörnsten RJ, Firestein BL. A novel role for snapin in dendrite patterning: interaction with cypin. Mol Biol Cell 2005; 16:5103-14. [PMID: 16120643 PMCID: PMC1266411 DOI: 10.1091/mbc.e05-02-0165] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Temporal and spatial assembly of signal transduction machinery determines dendrite branch patterning, a process crucial for proper synaptic transmission. Our laboratory previously cloned and characterized cypin, a protein that decreases PSD-95 family member localization and regulates dendrite number. Cypin contains zinc binding, collapsin response mediator protein (CRMP) homology, and PSD-95, Discs large, zona occludens-1 binding domains. Both the zinc binding and CRMP homology domains are needed for dendrite patterning. In addition, cypin binds tubulin via its CRMP homology domain to promote microtubule assembly. Using a yeast two-hybrid screen of a rat brain cDNA library with cypin lacking the carboxyl terminal eight amino acids as bait, we identified snapin as a cypin binding partner. Here, we show by affinity chromatography and coimmunoprecipitation that the carboxyl-terminal coiled-coil domain (H2) of snapin is required for cypin binding. In addition, snapin binds to cypin's CRMP homology domain, which is where tubulin binds. We also show that snapin competes with tubulin for binding to cypin, resulting in decreased microtubule assembly. Subsequently, overexpression of snapin in primary cultures of hippocampal neurons results in decreased primary dendrites present on these neurons and increased probability of branching. Together, our data suggest that snapin regulates dendrite number in developing neurons by modulating cypin-promoted microtubule assembly.
Collapse
Affiliation(s)
- Maxine Chen
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854-8082, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Arabo A, Lefebvre M, Fermanel M, Caston J. Administration of 17alpha-ethinylestradiol during pregnancy elicits modifications of maternal behavior and emotional alteration of the offspring in the rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 156:93-103. [PMID: 15862632 DOI: 10.1016/j.devbrainres.2005.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 02/07/2005] [Accepted: 02/08/2005] [Indexed: 11/15/2022]
Abstract
Intraperitoneal administration of 17alpha-ethinylestradiol (15 microg.kg(-1)) in pregnant rats, every day from day 9 to day 14 of pregnancy, elicited a high percentage of abortions. Quantification of maternal behavior showed that treated dams took better care of their pups than control dams, injected with the vehicle only, did. Postnatal reflexes, which reflect maturational rate, were established more promptly in the offspring of treated dams than in the offspring of control dams. However, when adult, the rats born from treated dams developed anxiety- and depressive-like behaviors. All these results are explained by the effects of the exogenous estrogen on the developing brain of the fetuses.
Collapse
Affiliation(s)
- A Arabo
- Université de Rouen, Faculté des Sciences, UPRES EA 1780 76821 Mont-Saint-Aignan Cedex, France
| | | | | | | |
Collapse
|
39
|
Gulinello M, Etgen AM. Sexually dimorphic hormonal regulation of the gap junction protein, CX43, in rats and altered female reproductive function in CX43+/- mice. Brain Res 2005; 1045:107-15. [PMID: 15910768 PMCID: PMC4169114 DOI: 10.1016/j.brainres.2005.03.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Revised: 03/10/2005] [Accepted: 03/15/2005] [Indexed: 11/19/2022]
Abstract
Astrocytic gap junctional communication is important in steroid hormone regulation of reproductive processes at the level of the hypothalamus, including estrous cyclicity and sexual behavior. We examined the effects of estradiol and progesterone on the abundance of the gap junctional protein, connexin 43 (CX43), which is highly expressed in astrocytes. Gonadectomized rats received hormone treatments that induce maximal sexual behavior and gonadotropin surges in females (estrogen for 48 h followed by progesterone, estrogen alone or progesterone alone). Control animals received vehicle (oil) injections. In the female rat preoptic area (POA), containing the gonadotropin-releasing hormone (GnRH) cell bodies, treatment with estrogen, progesterone or estrogen + progesterone significantly increased CX43 protein levels in immunoblots. In contrast, estrogen + progesterone significantly decreased CX43 levels in the male rat POA. This sexually dimorphic hormonal regulation of CX43 was not evident in the hypothalamus, which contains primarily GnRH nerve terminals. Treatment with estrogen + progesterone significantly decreased CX43 levels in both the male and female hypothalamus. To examine the role of CX43 in female reproductive function, we studied heterozygous female CX43 (CX43+/-) mice. Most mutant mice did not show normal estrous cycles. In addition, when compared to wild type females, CX43+/- mice had reduced lordosis behavior. These data suggest that hypothalamic CX43 expression is regulated by steroid hormones in a brain-region-specific and sexually dimorphic manner. Therefore, gap junctional communication in the POA and hypothalamus may be a factor regulating the estrous cycle and sexual behavior in female rodents.
