1
|
Calvo N, Einstein G. Steroid hormones: risk and resilience in women's Alzheimer disease. Front Aging Neurosci 2023; 15:1159435. [PMID: 37396653 PMCID: PMC10313425 DOI: 10.3389/fnagi.2023.1159435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
More women have Alzheimer disease (AD) than men, but the reasons for this phenomenon are still unknown. Including women in clinical research and studying their biology is key to understand not just their increased risk but also their resilience against the disease. In this sense, women are more affected by AD than men, but their reserve or resilience mechanisms might delay symptom onset. The aim of this review was to explore what is known about mechanisms underlying women's risk and resilience in AD and identify emerging themes in this area that merit further research. We conducted a review of studies analyzing molecular mechanisms that may induce neuroplasticity in women, as well as cognitive and brain reserve. We also analyzed how the loss of steroid hormones in aging may be linked to AD. We included empirical studies with human and animal models, literature reviews as well as meta-analyses. Our search identified the importance of 17-b-estradiol (E2) as a mechanism driving cognitive and brain reserve in women. More broadly, our analysis revealed the following emerging perspectives: (1) the importance of steroid hormones and their effects on both neurons and glia for the study of risk and resilience in AD, (2) E2's crucial role in women's brain reserve, (3) women's verbal memory advantage as a cognitive reserve factor, and (4) E2's potential role in linguistic experiences such as multilingualism and hearing loss. Future directions for research include analyzing the reserve mechanisms of steroid hormones on neuronal and glial plasticity, as well as identifying the links between steroid hormone loss in aging and risk for AD.
Collapse
Affiliation(s)
- Noelia Calvo
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
- Tema Genus, Linköping University, Linköping, Sweden
- Women’s College Research Institute, Toronto, ON, Canada
- Centre for Life Course and Aging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Abstract
As men grow older, circulating testosterone concentrations decline, while prevalence of cognitive impairment and dementia increase. Epidemiological studies of middle-aged and older men have demonstrated associations of lower testosterone concentrations with higher prevalence and incidence of cognitive decline and dementia, including Alzheimer's disease. In observational studies, men with prostate cancer treated by androgen deprivation therapy had a higher risk of dementia. Small intervention studies of testosterone using different measures of cognitive function have provided inconsistent results, with some suggesting improvement. A randomised placebo-controlled trial of one year's testosterone treatment conducted in 788 men aged ≥ 65 years, baseline testosterone < 9.54 nmol/L, showed an improvement in sexual function, but no improvement in cognitive function. There is a known association between diabetes and dementia risk. A randomised placebo-controlled trial of two year's testosterone treatment in 1,007 men aged 50-74 years, waist circumference ≥ 95 cm, baseline testosterone ≤ 14 nmol/L, showed an effect of testosterone in reducing type 2 diabetes risk. There were no cognitive endpoints in that trial. Additional research is warranted but at this stage lower testosterone concentrations in ageing men should be regarded as a biomarker rather than a proven therapeutic target for risk reduction of cognitive decline and dementia, including Alzheimer's disease.
Collapse
Affiliation(s)
- Bu B Yeap
- Medical School, University of Western Australia, Perth, Australia.
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia.
| | - Leon Flicker
- Medical School, University of Western Australia, Perth, Australia
- Western Australian Centre for Health and Ageing, University of Western Australia, Perth, Australia
- Department of Geriatric Medicine, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
3
|
Nemours S, Castro L, Ribatallada-Soriano D, Semidey ME, Aranda M, Ferrer M, Sanchez A, Morote J, Cantero-Recasens G, Meseguer A. Temporal and sex-dependent gene expression patterns in a renal ischemia-reperfusion injury and recovery pig model. Sci Rep 2022; 12:6926. [PMID: 35484379 PMCID: PMC9051203 DOI: 10.1038/s41598-022-10352-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/30/2022] [Indexed: 12/30/2022] Open
Abstract
Men are more prone to acute kidney injury (AKI) and chronic kidney disease (CKD), progressing to end-stage renal disease (ESRD) than women. Severity and capacity to regenerate after AKI are important determinants of CKD progression, and of patient morbidity and mortality in the hospital setting. To determine sex differences during injury and recovery we have generated a female and male renal ischemia/reperfusion injury (IRI) pig model, which represents a major cause of AKI. Although no differences were found in blood urea nitrogen (BUN) and serum creatinine (SCr) levels between both sexes, females exhibited higher mononuclear infiltrates at basal and recovery, while males showed more tubular damage at injury. Global transcriptomic analyses of kidney biopsies from our IRI pig model revealed a sexual dimorphism in the temporal regulation of genes and pathways relevant for kidney injury and repair, which was also detected in human samples. Enrichment analysis of gene sets revealed five temporal and four sexual patterns governing renal IRI and recovery. Overall, this study constitutes an extensive characterization of the time and sex differences occurring during renal IRI and recovery at gene expression level and offers a template of translational value for further study of sexual dimorphism in kidney diseases.
