1
|
Hussain MS, Altamimi ASA, Afzal M, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Shahwan M, Kukreti N, Wong LS, Kumarasamy V, Subramaniyan V. Kaempferol: Paving the path for advanced treatments in aging-related diseases. Exp Gerontol 2024; 188:112389. [PMID: 38432575 DOI: 10.1016/j.exger.2024.112389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/17/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Aging-related diseases (ARDs) are a major global health concern, and the development of effective therapies is urgently needed. Kaempferol, a flavonoid found in several plants, has emerged as a promising candidate for ameliorating ARDs. This comprehensive review examines Kaempferol's chemical properties, safety profile, and pharmacokinetics, and highlights its potential therapeutic utility against ARDs. Kaempferol's therapeutic potential is underpinned by its distinctive chemical structure, which confers antioxidative and anti-inflammatory properties. Kaempferol counteracts reactive oxygen species (ROS) and modulates crucial cellular pathways, thereby combating oxidative stress and inflammation, hallmarks of ARDs. Kaempferol's low toxicity and wide safety margins, as demonstrated by preclinical and clinical studies, further substantiate its therapeutic potential. Compelling evidence supports Kaempferol's substantial potential in addressing ARDs through several mechanisms, notably anti-inflammatory, antioxidant, and anti-apoptotic actions. Kaempferol exhibits a versatile neuroprotective effect by modulating various proinflammatory signaling pathways, including NF-kB, p38MAPK, AKT, and the β-catenin cascade. Additionally, it hinders the formation and aggregation of beta-amyloid protein and regulates brain-derived neurotrophic factors. In terms of its anticancer potential, kaempferol acts through diverse pathways, inducing apoptosis, arresting the cell cycle at the G2/M phase, suppressing epithelial-mesenchymal transition (EMT)-related markers, and affecting the phosphoinositide 3-kinase/protein kinase B signaling pathways. Subsequent studies should focus on refining dosage regimens, exploring innovative delivery systems, and conducting comprehensive clinical trials to translate these findings into effective therapeutic applications.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | | | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 346, United Arab Emirates
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 346, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia.
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
2
|
Rahul, Siddique YH. Neurodegenerative Diseases and Flavonoids: Special Reference to Kaempferol. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 20:327-342. [PMID: 33511932 DOI: 10.2174/1871527320666210129122033] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/21/2020] [Accepted: 09/13/2020] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, Huntington's disease, Multiple Sclerosis and Ischemic stroke have become a major health problem worldwide. Pre-clinical studies have demonstrated the beneficial effects of flavonoids on neurodegenerative diseases and suggest them to be used as therapeutic agents. Kaempferol is found in many plants such as tea, beans, broccoli, strawberries, and neuroprotective effects against the development of many neurodegenerative diseases such as Parkinson, Alzheimer's disease and Huntington's disease. The present study summarizes the neuroprotective effects of kaempferol in various models of neurodegenerative diseases. Kaempferol delays the initiation as well as the progression of neurodegenerative disorders by acting as a scavenger of free radicals and preserving the activity of various antioxidant enzymes. Kaempferol can cross the Blood-Brain Barrier (BBB), and therefore results in an enhanced protective effect. The multi-target property of kaempferol makes it a potential dietary supplement in preventing and treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Rahul
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Yasir H Siddique
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| |
Collapse
|
3
|
Ellis SN, Honeycutt JA. Sex Differences in Affective Dysfunction and Alterations in Parvalbumin in Rodent Models of Early Life Adversity. Front Behav Neurosci 2021; 15:741454. [PMID: 34803622 PMCID: PMC8600234 DOI: 10.3389/fnbeh.2021.741454] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023] Open
Abstract
The early life environment markedly influences brain and behavioral development, with adverse experiences associated with increased risk of anxiety and depressive phenotypes, particularly in females. Indeed, early life adversity (ELA) in humans (i.e., caregiver deprivation, maltreatment) and rodents (i.e., maternal separation, resource scarcity) is associated with sex-specific emergence of anxious and depressive behaviors. Although these disorders show clear sex differences in humans, little attention has been paid toward evaluating sex as a biological variable in models of affective dysfunction; however, recent rodent work suggests sex-specific effects. Two widely used rodent models of ELA approximate caregiver deprivation (i.e., maternal separation) and resource scarcity (i.e., limited bedding). While these approaches model aspects of ELA experienced in humans, they span different portions of the pre-weaning developmental period and may therefore differentially contribute to underlying mechanistic risk. This is borne out in the literature, where evidence suggests differences in trajectories of behavior depending on the type of ELA and/or sex; however, the neural underpinning of these differences is not well understood. Because anxiety and depression are thought to involve dysregulation in the balance of excitatory and inhibitory signaling in ELA-vulnerable brain regions (e.g., prefrontal cortex, amygdala, hippocampus), outcomes are likely driven by alterations in local and/or circuit-specific inhibitory activity. The most abundant GABAergic subtypes in the brain, accounting for approximately 40% of inhibitory neurons, contain the calcium-binding protein Parvalbumin (PV). As PV-expressing neurons have perisomatic and proximal dendritic targets on pyramidal neurons, they are well-positioned to regulate excitatory/inhibitory balance. Recent evidence suggests that PV outcomes following ELA are sex, age, and region-specific and may be influenced by the type and timing of ELA. Here, we suggest the possibility of a combined role of PV and sex hormones driving differences in behavioral outcomes associated with affective dysfunction following ELA. This review evaluates the literature across models of ELA to characterize neural (PV) and behavioral (anxiety- and depressive-like) outcomes as a function of sex and age. Additionally, we detail a putative mechanistic role of PV on ELA-related outcomes and discuss evidence suggesting hormone influences on PV expression/function which may help to explain sex differences in ELA outcomes.
Collapse
Affiliation(s)
- Seneca N Ellis
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States
| | - Jennifer A Honeycutt
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States.,Department of Psychology, Bowdoin College, Brunswick, ME, United States
| |
Collapse
|
4
|
Schiller CE, Johnson SL, Abate AC, Schmidt PJ, Rubinow DR. Reproductive Steroid Regulation of Mood and Behavior. Compr Physiol 2016; 6:1135-60. [PMID: 27347888 PMCID: PMC6309888 DOI: 10.1002/cphy.c150014] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this article, we examine evidence supporting the role of reproductive steroids in the regulation of mood and behavior in women and the nature of that role. In the first half of the article, we review evidence for the following: (i) the reproductive system is designed to regulate behavior; (ii) from the subcellular to cellular to circuit to behavior, reproductive steroids are powerful neuroregulators; (iii) affective disorders are disorders of behavioral state; and (iv) reproductive steroids affect virtually every system implicated in the pathophysiology of depression. In the second half of the article, we discuss the diagnosis of the three reproductive endocrine-related mood disorders (premenstrual dysphoric disorder, postpartum depression, and perimenopausal depression) and present evidence supporting the relevance of reproductive steroids to these conditions. Existing evidence suggests that changes in reproductive steroid levels during specific reproductive states (i.e., the premenstrual phase of the menstrual cycle, pregnancy, parturition, and the menopause transition) trigger affective dysregulation in susceptible women, thus suggesting the etiopathogenic relevance of these hormonal changes in reproductive mood disorders. Understanding the source of individual susceptibility is critical to both preventing the onset of illness and developing novel, individualized treatments for reproductive-related affective dysregulation. © 2016 American Physiological Society. Compr Physiol 6:1135-1160, 2016e.
Collapse
Affiliation(s)
- Crystal Edler Schiller
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah L. Johnson
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anna C. Abate
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter J. Schmidt
- Section on Behavioral Endocrinology, National Institute of Mental Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - David R. Rubinow
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Kudo D, Miyakoshi N, Hongo M, Matsumoto-Miyai K, Kasukawa Y, Misawa A, Ishikawa Y, Shimada Y. Nerve Growth Factor and Estrogen Receptor mRNA Expression in Paravertebral Muscles of Patients With Adolescent Idiopathic Scoliosis: A Preliminary Study. Spine Deform 2015; 3:122-127. [PMID: 27927302 DOI: 10.1016/j.jspd.2014.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 05/26/2014] [Accepted: 07/18/2014] [Indexed: 01/02/2023]
Abstract
STUDY DESIGN Comparison of nerve growth factor (NGF) and estrogen receptor (ER)α messenger ribonucleic acid (mRNA) expression in bilateral paravertebral muscles in adolescent idiopathic scoliosis (AIS). This expression in AIS was compared with that of normal control subjects. OBJECTIVES To investigate NGF and ERα mRNA expression in bilateral paravertebral muscles in AIS and control subjects to clarify its association with the development and progression of spinal curvature. SUMMARY OF BACKGROUND DATA Paravertebral muscle abnormalities in AIS patients have been investigated through various methods. Despite the roles of NGF and ER in human skeletal muscles, the association with idiopathic scoliosis is still unclear. METHODS A total of 14 AIS patients (average age, 15.9 ± 2.2 years; average Cobb angle, 48.2° ± 8.9°) and 8 controls (average age, 27.3 ± 9.3 years) were included. Muscle samples were harvested from bilateral paravertebral muscles at the apical vertebral level. Nerve growth factor and ERα mRNA expression was evaluated by the real-time polymerase chain reaction. The researchers compared expression levels in bilateral paravertebral muscles in each group. The expression ratio, the expression at the convex side relative to the concave side, was compared between groups and the correlation between Cobb angle and expression ratio was analyzed. RESULTS Nerve growth factor and ERα mRNA expression on the convex side was higher than on the concave side in the AIS group (p = .024 and .007, respectively) and the expression ratio of NGF and ERα in the AIS group was higher than that of control subjects (p = .004 and .017, respectively). The expression ratio of NGF and the Cobb angle were significantly correlated (r = -0.5728; p = .0323). CONCLUSIONS In the AIS group, both NGF and ERα mRNA expression was asymmetric. The AIS group had higher expression ratios than control group and the NGF expression ratio was positively correlated to the Cobb angle.
Collapse
Affiliation(s)
- Daisuke Kudo
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan.
| | - Naohisa Miyakoshi
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Michio Hongo
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Kazumasa Matsumoto-Miyai
- Department of Neurophysiology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Yuji Kasukawa
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Akiko Misawa
- Department of Orthopedic Surgery, Akita Prefectural Center on Development and Disability, 1-128 Aza-suwanosawa Kamikitate-momozaki, Akita 010-1407, Japan
| | - Yoshinori Ishikawa
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Yoichi Shimada
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| |
Collapse
|
6
|
Wessels JM, Leyland NA, Agarwal SK, Foster WG. Estrogen induced changes in uterine brain-derived neurotrophic factor and its receptors. Hum Reprod 2015; 30:925-36. [DOI: 10.1093/humrep/dev018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
7
|
Abstract
Despite decades of research aimed at identifying the causes of postpartum depression (PPD), PPD remains common, and the causes are poorly understood. Many have attributed the onset of PPD to the rapid perinatal change in reproductive hormones. Although a number of human and nonhuman animal studies support the role of reproductive hormones in PPD, several studies have failed to detect an association between hormone concentrations and PPD. The purpose of this review is to examine the hypothesis that fluctuations in reproductive hormone levels during pregnancy and the postpartum period trigger PPD in susceptible women. We discuss and integrate the literature on animal models of PPD and human studies of reproductive hormones and PPD. We also discuss alternative biological models of PPD to demonstrate the potential for multiple PPD phenotypes and to describe the complex interplay of changing reproductive hormones and alterations in thyroid function, immune function, hypothalamic-pituitary-adrenal (HPA) axis function, lactogenic hormones, and genetic expression that may contribute to affective dysfunction. There are 3 primary lines of inquiry that have addressed the role of reproductive hormones in PPD: nonhuman animal studies, correlational studies of postpartum hormone levels and mood symptoms, and hormone manipulation studies. Reproductive hormones influence virtually every biological system implicated in PPD, and a subgroup of women seem to be particularly sensitive to the effects of perinatal changes in hormone levels. We propose that these women constitute a "hormone-sensitive" PPD phenotype, which should be studied independent of other PPD phenotypes to identify underlying pathophysiology and develop novel treatment targets.
