1
|
Hong K, Cheng H, Zhuang Y, Yu Y. Sex-specific associations of the endocrine-disrupting chemicals with serum neurofilament light chain among US adults. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118272. [PMID: 40367613 DOI: 10.1016/j.ecoenv.2025.118272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/11/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
OBJECTIVE Endocrine-disrupting chemicals (EDCs) can interfere with endocrine function and lead to neurological damage. Neurofilament light chain (NfL) is a protein released into the blood after neuroaxonal damage, and it has become a dependable biomarker for neurological conditions. The study aimed to investigate the associations between single or combined EDCs exposure and serum NfL levels in adults. METHODS The 1372 participants included in the study were from the 2013-2014 National Health and Nutrition Examination Survey. Due to the difference in types of EDCs, participants were divided into two populations. Multiple linear regression models were used to assess the association between 32 EDCs and NfL. The least absolute shrinkage and selection operator regression model was used for EDCs selection and the weighted quantile sum (WQS) regression was used for examining the association of EDCs mixture with NfL and identify the predominant exposure. RESULTS Levels of urinary bisphenol S, mono(2-ethylhexyl) phthalate, dibutyl phosphate, glyphosate, and 3,5,6-trichloropyridinol were positively associated with serum NfL levels, while benzophenone-3, methylparaben, and propylparaben showed negative associations. In the WQS regression model, the changes of NfL were 0.154 (95 % CI: 0.014-0.294) and 0.164 (95 % CI: 0.033-0.296) for each quartile increase in WQS index of EDCs mixture in the two populations, respectively. Analysis of the subgroup with gender stratification suggested that the association between EDCs mixture and NfL was only significant in men. The positive mixture β was 0.219 (95 % CI: 0.056-0.380) and 0.257 (95 % CI: 0.082-0.433) in the two population, respectively. CONCLUSION The study suggested a potential association between single or combined exposure to EDCs and NfL levels. High-level EDCs exposure might be associated with more severe neurological damage, particularly in men.
Collapse
Affiliation(s)
- Kaiting Hong
- Zhoushan Women and Children Hospital, Zhoushan, China.
| | - Haoyue Cheng
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Yan Zhuang
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Yunxian Yu
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Zarei F, Moazedi AA, Salimi Z, Pourmotabbed A, Yousofvand N, Farshad M, Akrami MR. Activation of androgen receptors alters hippocampal synaptic plasticity and memory retention through modulation of L-type calcium channels. Life Sci 2023; 314:121155. [PMID: 36379312 DOI: 10.1016/j.lfs.2022.121155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/15/2022]
Abstract
AIMS It has been revealed that membrane androgen receptor activation modulates avoidance memory and synaptic plasticity. In a previous study, we showed that Calcineurin, a calcium dependent phosphatase, could be a potential mediator of these AR effects. Also, it is reported that AR activation leads to L-type calcium channel activation. The aim of the current study is to test whether L-type calcium channels are downstream of AR and whether this signal pathway mediates the impairment effect of androgenic steroids on passive avoidance memory and synaptic plasticity. MATERIALS AND METHODS We measured the effect of Nandrolone Decanoate (AR agonist), AR antagonist (Nilutamide) plus ND or L-type calcium channel inhibitor (Nifedipine) plus ND on passive avoidance performance of adolescent male rats. For extracellular field potential recordings hippocampal slices were perfused with ND, Nilutamide-ND or Nifedipine-ND. KEY FINDINGS Our results clarified that AR activation by ND could impair avoidance behavior as step through latency decreased in ND-treated group while application of both Nilutamide and Nifedipine reestablished normal avoidance behavior. Also, LTP induction in the CA1 area of hippocampus was diminished by ND perfusion and both AR antagonist and L-type calcium channel inhibitor application lead to normal LTP. These findings support our hypothesis that activation of L-type calcium channels are involved in ARs mechanism effects on both avoidance behavior and hippocampal synaptic plasticity. SIGNIFICANCE Understanding the biological effects of AR agonists on cognitive processes and its cellular mechanism may be a new/supplementary way to treating fear-related disorders.
Collapse
Affiliation(s)
- Fatemeh Zarei
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran.
| | - Ahmad Ali Moazedi
- Department of Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zahra Salimi
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, Centre for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada; Department of Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ali Pourmotabbed
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Namdar Yousofvand
- Department of Biology, Centre for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Moradpour Farshad
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, Centre for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Mohammad Reza Akrami
- Department of Neurosurgery, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Ysrraelit MC, Correale J. Impact of Andropause on Multiple Sclerosis. Front Neurol 2021; 12:766308. [PMID: 34803897 PMCID: PMC8602357 DOI: 10.3389/fneur.2021.766308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Andropause results from the natural decrease in testosterone levels that occurs with age. In contrast to menopause, which is a universal, well-characterized process associated with absolute gonadal failure, andropause ensues after gradual decline of both hypothalamic-pituitary-gonadal axis activity, as well as of testicular function, a process which usually develops over a period of many years. Increasing evidence on greater risk of Multiple sclerosis (MS) associated with lower testosterone levels is being reported. Likewise, epidemiological studies have shown a later age of onset of MS in men, relative to women, which could perhaps respond to the decline in protective testosterone levels. In this review, we will discuss the role of androgens in the development and function of the innate and adaptive immune response, as well as in neuroprotective mechanisms relevant to MS. Testosterone effects observed in different animal models and in epidemiological studies in humans will be discussed, as well as their correlation with physical disability and cognitive function levels. Finally, published and ongoing clinical trials exploring the role of androgens, particularly at key stages of sexual maturation, will be reviewed.
Collapse
Affiliation(s)
- Maria C Ysrraelit
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Jorge Correale
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
5
|
Bianchi VE, Rizzi L, Bresciani E, Omeljaniuk RJ, Torsello A. Androgen Therapy in Neurodegenerative Diseases. J Endocr Soc 2020; 4:bvaa120. [PMID: 33094209 PMCID: PMC7568521 DOI: 10.1210/jendso/bvaa120] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington disease, are characterized by the loss of neurons as well as neuronal function in multiple regions of the central and peripheral nervous systems. Several studies in animal models have shown that androgens have neuroprotective effects in the brain and stimulate axonal regeneration. The presence of neuronal androgen receptors in the peripheral and central nervous system suggests that androgen therapy might be useful in the treatment of neurodegenerative diseases. To illustrate, androgen therapy reduced inflammation, amyloid-β deposition, and cognitive impairment in patients with AD. As well, improvements in remyelination in MS have been reported; by comparison, only variable results are observed in androgen treatment of PD. In ALS, androgen administration stimulated motoneuron recovery from progressive damage and regenerated both axons and dendrites. Only a few clinical studies are available in human individuals despite the safety and low cost of androgen therapy. Clinical evaluations of the effects of androgen therapy on these devastating diseases using large populations of patients are strongly needed.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta, Falciano, San Marino
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
6
|
Giatti S, Diviccaro S, Falvo E, Garcia-Segura LM, Melcangi RC. Physiopathological role of the enzymatic complex 5α-reductase and 3α/β-hydroxysteroid oxidoreductase in the generation of progesterone and testosterone neuroactive metabolites. Front Neuroendocrinol 2020; 57:100836. [PMID: 32217094 DOI: 10.1016/j.yfrne.2020.100836] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022]
Abstract
The enzymatic complex 5α-reductase (5α-R) and 3α/3β-hydroxysteroid oxidoreductase (HSOR) is expressed in the nervous system, where it transforms progesterone (PROG) and testosterone (T) into neuroactive metabolites. These metabolites regulate myelination, brain maturation, neurotransmission, reproductive behavior and the stress response. The expression of 5α-R and 3α-HSOR and the levels of PROG and T reduced metabolites show regional and sex differences in the nervous system and are affected by changing physiological conditions as well as by neurodegenerative and psychiatric disorders. A decrease in their nervous tissue levels may negatively impact the course and outcome of some pathological events. However, in other pathological conditions their increased levels may have a negative impact. Thus, the use of synthetic analogues of these steroids or 5α-R modulation have been proposed as therapeutic approaches for several nervous system pathologies. However, further research is needed to fully understand the consequences of these manipulations, in particular with 5α-R inhibitors.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Eva Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
7
|
Jaeger ECB, Miller LE, Goins EC, Super CE, Chyr CU, Lower JW, Honican LS, Morrison DE, Ramdev RA, Spritzer MD. Testosterone replacement causes dose-dependent improvements in spatial memory among aged male rats. Psychoneuroendocrinology 2020; 113:104550. [PMID: 31901624 PMCID: PMC7080566 DOI: 10.1016/j.psyneuen.2019.104550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/16/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Testosterone has been shown to have dose-dependent effects on spatial memory in males, but the effects of aging upon this relationship remain unclear. Additionally, the mechanism by which testosterone regulates memory is unknown, but may involve changes in brain-derived neurotrophic factor (BDNF) within specific brain regions. We tested the effects of age and testosterone on spatial memory among male rats using two spatial memory tasks: an object-location memory task (OLMT) and the radial-arm maze (RAM). Castration had minimal effect on performance on the RAM, but young rats (2 months) performed significantly fewer working memory errors than aged rats (20 months), and aged rats performed significantly fewer reference memory errors. Both age and castration impaired performance on the OLMT, with only the young rats with intact gonads successfully performing the task. Subsequent experiments involved daily injections of either drug vehicle or one of four doses of testosterone propionate (0.125, 0.250, 0.500, and 1.00 mg/rat) given to castrated aged males. On the RAM, a low physiological dose (0.125 mg) and high doses (0.500-1.000 mg) of testosterone improved working memory, while an intermediate dose (0.250 mg) did not. On the OLMT, only the 0.250 mg T group showed a significant increase in exploration ratios from the exposure trials to the testing trials, indicating that this group remembered the position of the objects. Brain tissue (prefrontal cortex, hippocampus, and striatum) was collected from all subjects to assay BDNF. We found no evidence that testosterone influenced BDNF, indicating that it is unlikely that testosterone regulates spatial memory through changes in BDNF levels.