Collapse
Affiliation(s)
- Maria Gulinello
- Albert Einstein College of Medicine, Department of Neuroscience, 1300 Morris Park Avenue F113, Bronx, NY 10461, USA.
| | | |
Collapse
|
40
|
Topalli I, Etgen AM. Insulin-like growth factor-I receptor and estrogen receptor crosstalk mediates hormone-induced neurite outgrowth in PC12 cells. Brain Res 2005; 1030:116-24. [PMID: 15567343 DOI: 10.1016/j.brainres.2004.09.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2004] [Indexed: 01/08/2023]
Abstract
Estradiol (E(2)) and insulin-like growth factor-I (IGF-I) can act independently or in concert to promote neurite outgrowth in vivo and in cultured neurons. This study examined the role of crosstalk between estrogen receptor (ER)alpha and the IGF-I receptor as a critical mediator of hormone- and growth factor-dependent neurite outgrowth in a homogenous cell system. We used control PC12 cells and PC12 cells stably transfected with ER alpha, both of which express IGF-I receptor. Cells were treated for 1 week with vehicle, 1 nM E(2) or 100 ng/ml IGF-I alone or with E(2) or IGF-I in the presence of either the IGF-I receptor antagonist JB1 or the ER antagonist ICI 182,780. IGF-I significantly increased neurite outgrowth, as measured by the percentage of process-bearing cells, and absolute neurite length per cell in both control and ER alpha-transfected PC12 cells. In contrast, E(2) increased process formation and extension only in PC12 cells that were stably transfected with ER alpha. ICI 182,780 and JB1 blocked the IGF-I-induced increases in neurite length in both cell types. The efficacy of ICI 182,780 in control PC12 cells may have been due to the upregulation of ER alpha in these cells by the 7-day treatment with IGF-I. The ER and IGF-I receptor antagonists similarly blocked the E(2)-induced increase in neurite lengths in ER alpha-transfected cells. Immunofluorescent analysis of the cellular distribution of an axonal marker, phospho-neurofilament, verified that the processes extended by PC12 cells were neurites. These data suggest that receptor crosstalk between IGF-I receptors and ER alpha has an important role in neurite formation and extension even in a single-cell system.
Collapse
Affiliation(s)
- Ilir Topalli
- Department of Neuroscience, Forchheimer 113, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
41
|
Abstract
Previous work from our laboratory has shown that in cultures of hypothalamic neurons obtained from male fetuses at embryonic day 16 the axogenic response to estradiol (E2) is contingent upon culture with medium conditioned by astroglia from a target region for hypothalamic axons. E2 also induced increased levels of TrkB that were necessary for the axonal growth to occur. This convergence between estrogenic and neurotrophic signals prompted investigation of the mitogen activated protein kinase (MAPK) cascade. Analysis of the temporal course of MAPK activation showed increased levels of phosphorylated ERK up to 60 min after E2 exposure, with a maximal response at 5-15 min. UO126 (specific inhibitor of MEK 1/2) blocked E2 induced axonal elongation and ERK phosphorylation, confirming the involvement of ERK in the neuritogenic effect of E2. The membrane impermeable construct E2-BSA proved as effective as free E2 to induce axon elongation, suggesting that E2 exerted its effect through a membrane-associated receptor. This possibility received additional support from experiments showing that E2-BSA also increased ERK phosphorylation with the same time course than E2. These results indicate that ERK signaling is necessary for E2 to induce axon growth and this activation is mediated by a membrane bound estrogen receptor.