Collapse
|
8
|
Allopregnanolone as a mediator of affective switching in reproductive mood disorders. Psychopharmacology (Berl) 2014; 231:3557-67. [PMID: 24846476 PMCID: PMC4135022 DOI: 10.1007/s00213-014-3599-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/14/2014] [Indexed: 01/15/2023]
Abstract
RATIONALE Reproductive mood disorders, including premenstrual dysphoria (PMD) and postpartum depression (PPD), are characterized by affective dysregulation that occurs during specific reproductive states. The occurrence of illness onset during changes in reproductive endocrine function has generated interest in the role of gonadal steroids in the pathophysiology of reproductive mood disorders, yet the mechanisms by which the changing hormone milieu triggers depression in susceptible women remain poorly understood. OBJECTIVES This review focuses on one of the neurosteroid metabolites of progesterone - allopregnanolone (ALLO) - that acutely regulates neuronal function and may mediate affective dysregulation that occurs concomitant with changes in reproductive endocrine function. We describe the role of the "neuroactive" steroids estradiol and progesterone in reproductive endocrine-related mood disorders to highlight the potential mechanisms by which ALLO might contribute to their pathophysiology. Finally, using existing data, we test the hypothesis that changes in ALLO levels may trigger affective dysregulation in susceptible women. RESULTS Although there is no reliable evidence that basal ALLO levels distinguish those with PMD or PPD from those without, existing animal models suggest potential mechanisms by which specific reproductive states may unmask susceptibility to affective dysregulation. Consistent with these models, initially euthymic women with PMD and those with a history of PPD show a negative association between depressive symptoms and circulating ALLO levels following progesterone administration. CONCLUSIONS Existing animal models and our own preliminary data suggest that ALLO may play an important role in the pathophysiology of reproductive mood disorders by triggering affective dysregulation in susceptible women.
Collapse
|
9
|
Firozan B, Goudarzi I, Elahdadi Salmani M, Lashkarbolouki T, Rezaei A, Abrari K. Estradiol increases expression of the brain-derived neurotrophic factor after acute administration of ethanol in the neonatal rat cerebellum. Eur J Pharmacol 2014; 732:1-11. [DOI: 10.1016/j.ejphar.2014.02.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/10/2014] [Accepted: 02/23/2014] [Indexed: 11/16/2022]
|
10
|
Wilson ME. Stroke: understanding the differences between males and females. Pflugers Arch 2013; 465:595-600. [PMID: 23503729 DOI: 10.1007/s00424-013-1260-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 03/01/2013] [Accepted: 03/01/2013] [Indexed: 01/09/2023]
Abstract
Stroke is a significant cause of death and long-term disability in the USA. The incidence, mortality, and outcomes of stroke are significantly different between men and women. As with many diseases that affect men and women differently, an understanding on the reasons underlying those differences is critical to effective diagnosis and treatment. This review will examine the sex differences in stroke in both humans and animal models of stroke and review what is known about potential mechanisms underlying these differences. It is clear that there is a complex interaction between hormonal, genetic, and unknown factors at play in generating the sex differences in stroke.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, University of Kentucky, MS508 800 Rose St., Lexington, KY 40536, USA.
| |
Collapse
|
11
|
Schmidt PJ, Rubinow DR. Reproductive hormonal treatments for mood disorders in women. DIALOGUES IN CLINICAL NEUROSCIENCE 2012. [PMID: 22033644 PMCID: PMC3181679 DOI: 10.31887/dcns.2002.4.2/pschmidt] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There has been a century-long view in medicine that reproductive function in both men and women is intimately involved with mood regulation. The 19th century witnessed a proliferation of medical reports documenting beneficial effects on mood and behavior after medical or surgical manipulations of women's reproductive functíon. More recently, the results of several studies suggest that gonadal steroids do regulate mood in some women. Thus, there is considerable interest in the potential role of reproductive therapies in the management of depressive illness, including both classical and reproductive endocrine-related mood disorders. Future studies need to determine the predictors of response to hormonal therapies compared with traditional antidepressant agents, and to characterize the long-term safety and benefits of these therapies.
Collapse
Affiliation(s)
- Peter J Schmidt
- Behavioral Endocrinology Branch, National Institute of Mental Health, Bethesda, Md, USA
| | | |
Collapse
|
12
|
Vega Orozco A, Daneri C, Anesetti G, Cabrera R, Sosa Z, Rastrilla AM. Involvement of the oestrogenic receptors in superior mesenteric ganglion on the ovarian steroidogenesis in rat. Reproduction 2011; 143:183-93. [PMID: 22080140 DOI: 10.1530/rep-11-0056] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Oestradiol (E(2)) is a key hormone in the regulation of reproductive processes. The aims of this work were a) to examine the distributions of oestrogen receptor α (ERα) and ERβ in the neurons of the superior mesenteric ganglion (SMG) in the oestrus stage by immunohistochemistry, b) to demonstrate whether E(2) in the SMG modifies progesterone (P(4)), androstenedione (A(2)) and nitrite release in the ovarian compartment on oestrus day and c) to demonstrate whether E(2) in the ganglion modifies the activity and gene expression in the ovary of the steroidogenic enzymes 3β-hydroxysteroid dehydrogenase (3β-HSD) and 20α-hydroxysteroid dehydrogenase (20α-HSD). The ex vivo SMG-ovarian nervous plexus-ovary system was used. E(2), tamoxifen (Txf) and E(2) plus Txf were added in the ganglion to measure ovarian P(4) release, while E(2) alone was added to measure ovarian A(2) and nitrites release. Immunohistochemistry revealed cytoplasmic ERα immunoreactivity only in the neural somas in the SMG. E(2) increased ovarian P(4) and A(2) release at 15, 30 and 60 min but decreased nitrites. The activity and gene expression of 3β-HSD increased, while the activity and gene expression of 20α-HSD did not show changes with respect to the control. Txf in the ganglion diminished P(4) release only at 60 min. E(2) plus Txf in the ganglion reverted the effect of E(2) alone and the inhibitory effect of Txf. The results of this study demonstrate that ERα activation in the SMG has an impact on ovarian steroidogenesis in rats, thus providing evidence for the critical role of peripheral system neurons in the control of ovarian functions under normal and pathological conditions.
Collapse
Affiliation(s)
- Adriana Vega Orozco
- Laboratorio de Biología de la Reproducción (LABIR), Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina.
| | | | | | | | | | | |
Collapse
|
13
|
Johann S, Dahm M, Kipp M, Zahn U, Beyer C. Regulation of choline acetyltransferase expression by 17 β-oestradiol in NSC-34 cells and in the spinal cord. J Neuroendocrinol 2011; 23:839-48. [PMID: 21790808 DOI: 10.1111/j.1365-2826.2011.02192.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Motoneurones located in the ventral horn of the spinal cord conciliate cholinergic innervation of skeletal muscles. These neurones appear to be exceedingly affected in neurodegenerative diseases such as amyotrophic lateral sclerosis. The dysfunction of motoneurones is typically accompanied by alterations of cholinergic metabolism and signalling, as demonstrated by a decrease in choline acetyltransferase (ChAT) expression. 17 β-Oestradiol (E(2)) is generally accepted as neuroprotective factor in the brain under acute toxic and neurodegenerative conditions and also appears to exert a protective role for motoneurones. In the present study, we attempted to analyse the role of E(2) signalling on ChAT expression in the motoneurone-like cell line NSC-34 and in vivo. In a first step, we demonstrated the presence of oestrogen receptor α and β in NSC-34 cells, as well as in the cervical and lumbar parts, of the male mouse spinal cord. Subsequently, we investigated the effect of E(2) treatment on ChAT expression. The application of E(2) significantly increased the transcription of ChAT in NSC-34 cells and in the cervical but not lumbar part of the spinal cord. Our results indicate that E(2) can influence the cholinergic system by increasing ChAT expression in the mouse spinal cord. This mechanism might support motoneurones, in addition to survival-promoting mechanisms, in the temporal balance toxic or neurodegenerative challenges.
Collapse
Affiliation(s)
- S Johann
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany.
| | | | | | | | | |
Collapse
|
14
|
Mun'im A, Isoda H, Seki M, Negishi O, Ozawa T. Estrogenic and Acetylcholinesterase-Enhancement Activity of a New Isoflavone, 7,2',4'-Trihydroxyisoflavone-4'-O-beta-D-Glucopyranoside from Crotalaria Sessililflora. Cytotechnology 2011; 43:127-34. [PMID: 19003217 DOI: 10.1023/b:cyto.0000039899.67160.18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A new isoflavone, 7,2',4'-trihydroxyisoflavone-4'-O-beta-D-glucopyranoside has been isolated from the aerial part of Crotalaria sessiliflora. The isoflavone glucoside enhanced the proliferation of the MCF-7 human breast cancer cell line, which possesses estrogen receptor (ER) and responds to estrogen in culture. The estrogenic property of the isoflavone glucoside was blocked by the known ER antagonist tamoxifen, indicating the involvement of the ER. Furthermore, the isoflavone glucoside was found to enhance the acetylcholinesterase (AChE) activity of the rat neuronal cell line PC12 at low concentrations of nerve growth factor (NGF).
Collapse
Affiliation(s)
- Abdul Mun'im
- Institute of Applied Biochemistry, University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
| | | | | | | | | |
Collapse
|
15
|
Rubinow DR, Girdler SS. Hormones, heart disease, and health: individualized medicine versus throwing the baby out with the bathwater. Depress Anxiety 2011; 28:E1-E15. [PMID: 21648024 DOI: 10.1002/da.20833] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is increasingly axiomatic that depression has widespread adverse physiological effects and, conversely, that a variety of physiological systems impact the risk for developing depression. This convergence of depression and altered physiology is particularly dramatic during midlife--a time during which reproductive failure presages dramatic increases in prevalence of both heart disease and depression. The potentially meaningful and illuminating links between estrogen deficiency, cardiovascular disease (CVD), and depression have largely been obscured, first by assertions, subsequently repudiated, that the perimenopause was not a time of increased risk of depression, and more recently by the denegration of hormone replacement therapy by initial reports of the Women's Health Initiative. Increasingly, however, research has led to unavoidable conclusions that CVD and depression share common, mediating pathogenic processes and that these same processes are dramatically altered by the presence or absence of estrogen (E2). This review summarizes data supporting these contentions with the intent of placing depression and estrogen therapy in their proper physiologic context.