Collapse
Affiliation(s)
- Eliza C B Jaeger
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - L Erin Miller
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - Emily C Goins
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - Chloe E Super
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - Christina U Chyr
- Department of Biology, Middlebury College, Middlebury, VT, 05753, USA.
| | - John W Lower
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - Lauren S Honican
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - Daryl E Morrison
- Department of Biology, Middlebury College, Middlebury, VT, 05753, USA.
| | - Rajan A Ramdev
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA.
| | - Mark D Spritzer
- Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA; Department of Biology, Middlebury College, Middlebury, VT, 05753, USA.
| |
Collapse
|
8
|
Spritzer MD, Roy EA. Testosterone and Adult Neurogenesis. Biomolecules 2020; 10:biom10020225. [PMID: 32028656 PMCID: PMC7072323 DOI: 10.3390/biom10020225] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
It is now well established that neurogenesis occurs throughout adulthood in select brain regions, but the functional significance of adult neurogenesis remains unclear. There is considerable evidence that steroid hormones modulate various stages of adult neurogenesis, and this review provides a focused summary of the effects of testosterone on adult neurogenesis. Initial evidence came from field studies with birds and wild rodent populations. Subsequent experiments with laboratory rodents have tested the effects of testosterone and its steroid metabolites upon adult neurogenesis, as well as the functional consequences of induced changes in neurogenesis. These experiments have provided clear evidence that testosterone increases adult neurogenesis within the dentate gyrus region of the hippocampus through an androgen-dependent pathway. Most evidence indicates that androgens selectively enhance the survival of newly generated neurons, while having little effect on cell proliferation. Whether this is a result of androgens acting directly on receptors of new neurons remains unclear, and indirect routes involving brain-derived neurotrophic factor (BDNF) and glucocorticoids may be involved. In vitro experiments suggest that testosterone has broad-ranging neuroprotective effects, which will be briefly reviewed. A better understanding of the effects of testosterone upon adult neurogenesis could shed light on neurological diseases that show sex differences.
Collapse
Affiliation(s)
- Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
- Correspondence: ; Tel.: 802-443-5676
| | - Ethan A. Roy
- Graduate School of Education, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
9
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
10
|
Tenkorang MAA, Duong P, Cunningham RL. NADPH Oxidase Mediates Membrane Androgen Receptor-Induced Neurodegeneration. Endocrinology 2019; 160:947-963. [PMID: 30811529 PMCID: PMC6435014 DOI: 10.1210/en.2018-01079] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/22/2019] [Indexed: 12/21/2022]
Abstract
Oxidative stress (OS) is a common characteristic of several neurodegenerative disorders, including Parkinson disease (PD). PD is more prevalent in men than in women, indicating the possible involvement of androgens. Androgens can have either neuroprotective or neurodamaging effects, depending on the presence of OS. Specifically, in an OS environment, androgens via a membrane-associated androgen receptor (mAR) exacerbate OS-induced damage. To investigate the role of androgens on OS signaling and neurodegeneration, the effects of testosterone and androgen receptor activation on the major OS signaling cascades, the reduced form of NAD phosphate (NADPH) oxidase (NOX)1 and NOX2 and the Gαq/inositol trisphosphate receptor (InsP3R), were examined. To create an OS environment, an immortalized neuronal cell line was exposed to H2O2 prior to cell-permeable/cell-impermeable androgens. Different inhibitors were used to examine the role of G proteins, mAR, InsP3R, and NOX1/2 on OS generation and cell viability. Both testosterone and DHT/3-O-carboxymethyloxime (DHT)-BSA increased H2O2-induced OS and cell death, indicating the involvement of an mAR. Furthermore, classical AR antagonists did not block testosterone's negative effects in an OS environment. Because there are no known antagonists specific for mARs, an AR protein degrader, ASC-J9, was used to block mAR action. ASC-J9 blocked testosterone's negative effects. To determine OS-related signaling mediated by mAR, this study examined NOX1, NOX2, Gαq. NOX1, NOX2, and the Gαq complex with mAR. Only NOX inhibition blocked testosterone-induced cell loss and OS. No effects of blocking either Gαq or G protein activation were observed on testosterone's negative effects. These results indicate that androgen-induced OS is via the mAR-NOX complex and not the mAR-Gαq complex.
Collapse
Affiliation(s)
- Mavis A A Tenkorang
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas
| | - Phong Duong
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas
- Correspondence: Rebecca L. Cunningham, PhD, Department of Physiology and Anatomy, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, Texas 76107. E-mail:
| |
Collapse
|
11
|
Christensen A, Pike CJ. APOE genotype affects metabolic and Alzheimer-related outcomes induced by Western diet in female EFAD mice. FASEB J 2019; 33:4054-4066. [PMID: 30509127 PMCID: PMC6404574 DOI: 10.1096/fj.201801756r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/05/2018] [Indexed: 01/24/2023]
Abstract
Development of Alzheimer's disease (AD) is regulated by interactive effects of genetic and environmental risk factors. The most significant genetic risk factor for AD is the ε4 allele of apolipoprotein E ( APOE4), which has been shown to exert greater AD risk in women. An important modifiable AD risk factor is obesity and its associated metabolic dysfunctions. Whether APOE genotype might interact with obesity in females to regulate AD pathogenesis is unclear. To investigate this issue, we studied the effects of Western diet (WD) on female EFAD mice, a transgenic mouse model of AD that includes human APOE alleles ε3 (E3FAD) and ε4 (E4FAD). EFAD mice were fed either control (10% fat, 7% sugar) or WD (45% fat, 17% sugar), and both metabolic and neuropathologic outcomes were determined. Although E4FAD mice generally exhibited poorer metabolic status at baseline, E3FAD mice showed greater diet-induced metabolic impairments. Similarly, E4FAD mice exhibited higher levels of AD-related pathology overall, but only E3FAD showed significant increases on select measures of β-amyloid pathology after exposure to WD. These data demonstrate a gene-environment interaction between APOE and obesogenic diets in females. Understanding how AD-promoting effects of obesity are modulated by genetic factors will foster the identification of at-risk populations and development of preventive interventions.-Christensen, A., Pike, C. J. APOE genotype affects metabolic and Alzheimer-related outcomes induced by Western diet in female EFAD mice.
Collapse
Affiliation(s)
- Amy Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Christian J. Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
12
|
Nikray N, Karimi I, Siavashhaghighi Z, Becker LA, Mofatteh MM. An effort toward molecular biology of food deprivation induced food hoarding in gonadectomized NMRI mouse model: focus on neural oxidative status. BMC Neurosci 2018; 19:59. [PMID: 30249177 PMCID: PMC6154416 DOI: 10.1186/s12868-018-0461-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 09/20/2018] [Indexed: 12/03/2022] Open
Abstract
Background Environmental uncertainty, such as food deprivation, may alter internal milieu of nervous system through various mechanisms. In combination with circumstances of stress or aging, high consumption of unsaturated fatty acids and oxygen can make neural tissues sensitive to oxidative stress (OS). For adult rats, diminished level of gonadal steroid hormones accelerates OS and may result in special behavioral manifestations. This study was aimed to partially answer the question whether OS mediates trade-off between food hoarding and food intake (fat hoarding) in environmental uncertainty (e.g., fluctuations in food resource) within gonadectomized mouse model in the presence of food deprivation-induced food hoarding behavior. Results Hoarding behavior was not uniformly expressed in all male mice that exposed to food deprivation. Extended phenotypes including hoarder and non-hoarder mice stored higher and lower amounts of food respectively as compared to that of low-hoarder mice (normal phenotype) after food deprivation. Results showed that neural oxidative status was not changed in the presence of hoarding behavior in gonadectomized mice regardless of tissue type, however, glutathione levels of brain tissues were increased in the presence of hoarding behavior. Decreased superoxide dismutase activity in brain and spinal cord tissues and increased malondialdehyde in brain tissues of gonadectomized mice were also seen. Conclusions Although, food deprivation-induced hoarding behavior is a strategic response to food shortage in mice, it did not induce the same amount of hoarding across all colony mates. Hoarding behavior, in this case, is a response to the environmental uncertainty of food shortage, therefore is not an abnormal behavior. Hoarding behavior induced neural OS with regard to an increase in brain glutathione levels but failed to show other markers of neural OS. Decreased superoxide dismutase activity in brain and spinal cord tissues and increased malondialdehyde levels in brain tissues of gonadectomized mice could be a hallmark of debilitated antioxidative defense and more lipid peroxidation due to reduced amount of gonadal steroid hormones during aging.
Collapse
Affiliation(s)
- Noushin Nikray
- Laboratory of Molecular and Cellular Biology 1214, Department of Basic Veterinary Sciences, School of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Isaac Karimi
- Laboratory of Molecular and Cellular Biology 1214, Department of Basic Veterinary Sciences, School of Veterinary Medicine, Razi University, Kermanshah, Iran. .,Department of Biology, Faculty of Science, Razi University, Kermanshah, 67149-67346, Iran.
| | | | - Lora A Becker
- Department of Psychology, University of Evansville, Evansville, IN, 47722, USA
| | - Mohammad Mehdi Mofatteh
- Department of Accounting, School of Economics and Accounting, Islamic Azad University South Tehran Branch, Tehran, Iran
| |
Collapse
|
13
|
Iqbal R, Jain GK, Siraj F, Vohora D. Aromatase inhibition by letrozole attenuates kainic acid-induced seizures but not neurotoxicity in mice. Epilepsy Res 2018; 143:60-69. [PMID: 29665500 DOI: 10.1016/j.eplepsyres.2018.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/02/2018] [Accepted: 04/07/2018] [Indexed: 12/11/2022]
Abstract
Evidence shows neurosteroids play a key role in regulating epileptogenesis. Neurosteroids such as testosterone modulate seizure susceptibility through its transformation to metabolites which show proconvulsant and anticonvulsant effects, respectively. Reduction of testosterone by aromatase generates proconvulsant 17-β estradiol. Alternatively, testosterone is metabolized into 5α-dihydrotestosterone (5α-DHT) by 5α-reductase, which is then reduced by 3α-hydroxysteroid oxidoreductase enzyme (3α-HSOR) to form anticonvulsant metabolite 3α-androstanediol (3α-Diol), a potent GABAA receptor modulating neurosteroid. The present study evaluated whether inhibition of aromatase inhibitor letrozole protects against seizures and neuronal degeneration induced by kainic acid (KA) (10 mg/kg, i.p.) in Swiss albino mice. Letrozole (1 mg/kg, i.p.) administered one hour prior to KA significantly increased the onset time of seizures and reduced the% incidence of seizures. Pretreatment with finasteride, a selective inhibitor of 5α-reductase and indomethacin, a selective inhibitor of 3α-hydroxysteroid oxidoreductase enzyme (3α-HSOR), reversed the protective effects of letrozole in KA-induced seizures in mice. Microscopic examination using cresyl violet staining revealed that letrozole did not modify KA-induced neurotoxicity in the CA1, CA3 and DG region of the hippocampus. Letrozole treatment resulted in the reduced levels of 17-β estradiol and elevated the levels of 5α-dihydrotestosterone (DHT) and 3α-Diol in the hippocampus. Finasteride and indomethacin attenuated letrozole-induced elevations of 5α-DHT and 3α-Diol. Our results indicate the potential anticonvulsant effects of letrozole against KA-induced seizures in mice that might be mediated by inhibiting aromatization of testosterone to 17β-estradiol, a proconvulsant hormone and by redirecting the synthesis to anticonvulsant metabolites, 5α-DHT and 3α-Diol. Acute aromatase inhibition, thus, might be used as an adjuvant in the treatment of status epilepticus and can be pursued further.
