1
|
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]
|
2
|
Jurkowski MP, Bettio L, K. Woo E, Patten A, Yau SY, Gil-Mohapel J. Beyond the Hippocampus and the SVZ: Adult Neurogenesis Throughout the Brain. Front Cell Neurosci 2020; 14:576444. [PMID: 33132848 PMCID: PMC7550688 DOI: 10.3389/fncel.2020.576444] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Convincing evidence has repeatedly shown that new neurons are produced in the mammalian brain into adulthood. Adult neurogenesis has been best described in the hippocampus and the subventricular zone (SVZ), in which a series of distinct stages of neuronal development has been well characterized. However, more recently, new neurons have also been found in other brain regions of the adult mammalian brain, including the hypothalamus, striatum, substantia nigra, cortex, and amygdala. While some studies have suggested that these new neurons originate from endogenous stem cell pools located within these brain regions, others have shown the migration of neurons from the SVZ to these regions. Notably, it has been shown that the generation of new neurons in these brain regions is impacted by neurologic processes such as stroke/ischemia and neurodegenerative disorders. Furthermore, numerous factors such as neurotrophic support, pharmacologic interventions, environmental exposures, and stem cell therapy can modulate this endogenous process. While the presence and significance of adult neurogenesis in the human brain (and particularly outside of the classical neurogenic regions) is still an area of debate, this intrinsic neurogenic potential and its possible regulation through therapeutic measures present an exciting alternative for the treatment of several neurologic conditions. This review summarizes evidence in support of the classic and novel neurogenic zones present within the mammalian brain and discusses the functional significance of these new neurons as well as the factors that regulate their production. Finally, it also discusses the potential clinical applications of promoting neurogenesis outside of the classical neurogenic niches, particularly in the hypothalamus, cortex, striatum, substantia nigra, and amygdala.
Collapse
Affiliation(s)
- Michal P. Jurkowski
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Luis Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Emma K. Woo
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Anna Patten
- Centre for Interprofessional Clinical Simulation Learning (CICSL), Royal Jubilee Hospital, Victoria, BC, Canada
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Joana Gil-Mohapel
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
3
|
Abstract
Insulin-like growth factors (IGFs) bind specifically to the IGF1 receptor on the cell surface of targeted tissues. Ligand binding to the α subunit of the receptor leads to a conformational change in the β subunit, resulting in the activation of receptor tyrosine kinase activity. Activated receptor phosphorylates several substrates, including insulin receptor substrates (IRSs) and Src homology collagen (SHC). Phosphotyrosine residues in these substrates are recognized by certain Src homology 2 (SH2) domain-containing signaling molecules. These include, for example, an 85 kDa regulatory subunit (p85) of phosphatidylinositol 3-kinase (PI 3-kinase), growth factor receptor-bound 2 (GRB2) and SH2-containing protein tyrosine phosphatase 2 (SHP2/Syp). These bindings lead to the activation of downstream signaling pathways, PI 3-kinase pathway and Ras-mitogen-activated protein kinase (MAP kinase) pathway. Activation of these signaling pathways is known to be required for the induction of various bioactivities of IGFs, including cell proliferation, cell differentiation and cell survival. In this review, the well-established IGF1 receptor signaling pathways required for the induction of various bioactivities of IGFs are introduced. In addition, we will discuss how IGF signals are modulated by the other extracellular stimuli or by themselves based on our studies.
Collapse
Affiliation(s)
- Fumihiko Hakuno
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Takahashi
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Walser M, Schiöler L, Oscarsson J, Åberg MAI, Wickelgren R, Svensson J, Isgaard J, Åberg ND. Mode of GH administration and gene expression in the female rat brain. J Endocrinol 2017; 233:187-196. [PMID: 28275169 DOI: 10.1530/joe-16-0656] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/08/2017] [Indexed: 11/08/2022]
Abstract
The endogenous secretion of growth hormone (GH) is sexually dimorphic in rats with females having a more even and males a more pulsatile secretion and low trough levels. The mode of GH administration, mimicking the sexually dimorphic secretion, has different systemic effects. In the brains of male rats, we have previously found that the mode of GH administration differently affects neuron-haemoglobin beta (Hbb) expression whereas effects on other transcripts were moderate. The different modes of GH administration could have different effects on brain transcripts in female rats. Hypophysectomised female rats were given GH either as injections twice daily or as continuous infusion and GH-responsive transcripts were assessed by quantitative reverse transcription polymerase chain reaction in the hippocampus and parietal cortex (cortex). The different modes of GH-administration markedly increased Hbb and 5'-aminolevulinate synthase 2 (Alas2) in both brain regions. As other effects were relatively moderate, a mixed model analysis (MMA) was used to investigate general effects of the treatments. In the hippocampus, MMA showed that GH-infusion suppressed glia- and neuron-related transcript expression levels, whereas GH-injections increased expression levels. In the cortex, GH-infusion instead increased neuron-related transcripts, whereas GH-injections had no significant effect. Interestingly, this contrasts to previous results obtained from male rat cortex where GH-infusion generally decreased expression levels. In conclusion, the results indicate that there is a small but significant difference in response to mode of GH administration in the hippocampus as compared to the cortex. For both modes of GH administration, there was a robust effect on Hbb and Alas2.
Collapse
Affiliation(s)
- Marion Walser
- Department of Internal MedicineInstitute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Linus Schiöler
- Department for Public Health and Community MedicineThe Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | - Maria A I Åberg
- Department of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ruth Wickelgren
- Department of Clinical Chemistry and Transfusion MedicineThe Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal MedicineInstitute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörgen Isgaard
- Department of Internal MedicineInstitute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - N David Åberg
- Department of Internal MedicineInstitute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Daftary SS, Gore AC. IGF-1 in the Brain as a Regulator of Reproductive Neuroendocrine Function. Exp Biol Med (Maywood) 2016; 230:292-306. [PMID: 15855296 DOI: 10.1177/153537020523000503] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Given the close relationship among neuroendocrine systems, it Is likely that there may be common signals that coordinate the acquisition of adult reproductive function with other homeo-static processes. In this review, we focus on central nervous system insulin-like growth factor-1 (IGF-1) as a signal controlling reproductive function, with possible links to somatic growth, particularly during puberty. In vertebrates, the appropriate neurosecretion of the decapeptide gonadotropin-releas-ing hormone (GnRH) plays a critical role in the progression of puberty. Gonadotropin-releasing hormone is released in pulses from neuroterminals in the median eminence (ME), and each GnRH pulse triggers the production of the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). These pituitary hormones in turn stimulate the synthesis and release of sex steroids by the gonads. Any factor that affects GnRH or gonadotropin pulsatility is important for puberty and reproductive function and, among these factors, the neurotrophic factor IGF-1 is a strong candidate. Although IGF-1 is most commonly studied as the tertiary peripheral hormone in the somatotropic axis via its synthesis in the liver, IGF-1 Is also synthesIzed in the brain, within neurons and glia. In neuroendocrine brain regions, central IGF-1 plays roles in the regulation of neuroendocrine functions, including direct actions on GnRH neurons. Moreover, GnRH neurons themselves co-express IGF-1 and the IGF-1 receptor, and this expression is developmentally regulated. Here, we examine the role of IGF-1 acting in the hypothalamus as a critical link between reproductive and other neuroendocrine functions.
Collapse
Affiliation(s)
- Shabrine S Daftary
- Department of Pharmacology and Biological Chemistry, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
6
|
IGFBP-2: The dark horse in metabolism and cancer. Cytokine Growth Factor Rev 2015; 26:329-46. [DOI: 10.1016/j.cytogfr.2014.12.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/09/2014] [Indexed: 12/29/2022]
|
7
|
Walser M, Schiöler L, Oscarsson J, Aberg MAI, Svensson J, Aberg ND, Isgaard J. Different modes of GH administration influence gene expression in the male rat brain. J Endocrinol 2014; 222:181-90. [PMID: 24872576 DOI: 10.1530/joe-14-0223] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The endogenous secretion pattern in males of GH is episodic in rats and in humans, whereas GH administration is usually even. Different types of GH administration have different effects on body mass, longitudinal bone growth, and liver metabolism in rodents, whereas possible effects on brain plasticity have not been investigated. In this study, GH was administered as a continuous infusion or as two daily injections in hypophysectomized male rats. Thirteen transcripts previously known to respond to GH in the hippocampus and parietal cortex (cortex) were assessed by RT-PCR. To investigate the effects of type of GH administration on several transcripts with different variations, and categories of transcripts (neuron-, glia-, and GH-related), a mixed model analysis was applied. Accordingly, GH injections increased overall transcript abundance more than GH infusions (21% in the hippocampus, P<0.001 and 10% in the cortex, P=0.09). Specifically, GH infusions and injections robustly increased neuronal hemoglobin beta (Hbb) expression significantly (1.8- to 3.6-fold), and GH injections were more effective than GH infusions in increasing Hbb in the cortex (41%, P=0.02), whereas a 23% difference in the hippocampus was not significant. Also cortical connexin 43 was higher in the group with GH injections than in those with GH infusions (26%, P<0.007). Also, there were differences between GH injections and infusions in GH-related transcripts of the cortex (23%, P=0.04) and glia-related transcripts of the hippocampus (15%, P=0.02). Thus, with the exception of Hbb there is a moderate difference in responsiveness to different modes of GH administration.
Collapse
Affiliation(s)
- Marion Walser
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linus Schiöler
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Oscarsson
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria A I Aberg
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenLaboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - N David Aberg
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenLaboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jörgen Isgaard
- Laboratory of Experimental EndocrinologyDepartment of Internal Medicine, The Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Blå Stråket 5, SE-413 45 Gothenburg, SwedenDepartment for Public Health and Community MedicineThe Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenAstraZeneca R&DSE-431 83 Mölndal, Gothenburg, SwedenInstitute for Neuroscience and PhysiologyThe Sahlgrenska Academy, Center for Brain Repair and RehabilitationDepartment of Primary Health CareInstitute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Hoeflich A, Wirthgen E, David R, Classen CF, Spitschak M, Brenmoehl J. Control of IGFBP-2 Expression by Steroids and Peptide Hormones in Vertebrates. Front Endocrinol (Lausanne) 2014; 5:43. [PMID: 24778626 PMCID: PMC3985015 DOI: 10.3389/fendo.2014.00043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/20/2014] [Indexed: 12/03/2022] Open
Abstract
IGFBP-2 (1) has been described as a brain tumor oncogene (2) and is widely expressed in cancers from different origins (3-8). IGFBP-2 alone cannot cause malignant transformation, yet progression of brain tumors to higher grade (9) and also has been provided as a protective element in earlier stages of multistage colon carcinogenesis (10). Therefore, it is crucial to understand the factors that determine expression patterns of IGFBP-2 under normal and malignant conditions. The present review provides a comprehensive update of known factors that have an impact on expression of IGFBP-2.