Collapse
Affiliation(s)
- David R Rubinow
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina 27599. USA
| | | |
Collapse
|
16
|
Rubinow DR, Girdler SS. Hormones, heart disease, and health: individualized medicine versus throwing the baby out with the bathwater. Depress Anxiety 2011; 28:282-96. [PMID: 21456038 DOI: 10.1002/da.20810] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
It is increasingly axiomatic that depression has widespread adverse physiological effects, and conversely that a variety of physiological systems impact the risk for developing depression. This convergence of depression and altered physiology is particularly dramatic during midlife-a time during which reproductive failure presages dramatic increases in prevalence of both heart disease and depression. The potentially meaningful and illuminating links between estrogen (E2) deficiency, cardiovascular disease (CVD), and depression have largely been obscured, first by assertions, subsequently repudiated that the perimenopause was not a time of increased risk of depression, and more recently by the denegration of hormone replacement therapy by initial reports of the Women's Health Initiative. Increasingly, however, research has led to unavoidable conclusions that CVD and depression share common and mediating pathogenic processes and that these same processes are dramatically altered by the presence or absence of E2. This review summarizes data supporting this contention with the intent of placing depression and E2 therapy in their proper physiologic context.
Collapse
Affiliation(s)
- David R Rubinow
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA
| | | |
Collapse
|
17
|
Habauzit D, Flouriot G, Pakdel F, Saligaut C. Effects of estrogens and endocrine-disrupting chemicals on cell differentiation-survival-proliferation in brain: contributions of neuronal cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2011; 14:300-327. [PMID: 21790314 DOI: 10.1080/10937404.2011.578554] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Estrogens and estrogen receptors (ER) are key actors in the control of differentiation and survival and act on extrareproductive tissues such as brain. Thus, estrogens may display neuritogenic effects during development and neuroprotective effects in the pathophysiological context of brain ischemia and neurodegenerative pathologies like Alzheimer's disease or Parkinson's disease. Some of these effects require classical transcriptional "genomic" mechanisms through ER, whereas other effects appear to rely clearly on "membrane-initiated mechanisms" through cytoplasmic signal transduction pathways. Disturbances of these mechanisms by endocrine-disrupting chemicals (EDC) may exert adverse effects on brain. Some EDC may act via ER-independent mechanisms but might cross-react with endogenous estrogen. Other EDC may act through ER-dependent mechanisms and display agonistic/antagonistic estrogenic properties. Because of these potential effects of EDC, it is necessary to establish sensitive cell-based assays to determine EDC effects on brain. In the present review, some effects of estrogens and EDC are described with focus on ER-mediated effects in neuronal cells. Particular attention is given to PC12 cells, an interesting model to study the mechanisms underlying ER-mediated differentiating and neuroprotective effects of estrogens.
Collapse
Affiliation(s)
- Denis Habauzit
- UMR CNRS 6026 (Interactions Cellulaires et Moléculaires, Equipe RED), Université de Rennes 1, Rennes, France
| | | | | | | |
Collapse
|
18
|
Biasi E. Effects of postnatal dietary choline manipulation against MK-801 neurotoxicity in pre- and postadolescent rats. Brain Res 2010; 1362:117-32. [PMID: 20846509 DOI: 10.1016/j.brainres.2010.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 09/03/2010] [Accepted: 09/03/2010] [Indexed: 12/29/2022]
Abstract
Prenatal supplementation of rat dams with dietary choline has been shown to provide their offspring with neuroprotection against N-methyl-d-aspartate (NMDA) antagonist-mediated neurotoxicity. This study investigated whether postnatal dietary choline supplementation exposure for 30 and 60 days of rats starting in a pre-puberty age would also induce neuroprotection (without prenatal exposure). Male and female Sprague-Dawley rats (postnatal day 30 of age) were reared for 30 or 60 concurrent days on one of the four dietary levels of choline: 1) fully deficient choline, 2) 1/3 the normal level, 3) the normal level, or 4) seven times the normal level. After diet treatment, the rats received one injection of MK-801 (dizocilpine 3mg/kg) or saline control. Seventy-two hours later, the rats were anesthetized and transcardially perfused. Their brains were then postfixed for histology with Fluorojade-C (FJ-C) staining. Serial coronal sections were prepared from a rostrocaudal direction from 1.80 to 4.2mm posterior to the bregma to examine cell degeneration in the retrosplenial and piriform regions. MK-801, but not control saline, produced significant numbers of FJ-C positive neurons, indicating considerable neuronal degeneration. Dietary choline supplementation or deprivation in young animals reared for 30-60days did not alter NMDA antagonist-induced neurodegeneration in the retrosplenial region. An interesting finding is the absence of the piriform cortex involvement in young male rats and the complete absence of neurotoxicity in both hippocampus regions and DG. However, neurotoxicity in the piriform cortex of immature females treated for 60days appeared to be suppressed by low levels of dietary choline.
Collapse
Affiliation(s)
- Elisabetta Biasi
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
19
|
Abstract
The focus of this chapter is the relationship between the onset of depression in women and the reproductive events of the menopause transition. Epidemiologic studies have documented that the majority of women do not become depressed during the menopause transition. However, recent longitudinal studies suggest that in some women, the reproductive events related to the menopause transition could play a role in the onset of depression. No abnormality of ovarian hormones has been identified that distinguishes women with depression from those who remain asymptomatic during the menopause transition. Nonetheless, several findings suggest a role of ovarian hormones in the onset of these depressions. First, episodes of depression cluster during the stage of the menopause transition that is accompanied by estradiol withdrawal. Second, randomized controlled trials have documented the short-term (3-6 weeks) antidepressant efficacy of estradiol in depressed perimenopausal women. Third, experimentally induced estradiol withdrawal triggers mood symptoms in some women. Thus, although depression is not a uniform accompaniment of the menopause transition, in some women, age-related changes in ovarian estrogen production may alter central nervous system function and predispose them to develop depression.
Collapse
Affiliation(s)
- Peter J Schmidt
- Behavioral Endocrinology Branch, National Institute of Mental Health, Department of Health & Human Services, Bethesda, Maryland, USA.
| | | |
Collapse
|
20
|
Lanni C, Govoni S, Lucchelli A, Boselli C. Depression and antidepressants: molecular and cellular aspects. Cell Mol Life Sci 2009; 66:2985-3008. [PMID: 19521663 PMCID: PMC11115917 DOI: 10.1007/s00018-009-0055-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 04/28/2009] [Accepted: 05/20/2009] [Indexed: 01/05/2023]
Abstract
Clinical depression is viewed as a physical and psychic disease process having a neuropathological basis, although a clear understanding of its ethiopathology is still missing. The observation that depressive symptoms are influenced by pharmacological manipulation of monoamines led to the hypothesis that depression results from reduced availability or functional deficiency of monoaminergic transmitters in some cerebral regions. However, there are limitations to current monoamine theories related to mood disorders. Recently, a growing body of experimental data has showed that other classes of endogenous compounds, such as neuropeptides and amino acids, may play a significant role in the pathophysiology of affective disorders. With the development of neuroscience, neuronal networks and intracellular pathways have been identified and characterized, describing the existence of the interaction between monoamines and receptors in turn able to modulate the expression of intracellular proteins and neurotrophic factors, suggesting that depression/antidepressants may be intermingled with neurogenesis/neurodegenerative processes.
Collapse
Affiliation(s)
- Cristina Lanni
- Department of Experimental and Applied Pharmacology, Centre of Excellence in Applied Biology, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy
| | - Stefano Govoni
- Department of Experimental and Applied Pharmacology, Centre of Excellence in Applied Biology, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy
| | - Adele Lucchelli
- Department of Experimental and Applied Pharmacology, Centre of Excellence in Applied Biology, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy
| | - Cinzia Boselli
- Department of Experimental and Applied Pharmacology, Centre of Excellence in Applied Biology, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy
| |
Collapse
|
21
|
Marin R, Díaz M, Alonso R, Sanz A, Arévalo MA, Garcia-Segura LM. Role of estrogen receptor alpha in membrane-initiated signaling in neural cells: interaction with IGF-1 receptor. J Steroid Biochem Mol Biol 2009; 114:2-7. [PMID: 19167493 DOI: 10.1016/j.jsbmb.2008.12.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 12/31/2008] [Indexed: 12/25/2022]
Abstract
The mechanisms of action of estradiol in the nervous system involve nuclear-initiated steroid signaling and membrane-initiated steroid signaling. Estrogen receptors (ERs) are involved in both mechanisms. ERalpha interacts with the signaling of IGF-1 receptor in neural cells: ERalpha transcriptional activity is regulated by IGF-1 receptor signaling and estradiol regulates IGF-1 receptor signaling. The interaction between ERalpha and the IGF-1 receptor in the brain may occur at the plasma membrane of neurons and glial cells. Caveolin-1 may provide the scaffolding for the interaction of different membrane-associated molecules, including voltage-dependent anion channel, ERalpha and IGF-I receptor.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Physiology & Institute of Biomedical Technologies, University of La Laguna, School of Medicine, Santa Cruz de Tenerife, Spain
| | | | | | | | | | | |
Collapse
|
22
|
Lipov EG, Joshi JR, Sanders S, Slavin KV. A unifying theory linking the prolonged efficacy of the stellate ganglion block for the treatment of chronic regional pain syndrome (CRPS), hot flashes, and posttraumatic stress disorder (PTSD). Med Hypotheses 2009; 72:657-61. [PMID: 19237252 DOI: 10.1016/j.mehy.2009.01.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 01/08/2009] [Accepted: 01/12/2009] [Indexed: 11/19/2022]
Abstract
The mechanism of action of the stellate ganglion block (SGB) is still uncertain; however it has been used successfully in treatment of chronic regional pain syndrome (CRPS) for many years. Our new insights in to the mechanism of action of the stellate ganglion block were first reported in 2007 in our publication detailing the control of hot flashes with the use of stellate ganglion blockade. We have demonstrated very significant results in the treatment of hot flashes and our most recent application of this block has been for the treatment of posttraumatic stress disorder (PTSD). Stellate ganglion has been demonstrated to have second and third order neurons connections with the central nervous system nuclei that modulate body temperature, neuropathic pain, the manifestations of PTSD, and many other areas. We believe that the commonality between the CRPS, HF and PTSD is the trigger of increased nerve growth factor (NGF) leading to the increase in brain norepinephrine (NR), which in turn is affected by the SGB leading to a prolonged reduction of NGF and eventually a decrease in NR. This, in turn, leads to a reduction or elimination of many of the symptoms of CRPS, Hot flashes, and PTSD.
Collapse
Affiliation(s)
- Eugene G Lipov
- Advanced Pain Centers S.C., 2260 W. Higgins Rd., Ste. 101, Hoffman Estates, IL 60169, USA.
| | | | | | | |
Collapse
|
23
|
Abstract
Epidemiologic studies have documented that the majority of women do not become depressed during the menopause transition. However, recent longitudinal studies suggest that in some women, the events related to the menopause transition could play a role in the onset of depression. In this article we review evidence suggesting a relationship between the menopause transition and depression. Additionally, we describe several findings that suggest a role of ovarian hormones in the onset of these depressions, including the clustering of episodes of depression during the stage of the menopause transition that is accompanied by estradiol withdrawal, and the therapeutic effects of short-term estradiol in depressed perimenopausal women. Finally, we discuss possible causes of affective disturbances during the menopause transition.