Collapse
Affiliation(s)
- Ramsha Iqbal
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Gaurav K Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Fouzia Siraj
- National Institute of Pathology, Indian Council of Medical Research, New Delhi, 110029, India
| | - Divya Vohora
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
14
|
Lee JH, Byun MS, Yi D, Choe YM, Choi HJ, Baek H, Sohn BK, Lee JY, Kim HJ, Kim JW, Lee Y, Kim YK, Sohn CH, Woo JI, Lee DY. Sex-specific association of sex hormones and gonadotropins, with brain amyloid and hippocampal neurodegeneration. Neurobiol Aging 2017; 58:34-40. [DOI: 10.1016/j.neurobiolaging.2017.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/20/2017] [Accepted: 06/09/2017] [Indexed: 02/08/2023]
|
15
|
Bojar I, Pinkas J, Gujski M, Owoc A, Raczkiewicz D, Gustaw-Rothenberg K. Postmenopausal cognitive changes and androgen levels in the context of apolipoprotein E polymorphism. Arch Med Sci 2017; 13:1148-1159. [PMID: 28883857 PMCID: PMC5575214 DOI: 10.5114/aoms.2016.62869] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION The focus of this study was to assess cognitive functions in relation to androgens and specifically testosterone and dehydroepiandrosterone in postmenopausal women as well as the correlation between cognitive functions and these two androgens according to polymorphism of the apolipoprotein E gene (APOE). MATERIAL AND METHODS A group of 402 women was recruited to the study (minimum 2 years after the last menstruation, follicle-stimulating hormone (FSH) more than 30 U/ml and no dementia signs on Montreal Cognitive Assessment). The computerized battery of the Central Nervous System Vital Signs test was used to diagnose cognitive functions. APOE genotyping was performed by multiplex polymerase chain reaction (PCR). Testosterone (TTE) and dehydroepiandrosterone (DHEA) in the blood serum were assessed for further statistical correlations analysis. RESULTS In the group of postmenopausal women, higher testosterone concentration was associated with lower scores for Neurocognition Index (NCI) (p = 0.028), memory (p = 0.008) and psychomotor speed (p < 0.001). Presence of at least one APOE ε4 allele potentiated testosterone's negative influence on cognitive functions (p < 0.05). Woman with a high normal level of DHEA scored significantly better in verbal (p = 0.027) and visual memory (p < 0.001) than other participants. APOE polymorphism did not modify the relationship between DHEA concentration and scores for cognitive functions. CONCLUSIONS Hormonal balance variations after menopause may influence brain processes concerned with cognition, especially memory and psychomotor speed. The observed effects may be related to androgens' influence on higher cortical functions in the changed hormonal dynamics of the postmenopausal period.
Collapse
Affiliation(s)
- Iwona Bojar
- Department for Women Health, Institute of Rural Health, Lublin, Poland
| | - Jarosław Pinkas
- School of Public Health, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Mariusz Gujski
- Department of Prevention of Environmental Hazards and Allergology, Medical University of Warsaw, Warsaw, Poland
| | - Alfred Owoc
- Center for Public Health and Health Promotion, Institute of Rural Health, Lublin, Poland
| | - Dorota Raczkiewicz
- Institute of Statistics and Demography, Warsaw School of Economics, Warsaw, Poland
| | - Kasia Gustaw-Rothenberg
- Lou Ruvo Brain Wellness Center, Neurological Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Neurodegenerative Diseases, Institute of Rural Health, Lublin, Poland
| |
Collapse
|
16
|
Age-dependent regulation of obesity and Alzheimer-related outcomes by hormone therapy in female 3xTg-AD mice. PLoS One 2017; 12:e0178490. [PMID: 28575011 PMCID: PMC5456100 DOI: 10.1371/journal.pone.0178490] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/14/2017] [Indexed: 12/31/2022] Open
Abstract
Depletion of ovarian hormones at menopause is associated with increased Alzheimer's disease (AD) risk. Hormone loss also increases central adiposity, which promotes AD development. One strategy to improve health outcomes in postmenopausal women is estrogen-based hormone therapy (HT), though its efficacy is controversial. The window of opportunity hypothesis posits that HT is beneficial only if initiated near the onset of menopause. Here, we tested this hypothesis by assessing the efficacy of HT against diet-induced obesity and AD-related pathology in female 3xTg-AD mice at early versus late middle-age. HT protected against obesity and reduced β-amyloid burden only at early middle-age. One mechanism that contributes to AD pathogenesis is microglial activation, which is increased by obesity and reduced by estrogens. In parallel to its effects on β-amyloid accumulation, we observed that HT reduced morphological evidence of microglial activation in early but not late middle-age. These findings suggest that HT may be effective during human perimenopause in reducing indices of obesity and AD-related pathology, a conclusion consistent with the window of opportunity hypothesis.
Collapse
|
17
|
Crawford ED, Schally AV, Pinthus JH, Block NL, Rick FG, Garnick MB, Eckel RH, Keane TE, Shore ND, Dahdal DN, Beveridge TJR, Marshall DC. The potential role of follicle-stimulating hormone in the cardiovascular, metabolic, skeletal, and cognitive effects associated with androgen deprivation therapy. Urol Oncol 2017; 35:183-191. [PMID: 28325650 DOI: 10.1016/j.urolonc.2017.01.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 11/25/2022]
Abstract
PURPOSE To explore how follicle-stimulating hormone (FSH) may contribute to cardiovascular, metabolic, skeletal, and cognitive events in men treated for prostate cancer, with various forms of androgen deprivation therapy (ADT). MATERIALS AND METHODS A colloquium of prostate cancer experts was convened in May 2015, to discuss the role of FSH in the development of unwanted effects associated with ADT. Subsequently, a literature review (Medline, PubMed, and relevant congress abstract databases) was performed to further explore and evaluate the collected evidence. RESULTS It has become evident that, in the setting of ADT, FSH can promote the development of atherosclerotic plaque formation, metabolic syndrome, and insulin resistance. Data also suggest that FSH is an important mediator of bone remodeling, particularly bone resorption, and thereby increases the risk for bone fracture. Additional evidence implicates a role for FSH in bone metastasis as well. The influence of FSH on ADT-induced cognitive deficits awaits further elucidation; however, the possibility that FSH may be involved therein cannot be ruled out. CONCLUSIONS The widespread molecular and physiological consequences of FSH system activation in normal and pathological conditions are becoming better understood. Progress in this area has been achieved by the development of additional investigative and clinical measures to better evaluate specific adverse effects. More research is needed on FSH function in the development of cancer as well as its association with cardiovascular, metabolic, musculoskeletal, and cognitive effects in ADT.
Collapse
Affiliation(s)
- E David Crawford
- Department of Urologic Oncology, School of Medicine, University of Colorado, Denver, Denver, CO.
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Miami Veterans Affairs Medical Center, Miami, FL; Department of Pathology, University of Miami School of Medicine, Miami, FL; Department of Medicine, University of Miami School of Medicine, Miami, FL
| | - Jehonathan H Pinthus
- Department of Surgery, Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Norman L Block
- Endocrine, Polypeptide and Cancer Institute, Miami Veterans Affairs Medical Center, Miami, FL; Department of Pathology, University of Miami School of Medicine, Miami, FL; Department of Medicine, University of Miami School of Medicine, Miami, FL
| | - Ferenc G Rick
- Endocrine, Polypeptide and Cancer Institute, Miami Veterans Affairs Medical Center, Miami, FL; Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL
| | - Marc B Garnick
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado, Denver, CO
| | - Thomas E Keane
- Department of Urology, Medical University of South Carolina, Charleston, SC
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | | | | | | |
Collapse
|
18
|
Neri-Gómez T, Espinosa-Raya J, Díaz-Cintra S, Segura-Uribe J, Orozco-Suárez S, Gallardo JM, Guerra-Araiza C. Tibolone modulates neuronal plasticity through regulating Tau, GSK3β/Akt/PI3K pathway and CDK5 p35/p25 complexes in the hippocampus of aged male mice. Neural Regen Res 2017; 12:588-595. [PMID: 28553339 PMCID: PMC5436357 DOI: 10.4103/1673-5374.205098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Aging is a key risk factor for cognitive decline and age-related neurodegenerative disorders. Also, an age-related decrease in sex steroid hormones may have a negative impact on the formation of neurofibrillary tangles (NFTs); these hormones can regulate Tau phosphorylation and the principal kinase GSK3β involved in this process. Hormone replacement therapy decreases NFTs, but it increases the risk of some types of cancer. However, other synthetic hormones such as tibolone (TIB) have been used for hormone replacement therapy. The aim of this work was to evaluate the long-term effects of TIB (0.01 mg/kg and 1 mg/kg, intragastrically for 12 weeks) on the content of total and hyperphosphorylated Tau (PHF-1) proteins and the regulation of GSK3β/Akt/PI3K pathway and CDK5/p35/p25 complexes in the hippocampus of aged male mice. We observed that the content of PHF-1 decreased with TIB administration. In contrast, no changes were observed in the active form of GSK3β or PI3K. TIB decreased the expression of the total and phosphorylated form of Akt while increased that of p110 and p85. The content of CDK5 was differentially modified with TIB: it was increased at low doses and decreased at high doses. When we analyzed the content of CDK5 activators, an increase was found on p35; however, the content of p25 decreased with administration of low dose of TIB. Our results suggest a possible mechanism of action of TIB in the hippocampus of aged male mice. Through the regulation of Tau and GSK3β/Akt/PI3K pathway, and CDK5/p35/p25 complexes, TIB may modulate neuronal plasticity and regulate learning and memory processes.