Collapse
Affiliation(s)
- Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- *Correspondence: Andreas Hoeflich, Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, Dummerstorf 18196, Germany e-mail:
| | | | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock, Germany
| | | | - Marion Spitschak
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Julia Brenmoehl
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
9
|
Cheng MF. Hypothalamic neurogenesis in the adult brain. Front Neuroendocrinol 2013; 34:167-78. [PMID: 23684668 DOI: 10.1016/j.yfrne.2013.05.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 12/19/2022]
Abstract
Adult-born new neurons are continuously added to the hippocampus and the olfactory bulb to serve aspects of learning and perceptual functions. Recent evidence establishes a third neurogenic niche in the ventral hypothalamic parenchyma surrounding the third ventricle that ensures the plasticity of specific brain circuits to stabilize physiological functions such as the energy-balance regulatory system. Hypothalamic lesion studies have demonstrated that regions associated with reproduction-related functions are also capable of recruiting newborn neurons to restore physiological functions and courtship behavior. Induced by lesion or other stimulation, elevated neurotrophic factors trigger neurogenic cascades that contribute to remodeling of certain neural circuits to meet specific transient functions. This insight raises the possibility that event-specific changes, such as increased GnRH, may be mediated by courtship-sensitive neurotrophic factors. We will discuss the potentially integral and ubiquitous roles of neurogenesis in physiological and biological phenomena, roles that await future experimental exploration.
Collapse
Affiliation(s)
- Mei-Fang Cheng
- Department of Psychology, Rutgers University, 101 Warren Street, Newark, NJ, USA.
| |
Collapse
|
10
|
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
|
11
|
Traub ML, De Butte-Smith M, Zukin RS, Etgen AM. Oestradiol and insulin-like growth factor-1 reduce cell loss after global ischaemia in middle-aged female rats. J Neuroendocrinol 2009; 21:1038-44. [PMID: 19840235 PMCID: PMC2862345 DOI: 10.1111/j.1365-2826.2009.01927.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Whereas the ability of oestradiol and insulin-like growth factor (IGF)-1 to afford neuroprotection against ischaemia-induced neuronal death in young female and male rodents is well established, the impact of IGF-1 in middle-aged animals is largely unknown. The present study assessed the efficacy of oestradiol and IGF-1 with respect to reducing neuronal death after transient global ischaemia in middle-aged female rats after 8 weeks of hormone withdrawal. Rats were ovariohysterectomised and implanted 8 weeks later with an osmotic mini-pump delivering IGF-1 or saline into the lateral ventricle. Some rats also received physiological levels of oestradiol by subcutaneous pellet. Two weeks later, rats were subjected to global ischaemia or sham operation. Surviving hippocampal CA1 neurones were quantified. Ischaemia produced massive CA1 cell death compared to sham-operated animals, which was evident at 14 days. Significantly more neurones survived in animals treated with either oestradiol or IGF-1, but simultaneous treatment produced no additive effect. IGF-1, an endogenous growth factor, may be a clinically useful therapy in preventing human brain injury, with neuroprotective equivalence to oestradiol but without the harmful side-effects.
Collapse
Affiliation(s)
- Michael L. Traub
- Department of Obstetrics and Gynecology & Women’s Health, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| | - Maxine De Butte-Smith
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| | - Anne M. Etgen
- Department of Obstetrics and Gynecology & Women’s Health, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| |
Collapse
|
12
|
Veldhuis JD. Aging and hormones of the hypothalamo-pituitary axis: gonadotropic axis in men and somatotropic axes in men and women. Ageing Res Rev 2008; 7:189-208. [PMID: 18343203 DOI: 10.1016/j.arr.2007.12.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 12/18/2007] [Accepted: 12/19/2007] [Indexed: 10/22/2022]
Abstract
Neuroendocrinology of the aging (male) gonadal and (male and female) somatotropic axes will be reviewed. A companion chapter discusses reproductive hormonal changes in aging women. Both the gonadal and growth-hormone/insulin-like growth factor (GH/IGF-I) axes function as ensembles. The ensembles comprise tripartite interactions among the brain (hypothalamus), anterior pituitary gland (gonadotrope and somatotrope cells) and target organs (testis, liver, muscle, fat and brain). Compelling evidence indicates that combined hypothalamic and gonadal adaptations operate in the reproductive axis of older men, and multiple hypothalamic adaptations prevail in the GH axis of elderly men and women. Evolving investigative methods allow more precise parsing of the particular mechanisms that subserve such age-related changes, and suggest novel interventional strategies to evaluate the physiological impact of the dynamic alterations discerned in aging individuals.
Collapse
|
13
|
Kim SK, Kim JH, Han JH, Yoon YD. Inhibitory effect of tributyltin on expression of steroidogenic enzymes in mouse testis. Int J Toxicol 2008; 27:175-82. [PMID: 18404541 DOI: 10.1080/10915810801977906] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Tributyltin (TBT) is known to disrupt the development of reproductive organs, thereby reducing fertility. The aim of this study was to evaluate the acute toxicity of TBT on the testicular development and steroid hormone production. Immature (3-week-old) male mice were given a single administration of 25, 50, or 100 mg/kg of TBT by oral gavage. Lumen formation in seminiferous tubule was remarkably delayed, and the number of apoptotic germ cells found inside the tubules was increased in the TBT-exposed animals, whereas no apoptotic signal was observed in interstitial Leydig cells. Reduced serum testosterone concentration and down-regulated expressions of the mRNAs for cholesterol side-chain cleavage enzyme (P450scc), 17alpha -hydroxylase/C(17-20) lyase (P450(17alpha)), 3beta -hydroxysteroid-dehydrogenase (3beta -HSD), and 17beta -hydroxysteroid-dehydrogenase (17beta -HSD) were also observed after TBT exposure. Altogether, these findings demonstrate that exposure to TBT is associated with induced apoptosis of testicular germ cells and inhibition of steroidogenesis by reduction in the expression of steroidogenic enzymes in interstitial Leydig cells. These adverse effects of TBT would cause serious defects in testicular development and function.
Collapse
Affiliation(s)
- Suel-Kee Kim
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
14
|
Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, Tujague M, Ström A, Treuter E, Warner M, Gustafsson JA. Estrogen receptors: how do they signal and what are their targets. Physiol Rev 2007; 87:905-31. [PMID: 17615392 DOI: 10.1152/physrev.00026.2006] [Citation(s) in RCA: 1284] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
During the past decade there has been a substantial advance in our understanding of estrogen signaling both from a clinical as well as a preclinical perspective. Estrogen signaling is a balance between two opposing forces in the form of two distinct receptors (ER alpha and ER beta) and their splice variants. The prospect that these two pathways can be selectively stimulated or inhibited with subtype-selective drugs constitutes new and promising therapeutic opportunities in clinical areas as diverse as hormone replacement, autoimmune diseases, prostate and breast cancer, and depression. Molecular biological, biochemical, and structural studies have generated information which is invaluable for the development of more selective and effective ER ligands. We have also become aware that ERs do not function by themselves but require a number of coregulatory proteins whose cell-specific expression explains some of the distinct cellular actions of estrogen. Estrogen is an important morphogen, and many of its proliferative effects on the epithelial compartment of glands are mediated by growth factors secreted from the stromal compartment. Thus understanding the cross-talk between growth factor and estrogen signaling is essential for understanding both normal and malignant growth. In this review we focus on several of the interesting recent discoveries concerning estrogen receptors, on estrogen as a morphogen, and on the molecular mechanisms of anti-estrogen signaling.
Collapse
Affiliation(s)
- Nina Heldring
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
El-Bakri NK, Islam A, Suliman I, Lindgren U, Winblad B, Adem A. Ovariectomy and gonadal hormone treatment: effects on insulin-like growth factor-1 receptors in the rat brain. Growth Horm IGF Res 2004; 14:388-393. [PMID: 15336232 DOI: 10.1016/j.ghir.2004.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Revised: 04/27/2004] [Accepted: 04/28/2004] [Indexed: 02/04/2023]
Abstract
Various studies demonstrate that estradiol regulates structure and function of adult neurons. Long-term effect of estradiol in terms of neuroprotection is less documented compared to short-term one. It is well documented that estradiol interacts with insulin-like growth factor-1 (IGF-I) in the brain. The present study examines the effect of ovariectomy and two doses of ovarian hormone treatment on IGF-I receptor density in the adult rat by receptor autoradiography using (125)I-IGF-I as a ligand. Our result showed that ovariectomy decreased IGF-I receptor density in hippocampus, hypothalamus and parietal cortex compared to that of the sham-operated group. Treatment with low or high dose estrogen restored IGF-I receptor density to the control levels in nearly all areas studied in this investigation. It seems that low dose estrogen has more pronounced effect than the high dose in restoring IGF-I receptor density. On the other hand, progesterone treatment in high but not in low dose restored IGF-I receptor density to that of the control. These results demonstrate that both estrogen and progesterone significantly affects IGF-I receptor density in different areas of the brain. These effects indicate a dose-dependent modulator effect of ovarian hormones on IGF-I activity in the brain.