Collapse
Affiliation(s)
- Veronica Harsh
- National Institute of Mental Health, Section on Behavioral Endocrinology
| | | | | | - Peter J. Schmidt
- National Institute of Mental Health, Section on Behavioral Endocrinology
| |
Collapse
|
24
|
Anesetti G, Lombide P, Chávez-Genaro R. Prepubertal estrogen exposure modifies neurotrophin receptor expression in celiac neurons and alters ovarian innervation. Auton Neurosci 2009; 145:35-43. [DOI: 10.1016/j.autneu.2008.10.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 10/10/2008] [Accepted: 10/22/2008] [Indexed: 01/19/2023]
|
25
|
Kaur G, Janik J, Isaacson LG, Callahan P. Estrogen regulation of neurotrophin expression in sympathetic neurons and vascular targets. Brain Res 2007; 1139:6-14. [PMID: 17289002 DOI: 10.1016/j.brainres.2006.12.084] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Revised: 12/27/2006] [Accepted: 12/31/2006] [Indexed: 11/26/2022]
Abstract
We hypothesize that estrogen exerts a modulatory effect on sympathetic neurons to reduce neural cardiovascular tone and that these effects are modulated by nerve growth factor (NGF), a neurotrophin that regulates sympathetic neuron survival and maintenance. We examined the effects of estrogen on NGF and tyrosine hydroxylase (TH) protein content in specific vascular targets. Ovariectomized, adult Sprague-Dawley rats were implanted with placebo or 17beta-estradiol (release rate, 0.05 mg/day). Fourteen days later, NGF levels in the superior cervical ganglia (SCG) and its targets, the heart, external carotid artery, and the extracerebral blood vessels, as well as estrogen receptor alpha (ERalpha) content levels in the heart, were determined using semi-quantitative Western blot analysis. TH levels in the SCG and extracerebral blood vessels were determined by Western blotting and immunocytochemistry, respectively. Circulating levels of 17beta-estradiol and prolactin (PRL) were quantified by RIA. Estrogen replacement significantly decreased NGF protein in the SCG and its targets, the external carotid artery, heart and extracerebral blood vessels. TH protein associated with the extracerebral blood vessels was also significantly decreased, but ERalpha levels were significantly increased in the heart following estrogen replacement. These results indicate that estrogen reduces NGF protein content in sympathetic vascular targets, which may lead to decreased sympathetic innervations to these targets, and therefore reduced sympathetic regulation. In addition, the estrogen-induced increase in ERalpha levels in the heart, a target tissue of the SCG, suggests that estrogen may sensitize the heart to further estrogen modulation, and possibly increase vasodilation of the coronary vasculature.
Collapse
Affiliation(s)
- Gurjinder Kaur
- Center for Neuroscience and Behavior, Department of Zoology, Miami University, Oxford, OH 45056, USA.
| | | | | | | |
Collapse
|
26
|
Watson CS, Alyea RA, Hawkins BE, Thomas ML, Cunningham KA, Jakubas AA. Estradiol effects on the dopamine transporter - protein levels, subcellular location, and function. J Mol Signal 2006; 1:5. [PMID: 17224081 PMCID: PMC1769494 DOI: 10.1186/1750-2187-1-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Accepted: 12/05/2006] [Indexed: 01/18/2023] Open
Abstract
Background The effects of estrogens on dopamine (DA) transport may have important implications for the increased incidence of neurological disorders in women during life stages when hormonal fluctuations are prevalent, e.g. during menarche, reproductive cycling, pregnancy, and peri-menopause. Results The activity of the DA transporter (DAT) was measured by the specific uptake of 3H-DA. We found that low concentrations (10-14 to 10-8 M) of 17β-estradiol (E2) inhibit uptake via the DAT in PC12 cells over 30 minutes, with significant inhibition taking place due to E2 exposure during only the last five minutes of the uptake period. Such rapid action suggests a non-genomic, membrane-initiated estrogenic response mechanism. DAT and estrogen receptor-α (ERα) were elevated in cell extracts by a 20 ng/ml 2 day NGFβ treatment, while ERβ was not. DAT, ERα and ERβ were also detectable on the plasma membrane of unpermeabilized cells by immunocytochemical staining and by a fixed cell, quantitative antibody (Ab)-based plate assay. In addition, PC12 cells contained RNA coding for the alternative membrane ER GPR30; therefore, all 3 ER subtypes are candidates for mediating the rapid nongenomic actions of E2. At cell densities above 15,000 cells per well, the E2-induced inhibition of transport was reversed. Uptake activity oscillated with time after a 10 nM E2 treatment; in a slower room temperature assay, inhibition peaked at 9 min, while uptake activity increased at 3 and 20–30 min. Using an Ab recognizing the second extracellular loop of DAT (accessible only on the outside of unpermeabilized cells), our immunoassay measured membrane vs. intracellular/nonvesicular DAT; both were found to decline over a 5–60 min E2 treatment, though immunoblot analyses demonstrated no total cellular loss of protein. Conclusion Our results suggest that physiological levels of E2 may act to sequester DAT in intracellular compartments where the transporter's second extramembrane loop is inaccessible (inside vesicles) and that rapid estrogenic actions on this differentiated neuronal cell type may be regulated via membrane ERs of several types.
Collapse
Affiliation(s)
- Cheryl S Watson
- Department of Biochemistry & Molecular Biology, Univ. of Texas Medical Branch, Galveston TX 77555-0645, USA
| | - Rebecca A Alyea
- Department of Biochemistry & Molecular Biology, Univ. of Texas Medical Branch, Galveston TX 77555-0645, USA
| | - Bridget E Hawkins
- Department of Biochemistry & Molecular Biology, Univ. of Texas Medical Branch, Galveston TX 77555-0645, USA
| | - Mary L Thomas
- Department of Pharmacology & Toxicology, Univ. of Texas Medical Branch, Galveston TX 77555-1031, USA
| | - Kathryn A Cunningham
- Department of Pharmacology & Toxicology, Univ. of Texas Medical Branch, Galveston TX 77555-1031, USA
| | - Adrian A Jakubas
- Department of Biochemistry & Molecular Biology, Univ. of Texas Medical Branch, Galveston TX 77555-0645, USA
| |
Collapse
|
27
|
Bevan CL, Porter DM, Schumann CR, Bryleva EY, Hendershot TJ, Liu H, Howard MJ, Henderson LP. The endocrine-disrupting compound, nonylphenol, inhibits neurotrophin-dependent neurite outgrowth. Endocrinology 2006; 147:4192-204. [PMID: 16777973 DOI: 10.1210/en.2006-0581] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Endocrine-disrupting compounds (EDCs) may interfere with neuronal development due to high levels of accumulation in biological tissue and potentially aberrant steroid signaling. Treatment of dissociated embryonic Xenopus spinal cord neurons with the EDC, nonylphenol (NP), did not alter cell survival or neurite outgrowth but inhibited neurotrophin-induced neurite outgrowth, effects that were recapitulated by treatment with comparable concentrations of 17 beta-estradiol (E2) and beta-estradiol 6-(O-carboxy-methyl)oxime: BSA (E2-BSA), but not a synthetic androgen. Effects of NP were not inhibited by the nuclear estrogen receptor antagonist, ICI 182,780, but were inhibited by the G protein antagonist, pertussis toxin. Nerve growth factor (NGF)-induced neurite outgrowth in Xenopus neurons was shown to require MAPK signaling. NP did not affect TrkA expression, MAPK signaling, or phosphatidylinositol 3' kinase-Akt-glycogen synthase kinase 3 beta (PI3K-Akt-GSK3 beta) signaling in Xenopus. The ability of NP to inhibit NGF-induced neurite outgrowth without altering survival was recapitulated in the rat pheochromocytoma (PC12) cell line. As with Xenopus neurons, the inhibitory actions of NP in PC12 cells were not antagonized by ICI 182,780 and did not involve alterations in signaling along either the MAPK or PI3K-Akt-GSK3 beta pathways. NP did significantly inhibit the ability of NGF to increase protein kinase A activity in this cell line. These data have important implications with respect to potentially deleterious effects of NP exposure during early neural development and highlight the fact that bioaccumulation of EDCs, such as NP, may elicit very disparate effects along divergent signaling pathways than those that arise from the actions of physiological levels of endogenous estrogens.
Collapse
Affiliation(s)
- Cassandra L Bevan
- Department of Physiology, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Dumas J, Hancur-Bucci C, Naylor M, Sites C, Newhouse P. Estrogen treatment effects on anticholinergic-induced cognitive dysfunction in normal postmenopausal women. Neuropsychopharmacology 2006; 31:2065-78. [PMID: 16482084 DOI: 10.1038/sj.npp.1301042] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Estrogen has been shown to interact with the cholinergic system and influence cognition in animal models. This study investigated the interaction of estrogen and cholinergic system functioning and the effects of this interaction on cognitive task performance in healthy older women. Fifteen post-menopausal women were randomly and blindly placed on 1 mg of 17-beta estradiol or placebo for 3 months after which they participated in five anticholinergic challenge sessions, where they were administered one of two doses of the antimuscarinic drug scopolamine (SCOP) or the antinicotinic drug mecamylamine (MECA) or placebo. After the first challenge phase, they were crossed over to the other hormone treatment for another 3 months and repeated the challenges. Performance in multiple domains of cognition was assessed during anticholinergic drug challenge, including attention and verbal and nonverbal learning and memory. Results showed that estrogen pretreatment attenuated the anticholinergic drug-induced impairments on tests of attention and tasks with speed components. This study is the first to demonstrate the interaction of estrogen and the cholinergic system and the effects on cognitive performance in humans. The results suggest that estrogen status may affect cholinergic system tone and may be important for cholinergic system integrity.
Collapse
Affiliation(s)
- Julie Dumas
- Clinical Neuroscience Research Unit, Department of Psychiatry, University of Vermont College of Medicine, Burlington, VT 05401, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
The focus of this chapter is the relationship between the onset of depression in women and the reproductive events of the perimenopause. The perimenopause is a time of considerable variability in reproductive function, which appears in some women to be associated with an increased susceptibility to depression. Elevated gonadotropin levels occur during the late perimenopause, with plasma follicle-stimulating hormone levels increasing in association with impaired ovarian function and decreased estradiol secretion. It is unclear, however, whether the variability in ovarian hormone secretion during the perimenopause has a substantial causal role in the development of depression. Epidemiologic studies have documented that most women do not become depressed during the menopausal transition. However, recent longitudinal studies suggest that in some women, perimenopause-related reproductive events play a role in the onset of depression. Additionally, randomized controlled trials have documented the short-term (3-6 weeks) antidepressant efficacy of estradiol in depressed perimenopausal women. Thus, although depression is not a uniform accompaniment of the menopausal transition, in some women age-related changes in ovarian estrogen production may alter central nervous system function and predispose them to develop depression.