Collapse
Affiliation(s)
- H F Carrer
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Casilla de Correo 389, 5000 Córdoba, Argentina.
| | | | | |
Collapse
|
42
|
Zhang L, Sukhareva M, Barker JL, Maric D, Hao Y, Chang YH, Ma W, O'Shaughnessy T, Rubinow DR. Direct binding of estradiol enhances Slack (sequence like a calcium-activated potassium channel) channels’ activity. Neuroscience 2005; 131:275-82. [PMID: 15708472 DOI: 10.1016/j.neuroscience.2004.10.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2004] [Indexed: 10/25/2022]
Abstract
17Beta-estradiol (E2) is a major neuroregulator, exerting both genomic and non-genomic actions. E2 regulation of Slack (sequence like a calcium-activated potassium channel) potassium channels has not been identified in the CNS. We demonstrate E2-induced activation of Slack channels, which display a unitary conductance of about 60 pS, are inhibited by intracellular calcium, and are abundantly expressed in the nervous system. In lipid bilayers derived from rat cortical neuronal membranes, E2 increases Slack open probability and appears to decrease channel inactivation. Additionally, E2 binds to the Slack channel and activates outward currents in human embryonic kidney-293 cells that express Slack channels but not classical estrogen receptors (i.e. ERalpha or ERbeta). Neither E2-induced activation nor the binding intensity of E2 to the Slack channel is blocked by tamoxifen, an ER antagonist/agonist. Thus, E2 activates a potassium channel, Slack, through a non-traditional membrane binding site, adding to known non-genomic mechanisms by which E2 exerts pharmacological and toxicological effects in the CNS.
Collapse
Affiliation(s)
- L Zhang
- Behavioral Endocrinology Branch, NIMH/NIH, Building 10, Room 65340, MSC 1276, Bethesda, MD 20892-1276, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Brito VI, Carrer HF, Cambiasso MJ. Inhibition of tyrosine kinase receptor type B synthesis blocks axogenic effect of estradiol on rat hypothalamic neurones in vitro. Eur J Neurosci 2004; 20:331-7. [PMID: 15233742 DOI: 10.1111/j.1460-9568.2004.03485.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
17-beta-estradiol (E2) increases axonal growth and tyrosine kinase receptor (Trk)B levels of male-derived hypothalamic neurones in vitro. To investigate whether the axogenic response depends on the upregulation of TrkB, we analysed neuritic growth and neuronal polarization in cultures treated with an antisense oligonucleotide against TrkB mRNA. In cultures without E2, treatment with 7.5 or 10 micro m antisense reduced TrkB levels and the percentage of neurones showing an identifiable axon; the number and length of minor processes were increased. In cultures treated with 5 micro m antisense, morphometric parameters were normal although total TrkB levels were reduced. The same dose prevented the E2-dependent increase of TrkB levels and suppressed the axogenic effect of E2. These results indicate that TrkB is necessary for normal neuronal growth and maturation and further suggest that an increase in TrkB is necessary for E2 to exert its axogenic effect in male-derived neurones.
Collapse
Affiliation(s)
- V I Brito
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Casilla de Correo 389, 5000 Córdoba, Argentina
| | | | | |
Collapse
|
44
|
Carrer HF, Cambiasso MJ, Brito V, Gorosito S. Neurotrophic Factors and Estradiol Interact To Control Axogenic Growth in Hypothalamic Neurons. Ann N Y Acad Sci 2003; 1007:306-16. [PMID: 14993063 DOI: 10.1196/annals.1286.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Previous work from our laboratory has shown that in cultures of hypothalamic neurons obtained from male fetuses at embryonic day 16, the axogenic response to estrogen (E2) is contingent on coculture with target glia or target glia-conditioned media (CM). Neither the estrogen receptor blockers tamoxifen nor ICI 182,780 prevented the axogenic effects of the hormone. Estradiol made membrane-impermeable by conjugation to a protein of high molecular weight (E2-BSA) preserved its axogenic capacity, suggesting the possibility of a membrane effect responsible for the action of E2. Western blot analysis of extracts from homogenates of cultured neurons grown with E2 and CM from target glia had more TrkB than cultures with CM alone or E2 alone. To further investigate the interaction between E2 and the neurotrophin receptors, we used a specific antisense oligonucleotide (AS) to prevent the estradiol-induced increase of TrkB. The effect of E2 was suppressed in cultures in which TrkB was down-regulated by the AS, showing decreased axonal elongation when compared with neurons treated with E2 without AS or with sense TrkB. In cultures grown with AS, the axonal length of E2-treated cultures was not different from cultures without E2. Evidence suggesting cross-talk between E2 and neurotrophic factor(s) prompted investigation of signaling along the MAPK cascade. Immuno blotting of E2-treated cultures showed increased levels of phosphorylated ERK1 and ERK2. UO126 but not LY294002 blocked E2-induced axonal elongation, suggesting that the MAPKs are involved in this response.