Collapse
Affiliation(s)
- Stéphane Nemours
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Luis Castro
- Biomedical Research in Urology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Didac Ribatallada-Soriano
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Maria E Semidey
- Department of Pathology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Miguel Aranda
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Marina Ferrer
- Rodent Platform, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sanchez
- Unitat d'Estadística I Bioinformàtica, (UEB), Vall d'Hebron Research Institute, Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | - Joan Morote
- Biomedical Research in Urology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Gerard Cantero-Recasens
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Meseguer
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Bioquímica I Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Red de Investigación Renal (REDINREN), Instituto Carlos III-FEDER, Madrid, Spain.
| |
Collapse
|
4
|
Wen J, Patel C, Diglio F, Baker K, Marshall G, Li S, Cole PD. Cognitive impairment persists at least 1 year after juvenile rats are treated with methotrexate. Neuropharmacology 2022; 206:108939. [PMID: 34986414 PMCID: PMC8792316 DOI: 10.1016/j.neuropharm.2021.108939] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/21/2021] [Accepted: 12/28/2021] [Indexed: 11/24/2022]
Abstract
Methotrexate (MTX) is widely employed for children with cancer, but is also associated with persistent cognitive deficits among survivors. The present study investigated the mechanisms behind long-term cognitive dysfunction after juvenile animals are treated with MTX. Male and female Long-Evans rats were treated with a combination of 6 systemic doses (0.5 mg/kg/dose intraperitoneally) and 4 intrathecal doses (1 mg/kg) beginning at post-natal age 3 weeks, a schedule designed to mimic repeated exposure given to children with leukemia. Behavioral testing was conducted at 60-61 weeks of age, followed by analysis of brain histolopathology. This MTX regimen had no acute toxicity and no effect on growth. The spatial memory and visual memory deficits observed at 13 and 17 weeks of age persisted 1 year after MTX exposure in both females and males. Significantly decreased cell proliferation and increased hippocampal microglial activation were observed in MTX-treated females when compared to the controls, with a similar trend in the male groups. In addition, MTX treatment significantly increased the number of TUNEL positive cells in the periventricular area. Our study demonstrates that a clinically relevant regimen of systemic and intrathecal MTX induces persistent deficits in cognition, lasting approximately 1 year after the last injection. The mechanisms behind MTX-induced deficits are likely multifactorial, including suppression of neurogenesis, microglial activation, and increased brain cell apoptosis. Our study suggests female and male animals differ in susceptibility to MTX-induced neurotoxicity and provides insights for developing therapeutic approaches to prevent treatment related cognitive impairment among children with ALL.