Collapse
Affiliation(s)
- Peter J Schmidt
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bldg. 10-CRC, Rm. 6-5340, 10 Center Dr., MSC 1276, Bethesda, MD 20892-1276, USA.
| |
Collapse
|
30
|
Richeri A, Bianchimano P, Mármol NM, Viettro L, Cowen T, Brauer MM. Plasticity in rat uterine sympathetic nerves: the role of TrkA and p75 nerve growth factor receptors. J Anat 2005; 207:125-34. [PMID: 16050899 PMCID: PMC1571519 DOI: 10.1111/j.1469-7580.2005.00435.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Uterine sympathetic innervation undergoes profound remodelling in response to physiological and experimental changes in the circulating levels of sex hormones. It is not known, however, whether this plasticity results from changes in the innervating neurons, the neuritogenic properties of the target tissue or both. Using densitometric immunohistochemistry, we analysed the effects of prepubertal chronic oestrogen treatment (three subcutaneous injections of 20 microg of beta-oestradiol 17-cypionate on days 25, 27 and 29 after birth), natural peripubertal transition and late pregnancy (19-20 days post coitum) on the levels of TrkA and p75 nerve growth factor receptors in uterine-projecting sympathetic neurons of the thoraco-lumbar paravertebral sympathetic chain (T7-L2) identified using the retrograde tracer Fluorogold. For comparative purposes, levels of TrkA and p75 were assessed in the superior cervical ganglion (SCG) following prepubertal chronic oestrogen treatment. These studies showed that the vast majority of uterine-projecting neurons expressed both TrkA and p75. Both prepubertal chronic oestrogen treatment and the peripubertal transition increased the ratio p75 to TrkA in uterine-projecting neurons, whereas pregnancy elicited the opposite effect. Prepubertal chronic oestrogen treatment had no effects on levels of TrkA or p75 in sympathetic neurons of the SCG. Taken together, our data suggest that neurotrophin receptor-mediated events may contribute to regulate sex hormone-induced plasticity in uterine sympathetic nerves, and are in line with the idea that, in vivo, plasticity in uterine nerves involves changes in both the target and the innervating neurons.
Collapse
Affiliation(s)
- Analía Richeri
- Laboratorio de Biología Celular, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | | | | | | | | | | |
Collapse
|
31
|
Amantea D, Russo R, Bagetta G, Corasaniti MT. From clinical evidence to molecular mechanisms underlying neuroprotection afforded by estrogens. Pharmacol Res 2005; 52:119-32. [PMID: 15967377 DOI: 10.1016/j.phrs.2005.03.002] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Accepted: 03/14/2005] [Indexed: 11/15/2022]
Abstract
Recent studies have highlighted that female sex hormones represent potential neuroprotective agents against damage produced by acute and chronic injuries in the adult brain. Clinical reports have documented the effectiveness of estrogens to attenuate symptoms associated with Parkinson's disease, and to reduce the risk of Alzheimer's disease and cerebrovascular stroke. This evidence is corroborated by numerous experimental studies documenting the protective role of female sex hormones both in vitro and in vivo. Accordingly, estrogens have been shown to promote survival and differentiation of several neuronal populations maintained in culture, and to reduce cell death associated with excitotoxicity, oxidative stress, serum deprivation or exposure to beta-amyloid. The neuroprotective effects of estrogens have been widely documented in animal models of neurological disorders, such as Alzheimer's and Parkinson's diseases, as well as cerebral ischemia. Although estrogens are known to exert several direct effects on neurones, the cellular and molecular mechanisms implicated in their protective actions on the brain are not completely understood. Thus, on the basis of clinical and experimental evidence, in this review, we discuss recent findings concerning the neuronal effects of estrogens that may contribute to their neuroprotective actions. Both estrogen receptor-dependent and -independent mechanisms will be described. These include modulation of cell death regulators, such as Bcl-2, Akt and calpain, as well as interaction with growth factors, such as BDNF, NGF, IGF-I and their receptors. The anti-inflammatory effects of estrogens will also be described, namely their ability to reduce brain levels of inflammatory mediators, cytokines and chemokines. Finally, a brief overview about receptor-independent mechanisms of neuroprotection will aim at describing the antioxidant effects of estrogens, as well as their ability to modulate neurotransmission.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacobiology, University of Calabria, Via P. Bucci, Ed. Polifunzionale, Arcavacata di Rende (CS), Italy
| | | | | | | |
Collapse
|
32
|
Nordell VL, Lewis DK, Bake S, Sohrabji F. The neurotrophin receptor p75NTR mediates early anti-inflammatory effects of estrogen in the forebrain of young adult rats. BMC Neurosci 2005; 6:58. [PMID: 16156894 PMCID: PMC1239918 DOI: 10.1186/1471-2202-6-58] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Accepted: 09/12/2005] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Estrogen suppresses microglial activation and extravasation of circulating monocytes in young animals, supporting an anti-inflammatory role for this hormone. However, the mechanisms underlying estrogen's anti-inflammatory effects, especially in vivo, are not well understood. The present study tests the hypothesis that anti-inflammatory effects of estrogen are mediated by the pan-neurotrophin receptor p75NTR. Previously, we reported that estrogen attenuated local increases of interleukin(IL)-1beta in the NMDA-lesioned olfactory bulb, while further increasing NGF expression. RESULTS The present studies show that this lesion enhances expression of the neurotrophin receptor p75NTR at the lesion site, and p75NTR expression is further enhanced by estrogen treatment to lesioned animals. Specifically, estrogen stimulates p75NTR expression in cells of microvessels adjacent to the lesion site. To determine the role of this receptor in mediating estrogen's anti-inflammatory effects, a p75NTR neutralizing antibody was administered at the same time the lesion was created (by stereotaxic injections of NMDA) and specific markers of the inflammatory cascade were measured. Olfactory bulb injections of NMDA+vehicle (preimmune serum) increased IL-1beta and activated the signaling molecule c-jun terminal kinase (JNK)-2 at 6 h. At 24 h, the lesion significantly increased matrix metalloproteinase (MMP)-9 and prostaglandin (PG)E2, a COX-2 mediated metabolite of arachadonic acid. All of these markers were significantly attenuated by estrogen in a time-dependent manner. However, estrogen's effects on all these markers were abolished in animals that received anti-p75NTR. CONCLUSION These data support the hypothesis that estrogen's anti-inflammatory effects may be, in part, mediated by this neurotrophin receptor. In view of the novel estrogen-dependent expression of p75NTR in cells associated with microvessels, these data also suggest that the blood brain barrier is a critical locus of estrogen's neuro-immune effects.
Collapse
Affiliation(s)
- Vanessa L Nordell
- Department of Human Anatomy and Medical Neurobiology, Texas A&M University System Health Science Center College of Medicine, College Station, TX, 77843 USA
| | - Danielle K Lewis
- Department of Human Anatomy and Medical Neurobiology, Texas A&M University System Health Science Center College of Medicine, College Station, TX, 77843 USA
| | - Shameena Bake
- Department of Human Anatomy and Medical Neurobiology, Texas A&M University System Health Science Center College of Medicine, College Station, TX, 77843 USA
| | - Farida Sohrabji
- Department of Human Anatomy and Medical Neurobiology, Texas A&M University System Health Science Center College of Medicine, College Station, TX, 77843 USA
| |
Collapse
|
33
|
Hasan W, Smith HJ, Ting AY, Smith PG. Estrogen alters trkA and p75 neurotrophin receptor expression within sympathetic neurons. ACTA ACUST UNITED AC 2005; 65:192-204. [PMID: 16118792 DOI: 10.1002/neu.20183] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Survival and growth of sympathetic neurons is regulated by nerve growth factor acting through trkA and p75NTR receptors. Sympathetic neurons are also affected by gonadal steroid hormones, particularly estrogen. To determine if estrogen may influence sympathetic neurons via altered neurotrophin receptor expression, we investigated effects of acute or chronic estrogen administration on levels of trkA and p75NTR proteins, numbers of immunoreactive neurons, and numbers of neurons expressing trkA, p75NTR, and estrogen receptor-alpha transcripts. Superior cervical ganglia from ovariectomized or estradiol-treated rats were processed for in situ hybridization or immunohistochemistry, and percentages of stained neurons quantitated or processed for Western blot analysis. In ovariectomized rats, approximately 50% of sympathetic neurons expressed trkA mRNA and protein. Acute estrogen administration did not affect trkA transcript expression, but reduced trkA protein significantly. Chronic treatment did not alter neuronal trkA expression. Approximately 70% of sympathetic neurons in ovariectomized rats expressed p75NTR transcripts and about 50% showed p75NTR immunoreactivity. Acute estrogen did not affect p75NTR expression. However, chronic estrogen reduced p75NTR mRNA and protein expression significantly. Fifty to sixty percent of sympathetic neurons in ovariectomized rats displayed estrogen receptor-alpha mRNA. After acute estrogen administration, estrogen receptor-alpha transcript expression increased by 35%, although this was not maintained chronically. These findings indicate that estrogen can influence sympathetic neuronal neurotrophin receptor expression as well as estrogen receptor-alpha. Reduced trkA expression after acute estrogen may transiently predispose neurons to degenerative events, while diminished p75NTR expression by chronic estrogen administration may exert long-term effects on survival or axonal outgrowth in sympathetic neurons.
Collapse
Affiliation(s)
- Wohaib Hasan
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Mail Stop 3051, Kansas City, Kansas 66160, USA.
| | | | | | | |
Collapse
|
34
|
Cheema ZF, Santillano DR, Wade SB, Newman JM, Miranda RC. The extracellular matrix, p53 and estrogen compete to regulate cell-surface Fas/Apo-1 suicide receptor expression in proliferating embryonic cerebral cortical precursors, and reciprocally, Fas-ligand modifies estrogen control of cell-cycle proteins. BMC Neurosci 2004; 5:11. [PMID: 15038834 PMCID: PMC395829 DOI: 10.1186/1471-2202-5-11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2003] [Accepted: 03/23/2004] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Apoptosis is important for normal cerebral cortical development. We previously showed that the Fas suicide receptor was expressed within the developing cerebral cortex, and that in vitro Fas activation resulted in caspase-dependent death. Alterations in cell-surface Fas expression may significantly influence cortical development. Therefore, in the following studies, we sought to identify developmentally relevant cell biological processes that regulate cell-surface Fas expression and reciprocal consequences of Fas receptor activation. RESULTS Flow-cytometric analyses identified two distinct neural sub-populations that expressed Fas on their cell surface at high (FasHi) or moderate (FasMod) levels. The anti-apoptotic protein FLIP further delineated a subset of Fas-expressing cells with potential apoptosis-resistance. FasMod precursors were mainly in G0, while FasHi precursors were largely apoptotic. However, birth-date analysis indicated that neuroblasts express the highest levels of cell-surface Fas at the end of S-phase, or after their final round of mitosis, suggesting that Fas expression is induced at cell cycle checkpoints or during interkinetic nuclear movements. FasHi expression was associated with loss of cell-matrix adhesion and anoikis. Activation of the transcription factor p53 was associated with induction of Fas expression, while the gonadal hormone estrogen antagonistically suppressed cell-surface Fas expression. Estrogen also induced entry into S-phase and decreased the number of Fas-expressing neuroblasts that were apoptotic. Concurrent exposure to estrogen and to soluble Fas-ligand (sFasL) suppressed p21/waf-1 and PCNA. In contrast, estrogen and sFasL, individually and together, induced cyclin-A expression, suggesting activation of compensatory survival mechanisms. CONCLUSIONS Embryonic cortical neuronal precursors are intrinsically heterogeneous with respect to Fas suicide-sensitivity. Competing intrinsic (p53, cell cycle, FLIP expression), proximal (extra-cellular matrix) and extrinsic factors (gonadal hormones) collectively regulate Fas suicide-sensitivity either during neurogenesis, or possibly during neuronal migration, and may ultimately determine which neuroblasts successfully contribute neurons to the differentiating cortical plate.