Collapse
Affiliation(s)
- H F Carrer
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Casilla de Correo 389, 5000 Cordóba, Argentina.
| | | | | | | |
Collapse
|
45
|
Etgen AM. Ovarian Steroid and Growth Factor Regulation of Female Reproductive Function Involves Modification of Hypothalamic α1-Adrenoceptor Signaling. Ann N Y Acad Sci 2003; 1007:153-61. [PMID: 14993049 DOI: 10.1196/annals.1286.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The ovarian steroids estradiol (E(2)) and progesterone (P) act on target neurons in the hypothalamus and preoptic area to coordinate the expression of female reproductive behaviors with the timing of the preovulatory luteinizing hormone (LH) surge. This chapter will summarize evidence that E(2) and P facilitation of the receptive component of female reproductive behavior, lordosis, involves changes in both the expression of and intracellular signal transduction pathways engaged by alpha(1)-adrenergic receptors in the hypothalamus and preoptic area. The alpha(1)-adrenoceptors are thought to mediate the facilitatory effects of the catecholamine neurotransmitter norepinephrine on both lordosis behavior and LH release. E(2) first induces the expression of the alpha(1B)-adrenergic receptor subtype in the hypothalamus and preoptic area. P then acts in an E(2)-dependent manner to promote linkage of hypothalamic alpha(1)-adrenoceptors to an intracellular signaling pathway involving nitric oxide and cyclic GMP. This chapter will also describe recent findings that implicate brain insulin-like growth factor-I (IGF-I) receptors as obligatory co-mediators of hormonal regulation of hypothalamic alpha(1)-adrenoceptors and female neuroendocrine function. Additional studies suggest that E(2) and IGF-I facilitate lordosis behavior by activating kinases traditionally associated with growth factor signal transduction (mitogen-activated protein kinases and phosphatidlyinositol-3-kinases). These molecular events are proposed to help coordinate the timing of ovulation with the expression of sexual receptivity, thereby maximizing reproductive success.
Collapse
Affiliation(s)
- Anne M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| |
Collapse
|
46
|
Non-neuronal cells in the nervous system: sources and targets of neuroactive steroids. ADVANCES IN MOLECULAR AND CELL BIOLOGY 2003. [DOI: 10.1016/s1569-2558(03)31024-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
47
|
Carrer HF, Cambiasso MJ. Sexual differentiation of the brain: genes, estrogen, and neurotrophic factors. Cell Mol Neurobiol 2002; 22:479-500. [PMID: 12585676 PMCID: PMC11533755 DOI: 10.1023/a:1021825317546] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Based on evidence obtained during the past 50 years, the current hypothesis to explain the sexual dimorphism of structure and function in the brain of vertebrates maintains that these differences are produced by the epigenetic action of gonadal hormones. However, evidence has progressively accumulated suggesting that genetic mechanisms controlling sexual-specific neuronal characteristics precede, or occur in parallel with, hormonal effects. 1. In cultures of hypothalamic neurons taken from gestation day 16 (GD16) embryos, treatment of sexually segregated cultures with estradiol (E2) induces axon growth in neurons from male neurons, but not from female neurons. In these cultures treatment with E2 increased the levels of tyrosine kinase type B (TrkB) and insulin-like growth factor I (IGF-I) receptors in male but not in female neurons. This and other sex differences cannot be explained by differences in hormonal environment, because the donor embryos were obtained when gonadal secretion of steroids is just beginning, before the perinatal surge of testosterone that determines development of the male brain beginning at GD17/18. 