Collapse
Affiliation(s)
- Jing Wen
- Department of Pediatrics, Goryeb Children's Hospital-Atlantic Health, Morristown, NJ, 07960, USA; Division of Pediatric Hematology/Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Chadni Patel
- Rutgers Graduate Program in Cellular and Molecular Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Frank Diglio
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Kayla Baker
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Gregory Marshall
- Rutgers Graduate Program in Cellular and Molecular Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Shengguo Li
- Division of Pediatric Hematology/Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Peter D Cole
- Division of Pediatric Hematology/Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
5
|
Reiss AB, Saeedullah U, Grossfeld DJ, Glass AD, Pinkhasov A, Katz AE. Prostate cancer treatment and the relationship of androgen deprivation therapy to cognitive function. Clin Transl Oncol 2021; 24:733-741. [PMID: 34743290 DOI: 10.1007/s12094-021-02727-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/23/2021] [Indexed: 02/01/2023]
Abstract
Prostate cancer is the second most common form of cancer in men. For advanced, high risk prostate cancer, androgen deprivation therapy (ADT) is the preferred treatment and can induce remission, but resistance to ADT brings biochemical recurrence and progression of cancer. ADT brings adverse effects such as erectile dysfunction, decreased libido, and diminished physical strength. It is estimated that between 25 and 50% of men on ADT manifest some form of cognitive dysfunction that may be self-reported or reported by a family member. There is concern that impaired cognitive function with ADT is due to loss of testosterone support. Testosterone and its metabolites are known to possess neuroprotective properties. While a direct causal relationship between ADT and cognitive decline in prostate cancer patients has not been established, this review describes the controversy surrounding the possible connection between ADT and neurocognitive deterioration. The cellular and molecular mechanisms believed to underlie the protection of neuronal integrity by androgens are discussed. Results from animal models and human clinical studies are presented. Finally, we call attention to lifestyle modifications that may minimize cognitive issues in prostate cancer patients.
Collapse
Affiliation(s)
- A B Reiss
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA.
| | - U Saeedullah
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - D J Grossfeld
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - A D Glass
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - A Pinkhasov
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - A E Katz
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| |
Collapse
|
6
|
Sumien N, Cunningham JT, Davis DL, Engelland R, Fadeyibi O, Farmer GE, Mabry S, Mensah-Kane P, Trinh OTP, Vann PH, Wilson EN, Cunningham RL. Neurodegenerative Disease: Roles for Sex, Hormones, and Oxidative Stress. Endocrinology 2021; 162:6360925. [PMID: 34467976 PMCID: PMC8462383 DOI: 10.1210/endocr/bqab185] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases cause severe impairments in cognitive and motor function. With an increasing aging population and the onset of these diseases between 50 and 70 years, the consequences are bound to be devastating. While age and longevity are the main risk factors for neurodegenerative diseases, sex is also an important risk factor. The characteristic of sex is multifaceted, encompassing sex chromosome complement, sex hormones (estrogens and androgens), and sex hormone receptors. Sex hormone receptors can induce various signaling cascades, ranging from genomic transcription to intracellular signaling pathways that are dependent on the health of the cell. Oxidative stress, associated with aging, can impact the health of the cell. Sex hormones can be neuroprotective under low oxidative stress conditions but not in high oxidative stress conditions. An understudied sex hormone receptor that can induce activation of oxidative stress signaling is the membrane androgen receptor (mAR). mAR can mediate nicotinamide adenine dinucleotide-phosphate (NADPH) oxidase (NOX)-generated oxidative stress that is associated with several neurodegenerative diseases, such as Alzheimer disease. Further complicating this is that aging can alter sex hormone signaling. Prior to menopause, women experience more estrogens than androgens. During menopause, this sex hormone profile switches in women due to the dramatic ovarian loss of 17β-estradiol with maintained ovarian androgen (testosterone, androstenedione) production. Indeed, aging men have higher estrogens than aging women due to aromatization of androgens to estrogens. Therefore, higher activation of mAR-NOX signaling could occur in menopausal women compared with aged men, mediating the observed sex differences. Understanding of these signaling cascades could provide therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Nathalie Sumien
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - J Thomas Cunningham
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Delaney L Davis
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rachel Engelland
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - George E Farmer
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Steve Mabry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Paapa Mensah-Kane
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Oanh T P Trinh
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: Rebecca L. Cunningham, PhD, Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, TX, USA, 76107-2699.