Collapse
Affiliation(s)
- Nahid K El-Bakri
- Neurotec, Section of Experimental Geriatrics, Karolinska Institute, Huddinge University Hospital, B-84 S-141 86 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
16
|
Hiney JK, Srivastava V, Dearth RK, Dees WL. Influence of estradiol on insulin-like growth factor-1-induced luteinizing hormone secretion. Brain Res 2004; 1013:91-7. [PMID: 15196971 DOI: 10.1016/j.brainres.2004.03.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2004] [Indexed: 12/01/2022]
Abstract
Several studies suggest an interrelationship between estradiol (E2) and insulin-like growth factor-1 (IGF-1) at the hypothalamic level. The present study was designed to discern if the capability of IGF-1 to release LH and influence the timing of female puberty is influenced by E2. Twenty-eight-day-old female rats were ovariectomized (OVEX), then implanted with a third ventricular (3V) cannula. Two weeks later, these animals received subcutaneous (s.c.) injection of oil, or either one or two injections of E2 in the form of estradiol benzoate (1 microg). Forty-eight hours later, four basal blood samples were drawn then the animals received IGF-1 (200 ng) or saline via the 3V and four more blood samples were taken. Results indicated that E2 replacement lowered basal LH levels and IGF-1 induced a significant LH release in only animals that had E2 levels above 20 pg/ml. These levels of E2 were also associated with increases (p<0.05) in the expression of both IGF-1 receptor (IGF-1R) mRNA and protein. In order to further support the hypothesis that the action of IGF-1 at the time of puberty is influenced by E2, 24-day-old intact female rats received s.c. injection of sesame oil or 0.1 microg of E2. The next day, the E2-treated animals also received twice daily s.c. injections of either IGF-1 (500 ng) or saline until vaginal opening (VO) occurred. The animals that received E2 plus IGF-1 showed VO at 31.1 days, which was 2.5 days earlier (p<0.01) than E2-treated animals and 4 days earlier (p<0.001) than IGF-1-treated and saline control animals. Taken together, these results indicate that the hypothalamic action of IGF-1 to stimulate LH release and advance female pubertal development is dependent upon the influence of E2.
Collapse
Affiliation(s)
- Jill K Hiney
- Department of Veterinary Anatomy and Public Health, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843-4458, USA
| | | | | | | |
Collapse
|
17
|
Quesada A, Micevych PE. Estrogen interacts with the IGF-1 system to protect nigrostriatal dopamine and maintain motoric behavior after 6-hydroxdopamine lesions. J Neurosci Res 2004; 75:107-16. [PMID: 14689453 DOI: 10.1002/jnr.10833] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The most prominent neurochemical hallmark of Parkinson's disease (PD) is the loss of nigrostriatal dopamine (DA). Animal models of PD have concentrated on depleting DA and therapies have focused on maintaining or restoring DA. Within this context estrogen protects against 6-hydroxdopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) lesions of the nigrostriatal DA pathway. Present studies tested the hypothesis that neuroprotective estrogen actions involve activation of the insulin-like growth factor-1 (IGF-1) system. Ovariectomized rats were treated with either a single subcutaneous injection of 17beta-estradiol benzoate or centrally or peripherally IGF-1. All rats were infused unilaterally with 6-OHDA into the medial forebrain bundle (MFB) to lesion the nigrostriatal DA pathway. Tyrosine hydroxylase (TH) immunocytochemistry confirmed that rats injected with 6-OHDA had a massive loss of TH immunoreactivity in both the ipsilateral substantia nigra compacta (60% loss) and the striatum (>95% loss) compared to the contralateral side. Loss of TH immunoreactivity was correlated with loss of asymmetric forelimb movements, a behavioral assay for motor deficits. Pretreatment with estrogen or IGF-1 significantly prevented 6-OHDA-induced loss of substantia nigra compacta neurons (20% loss) and TH immunoreactivity in DA fibers in the striatum (<20% loss) and prevented the loss of asymmetric forelimb use. Blockage of IGF-1 receptors by intracerebroventricular JB-1, an IGF-1 receptor antagonist, attenuated both estrogen and IGF-1 neuroprotection of nigrostriatal DA neurons and motor behavior. These findings suggest that IGF-1 and estrogen acting through the IGF-1 system may be critical for neuroprotective effects of estrogen on nigrostriatal DA neurons in this model of PD.
Collapse
Affiliation(s)
- Arnulfo Quesada
- Department of Neurobiology, Laboratory of Neuroendocrinology, Brain Research Institute, David Geffen School of Medicine UCLA, Los Angeles, California 90095-1763, USA.
| | | |
Collapse
|
18
|
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
|
19
|
Kasukawa Y, Stabnov L, Miyakoshi N, Baylink DJ, Mohan S. Insulin-like growth factor I effect on the number of osteoblast progenitors is impaired in ovariectomized mice. J Bone Miner Res 2002; 17:1579-87. [PMID: 12211427 DOI: 10.1359/jbmr.2002.17.9.1579] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Because insulin-like growth factor (IGF) I is an important regulator of bone formation, we proposed the hypothesis that IGF-I could contribute in regulating the number of osteoblast progenitors (colony-forming unit fibroblast with ALP activity [CFU-F/ALP+]). To test ex vivo and in vivo effects of IGF-I on the number of CFU-F/ALP+, bone marrow cells (BMCs) derived from normal mice, growth hormone (GH)-deficient lit/lit mice, or ovariectomized (OVX) mice were cultured and the CFU-F/ALP+ number was counted. Ex vivo treatment of IGF-I increased the CFU-F/ALP+ number in a dose-dependent manner compared with vehicle-treated control cultures. The CFU-F/ALP+ number was decreased by 20% (p < 0.01; n = 7-9) in GH-deficient lit/lit mice compared with age-matched control mice. Four weeks after OVX or sham operation, IGF-I (2 microg/g body wt) or vehicle was administered twice on day 1, and 5 days later, BMCs were removed from the femur and cultured for 10 days (n = 9-10 per group). IGF-I administration increased the CFU-F/ALP+ number by 63% (p < 0.01) and 19% (NS), respectively, in sham-operated (sham) and OVX mice compared with the vehicle-treated control group. The serum IGF-I level was similar in OVX mice compared with sham mice; this finding is different from that found in rats in which OVX increases the serum IGF-I level. This study showed that IGF-I is an important regulator of osteoblast-progenitor number in the BMCs of mice both ex vivo and in vivo and that the IGF-I response to increase the number of osteoblast progenitors was impaired in OVX mice.
Collapse
Affiliation(s)
- Yuji Kasukawa
- Musculoskeletal Disease Center, JL Pettis Veterans Administration Medical Center, Loma Linda, California 92357, USA
| | | | | | | | | |
Collapse
|
20
|
Zulu VC, Nakao T, Sawamukai Y. Insulin-like growth factor-I as a possible hormonal mediator of nutritional regulation of reproduction in cattle. J Vet Med Sci 2002; 64:657-65. [PMID: 12237508 DOI: 10.1292/jvms.64.657] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The current review aims to establish insulin-like growth factor-1 (IGF-I) as the factor that signals nutritional status to the reproductive axis, and show that assessment of IGF-I in blood early postpartum during the negative energy balance (NEB) period could be used to predict both nutritional and reproductive status in dairy cattle. The review also explores the effect of nutritional status on circulating IGF-I concentrations and the endocrine role of IGF-I on the reproductive axis. IGF-I plays an important role in gonadotropin-induced folliculogenesis, ovarian steroidogenesis and corpus luteum (CL) function. It also modulates pituitary and hypothalamus function. IGF-I clearly has an endocrine role on the reproductive axis. Severe under nutrition significantly reduces plasma IGF-I concentrations. During the critical period of NEB in high yielding dairy cattle early postpartum, IGF-I concentrations are low in blood and its levels are positively correlated to energy status and reproductive function during this period. Changes in circulating IGF-I immediately postpartum may help predict both nutritional and reproductive status in dairy cattle. IGF-I is therefore one of the long sought factors that signal nutritional status to the reproductive axis.
Collapse
Affiliation(s)
- Victor Chisha Zulu
- Department of Veterinary Reproduction and Obstetrics, School of Veterinary Medicine, Rakuno Gakuen University, Bunkyodai-Midorimachi, Ebetsu, Japan
| | | | | |
Collapse
|
21
|
Melcangi RC, Martini L, Galbiati M. Growth factors and steroid hormones: a complex interplay in the hypothalamic control of reproductive functions. Prog Neurobiol 2002; 67:421-49. [PMID: 12385863 DOI: 10.1016/s0301-0082(02)00060-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mechanisms through which LHRH-secreting neurons are controlled still represent a crucial and debated field of research in the neuroendocrine control of reproduction. In the present review, we have specifically considered two potential signals reaching these hypothalamic neurons: steroid hormones and growth factors. Examples of the relevant physiological role of the interactions between these two families of biologically acting molecules have been provided. In many cases, these interactions occur at the level of hypothalamic astrocytes, which are presently accepted as functional partners of the LHRH-secreting neurons. On the basis of the observations here summarized, we have formulated the hypothesis that a functional co-operation of steroid hormones and growth factors occurring in the hypothalamic astrocytic compartment represents a key factor in the neuroendocrine control of reproductive functions.
Collapse
Affiliation(s)
- Roberto C Melcangi
- Department of Endocrinology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | | | | |
Collapse
|
22
|
Functional interactions between estrogen and insulin-like growth factor-I in the regulation of alpha 1B-adrenoceptors and female reproductive function. J Neurosci 2002. [PMID: 11896179 DOI: 10.1523/jneurosci.22-06-02401.2002] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The ovarian hormone estradiol (E(2)) and insulin-like growth factor-I (IGF-I) interact in the CNS to regulate neuroendocrine function and synaptic remodeling. Previously, our laboratory showed that 2 d E(2) treatment induces alpha(1B)-adrenoceptor expression and promotes IGF-I enhancement of alpha(1)-adrenoceptor potentiation of cAMP accumulation in the preoptic area (POA) and hypothalamus (HYP). This study examined the hypothesis that E(2)-dependent aspects of female reproductive function, including alpha(1B)-adrenoceptor expression and function in the POA and HYP, are mediated by brain IGF-I receptors (IGF-IRs) in female rats. Ovariohysterectomized rats were implanted with a guide cannula aimed at the third ventricle and treated in vivo with vehicle or E(2) daily for 2 d before experimentation. Intracerebroventricular infusions of JB-1, a selective IGF-IR antagonist, were administered every 12 hr beginning 1 hr before the first E(2) injection. Administration of JB-1 during E(2) priming completely blocks hormone-induced luteinizing hormone release and partially inhibits hormone-dependent reproductive behavior. Reproductive behavior is restored by intracerebroventricular infusion of 8-bromo-cGMP, the second messenger implicated in alpha(1)-adrenergic facilitation of lordosis. In addition, blockade of IGF-IRs during E(2) priming prevents E(2)-induced increases in alpha(1B)-adenoceptor binding density and abolishes acute IGF-I enhancement of NE-stimulated cAMP accumulation in HYP and POA slices. These data document the existence of a novel mechanism by which IGF-I participates in the remodeling of noradrenergic receptor signaling in the HYP and POA after E(2) treatment. These events may help coordinate the timing of ovulation with the expression of sexual receptivity.