Collapse
Affiliation(s)
- Zulfiqar F Cheema
- Department of Human Anatomy & Medical Neurobiology, & Center for Environmental and Rural Health, 228 Reynolds Medical Bldg., Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA
- Department of General Surgery, William Beaumont Hospital, Royal Oaks, MI 48073, USA
| | - Daniel R Santillano
- Department of Human Anatomy & Medical Neurobiology, & Center for Environmental and Rural Health, 228 Reynolds Medical Bldg., Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA
| | - Stephen B Wade
- Department of Human Anatomy & Medical Neurobiology, & Center for Environmental and Rural Health, 228 Reynolds Medical Bldg., Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA
- Pediatrics, Wake Forest University Baptist Medical Center, Medical Center Boulevard, Winston Salem, NC 27157, USA
| | - Joseph M Newman
- Department of Human Anatomy & Medical Neurobiology, & Center for Environmental and Rural Health, 228 Reynolds Medical Bldg., Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA
| | - Rajesh C Miranda
- Department of Human Anatomy & Medical Neurobiology, & Center for Environmental and Rural Health, 228 Reynolds Medical Bldg., Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA
| |
Collapse
|
35
|
Tinkler GP, Tobin JR, Voytko ML. Effects of two years of estrogen loss or replacement on nucleus basalis cholinergic neurons and cholinergic fibers to the dorsolateral prefrontal and inferior parietal cortex of monkeys. J Comp Neurol 2004; 469:507-21. [PMID: 14755532 DOI: 10.1002/cne.11028] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The present study examined the long-term (2 years) effects of estrogen loss or estrogen replacement therapy (ERT) on cholinergic neurons in the nucleus basalis of Meynert and on cholinergic fibers in the prefrontal and parietal cortex of adult female cynomolgus monkeys. Cholinergic fiber density in layer II of the prefrontal cortex was decreased in monkeys who were ovariectomized and treated with placebo for 2 years. In contrast, ovariectomized monkeys receiving ERT for 2 years had fiber densities that were comparable to those of intact controls. No differences in parietal cholinergic fiber density or nucleus basalis cholinergic neuron number or volume were found among intact, ovariectomized, or ERT monkeys. Our results suggest that ERT is effective in preventing region-specific changes in cortical cholinergic fibers that result from the loss of circulating ovarian hormones. These modest but appreciable effects on cholinergic neurobiology following long-term estrogen loss and ERT may contribute to changes in visuospatial attention function that is mediated by the prefrontal cortex.
Collapse
Affiliation(s)
- Gregory Paul Tinkler
- Interdisciplinary Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | |
Collapse
|
36
|
Pongrac JL, Gibbs RB, Defranco DB. Estrogen-mediated regulation of cholinergic expression in basal forebrain neurons requires extracellular-signal-regulated kinase activity. Neuroscience 2004; 124:809-16. [PMID: 15026121 DOI: 10.1016/j.neuroscience.2004.01.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2004] [Indexed: 11/18/2022]
Abstract
Beyond the role estrogen plays in neuroendocrine feedback regulation involving hypothalamic neurons, other roles for estrogen in maintaining the function of CNS neurons remains poorly understood. Primary cultures of embryonic rat neurons together with radiometric assays were used to demonstrate how estrogen alters the cholinergic phenotype in basal forebrain by differentially regulating sodium-coupled high-affinity choline uptake and choline acetyltransferase activity. High-affinity choline uptake was significantly increased 37% in basal forebrain cholinergic neurons grown in the presence of a physiological dose of estrogen (5 nM) from 4 to 10 days in vitro whereas choline acetyltransferase activity was not significantly changed in the presence of 5 or 50 nM estrogen from 4 to 10 or 10 to 16 days in vitro. Newly-synthesized acetylcholine was significantly increased 35% following 6 days of estrogen treatment (10 days in vitro). These effects are in direct contrast to those found for nerve growth factor; that is, nerve growth factor can enhance the cholinergic phenotype through changes in choline acetyltransferase activity alone. This is most surprising given that mitogen-activated protein kinase and extracellular-signal-regulated kinase1/2, kinases also activated in the signaling pathway of nerve growth factor, were found to participate in the estrogen-mediated changes in the cholinergic phenotype. Likewise, general improvement in the viability of the cultures treated with estrogen does not account for the effects of estrogen as determined by lactate dehydrogenase release and nerve growth factor-responsiveness. These findings provide evidence that estrogen enhances the differentiated phenotype in basal forebrain cholinergic neurons through second messenger signaling in a manner distinct from nerve growth factor and independent of improved survival.
Collapse
Affiliation(s)
- J L Pongrac
- Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | |
Collapse
|
37
|
MacLusky NJ, Chalmers-Redman R, Kay G, Ju W, Nethrapalli IS, Tatton WG. Ovarian steroids reduce apoptosis induced by trophic insufficiency in nerve growth factor-differentiated PC12 cells and axotomized rat facial motoneurons. Neuroscience 2003; 118:741-54. [PMID: 12710981 DOI: 10.1016/s0306-4522(02)00940-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Previous studies have demonstrated that ovarian steroids exert neuroprotective effects in a variety of in vitro and in vivo systems. The mechanisms underlying these effects remain poorly understood. In the present study, the neuroprotective effects of estradiol (E(2)) and progesterone (P) were examined in two models of apoptosis induced by growth factor insufficiency: partially nerve growth factor (NGF)-differentiated PC12 cells, after serum and NGF withdrawal; and axotomized immature rat facial motor motoneurons. E(2) and P both increased the survival of trophically withdrawn NGF-differentiated PC12 cells, at physiologically relevant concentrations. However, neither steroid had a significant effect on the survival of PC12 cells that had not been NGF treated. Exposure to NGF had no effect on the expression of estrogen receptor (ER)beta, but markedly increased the levels of ERalpha and altered the expression of the progesterone receptor (PR) from predominantly PR-B in NGF naive cells, to predominantly PR-A after NGF. The survival promoting effects of E(2) and P were blocked by the specific steroid receptor antagonists Faslodex (ICI 182780) and onapristone (ZK98299), respectively. Inhibitors of RNA (actinomycin D) or protein (cycloheximide) synthesis also abrogated the protective effects of both steroids. In immature rats, E(2) and P both significantly increased the numbers of surviving facial motor neurons at 21 days after axotomy. These data demonstrate significant protective effects of E(2) and P in two well-characterized models of apoptosis induced by trophic withdrawal and suggest that, at least in PC12 cells, the effects of the steroids are mediated via interaction with nuclear steroid receptor systems. The lack of steroid responsiveness in NGF-naive PC12 cells despite the presence of abundant ERbeta and PR-B are consistent with the view that ERalpha and PR-A may be particularly important as mediators of the neuroprotective effects of their corresponding hormonal ligands.
Collapse
Affiliation(s)
- N J MacLusky
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia-Presbyterian Medical Center, 622 West 168th Street, New York, NY 10032-3702, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Bjorling DE, Beckman M, Saban R. Neurogenic inflammation of the bladder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 539:551-83. [PMID: 15176313 DOI: 10.1007/978-1-4419-8889-8_37] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Current evidence suggests multiple and redundant pathways through which the nervous system can initiate, amplify, and perpetuate inflammation. Many of the processes initiated by neurogenic inflammation have the capacity to recruit the participation of additional sensory nerves. These observations indicate that effective strategies for prevention or treatment of neurogenic inflammation of the bladder will entail or require intervention at multiple points. It has been observed that pain management in the future will be based on selective intervention tailored to the specific processes modulating pain perception in individual patients. It is exciting to contemplate the same approach to prevention and treatment of neurogenic bladder inflammation.
Collapse
Affiliation(s)
- Dale E Bjorling
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin, USA
| | | | | |
Collapse
|
39
|
Papka RE, Mowa CN. Estrogen Receptors in the Spinal Cord, Sensory Ganglia, and Pelvic Autonomic Ganglia. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 231:91-127. [PMID: 14713004 DOI: 10.1016/s0074-7696(03)31003-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Until relatively recently, most studies of the effects of estradiol in the nervous system focused on hypothalamic, limbic, and other brain centers involved in reproductive hormone output, feedback, and behaviors. Almost no studies addressed estradiol effects at the spinal cord or peripheral nervous system level. Prior to the mid-1960s-1970s, few studies examined neural components of reproductive endocrine organs (e.g., ovary or testis) or the genital organs (e.g., uterus or penis) because available data supported endocrine regulation of these structures. Over the last two decades interest in and studies on the innervation of the genital organs have burgeoned. Because of the responsiveness of genital organs to sex steroid hormones, these neural studies seeded interest in whether or not autonomic and sensory neurons that innervate these organs, along with their attendant spinal cord circuits, also are responsive to sex hormones. From the mid-1980s there has been a steady growth of interest in, and studies of the neuroanatomy, neurochemistry, neural connectivity, and neural functional aspects in reproductive organs and the response of these parameters to sex steroids. Thus, with the growth of probes and techniques, has come studies of anatomy, neurochemistry, and circuitry of sex hormone-responsive neurons and circuits in the spinal cord and peripheral nervous system. This review focuses on estrogen receptors in sensory, autonomic, and spinal cord neurons in locales that are associated with innervation of female reproductive organs.