| |
Collapse
|
7
|
Yawson EO, Akinola OB. Hippocampal cellular changes in androgen deprived insulin resistant rats. Metab Brain Dis 2021; 36:1037-1048. [PMID: 33666820 DOI: 10.1007/s11011-021-00678-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Androgen deprivation can be achieved through testosterone antagonists (chemical castration) with or without orchidectomy. We use a rat model to characterize hippocampal structural and functional changes that might be associated with a subset population of androgen deprived insulin-resistant patients. Adult male Wistar rats assigned into six (6) groups: control group (distilled water/sham), orchiectomy group (bilateral orchiectomy), flutamide group (oral flutamide; 11 mg/kg body weight), diabetes group (multiple low-dose of streptozotocin (STZ; 30 mg/kg body weight intraperitoneally), orchiectomy and diabetic group (bilateral orchiectomy with 30 mg/kg body weight of STZ), and orchiectomy/diabetic/flutamide group (bilateral orchiectomy with 30 mg/kg body weight of STZ with 11 mg/kg body weight of flutamide). Animals were sacrificed at 30 and 60 days respectively. Spatial learning and working memory behavior were assessed; while total plasma; testosterone, insulin levels, and fasting blood glucose were assayed; the Homeostasis model for insulin resistance was also calculated. Histological examinations by H&E and CFV, while immunohistochemical analysis of astrocytes, P53 protein, and NSE were performed. Androgen deprived insulin-resistant state caused altered learning and cognitive behavior through decreased percentage correct alternation to an increased escape latency period. Significant bidirectional correlates exist between the hormonal profiles relative to the control group (p < 0.05), especially in the 60 days post-orchiectomy. While histological and immunohistochemical data indicate microcellular derangement. That the summate effects of androgen deprivation and impaired insulin signaling exacerbate hippocampal neurodegenerative changes that merit further studies.
Collapse
Affiliation(s)
- Emmanuel O Yawson
- Neuroendocrinology division, Department of Anatomy, University of Ilorin, Ilorin, Nigeria
| | - Oluwole B Akinola
- Neuroendocrinology division, Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
8
|
Almaguer-Mederos LE, Aguilera-Rodríguez R, Almaguer-Gotay D, Hechavarría-Barzaga K, Álvarez-Sosa A, Chapman-Rodríguez Y, Silva-Ricardo Y, González-Zaldivar Y, Vázquez-Mojena Y, Cuello-Almarales D, Rodríguez-Estupiñán A. Testosterone Levels Are Decreased and Associated with Disease Duration in Male Spinocerebellar Ataxia Type 2 Patients. THE CEREBELLUM 2021; 19:597-604. [PMID: 32440846 DOI: 10.1007/s12311-020-01134-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Spinocerebellar ataxia type 2 (SCA2) is a progressive neurodegenerative disorder due to an unstable expansion of a CAG repeat in the ATXN2 gene. Despite clinical and experimental evidence indicating the relevance of the gonadotropic axis to the prognosis and therapeutics for several late-onset neurodegenerative disorders, its functioning and association with disease severity have not been previously explored in SCA2. To assess serum levels of testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH), and their clinical relevance in SCA2 patients. A case-control study involving 94 Cuban SCA2 patients and 101 gender- and age-matched healthy controls was conducted. Testosterone, LH, and FSH serum levels were determined by radioimmunoassay or immunoradiometric assay systems. Clinical outcomes included age at onset, disease duration, Scale for the Assessment and Rating of Ataxia (SARA) score, and progression rate. Univariate general linear models were generated. Testosterone, LH, and FSH serum levels were significantly reduced in male SCA2 patients relative to control individuals. On average, there was a 35% reduction in testosterone levels in male patients versus male control individuals. Testosterone levels were associated with disease duration (r = 0.383; p = 0.025) and age at onset (r = 0.414; p = 0.011) in male SCA2 patients, but no association was observed between testosterone and CAG expansion size, SARA score, or progression rate. Testosterone levels might be a biomarker of disease progression in male SCA2 patients. Further studies are needed to explore the effects of low testosterone levels on non-motor symptoms, and to assess the potential of testosterone replacement therapy in male SCA2 patients.