Collapse
|
23
|
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
|
24
|
Nagao T, Yoshimura S, Saito Y, Nakagomi M, Usumi K, Ono H. Reproductive effects in male and female rats from neonatal exposure to p-octylphenol. Reprod Toxicol 2001; 15:683-92. [PMID: 11738521 DOI: 10.1016/s0890-6238(01)00173-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A number of alkylphenolic compounds are used in a variety of commercial products and have been shown in in vitro studies to be weakly estrogenic, but in vivo data are not available addressing this issue in mammals. Human exposure to alkylphenols may occur not only from these environmental contaminants but also through contact with manufactured and metabolic breakdown products. In this study, Sprague-Dawley rats were exposed to octylphenol by oral gavage at doses of 0 (vehicle: corn oil), 12.5, 25, 50, or 100 mg/kg once daily on postnatal days 1 through 5 to examine its effects on male and female reproductive function after puberty. In addition, preputial separation and vaginal opening as endpoints of sexual maturation, estrous cycling, sperm count, serum testosterone concentration, and histopathologic changes of the reproductive organs of male and female rats were examined. Male reproductive organs were weighed at necropsy. Body weights of male and female rats exposed to octylphenol at 50 and 100 mg/kg throughout the study after the administration period, those of both sexes at 7 and 9 weeks of age in the 25 mg/kg group, and that of females at 9 weeks of age in the 12.5 mg/kg group were lower than those of controls. Significant delays in acquisition of puberty in males and females exposed to octylphenol at 50 and 100 mg/kg were observed. Estrous cycle, copulation and fertility, sperm count, and serum testosterone concentration were not affected by neonatal exposure to octylphenol. Significant decrease in absolute and relative prostate weight in the 12.5, 25, 50, and 100 mg/kg groups, and absolute epididymal weight in the 100 mg/kg group, increase in relative testes weight in the 100 mg/kg group, and relative seminal vesicle weights in the 50 and 100 mg/kg groups were found. Histopathologic analyses of reproductive organs in male and female rats exposed neonatally to octylphenol revealed no marked alterations. The results of this study indicate that early neonatal exposure to octylphenol by oral gavage did not cause dysfunction of reproductive performance (mating and fertility) in male or female rats, and no disruption of development of the reproductive tract was observed in male or female rats, while significant decreases in body weights in the 25 mg/kg and more groups, delays of sexual maturation in the 50 mg/kg and greater groups, and decrease in ventral prostate weights in all octylphenol-treated groups were found. Therefore, it is concluded that NOAEL (no-observed adverse effect level) for systemic toxicity was < or =12.5 mg/kg/day and that for reproductive toxicity was 100 mg/kg/day under the present experimental condition.
Collapse
Affiliation(s)
- T Nagao
- Department of Reproductive and Developmental Biology, Hatano Research Institute, Food and Drug Safety Center, Hadano, Kanagawa, Japan.
| | | | | | | | | | | |
Collapse
|
25
|
Etgen AM, Ansonoff MA, Quesada A. Mechanisms of ovarian steroid regulation of norepinephrine receptor-mediated signal transduction in the hypothalamus: implications for female reproductive physiology. Horm Behav 2001; 40:169-77. [PMID: 11534978 DOI: 10.1006/hbeh.2001.1676] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In many mammalian species, the ovarian steroid hormones estradiol (E(2)) and progesterone (P) act in the hypothalamus and preoptic area to coordinate the timing of female sexual receptivity with ovulation. We study lordosis behavior, an important component of sexual receptivity in rats, and its regulation by E(2) and P as a model system for understanding how hormonal modulation of synaptic neurotransmission influences reproductive physiology and behavior. Our findings suggest that E(2) and P extensively regulate synaptic communication involving the catecholamine norepinephrine (NE) in the hypothalamus. Estrogen priming shifts the balance of postsynaptic NE receptor signaling in the hypothalamus and preoptic area away from beta-adrenergic activation of cAMP synthesis toward alpha(1)-adrenergic signaling pathways. Attenuation of beta-adrenergic signal transduction is achieved by receptor-G-protein uncoupling, apparently due to stable receptor phosphorylation. E(2) modification of alpha(1)-adrenergic signaling includes both increased expression of the alpha(1B)-adrenoceptor subtype and a dramatic, P-induced reconfiguration of the biochemical responses initiated by agonist activation of alpha(1)-adrenoceptors. Among these is the emergence of alpha(1)-adrenergic receptor coupling to cGMP synthesis. We also present evidence that estrogen promotes novel, functional interactions between insulin-like growth factor-1 (IGF-1) and alpha(1)-adrenergic receptor signaling in the hypothalamus and preoptic area. Thus, estrogen amplification of signaling mediated by alpha(1)-adrenoceptors is multifaceted, involving changes in gene expression (of the alpha(1B)-adrenoceptor), switching of receptor linkage to previously inactive intracellular pathways, and the promotion of cross talk between IGF-1 and NE receptors. We propose that this hormone-dependent remodeling of hypothalamic responses to NE maximizes reproductive success by coordinating the timing of the preovulatory release of gonadotropins with the period of behavioral receptivity in female rodents.
Collapse
Affiliation(s)
- A M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA.
| | | | | |
Collapse
|
26
|
Miller BH, Gore AC. Alterations in hypothalamic insulin-like growth factor-I and its associations with gonadotropin releasing hormone neurones during reproductive development and ageing. J Neuroendocrinol 2001; 13:728-36. [PMID: 11489090 DOI: 10.1046/j.1365-2826.2001.00686.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Insulin-like growth factor-I (IGF-I) is thought to play a role in the onset of reproductive ability at puberty and the control of reproductive function throughout adult life. It is believed that these effects are mediated at least in part by the activation of gonadotropin releasing hormone (GnRH) neurones by IGF-I, but the interactions of IGF-I with GnRH neurones in vivo are largely unknown. We first examined the anatomical relationship between GnRH and IGF-I cells in neuroendocrine regions. Using double-label immunocytochemistry, we observed that in the preoptic area-anterior hypothalamus (POA-AH), the site of GnRH perikarya, the majority (78%) of GnRH cell bodies expressed IGF-I immunoreactivity. IGF-I immunoreactivity was also high in the median eminence, the site of GnRH release, and GnRH neuroterminals were seen to interweave among IGF-I-immunopositive cells. Due to this substantial overlap of GnRH and IGF-I immunoreactive elements, we then tested the hypothesis that changes in IGF-I may regulate the GnRH system. Animals were examined at the two important reproductive life transitions: puberty and reproductive senescence. IGF-I mRNA levels were measured in POA-AH and medial basal hypothalamus-median eminence (MBH-ME) and effects of IGF-I treatment on GnRH mRNA levels were quantified by RNase protection assay. Although IGF-I treatment did not alter GnRH gene expression, there were significant alterations in hypothalamic IGF-I gene expression at both puberty and reproductive senescence. During puberty, IGF-I mRNA levels in the MBH-ME of rats increased from the juvenile stage (P25) to the day of vaginal opening (P35), and from the day of vaginal opening to young adulthood (P45) in the POA-AH. During reproductive ageing, IGF-I mRNA levels were significantly lower in middle-aged than young rats, particularly in the MBH-ME. At all ages, IGF-I expression was greater in the MBH-ME than in the POA-AH. These experiments demonstrate that: (i) the majority of adult GnRH neurones are immunopositive for the IGF-I protein; (ii) hypothalamic IGF-I levels increase at the onset of reproductive function and decrease at reproductive senescence in a regionally specific manner; and (iii) despite the presence of IGF-I in GnRH perikarya, IGF-I does not affect GnRH gene expression, suggesting that IGF-I may act at the level of GnRH release rather than gene expression.
Collapse
Affiliation(s)
- B H Miller
- Fishberg Research Center for Neurobiology, Kastor Neurobiology of Aging Laboratories, and Schwartz Department of Geriatrics and Adult Development, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
27
|
Abstract
Although central and peripheral factors have been implicated in the neuromodulation of GnRH in PCOS, there are no definitive or conclusive data to establish a primary causal role for any one factor. Because increased GnRH pulse frequency is at least a contributor to the secretion of excess LH and insufficient FSH that are the proximate cause of chronic anovulation in PCOS, strategies to slow the GnRH pulse generator are likely to promote ovulation in women with PCOS. Several pharmacologic agents, such as dopamine agonists and antagonists, have been tried, but the lack of consistent effects in women with PCOS limits their clinical utility. Current treatment strategies include the use of the combined oral contraceptive pills, antiandrogens or androgen receptor blockers, and insulin sensitizers. Oral contraceptive preparations are effective in suppressing ovarian hyperandrogenemia, regulating menstrual cycles, and reducing the risk of endometrial hyperplasia. Androgen blockade and antiandrogens provide symptomatic relief from androgen-induced acne and hirsutism and have been reported to restore ovulation in women with PCOS. Whether this effect is mediated peripherally or centrally remains to be clarified. The most recent class of pharmacologic agents to gain popularity are the "insulin modifiers." With increasing evidence that insulin resistance constitutes a key metabolic element, it seems logical that improving insulin sensitivity and glucose disposal might wholly, or partially, reverse certain features of PCOS, including anovulation. To date, insulin modifiers have proved most promising in improving the clinical features and promoting fertility, but whether this effect is centrally mediated is yet to be elucidated.