Collapse
Affiliation(s)
- Teresa Neri-Gómez
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.,Laboratorio de Farmacología Conductual, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón Col. Sto. Tomás, Ciudad de México, México
| | - Judith Espinosa-Raya
- Laboratorio de Farmacología Conductual, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón Col. Sto. Tomás, Ciudad de México, México
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México Campus Juriquilla, Querétaro, Querétaro, México
| | - Julia Segura-Uribe
- Enfermedades Neurológicas (Neurological Diseases), Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Sandra Orozco-Suárez
- Enfermedades Neurológicas (Neurological Diseases), Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Juan Manuel Gallardo
- Enfermedades Nefrológicas (Kidney Diseases), Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| |
Collapse
|
19
|
Song J, Jung C, Kim OY. The Novel Implication of Androgen in Diabetes-induced Alzheimer's Disease. J Lipid Atheroscler 2017. [DOI: 10.12997/jla.2017.6.2.66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, Busan, Korea
| |
Collapse
|
20
|
Cai Y, Chew C, Muñoz F, Sengelaub DR. Neuroprotective effects of testosterone metabolites and dependency on receptor action on the morphology of somatic motoneurons following the death of neighboring motoneurons. Dev Neurobiol 2016; 77:691-707. [PMID: 27569375 DOI: 10.1002/dneu.22445] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 11/11/2022]
Abstract
Partial depletion of spinal motoneuron populations induces dendritic atrophy in neighboring motoneurons, and treatment with testosterone is neuroprotective, attenuating induced dendritic atrophy. In this study we examined whether the protective effects of testosterone could be mediated via its androgenic or estrogenic metabolites. Furthermore, to assess whether these neuroprotective effects were mediated through steroid hormone receptors, we used receptor antagonists to attempt to prevent the neuroprotective effects of hormones after partial motoneuron depletion. Motoneurons innervating the vastus medialis muscles of adult male rats were selectively killed by intramuscular injection of cholera toxin-conjugated saporin. Simultaneously, some saporin-injected rats were treated with either dihydrotestosterone or estradiol, alone or in combination with their respective receptor antagonists, or left untreated. Four weeks later, motoneurons innervating the ipsilateral vastus lateralis muscle were labeled with cholera toxin-conjugated horseradish peroxidase, and dendritic arbors were reconstructed in three dimensions. Compared with intact normal animals, partial motoneuron depletion resulted in decreased dendritic length in remaining quadriceps motoneurons. Dendritic atrophy was attenuated with both dihydrotestosterone and estradiol treatment to a degree similar to that seen with testosterone, and attenuation of atrophy was prevented by receptor blockade. Together, these findings suggest that neuroprotective effects on motoneurons can be mediated by either androgenic or estrogenic hormones and require action via steroid hormone receptors, further supporting a role for hormones as neurotherapeutic agents in the injured nervous system. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 691-707, 2017.
Collapse
Affiliation(s)
- Yi Cai
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, 47405
| | - Cory Chew
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, 47405
| | - Fernando Muñoz
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, 47405
| | - Dale R Sengelaub
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, 47405
| |
Collapse
|
21
|
Levels and actions of neuroactive steroids in the nervous system under physiological and pathological conditions: Sex-specific features. Neurosci Biobehav Rev 2016; 67:25-40. [DOI: 10.1016/j.neubiorev.2015.09.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 01/21/2023]
|
22
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
23
|
Pomara C, Neri M, Bello S, Fiore C, Riezzo I, Turillazzi E. Neurotoxicity by synthetic androgen steroids: oxidative stress, apoptosis, and neuropathology: A review. Curr Neuropharmacol 2015; 13:132-45. [PMID: 26074748 PMCID: PMC4462038 DOI: 10.2174/1570159x13666141210221434] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/30/2014] [Accepted: 10/25/2014] [Indexed: 12/25/2022] Open
Abstract
Anabolic-androgenic steroids (AAS) are synthetic substances derived from testosterone that are largely employed due to their trophic effect on muscle tissue of athletes at all levels. Since a great number of organs and systems are a target of AAS, their adverse effects are primarily on the following systems: reproductive, hepatic, musculoskeletal, endocrine, renal, immunological, infectious, cardiovascular, cerebrovascular, and hematological. Neuropsychiatric and behavioral effects as a result of AAS abuse are well known and described in the literature. Mounting evidence exists suggesting that in addition to psychiatric and behavioral effects, non-medical use of AAS carries neurodegenerative potential. Although, the nature of this association remains largely unexplored, recent animal studies have shown the recurrence of this AAS effect, ranging from neurotrophin unbalance to increased neuronal susceptibility to apoptotic stimuli. Experimental and animal studies strongly suggest that apoptotic mechanisms are at least in part involved in AAS-induced neurotoxicity. Furthermore, a great body of evidence is emerging suggesting that increased susceptibility to cellular oxidative stress could play a pivotal role in the pathogenesis of many neurodegenerative disorders and cognitive impairment. As in other drug-evoked encephalopathies, the key mechanisms involved in AAS - induced neuropathology could represent a target for future neuroprotective strategies. Progress in the understanding of these mechanisms will provide important insights into the complex pathophysiology of AAS-induced neurodegeneration, and will pave the way for forthcoming studies. Supplementary to abandoning the drug abuse that represents the first step in reducing the possibility of irreversible brain damage in AAS abusers, neuroprotective strategies have to be developed and implemented in future.
Collapse
Affiliation(s)
- Cristoforo Pomara
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- Department of Anatomy, University of Malta. Msida, Malta
| | - Margherita Neri
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stefania Bello
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Carmela Fiore
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Irene Riezzo
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Emanuela Turillazzi
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
24
|
Zup SL, Edwards NS, McCarthy MM. Sex- and age-dependent effects of androgens on glutamate-induced cell death and intracellular calcium regulation in the developing hippocampus. Neuroscience 2014; 281:77-87. [PMID: 25264034 DOI: 10.1016/j.neuroscience.2014.09.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 08/31/2014] [Accepted: 09/18/2014] [Indexed: 12/12/2022]
Abstract
Hippocampal neurons must maintain control over cytosolic calcium levels, especially during development, as excitation and calcium flux are necessary for proper growth and function. But excessive calcium can lead to excitotoxic cell death. Previous work suggests that neonatal male and female hippocampal neurons regulate cytosolic calcium differently, thereby leading to differential susceptibility to excitotoxic damage. Hippocampal neurons are also exposed to gonadal hormones during development and express high levels of androgen receptors. Androgens have both neuroprotective and neurotoxic effects in adults and developing animals. The present study sought to examine the effect of androgen on cell survival after an excitatory stimulus in the developing hippocampus, and whether androgen-mediated calcium regulation was the governing mechanism. We observed that glutamate did not induce robust or sexually dimorphic apoptosis in cultured hippocampal neurons at an early neonatal time point, but did 5days later - only in males. Further, pretreatment with the androgen dihydrotestosterone (DHT) protected males from apoptosis during this time, but had no effect on females. Calcium imaging of sex-specific cultures revealed that DHT decreased the peak of intracellular calcium induced by glutamate, but only in males. To determine a possible mechanism for this androgen neuroprotection and calcium regulation, we quantified three calcium regulatory proteins, plasma membrane calcium ATPase1 (PMCA1), sodium/calcium exchanger1 (NCX1), and the sarco/endoplasmic reticulum calcium ATPase 2 (SERCA2). Surprisingly, there was no sex difference in the level of any of the three proteins. Treatment with DHT significantly decreased PMCA1 and NCX1, but increased SERCA2 protein levels in very young animals but not at a later timepoint. Taken together, these data suggest a complex interaction of sex, hormones, calcium regulation and developmental age; however androgens acting during the first week of life are implicated in regulation of hippocampal cell death and may be an underlying mechanism for sexually dimorphic apoptosis.
Collapse
Affiliation(s)
- S L Zup
- Program in Developmental and Brain Sciences, University of Massachusetts Boston, Boston, MA 02125, United States; Department of Psychology, University of Massachusetts Boston, Boston, MA 02125, United States.
| | - N S Edwards
- Department of Pharmacology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - M M McCarthy
- Department of Pharmacology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| |
Collapse
|
25
|
Braun CMJ, Roberge C. Gender-related protection from or vulnerability to severe CNS diseases: gonado-structural and/or gonado-activational? A meta-analysis of relevant epidemiological studies. Int J Dev Neurosci 2014; 38:36-51. [PMID: 25109841 DOI: 10.1016/j.ijdevneu.2014.07.009] [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: 06/23/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A vast scientific literature has dealt with gender-specific risk for brain disorder. That field is evolving toward a consensus to the effect that the estrogen hormone family is outstandingly and uniquely neuroprotective. However, the epidemiology relevant to this general outlook remains piecemeal. METHOD The present investigation strategically formats the relevant epidemiological findings around the world in order to quantitatively meta-analyze gender ratio of risk for a variety of relevant severe central nervous system (CNS) diseases at all three gonadal stages of the life cycle, pre pubertal, post adolescent/pre menopausal, and post menopausal. RESULTS The data quantitatively establish that (1) no single epidemiological study should be cited as evidence of gender-specific neuroprotection against the most common severe CNS diseases because the gender-specific risk ratios are contradictory from one study to the other; (2) risk for severe CNS disease is indeed significantly gender-specific, but either gender can be protected: it depends on the disease, not at all on the age bracket. CONCLUSION Our assay of gender-specific risk for severe brain disease around the world has not been able to support the idea according to which any one gender-prevalent gonadal steroid hormone dominates as a neuroprotective agent at natural concentrations.