Collapse
Affiliation(s)
- Luis E Almaguer-Mederos
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba.
| | - Raúl Aguilera-Rodríguez
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | - Dennis Almaguer-Gotay
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | | | | | | | | | | | - Yaimé Vázquez-Mojena
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | - Dany Cuello-Almarales
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | | |
Collapse
|
9
|
Wen C, Yu Y, Gao C, Qi X, Cardona CJ, Xing Z. RIPK3-Dependent Necroptosis Is Induced and Restricts Viral Replication in Human Astrocytes Infected With Zika Virus. Front Cell Infect Microbiol 2021; 11:637710. [PMID: 33796483 PMCID: PMC8007970 DOI: 10.3389/fcimb.2021.637710] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
Apoptosis, pyroptosis and necroptosis are regulated processes of cell death which can be crucial for viral disease outcomes in hosts because of their effects on viral pathogenicity and host resistance. Zika virus (ZIKV) is a mosquito-borne flavivirus, which infects humans and can cause neurological disorders. Neural developmental disorders and microcephaly could occur in infected fetuses. Several types of nervous cells have been reported to be susceptible to ZIKV infection. Human astrocytes play important roles in the nutritional support and defense of neurons. In this study, we show that human astrocytes are susceptible to ZIKV infection and undergo progressive cell death after infection. In infected astrocytes we detected no cleavage or activation of pro-caspase-3 and pro-caspase-1. Apoptotic substrates and increased secretion of interleukin (IL)-1β or IL-18 were not detected, either. These ruled out the occurrence of apoptosis or pyroptosis in ZIKV-infected astrocytes. We detected, however, an increase of phosphorylated receptor-interacting serine/threonine-protein kinase (RIPK)1, RIPK3, and mixed lineage kinase domain-like (MLKL) protein, indicating that programmed necrosis, or necroptosis, was induced in infected astrocytes. The phosphorylation and cell death were inhibited in cells pre-treated with GSK’872, an inhibitor of RIPK3, while inhibition of RIPK1 with an inhibitor, Necrostatin-1, had no effect, suggesting that ZIKV-induced necroptosis was RIPK1-independent in astrocytes. Consistent with this finding, the inhibition of RIPK1 had no effect on the phosphorylation of MLKL. We showed evidence that MLKL phosphorylation was RIPK3-dependent and ZBP-1, which could stimulate RIPK3, was upregulated in ZIKV-infected astrocytes. Finally, we demonstrated that in GSK’872-pre-treated astrocytes, viral replication increased significantly, which indicates that necroptosis may be protective against viral replication in astrocytes. Our finding that astrocytes uniquely underwent necroptosis in response to ZIKV infection provides insight and helps us better understand the viral pathogenesis in the ZIKV-infected central nervous system.
Collapse
Affiliation(s)
- Chunxia Wen
- Medical School, Jiangsu Provincial Key Laboratory of Medicine, and the State Key Laboratory of Pharmaceutical Technology, Nanjing University, Nanjing, China
| | - Yufeng Yu
- Medical School, Jiangsu Provincial Key Laboratory of Medicine, and the State Key Laboratory of Pharmaceutical Technology, Nanjing University, Nanjing, China
| | - Chengfeng Gao
- Medical School, Jiangsu Provincial Key Laboratory of Medicine, and the State Key Laboratory of Pharmaceutical Technology, Nanjing University, Nanjing, China
| | - Xian Qi
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Carol J Cardona
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Minnesota at Twin Cities, Saint Paul, MN, United States
| | - Zheng Xing
- Medical School, Jiangsu Provincial Key Laboratory of Medicine, and the State Key Laboratory of Pharmaceutical Technology, Nanjing University, Nanjing, China.,Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Minnesota at Twin Cities, Saint Paul, MN, United States
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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: 38] [Impact Index Per Article: 7.6] [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
|
14
|
Fattoretti P, Malatesta M, Mariotti R, Zancanaro C. Testosterone administration increases synaptic density in the gyrus dentatus of old mice independently of physical exercise. Exp Gerontol 2019; 125:110664. [DOI: 10.1016/j.exger.2019.110664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/17/2019] [Accepted: 07/15/2019] [Indexed: 10/26/2022]
|
15
|