Collapse
Affiliation(s)
- R E Papka
- Department of Neurobiology, Northeastern Ohio Universities College of Medicine, Rootstown, Ohio 44272, USA
| | | |
Collapse
|
40
|
Carrer HF, Cambiasso MJ. Sexual differentiation of the brain: genes, estrogen, and neurotrophic factors. Cell Mol Neurobiol 2002; 22:479-500. [PMID: 12585676 DOI: 10.1023/a:1021825317546] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Based on evidence obtained during the past 50 years, the current hypothesis to explain the sexual dimorphism of structure and function in the brain of vertebrates maintains that these differences are produced by the epigenetic action of gonadal hormones. However, evidence has progressively accumulated suggesting that genetic mechanisms controlling sexual-specific neuronal characteristics precede, or occur in parallel with, hormonal effects. 1. In cultures of hypothalamic neurons taken from gestation day 16 (GD16) embryos, treatment of sexually segregated cultures with estradiol (E2) induces axon growth in neurons from male neurons, but not from female neurons. In these cultures treatment with E2 increased the levels of tyrosine kinase type B (TrkB) and insulin-like growth factor I (IGF-I) receptors in male but not in female neurons. This and other sex differences cannot be explained by differences in hormonal environment, because the donor embryos were obtained when gonadal secretion of steroids is just beginning, before the perinatal surge of testosterone that determines development of the male brain beginning at GD17/18. 2. The response to estrogen is contingent upon coculture with heterotopic glia (mostly astrocytes) from a target region (amygdala) harvested from same-sex fetuses at GD16, whereas in the presence of homotopic glia or in cultures without glia, E2 had no effect. It was concluded that the axogenic effect of E2 depends on interaction between neurons and glia from a target region and that neurons from fetal male donors appear to mature earlier than neurons from females, a differentiated response that takes place prior to divergent exposure to gonadal secretions. 3. The effects of target and nontarget glia-conditioned media (CM) on the E2-induced growth of neuronal processes of hypothalamic neurons obtained from sexually segregated fetal donors were also studied. Estrogen added to media conditioned by target glia modified the number of primary neurites and the growth of axons of hypothalamic neurons of males but not of females. 4. Neither the Type III steroidal receptor blocker tamoxifen nor Type I antiestrogen ICI 182,780 prevented the axogenic effects of the hormone. Estradiol made membrane-impermeable by conjugation to a protein of high molecular weight (E2-BSA) preserved its axogenic capacity, suggesting the possibility of a membrane effect responsible for the action of E2. 5. Western blot analysis of the tyrosine kinase type A (TrkA), type B (TrkB), type C (TrkC), and insulin-like growth factor (IGF-I R) receptors in extracts from homogenates of cultured hypothalamic neurons showed that in cultures of male-derived neurons grown with E2 and CM from target glia, the amounts of TrkB and IGF-I R increased notably. Densitometric quantification showed that these cultures had more TrkB than cultures with CM alone or E2 alone. On the contrary, in cultures of female-derived neurons, the presence of CM alone induced maximal levels of TrkB, which were not further increased by E2; female-derived neurons in all conditions did not contain IGF-I R. Levels of TrkC were not modified by any experimental condition in male- or female-derived cultures and Trk A was not found in the homogenates. These results are compared with similar data from other laboratories and integrated in a model for the confluent interaction of estrogen and neurotrophic factors released by glia that may contribute to the sexual differentiation of the brain.
Collapse
Affiliation(s)
- Hugo F Carrer
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Casilla de Correo 389, Córdoba 5000, Argentina.
| | | |
Collapse
|
41
|
Krizsan-Agbas D, Smith PG. Estrogen modulates myometrium-induced sympathetic neurite formation through actions on target and ganglion. Neuroscience 2002; 114:339-47. [PMID: 12204203 DOI: 10.1016/s0306-4522(02)00262-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Estrogen induces rapid and extensive degeneration of rodent uterine myometrial sympathetic innervation. To clarify the underlying mechanisms, we used explant cultures to assess whether estrogen affects the myometrium's ability to induce sympathetic neuritogenesis and the sympathetic neuron's ability to respond. Superior cervical ganglion explants from ovariectomized adult donors extended neurites when cultured alone in serum-free medium, and their numbers increased 2.3-fold in the presence of myometrial explants from ovariectomized adult rats. The myometrium's ability to induce neuritogenesis was abolished by injection of myometrium donors with 17beta-estradiol 24 h prior to tissue harvest. Myometrial neurite-promoting effects were also abolished by adding 2x10(-8)M estradiol to the culture medium. Because outgrowth from ganglia of ovariectomized rats cultured alone was not affected by estrogen in the culture medium, this indicates that estrogen acts directly on myometrium to abrogate its neurite-promoting effects. However, estrogen injection of ganglion donor rats also inhibited neurite extension toward ovariectomized myometrium, suggesting that some factor in ovariectomized rats normally acts on the ganglion to prevent estrogen from inhibiting neurite outgrowth. When ganglia from hypophysectomized ovariectomized donors were cultured alone, neuritogenesis was normal but estrogen added to the culture medium now attenuated outgrowth. Prolactin but not other pituitary-derived hormones reversed the suppression of neuritogenesis induced by estrogen. We conclude that estrogen acts directly on myometrium to inhibit its neuritogenic effects on sympathetic neurons. Estrogen can also attenuate neurite formation by acting directly on the ganglion; this effect normally is not apparent at low estrogen levels because a pituitary hormone (possibly prolactin) prevents the ganglion from responding fully to estrogen. With high in vivo estrogen, this pituitary hormone's effects are abated, possibly through diminished release, and estrogen directly reduces ganglion neuritogenesis. Thus, estrogen regulates uterine sympathetic nerve remodeling through actions on myometrium, ganglion, and intermediary pituitary factors.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cells, Cultured
- Coculture Techniques
- Culture Media, Conditioned/pharmacology
- Estrogens/metabolism
- Estrogens/pharmacology
- Estrous Cycle/metabolism
- Feedback/drug effects
- Feedback/physiology
- Female
- Ganglia, Sympathetic/cytology
- Ganglia, Sympathetic/drug effects
- Ganglia, Sympathetic/growth & development
- Myometrium/innervation
- Myometrium/metabolism
- Nerve Degeneration/chemically induced
- Nerve Degeneration/metabolism
- Neurites/drug effects
- Neurites/metabolism
- Neurites/ultrastructure
- Neuronal Plasticity/drug effects
- Neuronal Plasticity/physiology
- Ovariectomy
- Pituitary Gland, Anterior/metabolism
- Pituitary Hormones/metabolism
- Pituitary Hormones/pharmacology
- Rats
- Rats, Sprague-Dawley
- Sympathetic Fibers, Postganglionic/cytology
- Sympathetic Fibers, Postganglionic/drug effects
- Sympathetic Fibers, Postganglionic/growth & development
Collapse
Affiliation(s)
- D Krizsan-Agbas
- Department of Molecular and Integrative Physiology, R.L. Smith Mental Retardation Research Center, Kansas University Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160-7401, USA
| | | |
Collapse
|
42
|
Isoda H, Talorete TPN, Kimura M, Maekawa T, Inamori Y, Nakajima N, Seki H. Phytoestrogens genistein and daidzin enhance the acetylcholinesterase activity of the rat pheochromocytoma cell line PC12 by binding to the estrogen receptor. Cytotechnology 2002; 40:117-23. [PMID: 19003112 PMCID: PMC3449519 DOI: 10.1023/a:1023903220539] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Some compounds derived from plants have been known to possess estrogenic properties and can thus alter the physiology of higher organisms. Genistein and daidzin are examples of these phytoestrogens, which have recently been the subject of extensive research. In this study, genistein and daidzin were found to enhance the acetylcholinesterase (AChE) activity of the rat neuronal cell line PC12 at concentrations as low as 0.08 muM by binding to the estrogen receptor (ER). Results have shown that this enhancement was effectively blocked by the known estrogen receptor antagonist tamoxifen, indicating the involvement of the ER in AChE induction. That genistein and daidzin are estrogenic were confirmed in a cell proliferation assay using the human breast cancer cell line MCF7. This proliferation was also blocked by tamoxifen, again indicating the involvement of the ER. On the other hand, incubating the PC12 cells in increasing concentrations of 17 beta-estradiol (E2) did not lead to enhanced AChE activity, even in the presence of genistein or daidzin. This suggests that mere binding of an estrogenic compound to the ER does not necessarily lead to enhanced AChE activity. Moreover, the effect of the phytoestrogens on AChE activity cannot be expressed in the presence of E2 since they either could not compete with the natural ligand in binding to the ER or that E2 down-regulates its own receptor. This study clearly suggests that genistein and daidzin enhance AChE activityin PC12 cells by binding to the ER; however, the actual mechanism of enhancement is not known.
Collapse
Affiliation(s)
- Hiroko Isoda
- National Institute of Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Cossette LJ, Gaumond I, Martinoli MG. Combined effect of xenoestrogens and growth factors in two estrogen-responsive cell lines. Endocrine 2002; 18:303-8. [PMID: 12450323 DOI: 10.1385/endo:18:3:303] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It is now well recognized that estrogenic signaling mechanisms are far more complex than once thought. Several crosstalks between the estrogen receptor and other signaling pathways may influence the estrogenic stimulation of cell growth. Thus, the estrogenic effects of several environmental contaminants, now suspected to act as endocrine disrupters, may be influenced by a simultaneous stimulation of other signaling pathways. The aim of this study was to investigate whether the growth response of two estrogen-responsive cell lines, MCF-7 and GH3, treated with xenoestrogens might be affected by the addition of growth factors to their culture medium. Cells were treated with two known xenoestrogens, endosulfan and chlordane, alone or in the presence of insulin-like growth factor-1 and epidermal growth factor, respectively, and their growth was measured using the 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxyanilide proliferation assay. Our results show that treatment with endosulfan or chlordane as well as treatment with growth factors increased cell growth, while the administration of xenoestrogens together with growth factors triggered a partly additive response with no antagonist or synergistic effect. These results sustain a role for xenoestrogens in cellular growth.
Collapse
Affiliation(s)
- Louis J Cossette
- Department of Biochemistry, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | | | | |
Collapse
|
44
|
McMillan PJ, LeMaster AM, Dorsa DM. Tamoxifen enhances choline acetyltransferase mRNA expression in rat basal forebrain cholinergic neurons. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 103:140-5. [PMID: 12106699 DOI: 10.1016/s0169-328x(02)00195-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Novel estrogen-like molecules known as SERMs (selective estrogen receptor modulators) produce many of the beneficial estrogen-like actions without the detrimental side-effects. The SERM, tamoxifen, an estrogen-like molecule with both agonist and antagonist properties, is widely prescribed for the treatment of breast cancer. While the effects of tamoxifen are being evaluated in many peripheral tissues, its effects in the central nervous system (CNS) have been largely ignored. In the present study, we begin to evaluate the effects of tamoxifen in the rat basal forebrain, a region known to be highly responsive to estrogen. We compared the effects of short-term (24 h) tamoxifen treatment to that of estrogen on ChAT mRNA expression in cholinergic neurons. In addition, we examined the effect of tamoxifen in the presence and absence of estrogen. Our results indicate that tamoxifen enhances ChAT expression in a manner similar to that of estrogen in several basal forebrain regions. In contrast, tamoxifen exhibits antagonist properties with respect to estrogen-induction of progesterone receptor mRNA in the medial preoptic nucleus. These results indicate tamoxifen has estrogenic properties with respect to cholinergic neurons, suggesting a previously unidentified effect of this agent in the CNS.