Collapse
Affiliation(s)
- Claude M J Braun
- Department of Psychology, Université du Québec à Montréal, Canada.
| | - Carl Roberge
- Department of Psychology, Université du Québec à Montréal, Canada
| |
Collapse
|
26
|
Li R, Singh M. Sex differences in cognitive impairment and Alzheimer's disease. Front Neuroendocrinol 2014; 35:385-403. [PMID: 24434111 PMCID: PMC4087048 DOI: 10.1016/j.yfrne.2014.01.002] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/31/2013] [Accepted: 01/06/2014] [Indexed: 12/15/2022]
Abstract
Studies have shown differences in specific cognitive ability domains and risk of Alzheimer's disease between the men and women at later age. However it is important to know that sex differences in cognitive function during adulthood may have their basis in both organizational effects, i.e., occurring as early as during the neuronal development period, as well as in activational effects, where the influence of the sex steroids influence brain function in adulthood. Further, the rate of cognitive decline with aging is also different between the sexes. Understanding the biology of sex differences in cognitive function will not only provide insight into Alzheimer's disease prevention, but also is integral to the development of personalized, gender-specific medicine. This review draws on epidemiological, translational, clinical, and basic science studies to assess the impact of sex differences in cognitive function from young to old, and examines the effects of sex hormone treatments on Alzheimer's disease in men and women.
Collapse
Affiliation(s)
- Rena Li
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, United States.
| | - Meharvan Singh
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research (IAADR), Center FOR HER, University of North Texas, Health Science Center, Fort Worth, TX 76107, United States
| |
Collapse
|
27
|
Jayaraman A, Christensen A, Moser VA, Vest RS, Miller CP, Hattersley G, Pike CJ. Selective androgen receptor modulator RAD140 is neuroprotective in cultured neurons and kainate-lesioned male rats. Endocrinology 2014; 155:1398-406. [PMID: 24428527 PMCID: PMC3959610 DOI: 10.1210/en.2013-1725] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The decline in testosterone levels in men during normal aging increases risks of dysfunction and disease in androgen-responsive tissues, including brain. The use of testosterone therapy has the potential to increase the risks for developing prostate cancer and or accelerating its progression. To overcome this limitation, novel compounds termed "selective androgen receptor modulators" (SARMs) have been developed that lack significant androgen action in prostate but exert agonist effects in select androgen-responsive tissues. The efficacy of SARMs in brain is largely unknown. In this study, we investigate the SARM RAD140 in cultured rat neurons and male rat brain for its ability to provide neuroprotection, an important neural action of endogenous androgens that is relevant to neural health and resilience to neurodegenerative diseases. In cultured hippocampal neurons, RAD140 was as effective as testosterone in reducing cell death induced by apoptotic insults. Mechanistically, RAD140 neuroprotection was dependent upon MAPK signaling, as evidenced by elevation of ERK phosphorylation and inhibition of protection by the MAPK kinase inhibitor U0126. Importantly, RAD140 was also neuroprotective in vivo using the rat kainate lesion model. In experiments with gonadectomized, adult male rats, RAD140 was shown to exhibit peripheral tissue-specific androgen action that largely spared prostate, neural efficacy as demonstrated by activation of androgenic gene regulation effects, and neuroprotection of hippocampal neurons against cell death caused by systemic administration of the excitotoxin kainate. These novel findings demonstrate initial preclinical efficacy of a SARM in neuroprotective actions relevant to Alzheimer's disease and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Anusha Jayaraman
- Davis School of Gerontology (A.J., A.C., R.S.V., C.J.P.) and Neuroscience Graduate Program (V.A.M., C.J.P.), University of Southern California, Los Angeles, California 90089; and Radius Health, Inc. (C.P.M., G.H.), Cambridge, Massachusetts 02139
| | | | | | | | | | | | | |
Collapse
|
28
|
Quillinan N, Deng G, Grewal H, Herson PS. Androgens and stroke: good, bad or indifferent? Exp Neurol 2014; 259:10-5. [PMID: 24512750 DOI: 10.1016/j.expneurol.2014.02.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/28/2014] [Accepted: 02/02/2014] [Indexed: 12/16/2022]
Abstract
Cerebral ischemia caused by loss of blood supply to the brain during cardiac arrest or stroke are major causes of death and disability. Biological sex is an important factor in predicting vulnerability of the brain to an ischemic insult, with males being at higher risk for cardio-cerebrovascular events than females of the same age. However, relative incidence of stroke between the genders appears to normalize at advanced ages. Therefore, many scientists have focused on the mechanisms of sex differences in outcome following brain ischemic injury, with a particular emphasis on the role of sex steroids. The majority of studies indicate that female sex steroids, such as estrogen and progesterone, play important roles in the relative neuroprotection following cerebral ischemia observed in females. However, less is known about male sex steroids and brain damage. This review describes the state of our knowledge of androgen-related contributions to neurological injury and recovery following cerebral ischemia that occurs following stroke. Experimental studies examining the effects of castration, androgenic agonists and antagonists and aging provide valuable insights into the role of androgens in clinical outcome following cerebrovascular events.
Collapse
Affiliation(s)
- Nidia Quillinan
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, USA
| | - Guiying Deng
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, USA
| | - Himmat Grewal
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, USA
| | - Paco S Herson
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO 80045, USA.
| |
Collapse
|
29
|
Basualto-Alarcón C, Varela D, Duran J, Maass R, Estrada M. Sarcopenia and Androgens: A Link between Pathology and Treatment. Front Endocrinol (Lausanne) 2014; 5:217. [PMID: 25566189 PMCID: PMC4270249 DOI: 10.3389/fendo.2014.00217] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022] Open
Abstract
Sarcopenia, the age-related loss of skeletal muscle mass and function, is becoming more prevalent as the lifespan continues to increase in most populations. As sarcopenia is highly disabling, being associated with increased risk of dependence, falls, fractures, weakness, disability, and death, development of approaches to its prevention and treatment are required. Androgens are the main physiologic anabolic steroid hormones and normal testosterone levels are necessary for a range of developmental and biological processes, including maintenance of muscle mass. Testosterone concentrations decline as age increase, suggesting that low plasma testosterone levels can cause or accelerate muscle- and age-related diseases, as sarcopenia. Currently, there is increasing interest on the anabolic properties of testosterone for therapeutic use in muscle diseases including sarcopenia. However, the pathophysiological mechanisms underlying this muscle syndrome and its relationship with plasma level of androgens are not completely understood. This review discusses the recent findings regarding sarcopenia, the intrinsic, and extrinsic mechanisms involved in the onset and progression of this disease and the treatment approaches that have been developed based on testosterone deficiency and their implications.
Collapse
Affiliation(s)
- Carla Basualto-Alarcón
- Programa de Anatomía y Biología del Desarrollo, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Diego Varela
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Javier Duran
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Rodrigo Maass
- Facultad de Medicina, Departamento de Morfofunción, Universidad Diego Portales, Santiago, Chile
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
- *Correspondence: Manuel Estrada, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Independencia 1027, Santiago 8389100, Chile e-mail:
| |
Collapse
|
30
|
Vest RS, Pike CJ. Gender, sex steroid hormones, and Alzheimer's disease. Horm Behav 2013; 63:301-7. [PMID: 22554955 PMCID: PMC3413783 DOI: 10.1016/j.yhbeh.2012.04.006] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 03/24/2012] [Accepted: 04/11/2012] [Indexed: 02/07/2023]
Abstract
Age-related loss of sex steroid hormones is a established risk factor for the development of Alzheimer's disease (AD) in women and men. While the relationships between the sex steroid hormones and AD are not fully understood, findings from both human and experimental paradigms indicate that depletion of estrogens in women and androgens in men increases vulnerability of the aging brain to AD pathogenesis. We review evidence of a wide range of beneficial neural actions of sex steroid hormones that may contribute to their hypothesized protective roles against AD. Both estrogens and androgens exert general neuroprotective actions relevant to a several neurodegenerative conditions, some in a sex-specific manner, including protection from neuron death and promotion of select aspects of neural plasticity. In addition, estrogens and androgens regulate key processes implicated in AD pathogenesis, in particular the accumulation of β-amyloid protein. We discuss evidence of hormone-specific mechanisms related to the regulation of the production and clearance of β-amyloid as critical protective pathways. Continued elucidation of these pathways promises to yield effective hormone-based strategies to delay development of AD.
Collapse
Affiliation(s)
- Rebekah S Vest
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | | |
Collapse
|
31
|
Drummond ES, Martins RN, Handelsman DJ, Harvey AR. Altered expression of Alzheimer's disease-related proteins in male hypogonadal mice. Endocrinology 2012; 153:2789-99. [PMID: 22514046 DOI: 10.1210/en.2011-2003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Age-related depletion of estrogens and androgens is associated with an increase in Alzheimer's disease (AD) brain pathology and diminished cognitive function. Here we investigated AD-associated molecular and cellular changes in brains of aged hypogonadal (hpg) male and female mice. hpg Mice have a spontaneous, inactivating genetic mutation in the GnRH gene resulting in life-long deficiency of gonadotropins and gonadal sex hormones. Western blot analysis revealed low levels of amyloid precursor protein and high levels of presenilin 1, amyloid precursor protein C-terminal fragment, and β-amyloid 42 in brains of aged male, but not female, hpg mice. Changes were confined to the hippocampus and were not evident in the cerebellum or other brain tissues. Male hpg mice tended to have lower levels of IL-1β protein than male littermate controls. Immunohistochemical staining of the basal forebrain revealed that male hpg mice had lower choline acetyltransferase levels per neuron compared with controls. These AD-like changes specific to male hpg mice supports a link between androgen depletion and the development of AD pathology.
Collapse
Affiliation(s)
- Eleanor S Drummond
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Western Australia, 6009 Australia.
| | | | | | | |
Collapse
|
32
|
Rosario ER, Carroll JC, Pike CJ. Evaluation of the effects of testosterone and luteinizing hormone on regulation of β-amyloid in male 3xTg-AD mice. Brain Res 2012; 1466:137-45. [PMID: 22587890 DOI: 10.1016/j.brainres.2012.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 04/12/2012] [Accepted: 05/05/2012] [Indexed: 01/15/2023]
Abstract
During normal aging, men experience a significant decline in testosterone levels and a compensatory elevation in levels of gonadotropin luteinizing hormone (LH). Both low testosterone and elevated LH have been identified as significant risk factors for the development of Alzheimer's disease (AD) in men. It is unclear whether changes in testosterone or LH primarily underlie the relationship with AD, and therefore may be a more suitable therapeutic target. To examine this issue, we compared levels of β-amyloid (Aβ) immunoreactivity in male 3xTg-AD mice under varying experimental conditions associated with relatively low or high levels of testosterone and/or LH. In gonadally intact mice, Aβ accumulation increased after treatment with the gonadotropin-releasing hormone agonist leuprolide, which inhibits the hypothalamic-pituitary-gonadal (HPG) axis and reduces both testosterone and LH levels. In gonadectomized (GDX) mice with low testosterone and high LH, we also observed increased Aβ levels. Treatment of GDX mice with testosterone significantly reduced Aβ levels. In contrast, leuprolide did not significantly decrease Aβ levels and moreover, inhibited the Aβ-lowering effect of testosterone. Evaluation of hippocampal-dependent behavior revealed parallel findings, with performance in GDX mice improved by testosterone but not leuprolide. These data suggest that Aβ-lowering actions of testosterone are mediated directly by androgen pathways rather than indirectly via regulation of LH and the HPG axis. These findings support the clinical evaluation of androgen therapy in the prevention and perhaps treatment of AD in hypogonadal men.