Collapse
Affiliation(s)
- B N Kalro
- Departments of Obstetrics, Gynecology, and Reproductive Services, Magee-Womens Hospital, University of Pittsburgh School of Medicine, USA
| | | | | |
Collapse
|
28
|
Quesada A, Etgen AM. Insulin-like growth factor-1 regulation of alpha(1)-adrenergic receptor signaling is estradiol dependent in the preoptic area and hypothalamus of female rats. Endocrinology 2001; 142:599-607. [PMID: 11159830 DOI: 10.1210/endo.142.2.7946] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently, we demonstrated that estradiol (E(2)) modulates cross-talk between protein tyrosine kinases and norepinephrine (NE) receptor signaling in the hypothalamus (HYP) and preoptic area (POA), brain areas that govern female reproductive function. We are now investigating the identity of protein tyrosine kinase(s) that modify NE receptor signaling in the HYP and POA. Incubation of POA and HYP slices with insulin-like growth factor I (IGF-I), which signals via a receptor (IGF-IR) with endogenous tyrosine kinase activity, enhances NE-stimulated cAMP accumulation only in tissue derived from ovariectomized, E(2)-primed animals. JB-1, an antagonist for IGF-IR, prevents the IGF-I enhancement of NE-stimulated cAMP accumulation in both POA and HYP slices. IGF-I enhances NE-stimulated cAMP accumulation via modulation of alpha(1)-adrenoceptor potentiation of adenylyl cyclase. Binding studies in membranes demonstrate that ovariectomized, E(2)-primed animals show a significant increase in the density of [(125)I]IGF-I-binding sites in both POA and HYP compared with ovariectomized control animals. Neither the IC(50) for [(125)I]IGF-I displacement by IGF-I nor the levels of IGF-I binding proteins in serum or brain tissue are affected by E(2). RIA results showed that E(2) does not modify serum or brain IGF-I levels. These results indicate that E(2) regulation of NE receptor function in the POA and HYP involves increased expression of IGF-IR, and that after E(2) treatment, IGF-IR activation augments alpha(1)-adrenoceptor signaling.
Collapse
Affiliation(s)
- A Quesada
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | |
Collapse
|
29
|
Abstract
This review highlights recent evidence from clinical and basic science studies supporting a role for estrogen in neuroprotection. Accumulated clinical evidence suggests that estrogen exposure decreases the risk and delays the onset and progression of Alzheimer's disease and schizophrenia, and may also enhance recovery from traumatic neurological injury such as stroke. Recent basic science studies show that not only does exogenous estradiol decrease the response to various forms of insult, but the brain itself upregulates both estrogen synthesis and estrogen receptor expression at sites of injury. Thus, our view of the role of estrogen in neural function must be broadened to include not only its function in neuroendocrine regulation and reproductive behaviors, but also to include a direct protective role in response to degenerative disease or injury. Estrogen may play this protective role through several routes. Key among these are estrogen dependent alterations in cell survival, axonal sprouting, regenerative responses, enhanced synaptic transmission and enhanced neurogenesis. Some of the mechanisms underlying these effects are independent of the classically defined nuclear estrogen receptors and involve unidentified membrane receptors, direct modulation of neurotransmitter receptor function, or the known anti-oxidant activities of estrogen. Other neuroprotective effects of estrogen do depend on the classical nuclear estrogen receptor, through which estrogen alters expression of estrogen responsive genes that play a role in apoptosis, axonal regeneration, or general trophic support. Yet another possibility is that estrogen receptors in the membrane or cytoplasm alter phosphorylation cascades through direct interactions with protein kinases or that estrogen receptor signaling may converge with signaling by other trophic molecules to confer resistance to injury. Although there is clear evidence that estradiol exposure can be deleterious to some neuronal populations, the potential clinical benefits of estrogen treatment for enhancing cognitive function may outweigh the associated central and peripheral risks. Exciting and important avenues for future investigation into the protective effects of estrogen include the optimal ligand and doses that can be used clinically to confer benefit without undue risk, modulation of neurotrophin and neurotrophin receptor expression, interaction of estrogen with regulated cofactors and coactivators that couple estrogen receptors to basal transcriptional machinery, interactions of estrogen with other survival and regeneration promoting factors, potential estrogenic effects on neuronal replenishment, and modulation of phenotypic choices by neural stem cells.
Collapse
|
30
|
Cambiasso MJ, Colombo JA, Carrer HF. Differential effect of oestradiol and astroglia-conditioned media on the growth of hypothalamic neurons from male and female rat brains. Eur J Neurosci 2000; 12:2291-8. [PMID: 10947808 DOI: 10.1046/j.1460-9568.2000.00120.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To determine whether soluble products from different CNS regions differ in their ability to support oestrogen-stimulated neurite growth, hypothalamic neurons from sexually segregated embryos were cultured with astroglia-conditioned medium (CM) derived from cortex, striatum and mesencephalon, with or without 17-beta-oestradiol 100 nM added to the medium. After 48 h in vitro, neurite outgrowth was quantified by morphometric analysis. Astroglia-CM from mesencephalon (a target for the axons of hypothalamic neurons) induced the greatest axogenic response in males and in this case only a neuritogenic effect could be demonstrated for oestradiol. On the other hand, astroglia-CM from regions that do not receive projections from ventromedial hypothalamus inhibited axon growth. A sexual difference in the response of hypothalamic neurons to astroglia-CM and oestradiol was found; growth of neurons from female foetuses was increased by astroglia-CM from mesencephalon, but no neuritogenic effect could be demonstrated for oestradiol in these cultures. Blot immunobinding demonstrated the presence of receptors for neurotrophic factors in cultures of hypothalamic neurons; Western blot analysis of these cultures demonstrated that oestradiol increased the concentration of trkB and IGF-I Rbeta, whereas trkA was not detected and the concentration of trkC was not modified. These results support the hypothesis that target regions produce some factor(s) that stimulate the growth of axons from projecting neurons and further indicate that in the case of males this effect is modulated by oestradiol, perhaps mediated through the upregulation of trkB and IGF-I receptors.
Collapse
Affiliation(s)
- M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Córdoba, Argentina
| | | | | |
Collapse
|
31
|
Cardona-Gómez GP, Chowen JA, Garcia-Segura LM. Estradiol and progesterone regulate the expression of insulin-like growth factor-I receptor and insulin-like growth factor binding protein-2 in the hypothalamus of adult female rats. JOURNAL OF NEUROBIOLOGY 2000; 43:269-81. [PMID: 10842239 DOI: 10.1002/(sici)1097-4695(20000605)43:3<269::aid-neu5>3.0.co;2-d] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Gonadal hormones interact with insulin-like growthfactor-I (IGF-I) to regulate synaptic plasticity during the estrous cycle in the rat mediobasal hypothalamus. It has been proposed that tanycytes, specialized glial cells lining the ventral region of the third ventricle, may regulate the availability of IGF-I to hypothalamic neurons. IGF-I levels in tanycytes fluctuate during the estrous cycle. Furthermore, estrogen administration to ovariectomized rats increases IGF-I levels in tanycytes, while progesterone, injected simultaneously with estrogen, blocks the estrogen-induced increase of IGF-I levels in tanycytes. To test whether hormonal regulation of IGF-I receptor (IGF-IR) and IGF binding protein-2 (IGFBP-2) may be involved in the accumulation of IGF-I in tanycytes, we assessed the effect of ovarian hormones on the levels of these molecules in the mediobasal hypothalamus of adult female rats. Ovariectomized animals were treated with either oil, estrogen, progesterone, or estrogen and progesterone simultaneously and then killed 6 or 24 h later. Some neurons, some astrocytes, and many tanycytes in the mediobasal hypothalamus were found by confocal microscopy to be immunoreactive for IGF-IR. IGFBP-2 immunoreactivity was restricted almost exclusively to tanycytes and ependymal cells and was colocalized with IGF-IR immunoreactivity in tanycytes. By electron microscope immunocytochemistry using colloidal gold labeling, IGF-IR and IGFBP-2 immunoreactivities were observed in the microvilli of tanycytes in the lumen of the third ventricle. IGF-IR and IGFBP-2 immunoreactive levels on the apical surface of tanycytes were significantly decreased by the administration of progesterone, either alone or in the presence of estradiol. IGF-IR levels in the mediobasal hypothalamus, measured by Western blotting, were not significantly affected by the separate administration of estradiol or progesterone to ovariectomized rats. However, the simultaneous administration of both hormones resulted in a marked decrease in IGF-IR protein levels. Estradiol administration to ovariectomized rats increased IGFBP-2 immunoreactive levels in the hypothalamus. While progesterone did not significantly affect IGFBP-2 expression, the simultaneous injection of estradiol and progesterone resulted in a marked decrease in IGFBP-2 protein levels. The effect of estradiol on IGFBP-2 was observed both in protein and mRNA levels, suggesting a transcriptional regulation. However, the simultaneous administration of progesterone and estradiol had different effects on IGF-IR protein and IGF-IR mRNA levels, as well as on IGFBP-2 protein and IGFBP-2 mRNA levels, suggesting a postranscriptional action. These findings indicate that estradiol and progesterone regulate the expression of IGF-IR and IGFBP-2 in the mediobasal hypothalamus of adult female rats. Regulation of the hypothalamic IGF-I system by ovarian hormones may be physiologically relevant for neuroendocrine regulation and for synaptic plasticity during the estrous cycle. These results do not support the hypothesis that estrogen-induced accumulation of IGF-I by tanycytes is mediated by the hormonal regulation of IGF-IR. However, estrogen-induced up-regulation of IGFBP-2 and progesterone-induced down-regulation of IGF-IR and IGFBP-2 levels in the apical plasma membrane of tanycytes may be involved in the fluctuation of IGF-I levels in the mediobasal hypothalamus during the estrous cycle.
Collapse
|
32
|
Cardona-Gómez GP, Trejo JL, Fernandez AM, Garcia-Segura LM. Estrogen receptors and insulin-like growth factor-I receptors mediate estrogen-dependent synaptic plasticity. Neuroreport 2000; 11:1735-8. [PMID: 10852234 DOI: 10.1097/00001756-200006050-00027] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Previous studies have shown that estradiol induces a transient disconnection of axo-somatic inhibitory synapses in the hypothalamic arcuate nucleus of adult ovariectomized rats. The synaptic disconnection is accompanied by an increase in the levels of insulin-like growth factor-I (IGF-I) in the arcuate nucleus, suggesting that IGF-I signaling may be involved in the estrogen-induced synaptic plasticity. The role of estrogen receptors and IGF-I receptors in the synaptic changes has been studied by assessing the number of axo-somatic synapses in ovariectomized rats treated with intracerebroventricular administration of the estrogen receptor antagonist ICI 182,780 and the IGF-I receptor antagonist JBI to ovariectomized rats. Estradiol administration resulted in a significant decrease in the number of axo-somatic synapses on arcuate neurons in control ovariectomized rats. Both the estrogen receptor antagonist and the IGF-I receptor antagonist blocked the estrogen-induced synaptic decrease. This finding suggest that estrogen-induced synaptic plasticity in the arcuate nucleus is dependent on the activation of both estrogen receptors and IGF-I receptors.