Babahajian A, Sarveazad A, Golab F, Vahabzadeh G, Alizadeh A, Rasoolijazi H, Amini N, Entezari M, Soleimani M, Katebi M, Haramshahi SMA. Neuroprotective Effects of Trolox, Human Chorionic Gonadotropin, and Carnosic Acid on Hippocampal Neurodegeneration After Ischemiareperfusion Injury. Curr Stem Cell Res Ther 2019; 14:177-183. [PMID: 30227824 DOI: 10.2174/1574888x13666180918093822] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/20/2018] [Accepted: 08/21/2018] [Indexed: 11/22/2022]
Abstract
INTRODUCTION One of the serious complications of stroke is memory impairment, which is considered as one of the complications of reperfusion of tissue. The present study was designed to compare the effect of administration of Trolox, carnosic acid and Human Chorionic Gonadotropin (HCG) immediately after reperfusion of the stroke tissue on the memory and hippocampal histology. METHOD Ischemia-Reperfusion Model (IRI) was created by bilateral occlusion of the common carotid artery for 15 minutes and the first dose was administered immediately after reperfusion. 10 days after ischemia, passive avoidance memory test and apoptotic protein levels were evaluated. RESULTS Cerebral Ischemia perfusion reduced the time of latency in entering the dark box in the ischemic group. Administration of Trolox and HCG increased this latency time, while treatment with carnosic acid had no effect. Also, IRI significantly reduced the number of healthy cells in the hippocampus. Administration of Trolox, carnosic acid and HCG increased the number of healthy cells and decreased the expression of Caspase-3 and Bax, but significantly increased the expression of Bcl-2 compared to the ischemic group. CONCLUSION Findings indicate the beneficial effects of HCG and Trolox on the improvement of memory and the number of healthy cells in the hippocampal region. It is worth noting that the amount of apoptosis in the hippocampus was significantly reduced by Trolox, HCG and Carnosic acid.
Collapse
Affiliation(s)
- Asrin Babahajian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Liver and Digestive Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Gelareh Vahabzadeh
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Homa Rasoolijazi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maedeh Entezari
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Katebi
- Department of Anatomy, Faculty of Medicine, Hormozgan University of Medical Sciences, Hormozgan, Iran
| | - Seyed Mohammad Amin Haramshahi
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Ciprés-Flores FJ, Segura-Uribe JJ, Orozco-Suárez S, Guerra-Araiza C, Guevara-Salazar JA, Castillo-García EL, Soriano-Ursúa MA, Farfán-García ED. Beta-blockers and salbutamol limited emotional memory disturbance and damage induced by orchiectomy in the rat hippocampus. Life Sci 2019; 224:128-137. [PMID: 30905783 DOI: 10.1016/j.lfs.2019.03.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
AIM To evaluate the therapeutic potential of ligands of beta-adrenoceptors in cognitive disorders. Testosterone and adrenergic pathways are involved in hippocampal and emotional memory. Moreover, is strongly suggested that androgen diminishing in aging is involved in cognitive deficit, as well as beta-adrenoceptors, particularly beta2-adrenoceptor, participate in the adrenergic modulation of memory. In this regard, some animal models of memory disruption have shown improved performance after beta-drug administration. MATERIAL AND METHODS In this work, we evaluated the effects of agonists (isoproterenol and salbutamol) and antagonists (propranolol and carvedilol) on beta-adrenoceptors in orchiectomized rats, as well as their effects in the performance on avoidance task and damage in hippocampal neurons by immunohistochemistry assays. KEY FINDINGS Surprisingly, we found that both antagonists and salbutamol (but not isoproterenol) modulate the effects of hormone deprivation, improving memory and decreasing neuronal death and amyloid-beta related changes in some regions (particularly CA1-3 and dentate gyrus) of rat hippocampus. SIGNIFICANCE Two β-antagonists and one β2-agonist modulated the effects of hormone deprivation on memory and damage in brain. The mechanisms of signaling of these drugs for beneficial effects remain unclear, even if used β-ARs ligands share a weak activity on β-arrestin/ERK-pathway activation which can be involved in these effects as we proposed in this manuscript. Our observations could be useful for understanding effects suggested of adrenergic drugs to modulate emotional memory. But also, our results could be related to other pathologies involving neuronal death and Aβ accumulation.