Collapse
Affiliation(s)
- Emily R Rosario
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | |
Collapse
|
33
|
Aras R, Barron AM, Pike CJ. Caspase activation contributes to astrogliosis. Brain Res 2012; 1450:102-15. [PMID: 22436850 PMCID: PMC3319728 DOI: 10.1016/j.brainres.2012.02.056] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 01/30/2012] [Accepted: 02/04/2012] [Indexed: 10/28/2022]
Abstract
Caspases, a family of cysteine proteases, are widely activated in neurons and glia in the injured brain, a response thought to induce apoptosis. However, caspase activation in astrocytes following injury is not strongly associated with apoptosis. The present study investigates the potential role of caspase activation in astrocytes with another characteristic response to neural injury, astrogliosis. Caspase activity and morphological and biochemical indices of astrogliosis and apoptosis were assessed in (i) cultured neonatal rat astrocytes treated with astrogliosis-inducing stimuli (dibutryl cAMP, β-amyloid peptide), and (ii) cultures of adult rat hippocampal astrocytes generated from control and kainate-lesioned rats. The effects of broad spectrum and specific pharmacological caspase inhibitors were assessed on indicators of astrogliosis, including stellate morphology and expression of glutamine synthetase and fibroblast growth factor-2. Reactive neonatal and adult astrocytes demonstrated an increase in total caspase activity with a corresponding increase in the expression of active caspase-3 in the absence of cell death. Broad spectrum caspase inhibition with zVAD significantly attenuated increases in glutamine synthetase and fibroblast growth factor-2 in the reactive astrocytes. In the reactive neonatal astrocyte cultures, specific inhibition of caspases-3 and -11 also attenuated glutamine synthetase and fibroblast growth factor-2 expression, but did not reverse the morphological reactive phenotype. Astrogliosis is observed in all forms of brain injury and despite extensive study, its molecular triggers remain largely unknown. While previous studies have demonstrated active caspases in astrocytes following acute brain injury, here we present evidence functionally implicating the caspases in astrogliosis.
Collapse
Affiliation(s)
- Radha Aras
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089
| | - Anna M. Barron
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
34
|
Ota H, Akishita M, Akiyoshi T, Kahyo T, Setou M, Ogawa S, Iijima K, Eto M, Ouchi Y. Testosterone deficiency accelerates neuronal and vascular aging of SAMP8 mice: protective role of eNOS and SIRT1. PLoS One 2012; 7:e29598. [PMID: 22238626 PMCID: PMC3251570 DOI: 10.1371/journal.pone.0029598] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 12/01/2011] [Indexed: 01/14/2023] Open
Abstract
Oxidative stress and atherosclerosis-related vascular disorders are risk factors for cognitive decline with aging. In a small clinical study in men, testosterone improved cognitive function; however, it is unknown how testosterone ameliorates the pathogenesis of cognitive decline with aging. Here, we investigated whether the cognitive decline in senescence-accelerated mouse prone 8 (SAMP8), which exhibits cognitive impairment and hypogonadism, could be reversed by testosterone, and the mechanism by which testosterone inhibits cognitive decline. We found that treatment with testosterone ameliorated cognitive function and inhibited senescence of hippocampal vascular endothelial cells of SAMP8. Notably, SAMP8 showed enhancement of oxidative stress in the hippocampus. We observed that an NAD+-dependent deacetylase, SIRT1, played an important role in the protective effect of testosterone against oxidative stress-induced endothelial senescence. Testosterone increased eNOS activity and subsequently induced SIRT1 expression. SIRT1 inhibited endothelial senescence via up-regulation of eNOS. Finally, we showed, using co-culture system, that senescent endothelial cells promoted neuronal senescence through humoral factors. Our results suggest a critical role of testosterone and SIRT1 in the prevention of vascular and neuronal aging.
Collapse
Affiliation(s)
- Hidetaka Ota
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| | - Takuyu Akiyoshi
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomoaki Kahyo
- Hamamatsu University School of Medicine, Department of Molecular Anatomy, Hamamatsu, Shizuoka, Japan
| | - Mitsutoshi Setou
- Hamamatsu University School of Medicine, Department of Molecular Anatomy, Hamamatsu, Shizuoka, Japan
| | - Sumito Ogawa
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Katsuya Iijima
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masato Eto
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yasuyoshi Ouchi
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
35
|
Cunningham RL, Macheda T, Watts LT, Poteet E, Singh M, Roberts JL, Giuffrida A. Androgens exacerbate motor asymmetry in male rats with unilateral 6-hydroxydopamine lesion. Horm Behav 2011; 60:617-24. [PMID: 21907204 PMCID: PMC3210335 DOI: 10.1016/j.yhbeh.2011.08.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 08/12/2011] [Accepted: 08/19/2011] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by dopamine neuron loss in the nigrostriatal pathway that shows greater incidence in men than women. The mechanisms underlying this gender bias remain elusive, although one possibility is that androgens may increase dopamine neuronal vulnerability to oxidative stress. Motor impairment can be modeled in rats receiving a unilateral injection of 6-hydroxydopamine (6-OHDA), a neurotoxin producing nigrostriatal degeneration. To investigate the role of androgens in PD, we compared young (2 months) and aged (24 months) male rats receiving gonadectomy (GDX) and their corresponding intact controls. One month after GDX, rats were unilaterally injected with 6-OHDA, and their motor impairment and asymmetry were assessed 2 weeks later using the cylinder test and the amphetamine-induced rotation test. Plasma samples were also collected to assess the concentration of testosterone and advanced oxidation protein products, a product of oxidative stress. GDX decreased lesion-induced asymmetry along with oxidative stress and increased amphetamine-induced rotations. These results show that GDX improves motor behaviors by decreasing motor asymmetry in 6-OHDA-treated rats, an effect that may be ascribed to increased release of striatal dopamine and decreased oxidative stress. Collectively, the data support the hypothesis that androgens may underlie the gender bias observed in PD.
Collapse
Affiliation(s)
- Rebecca L Cunningham
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Shiota M, Yokomizo A, Naito S. Oxidative stress and androgen receptor signaling in the development and progression of castration-resistant prostate cancer. Free Radic Biol Med 2011; 51:1320-8. [PMID: 21820046 DOI: 10.1016/j.freeradbiomed.2011.07.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 07/09/2011] [Accepted: 07/13/2011] [Indexed: 11/25/2022]
Abstract
Aberrant androgen receptor (AR) signaling plays a critical role in androgen-dependent prostate cancer (PCa), as well as in castration-resistant PCa (CRPC). Oxidative stress seems to contribute to the tumorigenesis and progression of PCa, as well as the development of CRPC, via activation of AR signaling. This notion is supported by the fact that there is an aberrant or improper regulation of the redox status in these disorders. Additionally, androgen-deprivation-induced oxidative stress seems to be involved in the pathogenesis of several disorders caused by androgen-deprivation therapy (ADT), including osteoporosis, neurodegenerative disease, and cardiovascular disease. Oxidative stress can be suppressed with antioxidants or via a reduction in reactive oxygen species production. Thus, developing new therapeutic agents that reduce oxidative stress might be useful in preventing the conversion of androgen-dependent PCa into CRPC, as well as reducing the adverse effects associated with ADT. The objective of this review is to provide an overview regarding the relationship between oxidative stress and AR signaling in the context of PCa and especially CRPC. Additionally, we discuss the potential use of antioxidant therapies in the treatment of PCa.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | |
Collapse
|
37
|
Thompson CK. Cell death and the song control system: A model for how sex steroid hormones regulate naturally-occurring neurodegeneration. Dev Growth Differ 2011; 53:213-24. [DOI: 10.1111/j.1440-169x.2011.01257.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
38
|
Thompson CK, Brenowitz EA. Neuroprotective effects of testosterone in a naturally occurring model of neurodegeneration in the adult avian song control system. J Comp Neurol 2010; 518:4760-70. [PMID: 20963827 PMCID: PMC2963470 DOI: 10.1002/cne.22486] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Seasonal regression of the avian song control system, a series of discrete brain nuclei that regulate song learning and production, serves as a useful model for investigating the neuroprotective effects of steroids. In seasonally breeding male songbirds, the song control system regresses rapidly when males are transferred from breeding to nonbreeding physiological conditions. One nucleus in particular, the HVC, regresses in volume by 22% within days of castration and transfer to a nonbreeding photoperiod. This regression is mediated primarily by a 30% decrease in neuron number, a result of a caspase-dependent process of programmed cell death. Here we examine whether testosterone (T) can act locally in the brain to prevent seasonal-like neurodegeneration in HVC. We began to infuse T intracerebrally near HVC on one side of the brain in breeding-condition male white-crowned sparrows 2 days prior to T withdrawal and shifting them to short-day photoperiods. The birds were killed 3 or 7 days later. Local T infusion significantly protected ipsilateral HVC from volume regression and neuron loss. In addition, T infusion significantly reduced the number, density, and number/1,000 neurons of activated caspase-3 cells and cells positive for cleaved PARP, both markers for programmed cell death, in the ipsilateral HVC. T infusion near HVC also prevented regression of ipsilateral efferent targets of HVC neurons, including the volumes of robust nucleus of the arcopallium (RA) and Area X and the soma area and density of RA neurons. Thus T can act locally in the brain to have a neuroprotective effect and act transsynaptically to prevent regression of efferent nuclei.