Collapse
Affiliation(s)
- Pamela J McMillan
- Departments of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA.
| | | | | |
Collapse
|
45
|
Wilson ME, Liu Y, Wise PM. Estradiol enhances Akt activation in cortical explant cultures following neuronal injury. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 102:48-54. [PMID: 12191493 DOI: 10.1016/s0169-328x(02)00181-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have previously demonstrated that estradiol reduces cell death in cortical explant cultures following injury induced by metabolic inhibition in a receptor-dependent fashion. In this study, we examined whether cell death involves apoptosis and assessed the potential mediators of estradiol's actions. Cortical explant cultures were generated from postnatal day 3 rat pups. On day 7 in vitro, explants were injured by exposure to 1 mM 2-DG/2 mM KCN for 2 h to model the metabolic inhibition observed during ischemia. Explants were fixed in 4% paraformaldehyde at 2, 6, 10 and 24 h following the injury period and 18-microm thick sections were cut on a cryostat and stained with cresyl violet to assess cell death. The same sections were also labeled by TUNEL to determine whether cell death occurred by apoptosis. Other sections were used for immunohistochemistry to determine whether cells that stained positive for activated caspase 3 were also immunopositive for NeuN, a neuronal marker, or GFAP, an astrocyte marker. Protein was extracted for Western blot analysis from a separate set of explants collected at 0, 0.5, 1, 2 and 4 h following the conclusion of the injury. Estradiol treatment significantly reduced the number of cells undergoing apoptotic cell death as indicated by nuclear condensation visualized by cresyl violet staining (P<0.05). TUNEL staining revealed that the majority of pyknotic and fragmented nuclei were also TUNEL positive. Furthermore, caspase 3 activation appeared to be restricted to neurons. To examine a possible mechanism by which estradiol prevents apoptosis, we examined the level of activation of Akt kinase, which mediates antiapoptotic signals. Potential activation was measured by phosphorylation of Akt at Ser473 by Western blot analysis. In the absence of estradiol, pAkt levels were significantly increased at 2 h following the termination of injury. Explants that were pretreated with estradiol exhibited elevated levels of pAkt at 1 h following injury. Treatment with ICI 182,780 prevented the effect of estradiol. These studies suggest that estradiol prevents injury-induced apoptosis and that Akt activation may mediate these protective effects.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536, USA.
| | | | | |
Collapse
|
46
|
Bjorling DE, Beckman M, Clayton MK, Wang ZY. Modulation of nerve growth factor in peripheral organs by estrogen and progesterone. Neuroscience 2002; 110:155-67. [PMID: 11882380 DOI: 10.1016/s0306-4522(01)00568-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nerve growth factor (NGF) synthesized in peripheral organs plays a critical role in the development and maintenance of the nervous system and also participates in processing nociceptive stimuli. Previous studies suggest that reproductive hormones may regulate the expression of NGF. Ovariectomies were performed on female mice, and mice were killed 24 h after hormone replacement to evaluate the effects of estrogen and progesterone on NGF in peripheral organs, specifically the uterus, bladder, heart, and salivary gland. Sham-operated intact mice and untreated ovariectomized mice served as controls. Immunohistochemistry demonstrated the presence of NGF, estrogen receptor-alpha, estrogen receptor-beta, and progesterone receptors in these organs. Ovariectomy caused a significant decrease in NGF protein content in the uterus, and short term treatment of ovariectomized mice with estrogen and/or progesterone increased uterine NGF mRNA and restored NGF protein to concentrations similar to intact control mice. Ovariectomy did not affect NGF protein concentrations in the salivary gland, but treatment of ovariectomized mice with estrogen alone or in conjunction with progesterone stimulated concentrations of NGF protein that exceeded those observed in intact control or ovariectomized, untreated mice. NGF mRNA was increased in salivary glands from ovariectomized mice treated with progesterone alone or in combination with estrogen relative to other groups. NGF protein content of the hearts of ovariectomized mice treated with estrogen alone or in conjunction with progesterone was increased relative to intact controls and ovariectomized, untreated mice, but neither ovariectomy or hormone replacement affected NGF mRNA content in the heart. NGF protein content of the bladder was unaffected by ovariectomy or hormone treatment, and bladder NGF mRNA was unaffected by ovariectomy or hormone treatment. Collectively, these results indicate that reproductive hormones have the capacity to regulate NGF message and protein in a manner that varies among organs. Fluctuations in the expression of NGF, in conjunction with other factors, may help to explain gender differences in pain sensation and inflammatory response.
Collapse
Affiliation(s)
- D E Bjorling
- Department of Surgical Sciences, School of Veterinary Medicine, The University of Wisconsin, 2015 Linden Drive West, Madison, WI 53706, USA.
| | | | | | | |
Collapse
|
47
|
Estrogen regulates the development of brain-derived neurotrophic factor mRNA and protein in the rat hippocampus. J Neurosci 2002. [PMID: 11923430 DOI: 10.1523/jneurosci.22-07-02650.2002] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During development, estrogen has a variety of effects on morphological and electrophysiological properties of hippocampal neurons. Brain-derived neurotrophic factor (BDNF) also plays an important role in the survival and differentiation of neurons during development. We examined the effects of gonadectomy with and without estrogen replacement on the mRNA and protein of BDNF and its receptor, trkB, during early postnatal development of the rat hippocampus. We used immunocytochemistry to demonstrate that estrogen receptor alpha (ERalpha) and BDNF were localized to the same cells within the developing hippocampus. BDNF and ERalpha were colocalized in pyramidal cells of the CA3 subregion and to a lesser extent in CA1. To determine whether BDNF mRNA was regulated by estrogen during development, we gonadectomized male rat pups at postnatal day 0 (P0) and examined mRNA and protein levels from P0 to P25 using real-time reverse transcription-PCR and Western blot analysis. After gonadectomy, BDNF mRNA levels are significantly reduced on P7, but after treatment of gonadectomized animals with estradiol benzoate on P0, levels at all ages were similar to those in intact animals. BDNF mRNA changes after gonadectomy are accompanied by an increase in the levels of BDNF protein, which were reduced by estrogen treatment at P0. We also examined the effect of postnatal estrogen treatment on trkB. There were no significant changes in trkB mRNA or protein in gonadectomized or estrogen-replaced animals. These results suggest that a direct interaction may exist between ERalpha and BDNF to alter hippocampal physiology during development in the rat.
Collapse
|
48
|
Estrogen protects against global ischemia-induced neuronal death and prevents activation of apoptotic signaling cascades in the hippocampal CA1. J Neurosci 2002. [PMID: 11896151 DOI: 10.1523/jneurosci.22-06-02115.2002] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The importance of postmenopausal estrogen replacement therapy in affording protection against the selective and delayed neuronal death associated with cardiac arrest or cardiac surgery in women remains controversial. Here we report that exogenous estrogen at levels that are physiological for hormone replacement in postmenopausal women affords protection against global ischemia-induced neuronal death and prevents activation of apoptotic signaling cascades in the hippocampal CA1 of male gerbils. Global ischemia induced a marked increase in activated caspase-3 in CA1, evident at 6 hr after ischemia. Global ischemia induced a marked upregulation of the proapoptotic neurotrophin receptor p75(NTR) in CA1, evident at 48 hr. p75(NTR) expression was induced primarily in terminal deoxynucleotidyl transferase-mediated UTP nick-end labeling-positive cells, indicating expression in neurons undergoing apoptosis. Global ischemia also induced a marked downregulation of mRNA encoding the AMPA receptor GluR2 subunit in CA1. Caspase-3, p75(NTR), and GluR2 were not significantly changed in CA3 and dentate gyrus, indicating that the ischemia-induced changes in gene expression were cell specific. Exogenous estrogen attenuated the ischemia-induced increases in activated caspase-3 and blocked the increase in p75(NTR) in post-ischemic CA1 neurons but did not prevent ischemia-induced downregulation of GluR2. These findings demonstrate that long-term estrogen at physiological levels ameliorates ischemia-induced hippocampal injury and indicate that estrogen intervenes at the level of apoptotic signaling cascades to prevent onset of death in neurons otherwise "destined to die."
Collapse
|
49
|
Gollapudi L, Oblinger MM. Estrogen effects on neurite outgrowth and cytoskeletal gene expression in ERalpha-transfected PC12 cell lines. Exp Neurol 2001; 171:308-16. [PMID: 11573983 DOI: 10.1006/exnr.2001.7744] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The potential of gonadal steroids like estrogen (E) to promote neurite sprouting is of interest in development and aging, as well as after neural trauma. The specific roles of the two main estrogen receptors, ERalpha and ERbeta, in neuronal sprouting are not yet well understood. We examined the hypothesis that E can enhance nerve growth factor (NGF)-stimulated neurite sprouting in an ERalpha-dependent manner. PC12 cells that were stably transfected with the full-length rat ERalpha gene (PCER) and a control line of cells transfected with vector DNA alone (PCCON) were compared. Both cell lines vigorously differentiate neurites when treated with NGF. We determined that both lines show basal expression of ERbeta mRNA, but only the PCER cells express ERalpha mRNA. Estrogen treatment markedly enhanced NGF-stimulated neurite outgrowth from PCER but not from PCCON cells. Significantly larger proportions of PCER cells (34 and 53% at 24 and 48 h, respectively) had neurites than did the PCCON cells (17 and 26% at 24 and 48 h) after E plus NGF treatment. We also examined the effects of E and NGF treatment of PCER and PCCON cells on peripherin, alpha-tubulin, and tau mRNA expression. In undifferentiated PCER cells, E treatment increased peripherin, reduced alpha-tubulin, and did not alter tau mRNA levels. No changes in these mRNAs were observed in the controls (undifferentiated PCCON cells) after E treatment. NGF treatment markedly stimulated expression of peripherin, alpha-tubulin, and tau mRNAs in both PCER and PCCON cells. From these observations we conclude that E synergizes with NGF and stimulates neurite sprouting and also modulates expression of several cytoskeletal mRNAs through ERalpha.
Collapse
Affiliation(s)
- L Gollapudi
- Department of Cell Biology and Anatomy, Chicago Medical School, North Chicago, Illinois 60064, USA
| | | |
Collapse
|
50
|
Liuzzi FJ, Bufton SM, Scoville SA. Short-term estrogen replacement increases beta-preprotachykinin mRNA levels in uninjured dorsal root ganglion neurons, but not in axotomized neurons. Exp Neurol 2001; 170:101-8. [PMID: 11421587 DOI: 10.1006/exnr.2001.7697] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Dorsal root ganglion (DRG) neurons that mediate nociception express the high affinity NGF receptor (trkA) gene and the preprotachykinin (PPT) gene. NGF has been shown to regulate both of these DRG neuronal genes. Our laboratory has shown that these genes are also regulated by estrogen. Long-term daily estrogen replacement, in adult ovariectomized (OVX) rats, causes a coordinate decline in trkA and beta-PPT mRNA levels in lumbar DRG neurons, while short-term estrogen replacement increases trkA mRNA levels in uninjured as well as in axotomized lumbar DRG neurons. The purpose of the current study was to test the hypothesis that short-term estrogen replacement increases DRG beta-PPT mRNA levels in lumbar DRG neurons of OVX rats and that the increase is dependent on target-derived NGF. Sciatic nerve transection (SNT) was used to eliminate target-derived NGF in L4 and L5 DRGs in adult OVX rats. Seven days later, one-half of the SNT and one-half of the animals that had received sham sciatic nerve transactions (SHAM) received two daily injections of estradiol benzoate (EB). The remaining rats received two daily injections of vehicle alone. Quantitative in situ hybridization analyses of sections from L4 and L5 DRGs showed that two daily injections of EB significantly increased beta-PPT mRNA levels in DRGs of SHAM animals, but had no effect on beta-PPT mRNA levels in DRGs from SNT animals. These data coupled with our earlier observations of the effect of short-term estrogen replacement on DRG trkA mRNA levels, indicate that the regulation of DRG beta-PPT mRNA levels by estrogen requires target-derived NGF.
Collapse
Affiliation(s)
- F J Liuzzi
- Division of Anatomy, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | | | | |
Collapse
|