Collapse
Affiliation(s)
- Fabiola J Ciprés-Flores
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Mexico City, Mexico
| | - Julia J Segura-Uribe
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Mexico City, Mexico; Unidad de Investigación Médica en Enfermedades Neurológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Sandra Orozco-Suárez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Juan A Guevara-Salazar
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Mexico City, Mexico
| | - Emily L Castillo-García
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Mexico City, Mexico
| | - Marvin A Soriano-Ursúa
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Mexico City, Mexico.
| | - Eunice D Farfán-García
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Mexico City, Mexico.
| |
Collapse
|
17
|
Lei Y, Renyuan Z. Effects of Androgens on the Amyloid-β Protein in Alzheimer's Disease. Endocrinology 2018; 159:3885-3894. [PMID: 30215697 DOI: 10.1210/en.2018-00660] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/06/2018] [Indexed: 12/24/2022]
Abstract
Age-related androgen depletion has been implicated in compromised neuroprotection and is involved in the pathogenesis of neurodegenerative disease, including Alzheimer's disease (AD), the leading cause of dementia. Emerging data revealed that reduction of both serum and brain androgen levels in males is associated with increased amyloid-β (Aβ) accumulation, a putative cause of AD. It has been demonstrated that androgens can function as the endogenous negative regulators of Aβ. However, the mechanisms by which androgens regulate Aβ production, degradation, and clearance, as well as the Aβ-induced pathological process in AD, are still elusive. This review emphasizes the contributions of androgen to Aβ metabolism and toxicity in AD and thus may provide novel strategies for prevention and therapeutics.
Collapse
Affiliation(s)
- Yang Lei
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Zhou Renyuan
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Cardiomyocyte-specific disruption of Cathepsin K protects against doxorubicin-induced cardiotoxicity. Cell Death Dis 2018; 9:692. [PMID: 29880809 PMCID: PMC5992138 DOI: 10.1038/s41419-018-0727-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
The lysosomal cysteine protease Cathepsin K is elevated in humans and animal models of heart failure. Our recent studies show that whole-body deletion of Cathepsin K protects mice against cardiac dysfunction. Whether this is attributable to a direct effect on cardiomyocytes or is a consequence of the global metabolic alterations associated with Cathepsin K deletion is unknown. To determine the role of Cathepsin K in cardiomyocytes, we developed a cardiomyocyte-specific Cathepsin K-deficient mouse model and tested the hypothesis that ablation of Cathepsin K in cardiomyocytes would ameliorate the cardiotoxic side-effects of the anticancer drug doxorubicin. We used an α-myosin heavy chain promoter to drive expression of Cre, which resulted in over 80% reduction in protein and mRNA levels of cardiac Cathepsin K at baseline. Four-month-old control (Myh-Cre-; Ctskfl/fl) and Cathepsin K knockout (Myh-Cre+; Ctskfl/fl) mice received intraperitoneal injections of doxorubicin or vehicle, 1 week following which, body and tissue weight, echocardiographic properties, cardiomyocyte contractile function and Ca2+-handling were evaluated. Control mice treated with doxorubicin exhibited a marked increase in cardiac Cathepsin K, which was associated with an impairment in cardiac structure and function, evidenced as an increase in end-systolic and end-diastolic diameters, decreased fractional shortening and wall thickness, disruption in cardiac sarcomere and microfilaments and impaired intracellular Ca2+ homeostasis. In contrast, the aforementioned cardiotoxic effects of doxorubicin were attenuated or reversed in mice lacking cardiac Cathepsin K. Mechanistically, Cathepsin K-deficiency reconciled the disturbance in cardiac energy homeostasis and attenuated NF-κB signaling and apoptosis to ameliorate doxorubicin-induced cardiotoxicity. Cathepsin K may represent a viable drug target to treat cardiac disease.
Collapse
|
19
|
Mhaouty-Kodja S. Role of the androgen receptor in the central nervous system. Mol Cell Endocrinol 2018; 465:103-112. [PMID: 28826929 DOI: 10.1016/j.mce.2017.08.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/30/2017] [Accepted: 08/02/2017] [Indexed: 11/17/2022]
Abstract
The involvement of gonadal androgens in functions of the central nervous system was suggested for the first time about half a century ago. Since then, the number of functions attributed to androgens has steadily increased, ranging from regulation of the hypothalamic-pituitary-gonadal axis and reproductive behaviors to modulation of cognition, anxiety and other non-reproductive functions. This review focuses on the implication of the neural androgen receptor in these androgen-sensitive functions and behaviors.