Collapse
Affiliation(s)
- Christopher K Thompson
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington 98195-1525, USA.
| | | |
Collapse
|
39
|
Wagner J, Dusick JR, McArthur DL, Cohan P, Wang C, Swerdloff R, Boscardin WJ, Kelly DF. Acute gonadotroph and somatotroph hormonal suppression after traumatic brain injury. J Neurotrauma 2010; 27:1007-19. [PMID: 20214417 DOI: 10.1089/neu.2009.1092] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hormonal dysfunction is a known consequence of moderate and severe traumatic brain injury (TBI). In this study we determined the incidence, time course, and clinical correlates of acute post-TBI gonadotroph and somatotroph dysfunction. Patients had daily measurement of serum luteinizing hormone (LH), follicle-stimulating hormone (FSH), testosterone, estradiol, growth hormone, and insulin-like growth factor-1 (IGF-1) for up to 10 days post-injury. Values below the fifth percentile of a healthy cohort were considered abnormal, as were non-measurable growth hormone (GH) values. Outcome measures were frequency and time course of hormonal suppression, injury characteristics, and Glasgow Outcome Scale (GOS) score. The cohort consisted of 101 patients (82% males; mean age 35 years; Glasgow Coma Scale [GCS] score <or=8 in 87%). In men, 100% had at least one low testosterone value, and 93% of all values were low; in premenopausal women, 43% had at least one low estradiol value, and 39% of all values were low. Non-measurable GH levels occurred in 38% of patients, while low IGF-1 levels were observed in 77% of patients, but tended to normalize within 10 days. Multivariate analysis revealed associations of younger age with low FSH and low IGF-1, acute anemia with low IGF-1, and older age and higher body mass index (BMI) with low GH. Hormonal suppression was not predictive of GOS score. These results indicate that within 10 days of complicated mild, moderate, and severe TBI, testosterone suppression occurs in all men and estrogen suppression occurs in over 40% of women. Transient somatotroph suppression occurs in over 75% of patients. Although this acute neuroendocrine dysfunction may not be TBI-specific, low gonadal steroids, IGF-1, and GH may be important given their putative neuroprotective functions.
Collapse
Affiliation(s)
- Justin Wagner
- University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Androgens can protect neurones from injury, although androgen neuroprotection is not well characterised in terms of either specificity or mechanism. In the present study, we compared the ability of androgens to protect neurones against a panel of insults, empirically determined to induce cell death by apoptotic or non-apoptotic mechanisms. Three criteria defining but not inclusive of apoptosis are: protection by caspase inhibition, protection by protein synthesis inhibition and the presence of pyknotic nuclei. According to these criteria, beta-amyloid, staurosporine, and Apoptosis Activator II induced cell death involving apoptosis, whereas hydrogen peroxide (H(2)O(2)), iron, calcium ionophore and 3-nitropropionic acid induced cell death featuring non-apoptotic characteristics. Pretreatment of hippocampal neurones with testosterone or dihydrotestosterone attenuated cell death induced by beta-amyloid, staurosporine and Apoptosis Activator II, but none of the other insults. The anti-oxidant Trolox did not reduce cell death induced by beta-amyloid, staurosporine and Apoptosis Activator II, but did protect against H(2)O(2) and iron. Similarly, a supra-physiological concentration of oestrogen reduced cell death induced by H(2)O(2) and iron, an effect not observed with androgens. We also show that activation of oestrogen pathways was not necessary for androgen neuroprotection. These data suggest that androgens directly activate a neuroprotective mechanism specific to inhibition of cell death involving apoptosis. Determining the specificity of androgen neuroprotection may enable the development of androgen compounds for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- T V Nguyen
- Neuroscience Graduate Programme and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089-0191, USA
| | | | | | | |
Collapse
|
41
|
Rosario ER, Carroll J, Pike CJ. Testosterone regulation of Alzheimer-like neuropathology in male 3xTg-AD mice involves both estrogen and androgen pathways. Brain Res 2010; 1359:281-90. [PMID: 20807511 DOI: 10.1016/j.brainres.2010.08.068] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 08/20/2010] [Accepted: 08/22/2010] [Indexed: 01/20/2023]
Abstract
Normal, age-related depletion of the androgen testosterone is a risk factor for Alzheimer's disease (AD) in men. Previously, we reported that experimental androgen depletion significantly accelerates development of AD-like neuropathology in the 3xTg-AD triple-transgenic mouse model of AD, an effect prevented by androgen treatment. Because testosterone is metabolized in brain into both the androgen dihydrotestosterone (DHT) and the estrogen 17β-estradiol (E2), testosterone can mediate its effects through androgen and or estrogen pathways. To define the role of androgen and estrogen pathways in regulation of AD-like neuropathology, we compared the effects of testosterone (T) and its metabolites DHT and E2 in male 3xTg-AD mice depleted of endogenous sex steroid hormones by gonadectomy (GDX). Male 3xTg-AD mice were sham GDX or GDX, immediately treated with vehicle, T, DHT, or E2, and 4 months later evaluated for two indices of AD-like neuropathology, β-amyloid (Aβ) accumulation and tau hyperphosphorylation. In comparison to sham GDX mice, we observed a significant increase in Aβ accumulation in GDX mice in subiculum, hippocampus, and amygdala. Treatment of GDX mice with T prevented the increased Aβ accumulation in all three brain regions. DHT treatment yielded similar results, significantly reducing Aβ accumulation across brain regions. Interestingly, E2 prevented Aβ accumulation in hippocampus but exerted only partial effects in subiculum and amygdala. Levels of tau hyperphosphorylation in sham GDX male 3xTg-AD mice were modest and only slightly increased by GDX. Treatment of GDX mice with T or E2 but not DHT reduced tau hyperphosphorylation to levels lower than observed in sham animals. These data suggest that testosterone regulates Aβ pathology through androgen and estrogen pathways and reduces tau pathology largely through estrogen pathways. These findings further define hormone pathways involved in regulation of AD-related pathology, information that is important for understanding disease etiology and developing pathway-specific hormone interventions.
Collapse
Affiliation(s)
- Emily R Rosario
- Neuroscience Graduate Program, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | | | | |
Collapse
|
42
|
Gillies GE, McArthur S. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol Rev 2010; 62:155-98. [PMID: 20392807 PMCID: PMC2879914 DOI: 10.1124/pr.109.002071] [Citation(s) in RCA: 511] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The classic view of estrogen actions in the brain was confined to regulation of ovulation and reproductive behavior in the female of all mammalian species studied, including humans. Burgeoning evidence now documents profound effects of estrogens on learning, memory, and mood as well as neurodevelopmental and neurodegenerative processes. Most data derive from studies in females, but there is mounting recognition that estrogens play important roles in the male brain, where they can be generated from circulating testosterone by local aromatase enzymes or synthesized de novo by neurons and glia. Estrogen-based therapy therefore holds considerable promise for brain disorders that affect both men and women. However, as investigations are beginning to consider the role of estrogens in the male brain more carefully, it emerges that they have different, even opposite, effects as well as similar effects in male and female brains. This review focuses on these differences, including sex dimorphisms in the ability of estradiol to influence synaptic plasticity, neurotransmission, neurodegeneration, and cognition, which, we argue, are due in a large part to sex differences in the organization of the underlying circuitry. There are notable sex differences in the incidence and manifestations of virtually all central nervous system disorders, including neurodegenerative disease (Parkinson's and Alzheimer's), drug abuse, anxiety, and depression. Understanding the cellular and molecular basis of sex differences in brain physiology and responses to estrogen and estrogen mimics is, therefore, vitally important for understanding the nature and origins of sex-specific pathological conditions and for designing novel hormone-based therapeutic agents that will have optimal effectiveness in men or women.
Collapse
Affiliation(s)
- Glenda E Gillies
- Centre for Neuroscience, Department of Medicine, Hammersmith Hospital, Imperial College Faculty of Medicine, DuCane Road, London W12ONN, UK.
| | | |
Collapse
|
43
|
Cunningham RL, Giuffrida A, Roberts JL. Androgens induce dopaminergic neurotoxicity via caspase-3-dependent activation of protein kinase Cdelta. Endocrinology 2009; 150:5539-48. [PMID: 19837873 PMCID: PMC2795716 DOI: 10.1210/en.2009-0640] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aged men have a greater incidence of Parkinson's disease (PD) than women. PD is a neurodegenerative condition associated with the loss of dopamine neurons in the nigrostriatal pathway. This study examined the neurotoxic effects of androgens in a dopaminergic cell line (N27 cells) and the downstream signaling pathways activated by androgens. Treatment of N27 cells with testosterone- and dihydrotestosterone-induced mitochondrial dysfunction, protein kinase C (PKC)-delta cleavage, and apoptosis in dopaminergic neuronal cells. Inhibition of caspase-3 prevented the cleavage of PKCdelta from the full-length element to the catalytic fragment and apoptosis in N27 cells, suggesting that androgen-induced apoptosis is mediated by caspase-3-dependent activation of PKCdelta. Androgen-induced apoptosis may be specific to dopamine neurons as evidenced by a lack of testosterone-induced apoptosis in GnRH neurons. These results support a neurotoxic consequence of testosterone on dopaminergic neurons and may provide insight into the gender bias found in PD.