Collapse
|
33
|
Quesada A, Etgen AM. Tyrosine kinase effects on adrenoceptor-stimulated cyclic AMP accumulation in preoptic area and hypothalamus of female rats: modulation by estradiol. Brain Res 2000; 861:117-25. [PMID: 10751571 DOI: 10.1016/s0006-8993(00)02052-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
These studies examined the functional interactions between adrenergic G-protein coupled receptors and protein tyrosine kinases in the preoptic area and hypothalamus, brain regions that regulate reproductive function in female rats, and evaluated whether in vivo treatment with estradiol for 2 days modulates the cross-talk between these two signaling pathways. In hypothalamic slices genistein, a general tyrosine kinase inhibitor, enhances norepinephrine-stimulated cAMP synthesis independent of estradiol treatment. Genistein appears to act by increasing beta-adrenoceptor signaling. At high norepinephrine concentrations, estradiol potentiates genistein enhancement of the cAMP response in hypothalamic slices. This interaction between estradiol and genistein appears to involve modification of alpha(2)-adrenoceptor signaling mechanisms. In preoptic area slices, genistein enhancement of norepinephrine-stimulated cAMP synthesis is only observed in estradiol-treated rats. In this brain region, genistein enhances cAMP accumulation by modifying alpha(1)- and/or alpha(2)-adrenoceptor rather than beta-adrenoceptor signaling. Genistein amplification of norepinephrine-stimulated cAMP synthesis is not mediated by interactions with estrogen receptors, or by regulation of adenylyl cyclase or phosphodiesterase activities. At the concentration used, genistein inhibits tyrosine phosphorylation in slices from both brain regions. Daidzein, an inactive analogue of genistein, fails to enhance the norepinephrine-stimulated cAMP response in either brain region independent of hormone treatment. These results suggest that protein tyrosine kinases regulate adrenergic responses in the hypothalamus and preoptic area. Moreover, the functional interaction between adrenergic G-protein coupled receptor signaling and protein tyrosine kinases is modified in a brain region and receptor subtype specific manner by estradiol.
Collapse
Affiliation(s)
- A Quesada
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, F113, Bronx, NY 10461, USA.
| | | |
Collapse
|
34
|
Lackey BR, Gray SL, Henricks DM. Physiological basis for use of insulin-like growth factors in reproductive applications: a review. Theriogenology 2000; 53:1147-56. [PMID: 10798491 DOI: 10.1016/s0093-691x(00)00259-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The insulin-like growth factors (IGF-I and -II) are ubiquitously expressed factors that regulate cell growth, differentiation and maintenance of differentiated cell function. All aspects of male and female reproduction are influenced by the IGF system. This review will examine the IGF system as it pertains to reproductive physiology and applications.
Collapse
Affiliation(s)
- B R Lackey
- Department of Animal and Veterinary Science Clemson University, SC 29634, USA
| | | | | |
Collapse
|
35
|
Azcoitia I, Sierra A, Garcia-Segura LM. Neuroprotective effects of estradiol in the adult rat hippocampus: interaction with insulin-like growth factor-I signalling. J Neurosci Res 1999; 58:815-22. [PMID: 10583912 DOI: 10.1002/(sici)1097-4547(19991215)58:6<815::aid-jnr8>3.0.co;2-r] [Citation(s) in RCA: 159] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We have previously shown that 17-beta-estradiol protects neurons in the dentate gyrus from kainic acid-induced death in vivo. To analyse whether this effect is mediated through estrogen receptors and through cross-talk between steroid and insulin-like growth factor (IGF) systems, we have concomitantly administered antagonists of estrogen receptor (ICI 182,780) or the IGF-I receptor (JB1) with estradiol. In addition, we have also administered IGF-I with or without the estrogen receptor antagonist. JB1 (20 microg/ml), ICI 182,780 (10(-7) M), and IGF-I (100 microg/ml) were delivered into the left lateral ventricle of young ovariectomized rats via an Alzet osmotic minipump (0.5 microl/hr) for 2 weeks. All rats received kainic acid (7 mg/Kg b.w.) or vehicle i.p. injections at day 7 after minipump implant. Also on day 7, rats treated i.c. v.with only ICI 182,780 or JB1 received a single i.p. injection of 17-beta-estradiol (150 microg/rat) or vehicle. On day 14 after minipump implant, the rats were killed, brains processed, and the number of surviving hilar neurons estimated by the optical disector technique. Both IGF-I and estradiol treatments resulted in over 90% survival of hilar neurons. The neuroprotective action of estradiol was blocked by ICI 182,780 and by JB1. Furthermore, IGF-I enhancement of neuronal survival was significantly reduced by ICI 182,780. These results indicate that in this model of hippocampal lesion, the neuroprotective effect of estradiol depends both on estrogen receptors and IGF-I receptors, while the protection exerted by IGF-I depends also on estrogen receptors. In conclusion, an interaction of estrogen receptor and IGF-I receptor signalling may mediate neuroprotection in the adult rat hippocampus.
Collapse
Affiliation(s)
- I Azcoitia
- Department of Cell Biology, Faculty of Biology, Complutense University, Madrid, Spain.
| | | | | |
Collapse
|
36
|
Mong JA, McCarthy MM. Steroid-induced developmental plasticity in hypothalamic astrocytes: implications for synaptic patterning. JOURNAL OF NEUROBIOLOGY 1999; 40:602-19. [PMID: 10453059 DOI: 10.1002/(sici)1097-4695(19990915)40:4<602::aid-neu14>3.0.co;2-o] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have previously demonstrated that astrocytes in the developing arcuate nucleus of the rat hypothalamus exhibit a sexually dimorphic morphology as a result of differential exposure to gonadal steroids. Testosterone via its aromatized byproduct, estrogen, induces arcuate astrocytes to undergo differentiation during the first few days of life. These differentiated astrocytes exhibit a stellate morphology. Coincident with the steroid-induced increase in astrocyte differentiation is a reduction of dendritic spines on arcuate neurons. As a result, the arcuate nucleus of males has fewer axodendritic spine synapses than females and this dimorphism is retained throughout life. In the immediately adjacent ventromedial nucleus, neonatal astrocytes are immature and unresponsive to steroids. Neurons in this region show no change in dendritic spines in the first few days of life but do exhibit increased dendritic branching as a result of testosterone exposure. These findings illustrate the importance of distinct populations of astrocytes in restricted brain regions and their potential importance to the establishment of regionally specific synaptic patterning. Conflicting reports leave the site of steroid-mediated astrocyte responsiveness in the arcuate nucleus unresolved: Are gonadal steroids acting directly on astrocytes or are steroid-concentrating neurons mediating astrocytic responsiveness? In this review, we discuss the current understanding of astrocyte-neuron interactions and the possible mechanisms for steroid-mediated, astrocyte-directed synaptic patterning in the developing hypothalamus.
Collapse
Affiliation(s)
- J A Mong
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, 655 West Baltimore St., Baltimore, Maryland 21201, USA
| | | |
Collapse
|
37
|
Lackey BR, Gray SL, Henricks DM. The insulin-like growth factor (IGF) system and gonadotropin regulation: actions and interactions. Cytokine Growth Factor Rev 1999; 10:201-17. [PMID: 10647777 DOI: 10.1016/s1359-6101(99)00013-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factors (IGF) are polypeptides that regulate growth, differentiation and survival in a multitude of cells and tissues. The IGF system consists of ligands, receptors, binding proteins and binding protein proteases. The influence of the IGF system on reproductive parameters, specifically gonadotropin release and interactions between the IGF system and other effectors of gonadotropin release will be examined in this review.
Collapse
Affiliation(s)
- B R Lackey
- Department of Animal and Veterinary Science, Clemson University, SC 29634-0361, USA.
| | | | | |
Collapse
|
38
|
Longo KM, Sun Y, Gore AC. Insulin-like growth factor-I effects on gonadotropin-releasing hormone biosynthesis in GT1-7 cells. Endocrinology 1998; 139:1125-32. [PMID: 9492046 DOI: 10.1210/endo.139.3.5852] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The immortalized GT1-7 cell line synthesizes and secretes GnRH, the key hormone of reproduction. However, GT1-7 cells lack the normal inputs from neurotransmitters, growth factors, and steroids, which are involved in the maturation and maintenance of GnRH neurons in the brain. We examined the effects of the neurotrophic factor insulin-like growth factor-I (IGF-I) on GnRH gene expression and the mechanism for these changes. Initially, effects of IGF-I on GnRH gene expression were determined by ribonuclease protection assay. In time-course experiments, IGF-I treatment caused significant increases in nuclear GnRH primary transcript levels, an index of GnRH gene transcription, 4 and 8 h after initiation of IGF-I treatment. GnRH messenger RNA (mRNA) levels in the cytoplasm were stimulated by IGF-I at 24 h of treatment. IGF-I also affected GT1-7 cell morphology, with an increase in process extension and cell-cell contacts. In contrast, GnRH peptide levels in the medium were initially stimulated and then suppressed by IGF-I, indicating an uncoupling of biosynthesis and secretion. The increase in GnRH mRNA levels induced by IGF-I is probably caused by a transcriptional mechanism, as evidenced by the increase in GnRH primary transcript levels before a change in GnRH mRNA levels, as well as our finding of a similar GnRH mRNA half-life for both control and IGF-I-treated cells. Interestingly, GT1-7 cells themselves were observed to express IGF-I immunoreactivity, suggesting the possibility of autoregulation by this neurotrophic factor. It is concluded that IGF-I is an important modulator of GnRH gene expression and release in the GT1-7 cell line. The reported stimulatory effects of IGF-I in vivo, and its hypothesized role in the development of GnRH neurons in the brain, suggest that IGF-I may make the GT1-7 cells line more like a mature GnRH neuron, as a model for future studies.