2. The response to estrogen is contingent upon coculture with heterotopic glia (mostly astrocytes) from a target region (amygdala) harvested from same-sex fetuses at GD16, whereas in the presence of homotopic glia or in cultures without glia, E2 had no effect. It was concluded that the axogenic effect of E2 depends on interaction between neurons and glia from a target region and that neurons from fetal male donors appear to mature earlier than neurons from females, a differentiated response that takes place prior to divergent exposure to gonadal secretions. 3. The effects of target and nontarget glia-conditioned media (CM) on the E2-induced growth of neuronal processes of hypothalamic neurons obtained from sexually segregated fetal donors were also studied. Estrogen added to media conditioned by target glia modified the number of primary neurites and the growth of axons of hypothalamic neurons of males but not of females. 4. Neither the Type III steroidal receptor blocker tamoxifen nor Type I antiestrogen ICI 182,780 prevented the axogenic effects of the hormone. Estradiol made membrane-impermeable by conjugation to a protein of high molecular weight (E2-BSA) preserved its axogenic capacity, suggesting the possibility of a membrane effect responsible for the action of E2. 5. Western blot analysis of the tyrosine kinase type A (TrkA), type B (TrkB), type C (TrkC), and insulin-like growth factor (IGF-I R) receptors in extracts from homogenates of cultured hypothalamic neurons showed that in cultures of male-derived neurons grown with E2 and CM from target glia, the amounts of TrkB and IGF-I R increased notably. Densitometric quantification showed that these cultures had more TrkB than cultures with CM alone or E2 alone. On the contrary, in cultures of female-derived neurons, the presence of CM alone induced maximal levels of TrkB, which were not further increased by E2; female-derived neurons in all conditions did not contain IGF-I R. Levels of TrkC were not modified by any experimental condition in male- or female-derived cultures and Trk A was not found in the homogenates. These results are compared with similar data from other laboratories and integrated in a model for the confluent interaction of estrogen and neurotrophic factors released by glia that may contribute to the sexual differentiation of the brain.
Collapse
Affiliation(s)
- Hugo F Carrer
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Casilla de Correo 389, Córdoba 5000, Argentina.
| | | |
Collapse
|
48
|
Cardona-Gomez GP, Mendez P, Garcia-Segura LM. Synergistic interaction of estradiol and insulin-like growth factor-I in the activation of PI3K/Akt signaling in the adult rat hypothalamus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 107:80-8. [PMID: 12414126 DOI: 10.1016/s0169-328x(02)00449-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Estradiol and insulin-like growth factor-I (IGF-I) interact in the hypothalamus to regulate neuronal function, synaptic plasticity and neuroendocrine events. However, the molecular mechanisms involved in these interactions are still unknown. In the present study, the effect of estradiol on the signaling pathways of IGF-I receptor has been assessed in the hypothalamus of young adult ovariectomized rats, using specific antibodies for the phosphorylated forms of extracellular-signal regulated kinase (ERK) 1 and ERK2 and Akt/protein kinase B (Akt/PKB). Estradiol treatment resulted, between 6 and 24 h after systemic administration, in dose-dependent effects on the phosphorylation of ERK and Akt/PKB. Estradiol did not modify the level of ERK phosphorylation induced by intracerebroventricular administration of IGF-I. However, both hormones had a synergistic effect on the phosphorylation of Akt/PKB. These findings suggest that estrogen effects in the hypothalamus may be mediated in part by the activation of the signaling pathways of the IGF-I receptor.