Collapse
Affiliation(s)
- Rebecca L Cunningham
- Department of Pharmacology and the Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas 78229, USA.
| | | | | |
Collapse
|
44
|
Pereno G, Beltramino C. Differential role of gonadal hormones on kainic acid–induced neurodegeneration in medial amygdaloid nucleus of female and male rats. Neuroscience 2009; 163:952-63. [DOI: 10.1016/j.neuroscience.2009.06.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 06/24/2009] [Accepted: 06/27/2009] [Indexed: 11/30/2022]
|
45
|
Fargo KN, Foster AM, Sengelaub DR. Neuroprotective effect of testosterone treatment on motoneuron recruitment following the death of nearby motoneurons. Dev Neurobiol 2009; 69:825-35. [PMID: 19658088 PMCID: PMC2747250 DOI: 10.1002/dneu.20743] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Motoneuron loss is a significant medical problem, capable of causing severe movement disorders or even death. We have previously shown that motoneuron death induces marked dendritic atrophy in surviving nearby motoneurons. Additionally, in quadriceps motoneurons, this atrophy is accompanied by decreases in motor nerve activity. However, treatment with testosterone partially attenuates changes in both the morphology and activation of quadriceps motoneurons. Testosterone has an even larger neuroprotective effect on the morphology of motoneurons of the spinal nucleus of the bulbocavernosus (SNB), in which testosterone treatment can completely prevent dendritic atrophy. The present experiment was performed to determine whether the greater neuroprotective effect of testosterone on SNB motoneuron morphology was accompanied by a greater neuroprotective effect on motor activation. Right side SNB motoneurons were killed by intramuscular injection of cholera toxin-conjugated saporin in adult male Sprague-Dawley rats. Animals were either given Silastic testosterone implants or left untreated. Four weeks later, left side SNB motor activation was assessed with peripheral nerve recording. The death of right side SNB motoneurons resulted in several changes in the electrophysiological response properties of surviving left side SNB motoneurons, including decreased background activity, increased response latency, increased activity duration, and decreased motoneuron recruitment. Treatment with exogenous testosterone attenuated the increase in activity duration and completely prevented the decrease in motoneuron recruitment. These data provide a functional correlate to the known protective effects of testosterone treatment on the morphology of these motoneurons, and further support a role for testosterone as a therapeutic agent in the injured nervous system.
Collapse
Affiliation(s)
- Keith N Fargo
- Research and Development Service, Edward Hines, Jr. VA Hospital, Hines, Illinois 60141, USA.
| | | | | |
Collapse
|
46
|
Nguyen TVV, Yao M, Pike CJ. Dihydrotestosterone activates CREB signaling in cultured hippocampal neurons. Brain Res 2009; 1298:1-12. [PMID: 19729001 DOI: 10.1016/j.brainres.2009.08.066] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 08/20/2009] [Accepted: 08/24/2009] [Indexed: 12/17/2022]
Abstract
Although androgens induce numerous actions in brain, relatively little is known about which cell signaling pathways androgens activate in neurons. Recent work in our laboratory showed that the androgens testosterone and dihydrotestosterone (DHT) activate androgen receptor (AR)-dependent mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling. Since the transcription factor cyclic AMP response element binding protein (CREB) is a downstream effector of MAPK/ERK and androgens activate CREB in non-neuronal cells, we investigated whether androgens activate CREB signaling in neurons. First, we observed that DHT rapidly activates CREB in cultured hippocampal neurons, as evidenced by CREB phosphorylation. Further, we observed that DHT-induced CREB phosphorylation is AR-dependent, as it occurs in PC12 cells stably transfected with AR but in neither wild-type nor empty vector-transfected cells. Next, we sought to identify the signal transduction pathways upstream of CREB phosphorylation using pharmacological inhibitors. DHT-induced CREB phosphorylation in neurons was found to be dependent upon protein kinase C (PKC) signaling but independent of MAPK/ERK, phosphatidylinositol 3-kinase, protein kinase A, and Ca(2+)/calmodulin-dependent protein kinase IV. These results demonstrate that DHT induces PKC-dependent CREB signaling, which may contribute to androgen-mediated neural functions.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | | | | |
Collapse
|
47
|
Fargo KN, Foecking EM, Jones KJ, Sengelaub DR. Neuroprotective actions of androgens on motoneurons. Front Neuroendocrinol 2009; 30:130-41. [PMID: 19393684 PMCID: PMC2726741 DOI: 10.1016/j.yfrne.2009.04.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 04/03/2009] [Accepted: 04/14/2009] [Indexed: 01/30/2023]
Abstract
Androgens have a variety of protective and therapeutic effects in both the central and peripheral nervous systems. Here we review these effects as they related specifically to spinal and cranial motoneurons. Early in development, androgens are critical for the formation of important neuromuscular sex differences, decreasing the magnitude of normally occurring cell death in select motoneuron populations. Throughout the lifespan, androgens also protect against motoneuron death caused by axonal injury. Surviving motoneurons also display regressive changes to their neurites as a result of both direct axonal injury and loss of neighboring motoneurons. Androgen treatment enhances the ability of motoneurons to recover from these regressive changes and regenerate both axons and dendrites, restoring normal neuromuscular function. Androgens exert these protective effects by acting through a variety of molecular pathways. Recent work has begun to examine how androgen treatment can interact with other treatment strategies in promoting recovery from motoneuron injury.
Collapse
Affiliation(s)
- Keith N. Fargo
- Research and Development Service; Edward Hines, Jr. VA Hospital; Hines, Illinois USA
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago; Maywood, Illinois, USA
| | - Eileen M. Foecking
- Research and Development Service; Edward Hines, Jr. VA Hospital; Hines, Illinois USA
- Department of Otolaryngology--Head and Neck Surgery, Loyola University Chicago; Maywood, Illinois, USA
| | - Kathryn J. Jones
- Research and Development Service; Edward Hines, Jr. VA Hospital; Hines, Illinois USA
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago; Maywood, Illinois, USA
- Department of Otolaryngology--Head and Neck Surgery, Loyola University Chicago; Maywood, Illinois, USA
| | - Dale R. Sengelaub
- Department of Psychological and Brain Sciences, and Program in Neuroscience, Indiana University, Bloomington, Indiana USA
| |
Collapse
|
48
|
Pike CJ, Carroll JC, Rosario ER, Barron AM. Protective actions of sex steroid hormones in Alzheimer's disease. Front Neuroendocrinol 2009; 30:239-58. [PMID: 19427328 PMCID: PMC2728624 DOI: 10.1016/j.yfrne.2009.04.015] [Citation(s) in RCA: 390] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 04/25/2009] [Accepted: 04/28/2009] [Indexed: 12/19/2022]
Abstract
Risk for Alzheimer's disease (AD) is associated with age-related loss of sex steroid hormones in both women and men. In post-menopausal women, the precipitous depletion of estrogens and progestogens is hypothesized to increase susceptibility to AD pathogenesis, a concept largely supported by epidemiological evidence but refuted by some clinical findings. Experimental evidence suggests that estrogens have numerous neuroprotective actions relevant to prevention of AD, in particular promotion of neuron viability and reduction of beta-amyloid accumulation, a critical factor in the initiation and progression of AD. Recent findings suggest neural responsiveness to estrogen can diminish with age, reducing neuroprotective actions of estrogen and, consequently, potentially limiting the utility of hormone therapies in aged women. In addition, estrogen neuroprotective actions are also modulated by progestogens. Specifically, continuous progestogen exposure is associated with inhibition of estrogen actions whereas cyclic delivery of progestogens may enhance neural benefits of estrogen. In recent years, emerging literature has begun to elucidate a parallel relationship of sex steroid hormones and AD risk in men. Normal age-related testosterone loss in men is associated with increased risk to several diseases including AD. Like estrogen, testosterone has been established as an endogenous neuroprotective factor that not only increases neuronal resilience against AD-related insults, but also reduces beta-amyloid accumulation. Androgen neuroprotective effects are mediated both directly by activation of androgen pathways and indirectly by aromatization to estradiol and initiation of protective estrogen signaling mechanisms. The successful use of hormone therapies in aging men and women to delay, prevent, and or treat AD will require additional research to optimize key parameters of hormone therapy and may benefit from the continuing development of selective estrogen and androgen receptor modulators.
Collapse
Affiliation(s)
- Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW To discuss the relationship between androgens, cognition and Alzheimer's disease. RECENT FINDINGS It has been found that low circulating levels of androgens are a risk factor for Alzheimer's disease. Decreased circulating androgens are also associated with declining cognitive performance, particularly in memory-related tasks. Conversely, androgen supplementation to hypogonadal men results in improved memory performance. It has therefore been hypothesized that androgen supplementation may be beneficial in Alzheimer's disease. In recent studies, animal models have been used to elucidate the molecular mechanism behind this relationship between androgens and Alzheimer's disease. These studies have shown that androgen depletion results in increased levels of beta amyloid and hyperphosphorylated tau, changes which are thought to be associated with subsequent neuronal death. SUMMARY Androgen depletion results in molecular changes associated with Alzheimer's disease. Further human trials are needed to determine whether androgen modulating therapy for Alzheimer's disease has clinical significance.
Collapse
Affiliation(s)
- Eleanor S Drummond
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, Australia
| | | | | |
Collapse
|
50
|
Little CM, Coons KD, Sengelaub DR. Neuroprotective effects of testosterone on the morphology and function of somatic motoneurons following the death of neighboring motoneurons. J Comp Neurol 2009; 512:359-72. [PMID: 19003970 PMCID: PMC2592503 DOI: 10.1002/cne.21885] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Motoneuron loss is a significant medical problem, capable of causing severe movement disorders or even death. We have previously shown that partial depletion of motoneurons from sexually dimorphic, highly androgen-sensitive spinal motor populations induces dendritic atrophy in remaining motoneurons, and this atrophy is attenuated by treatment with testosterone. To test whether testosterone has similar effects in more typical motoneurons, we examined potential neuroprotective effects in motoneurons innervating muscles of the quadriceps. Motoneurons innervating the vastus medialis muscle were selectively killed by intramuscular injection of cholera toxin-conjugated saporin. Simultaneously, some saporin-injected rats were given implants containing testosterone or left untreated. Four weeks later, motoneurons innervating the ipsilateral vastus lateralis muscle were labeled with cholera toxin-conjugated horseradish peroxidase, and dendritic arbors were reconstructed in three dimensions. Compared with intact normal males, partial motoneuron depletion resulted in decreased dendritic length in remaining quadriceps motoneurons, and this atrophy was attenuated by testosterone treatment. To examine the functional consequences of the induced dendritic atrophy, and its attenuation with testosterone treatment, the activation of remaining quadriceps motoneurons was assessed using peripheral nerve recording. Partial motoneuron depletion resulted in decreased amplitudes of motor nerve activity, and these changes were attenuated by treatment with testosterone, providing a functional correlate to the neuroprotective effects of testosterone treatment on quadriceps motoneuron morphology. Together these findings suggest that testosterone has neuroprotective effects on morphology and function in both highly androgen-sensitive as well as more typical motoneuron populations, further supporting a role for testosterone as a neurotherapeutic agent in the injured nervous system.
Collapse
Affiliation(s)
- Christine M. Little
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405
| | - Kellie D. Coons
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405
| | - Dale R. Sengelaub
- Program in Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405
| |
Collapse
|