Collapse
Affiliation(s)
- K M Longo
- Arthur M. Fishberg Research Center for Neurobiology and Henry L. Schwartz Department of Geriatrics and Adult Development, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | |
Collapse
|
39
|
Abstract
1. Both the neuroendocrine system and the brain mechanisms underlying gender-specific behavior are known to be organized by steroid sex hormones, androgen and estrogen, during specific sensitive phases of early fetal and perinatal development. The factors that control these phasic effects of the hormones on brain development are still not understood. Processes of masculinization and defeminization are thought to be involved in the sex differentiation of mammalian reproductive behavior. 2. The P450 aromatase, converting androgen to estrogen, is a key enzyme in the development of neural systems, and the activity of this enzyme is likely to be one of the factors determining brain sex differentiation. 3. We have examined the localization and regulation of brain aromatase using the mouse as a model. Measurement of testosterone conversion to estradiol-17 beta, using a sensitive radiometric 3H2O assay, indicates that estrogens are formed more actively in the male mouse brain than in the female during both the prenatal and the neonatal periods. In primary cell cultures of embryonic mouse hypothalamus there are sex differences in aromatase activity during early and late embryogenesis, with a higher capacity for estrogen formation in the male than the female. These sex differences are regionally specific in the brain, since on gender differences in aromatase activity are detectable in cortical cells. 4. Aromatase activity in the mouse brain is neuronal rather than glial. Using a specific antibody to the mouse aromatase, immunoreactivity is restricted to neuronal soma and neurites in hypothalamic cultures. There are more neurons containing expressed aromatase in the male hypothalamus than in the female. Therefore, gender-specific differences in embryonic aromatase activity are neuronal. 5. Testosterone increases aromatase activity specifically in hypothalamic neurons, but has no effect on cortical cells. The neuronal aromatase activity appears to be sensitive to the inductive effects of androgen only in the later stages of embryonic development. Androgen also increases the numbers of aromatase-immunoreactive neurons in the hypothalamus. 6. This work suggests that the embryonic male hypothalamus and other androgen target areas contain a network of neurons which has the capacity to provide estrogen for the sexual differentiation of brain mechanisms of behavior. The phasic activity of the key enzyme, aromatase, during development is influenced by androgen. What determines the developmental action of androgen and the other factors involved in the regulation and expression of this neuronal enzyme still have to be established.
Collapse
Affiliation(s)
- J B Hutchison
- MRC Neuroendocrine Development and Behaviour Group, Babraham Institute, Cambridge, U.K
| |
Collapse
|
40
|
Fernandez-Galaz MC, Morschl E, Chowen JA, Torres-Aleman I, Naftolin F, Garcia-Segura LM. Role of astroglia and insulin-like growth factor-I in gonadal hormone-dependent synaptic plasticity. Brain Res Bull 1997; 44:525-31. [PMID: 9370220 DOI: 10.1016/s0361-9230(97)00238-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Gonadal hormones exert a critical influence over the architecture of specific brain areas affecting the formation of neuronal contacts. Cellular mechanisms mediating gonadal hormone actions on synapses have been studied extensively in the rat arcuate nucleus, a hypothalamic center involved in the feed-back regulation of gonadotropins. Gonadal steroids exert organizational and activational effects on arcuate nucleus synaptic connectivity. Perinatal testosterone induces a sexual dimorphic pattern of synaptic contacts. Furthermore, during the preovulatory and ovulatory phases of the estrous cycle there is a transient disconnection of inhibitory synaptic inputs to the somas of arcuate neurons. This synaptic remodeling is induced by estradiol, blocked by progesterone, and begins with the onset of puberty in females. Astroglia appear to play a significant role in the organizational and the activational hormone effects on neuronal connectivity by regulating the amount of neuronal membrane available for the formation of synaptic contacts and by releasing soluble factors, such as insulin-like growth factor I (IGF-I), which promote the differentiation of neural processes. Recent evidence indicates that gonadal steroids and IGF-I may interact in their trophic effects on the neuroendocrine hypothalamus. Estradiol and IGF-I promote the survival and morphological differentiation of rat hypothalamic neurons in primary cultures. The effect of estradiol depends on IGF-I, while the effects of both estradiol and IGF-I depend on estrogen receptors. Furthermore, estrogen activation of astroglia in hypothalamic tissue fragments depends on IGF-I receptors. These findings indicate that IGF-I may mediate some of the developmental and activational effects of gonadal steroids on the brain and suggest that IGF-I may activate the estrogen receptor to induce its neurotrophic effects on hypothalamic cells. In addition, IGF-I levels in the neuroendocrine hypothalamus are regulated by gonadal steroids. IGF-I levels in tanycytes, a specific astroglia cell type present in the arcuate nucleus and median eminence, increase at puberty, are affected by neonatal androgen levels, show sex differences, and fluctuate in accordance to the natural variations in plasma levels of ovarian steroids that are associated with the estrous cycle. These changes appear to be mediated by hormonal regulation of IGF-I uptake from blood or cerebrospinal fluid by tanycytes. These results suggest that tanycytes may be involved in the regulation of neuroendocrine events in adult rats by regulating the availability of IGF-I to hypothalamic neurons. In summary, IGF-I and different forms of neuron-astroglia communication are involved in the effects of estradiol on synaptic plasticity in the hypothalamic arcuate nucleus.
Collapse
|
41
|
Hutchison JB, Beyer C, Hutchison RE, Wozniak A. Sex differences in the regulation of embryonic brain aromatase. J Steroid Biochem Mol Biol 1997; 61:315-22. [PMID: 9365207 DOI: 10.1016/s0960-0760(97)80029-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oestrogen formed from androgen by aromatization plays a critical role in the sexual differentiation of the male brain and behaviour. A question which has still to be answered is what regulates the gender-specific changes in aromatase activity forming oestrogen during sensitive periods of brain growth. Using a primary cell culture technique and sexed embryos, we have shown that in the fetal mouse brain, oestrogen formation in the male is neuronal rather than glial and aromatase activity is regionally localized, being higher in the hypothalamus than in the cortex. The aromatase activity measured from cells in culture has the same enzyme binding affinity (apparent Km approximately 40 nM) as intact brain samples. Neurones developing in the embryonic male brain (embryonic day (ED) 15) contain higher aromatase activity (Vmax, 895 fmol/h/mg protein) than the female (Vmax, 604). Although a sex difference exists at early stages of embryonic development (ED 13), the embryonic aromatase system is regulated by steroids later in fetal development. The developing aromatase-containing neuroblasts probably form processes which connect to other aromatase neurones. Immunoreactive staining with an aromatase polyclonal antibody identifies an increase in numbers of aromatase-immunoreactive hypothalamic neuronal cell bodies following testosterone treatment. Testosterone treatment also causes both stimulation of neurite growth and branching as well as functional maturation of aromatase neurones. In particular, there is an increase in aromatase activity per neurone as well as a dramatic increase in the number of neurones expressing the enzyme. Both the functional and morphological changes depend on androgen receptor stimulation for several days in vitro. This conclusion is supported by colocalization studies which reveal a high number of fetal hypothalamic aromatase neurones co-expressing androgen receptor. We conclude that testosterone influences the growth of male hypothalamic neurones containing aromatase at a sensitive period of brain development. Endogenous steroid inhibitors of aromatase, probably formed within the neuroglia, also play a role in the control of oestrogen production. An endogenous 5alpha-reduced metabolite of testosterone, 5alpha-androstanedione, is almost as potent in inhibiting neuronal hypothalamic aromatase activity (Ki = 23 nM) as the synthetic non-steroidal inhibitors such as the imidazole, fadrozole, and the triazoles, arimidex and letrozole. It is clear that the oestrogen-forming capacity of the male hypothalamus has the special characteristics and plasticity of regulation which could affect brain differentiation at specific steroid-sensitive stages in ontogeny.
Collapse
Affiliation(s)
- J B Hutchison
- MRC Neuroendocrine Development and Behaviour Group, The Babraham Institute, Cambridge, U.K
| | | | | | | |
Collapse
|
42
|
D'Ercole AJ, Ye P, Calikoglu AS, Gutierrez-Ospina G. The role of the insulin-like growth factors in the central nervous system. Mol Neurobiol 1996; 13:227-55. [PMID: 8989772 DOI: 10.1007/bf02740625] [Citation(s) in RCA: 336] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Increasing evidence strongly supports a role for insulin-like growth factor-I (IGF-I) in central nervous system (CNS) development. IGF-I, IGF-II, the type IIGF receptor (the cell surface tyrosine kinase receptor that mediates IGF signals), and some IGF binding proteins (IGFBPs; secreted proteins that modulate IGF actions) are expressed in many regions of the CNS beginning in utero. The expression pattern of IGF system proteins during brain growth suggests highly regulated and developmentally timed IGF actions on specific neural cell populations. IGF-I expression is predominantly in neurons and, in many brain regions, peaks in a fashion temporally coincident with periods in development when neuron progenitor proliferation and/or neuritic outgrowth occurs. In contrast, IGF-II expression is confined mainly to cells of mesenchymal and neural crest origin. While expression of type I IGF receptors appears ubiquitous, that of IGFBPs is characterized by regional and developmental specificity, and often occurs coordinately with peaks of IGF expression. In vitro IGF-I has been shown to stimulate the proliferation of neuron progenitors and/or the survival of neurons and oligodendrocytes, and in some cultured neurons, to stimulate function. Transgenic (Tg) mice that overexpress IGF-I in the brain exhibit postnatal brain overgrowth without anatomic abnormality (20-85% increases in weight, depending on the magnitude of expression). In contrast, Tg mice that exhibit ectopic brain expression of IGFBP-1, an inhibitor of IGF action when present in molar excess, manifest postnatal brain growth retardation, and mice with ablated IGF-I gene expression, accomplished by homologous recombination, have brains that are 60% of normal size as adults. Taken together, these in vivo studies indicate that IGF-I can influence the development of most, if not all, brain regions, and suggest that the cerebral cortex and cerebellum are especially sensitive to IGF-I actions. IGF-I's growth-promoting in vivo actions result from its capacity to increase neuron number, at least in certain populations, and from its potent stimulation of myelination. These IGF-I actions, taken together with its neuroprotective effects following CNS and peripheral nerve injury, suggest that it may be of therapeutic benefit in a wide variety of disorders affecting the nervous system.