Collapse
|
49
|
Cambiasso MJ, Carrer HF. Nongenomic mechanism mediates estradiol stimulation of axon growth in male rat hypothalamic neurons in vitro. J Neurosci Res 2001; 66:475-81. [PMID: 11746365 DOI: 10.1002/jnr.1238] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The purpose of the present work was to investigate the participation of estradiol receptors (ER) in estrogen-induced axon growth in vitro. Hypothalamic neurons from 16 day (E16) male rat fetuses were cultured with or without 17-beta-estradiol at 1 x 10(-7) M in basal medium or medium conditioned by astroglia derived from ventral mesencephalon (CM). After 48 hr in vitro, neurite outgrowth was quantified by morphometric analysis. An axogenic effect could be demonstrated for estradiol added to CM. With RT-PCR, the mRNA transcript for ERalpha was found in the donor tissues as well as in the neuron cultures. In this model two specific nuclear ER blockers (tamoxifen and ICI 182,780) were ineffective in blocking the neuritogenic effect, and a membrane-impermeable estrogen-albumin construct (E2-BSA) was as effective as estradiol. These results indicate that the axogenic effect of estradiol at E16 is not exerted through the classical intracellular receptor signal transduction system and suggest the possibility of a membrane-mediated mechanism. The data are discussed in light of our previous findings pointing to the interdependent activation of the estrogenic and the trophic factor signaling pathways that mediate stimulated axon growth.
Collapse
Affiliation(s)
- M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Córdoba, Argentina
| | | |
Collapse
|
50
|
Cardona-Gómez GP, Mendez P, DonCarlos LL, Azcoitia I, Garcia-Segura LM. Interactions of estrogens and insulin-like growth factor-I in the brain: implications for neuroprotection. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2001; 37:320-34. [PMID: 11744097 DOI: 10.1016/s0165-0173(01)00137-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Data from epidemiological studies suggest that the decline in estrogen following menopause could increase the risk of neurodegenerative diseases. Furthermore, experimental studies on different animal models have shown that estrogen is neuroprotective. The mechanisms involved in the neuroprotective effects of estrogen are still unclear. Anti-oxidant effects, activation of different membrane-associated intracellular signaling pathways, and activation of classical nuclear estrogen receptors (ERs) could contribute to neuroprotection. Interactions with neurotrophins and other growth factors may also be important for the neuroprotective effects of estradiol. In this review we focus on the interaction between insulin-like growth factor-I (IGF-I) and estrogen signaling in the brain and on the implications of this interaction for neuroprotection. During the development of the nervous system, IGF-I promotes the differentiation and survival of specific neuronal populations. In the adult brain, IGF-I is a neuromodulator, regulates synaptic plasticity, is involved in the response of neural tissue to injury and protects neurons against different neurodegenerative stimuli. As an endocrine signal, IGF-I represents a link between the growth and reproductive axes and the interaction between estradiol and IGF-I is of particular physiological relevance for the regulation of growth, sexual maturation and adult neuroendocrine function. There are several potential points of convergence between estradiol and IGF-I receptor (IGF-IR) signaling in the brain. Estrogen activates the mitogen-activated protein kinase (MAPK) pathway and has a synergistic effect with IGF-I on the activation of Akt, a kinase downstream of phosphoinositol-3 kinase. In addition, IGF-IR is necessary for the estradiol induced expression of the anti-apoptotic molecule Bcl-2 in hypothalamic neurons. The interaction of ERs and IGF-IR in the brain may depend on interactions between neural cells expressing ERs with neural cells expressing IGF-IR, or on direct interactions of the signaling pathways of alpha and beta ERs and IGF-IR in the same cell, since most neurons expressing IGF-IR also express at least one of the ER subtypes. In addition, studies on adult ovariectomized rats given intracerebroventricular (i.c.v.) infusions with antagonists for ERs or IGF-IR or with IGF-I have shown that there is a cross-regulation of the expression of ERs and IGF-IR in the brain. The interaction of estradiol and IGF-I and their receptors may be involved in different neural events. In the developing brain, ERs and IGF-IR are interdependent in the promotion of neuronal differentiation. In the adult, ERs and IGF-IR interact in the induction of synaptic plasticity. Furthermore, both in vitro and in vivo studies have shown that there is an interaction between ERs and IGF-IR in the promotion of neuronal survival and in the response of neural tissue to injury, suggesting that a parallel activation or co-activation of ERs and IGF-IR mediates neuroprotection.
Collapse
Affiliation(s)
- G P Cardona-Gómez
- Instituto Cajal, C.S.I.C., Avenida Doctor Arce 37, E-28002, Madrid, Spain
| | | | | | | | | |
Collapse
|