Collapse
Affiliation(s)
- A J D'Ercole
- Department of Pediatrics CB# 7220, University of North Carolina, Chapel Hill 27599-7220, USA
| | | | | | | |
Collapse
|
43
|
Duenas M, Torres-Aleman I, Naftolin F, Garcia-Segura LM. Interaction of insulin-like growth factor-I and estradiol signaling pathways on hypothalamic neuronal differentiation. Neuroscience 1996; 74:531-9. [PMID: 8865203 DOI: 10.1016/0306-4522(96)00142-x] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neurotrophic effects of estradiol and insulin-like growth factor-I were assessed in primary cultures from fetal rat hypothalamus. Cultured neurons were immunostained with an antibody for the microtubule-associated protein-2. While both estradiol and insulin-like growth factor-I increased the number of microtubule-associated protein-2-immunoreactive neurons and the extension of immunoreactive processes, the effect of these two factors was not additive. The estradiol-induced increases in neuronal numbers and extension of neuronal processes were blocked by either the estrogen receptor antagonist ICI 182,780 or by an anti-sense oligonucleotide to the estrogen receptor. Furthermore, incubation of the cultures with an anti-sense oligonucleotide directed against the insulin-like growth factor-I messenger RNA also blocked the effect of estradiol. In turn, the effects of insulin-like growth factor-I were blocked by the estrogen receptor antagonist ICI 182,780 and by the anti-sense oligonucleotide to the estrogen receptor. These findings suggest that estradiol-induced activation of the estrogen receptor in developing hypothalamic cells requires the presence of insulin-like growth factor-I, and that both estradiol and insulin-like growth factor-I use the estrogen receptor as a mediator of their trophic effects on hypothalamic neurons.
Collapse
Affiliation(s)
- M Duenas
- Instituto Cajal, C.S.I.C., Madrid, Spain
| | | | | | | |
Collapse
|
44
|
Naftolin F, Leranth C, Horvath TL, Garcia-Segura LM. Potential neuronal mechanisms of estrogen actions in synaptogenesis and synaptic plasticity. Cell Mol Neurobiol 1996; 16:213-23. [PMID: 8743970 PMCID: PMC11563122 DOI: 10.1007/bf02088177] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/1995] [Accepted: 05/15/1995] [Indexed: 02/01/2023]
Abstract
1. Studies conducted on the rat arcuate nucleus, an area involved in the development and control of LH and FSH secretion, have shown the existence of hormonally regulated developmental sex differences in synaptic patterns and estrogen-induced synaptic plasticity during adult life. Several questions raised by these findings are examined in this review: 2. The mechanisms of estrogen-regulated developmental synaptogenesis. These include the role of glycocalyx glycoproteins in neuronal membranes, neural cell adhesion molecules, and insulin-like growth factor I. 3. The relationship among circulating estrogen, gonadotropin levels, and hypothalamic synaptic plasticity. Recent evidence for the role of GABAergic and dopaminergic synaptic inputs and POMC projections from the arcuate nucleus to the GnRH cells is discussed. 4. The synaptologic basis of age-related failure of positive feedback. The role of the cumulative effect of repeated preovulatory synaptic retraction and reapplication cycles on sensescent constant estrus is analyzed.
Collapse
Affiliation(s)
- F Naftolin
- Department of Obstetrics and Gynecology, Yale University, School of Medicine, New Haven, Connecticut 06520-8063, USA
| | | | | | | |
Collapse
|
45
|
Garcia-Segura LM, Chowen JA, Dueñas M, Parducz A, Naftolin F. Gonadal steroids and astroglial plasticity. Cell Mol Neurobiol 1996; 16:225-37. [PMID: 8743971 PMCID: PMC11563075 DOI: 10.1007/bf02088178] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/1995] [Accepted: 06/29/1995] [Indexed: 02/01/2023]
Abstract
1. Recent evidence indicates that astroglia participate in the metabolism of gonadal hormones, in the synthesis of neurosteroids, and in the plastic responses of neurons to gonadal steroids. The role of astroglia on plastic responses of neural tissue to gonadal hormones and neurosteroids is examined in this review. 2. Gonadal steroids and neurosteroids promote astroglia plasticity in several areas of the central nervous system, including the hypothalamus, the striatum, and the hippocampus. 3. Gonadal steroids and neurosteroids modulate astroglia proliferation and the formation of reactive astroglia after brain injury. 4. Astroglia is a source of trophic factors that may mediate effects of gonadal steroids on neural tissue. 5. Astroglia is involved in the promotion of synaptic plastic changes by gonadal hormones. 6. The effect of gonadal hormones on astroglial plasticity is dependent on specific membrane interactions with neurons and on the expression of the embryonic highly polysialylated isoform of the neural cell adhesion molecule on neuronal membranes. 7. In conclusion, coordinated responses of neurons and astroglia appear to be involved in the modulation of neural function and response to injury by gonadal hormones and neurosteroids.
Collapse
|
46
|
Kawata M. Roles of steroid hormones and their receptors in structural organization in the nervous system. Neurosci Res 1995; 24:1-46. [PMID: 8848287 DOI: 10.1016/0168-0102(96)81278-8] [Citation(s) in RCA: 225] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Due to their chemical properties, steroid hormones cross the blood-brain barrier where they have profound effects on neuronal development and reorganization both in invertebrates and vertebrates, including humans mediated through their receptors. Steroids play a crucial role in the organizational actions of cellular differentiation representing sexual dimorphism and apoptosis, and in the activational effects of phenotypic changes in association with structural plasticity. Their sites of action are primarily the genes themselves but some are coupled with membrane-bound receptor/ion channels. The effects of steroid hormones on gene transcription are not direct, and other cellular components interfere with their receptors through cross-talk and convergence of the signaling pathways in neurons. These genomic and non-genomic actions account for the divergent effects of steroid hormones on brain function as well as on their structure. This review looks again at and updates the tremendous advances made in recent decades on the study of the role of steroid (gonadal and adrenal) hormones and their receptors on developmental processes and plastic changes in the nervous system.
Collapse
Affiliation(s)
- M Kawata
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Japan
| |
Collapse
|
47
|
García-Segura LM, Chowen JA, Párducz A, Naftolin F. Gonadal hormones as promoters of structural synaptic plasticity: cellular mechanisms. Prog Neurobiol 1994; 44:279-307. [PMID: 7886228 DOI: 10.1016/0301-0082(94)90042-6] [Citation(s) in RCA: 202] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It is now obvious that the CNS is capable of undergoing a variety of plastic changes at all stages of development. Although the magnitude and distribution of these changes may be more dramatic in the immature animal, the adult brain retains a remarkable capacity for undergoing morphological and functional modifications. Throughout development, as well as in the postpubertal animal, gonadal steroids exert an important influence over the architecture of specific sex steroid-responsive areas, resulting in sexual dimorphisms at both morphological and physiological levels. We are only now beginning to gain insight into the mechanisms involved in gonadal steroid-induced synaptic changes. The number of synaptic inputs to specific neuronal populations is sexually dimorphic and this can be modulated by changes in the sex steroid environment. These modifications can be correlated with other morphological changes, such as glial cell activation, that are occurring simultaneously in the same anatomical area. Indeed, the close physical relationship between glial cells and neuronal synaptic contacts makes them an ideal candidate for participating in this process. Interestingly, not only can the morphology and immunoreactivity of glial cells be modulated by gonadal steroids, but a close negative correlation between the number of synapses and the amount of glial ensheathing of a neuron has been demonstrated, suggesting an active participation of these cells in this process. Glia have sex steroid receptors, are capable of producing and metabolizing steroids, and can produce other neuronal trophic factors in response to sex steroids. Hence, their role in gonadal steroid-induced synaptic plasticity is becoming more apparent. In addition, there is recent evidence that this process may involve certain cell surface molecules, such as the N-CAMs, since a specific isoform of this molecule, previously referred to as the embryonic form, is found in those areas of the brain which maintain the capacity to undergo synaptic remodelling. However, there is much work to be done in order to fully understand this phenomenon and before bringing it into a clinical setting in hopes of treating neurodegenerative diseases or injuries to the nervous system.
Collapse
|
48
|
Pilgrim C, Hutchison JB. Developmental regulation of sex differences in the brain: can the role of gonadal steroids be redefined? Neuroscience 1994; 60:843-55. [PMID: 7936207 DOI: 10.1016/0306-4522(94)90267-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
49
|
Garcia-Segura LM, Chowen JA, Dueñas M, Torres-Aleman I, Naftolin F. Gonadal steroids as promoters of neuro-glial plasticity. Psychoneuroendocrinology 1994; 19:445-53. [PMID: 7938345 DOI: 10.1016/0306-4530(94)90031-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Estradiol induces coordinated modifications in the extension of glial and neuronal processes in the arcuate nucleus of the hypothalamus of adult female rats. This hormonal effect results in natural fluctuations in the ensheathing of arcuate neurons by glial processes and these glial changes are linked to a remodelling of inhibitory GABAergic synapses during the estrous cycle. Hormonally induced glial and synaptic changes appear to be dependent on specific recognition or adhesion molecules on the neuronal and/or glial membranes.
Collapse
|
50
|
Garcia-Segura LM, Luquín S, Párducz A, Naftolin F. Gonadal hormone regulation of glial fibrillary acidic protein immunoreactivity and glial ultrastructure in the rat neuroendocrine hypothalamus. Glia 1994; 10:59-69. [PMID: 8300192 DOI: 10.1002/glia.440100108] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The influence of gonadal steroids on the ultrastructure of glial cells and on the immunoreactivity for the specific astrocytic marker glial fibrillary acidic protein (GFAP) has been assessed in the neuroendocrine hypothalamus. The following parameters were analyzed in the arcuate nucleus of adult female rats: the number and the surface density of cells immunoreactive for GFAP, the number of glial profiles showing bundles of glial filaments, the size of the bundles of glial filaments, and the proportion of neuronal perikaryal membrane apposed by glial processes. These parameters were studied during the different phases of the estrous cycle, after ovariectomy, and after the administration of estradiol or progesterone to ovariectomized rats. No significant differences were detected in the number of GFAP-immunoreactive cells among the different experimental groups. The surface density of GFAP-immunoreactive material, the number of glial profiles in the neuropil, and the proportion of neuronal perikaryal membrane covered by glia were increased in the afternoon of proestrus and in the morning of estrus compared with other phases of the estrous cycle or to ovariectomized rats and showed a rapid (5 h) and reversible increase in ovariectomized rats injected with 17 beta estradiol, with a maximal effect by 24 h after the administration of the hormone. In contrast, the size of the bundles of glial filaments was decreased in the afternoon of proestrus, in the morning of estrus, and by the administration of estradiol to ovariectomized rats. The parameters studied were not affected by the administration of progesterone. However, progesterone (300 micrograms/rat) blocked the effects of 17 beta estradiol (1, 10, and 300 micrograms). The results suggest that glial cells may be actively involved in the modulation of neuroendocrine events by the hypothalamus.
Collapse
|