Collapse
Affiliation(s)
- Sakina Mhaouty-Kodja
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 7 Quai St Bernard, 75005 Paris, France.
| |
Collapse
|
20
|
Massa MG, David C, Jörg S, Berg J, Gisevius B, Hirschberg S, Linker RA, Gold R, Haghikia A. Testosterone Differentially Affects T Cells and Neurons in Murine and Human Models of Neuroinflammation and Neurodegeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2017. [PMID: 28634006 DOI: 10.1016/j.ajpath.2017.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The high female-to-male sex ratio of multiple sclerosis (MS) prevalence has continuously confounded researchers, especially in light of male patients' accelerated disease course at later stages of MS. Although multiple studies have concentrated on estrogenic mechanisms of disease modulation, fairly little attention has been paid to androgenic effects in a female system, and even fewer studies have attempted to dissociate hormonal effects on the neurodegenerative and neuroinflammatory processes of MS. Herein, we demonstrate the differential effects of hormone treatment on the acute inflammatory and chronic neurodegenerative phases of murine experimental autoimmune encephalomyelitis. Although s.c. treatment with testosterone and aromatase inhibitor applied beginning on the day of immunization ameliorated initial course of disease, similar treatment administered therapeutically exacerbated chronic disease course. Spinal cord analyses of axonal densities reflected the clinical scores of the chronic phase. In vitro, testosterone treatment not only decreased Th1 and Th17 differentiation in an aromatase-independent fashion, but also exacerbated cell death in induced pluripotent stem cell-derived primary human neurons under oxidative stress conditions in an aromatase inhibitor-dependent manner. Thus, through the alleviation of inflammatory processes and the exacerbation of neurodegenerative processes, androgens may contribute to the epidemiologic sex differentials observed in MS prevalence and course.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Christina David
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Stefanie Jörg
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Berg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Barbara Gisevius
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Sarah Hirschberg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Ralf A Linker
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany.
| |
Collapse
|
21
|
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
|
22
|
Ruan Q, D'onofrio G, Wu T, Greco A, Sancarlo D, Yu Z. Sexual dimorphism of frailty and cognitive impairment: Potential underlying mechanisms (Review). Mol Med Rep 2017; 16:3023-3033. [PMID: 28713963 DOI: 10.3892/mmr.2017.6988] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 01/01/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to assess systematically gender differences in susceptibility to frailty and cognitive performance decline, and the underlying mechanisms. A systematic assessment was performed of the identified reviews of cohort, mechanistic and epidemiological studies. The selection criteria of the present study included: i) Sexual dimorphism of frailty, ii) sexual dimorphism of subjective memory decline (impairment) and atrophy of hippocampus during early life, iii) sexual dimorphism of late‑onset Alzheimer's disease and iv) sexual dimorphism mechanisms underlying frailty and cognitive impairment. Males exhibit a susceptibility to poor memory performance and a severe atrophy of the hippocampus during early life and females demonstrate a higher prevalence for frailty and late‑life dementia. The different alterations within the hypothalamic‑pituitary‑gonadal/adrenal axis, particularly with regard to gonadal hormones, cortisol and dehydroepiandrosterone/sulfate‑bound dehydroepiandrosterone prior to and following andropause in males and menopause in females, serve important roles in sexual dimorphism of frailty and cognitive impairment. These endocrine changes may accelerate immunosenescence, weaken neuroprotective and neurotrophic effects, and promote muscle catabolism. The present study suggested that these age‑associated endocrine alterations interact with gender‑specific genetic and epigenetic factors, together with immunosenescence and iron accumulation. Environment factors, including psychological factors, are additional potential causes of the sexual dimorphism of frailty and cognitive impairment.
Collapse
Affiliation(s)
- Qingwei Ruan
- Department of Geriatrics, Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
| | - Grazia D'onofrio
- Department of Medical Sciences, Geriatric Unit and Laboratory of Gerontology and Geriatrics, The Scientific Institute for Research and Health Care, Home for Relief of the Suffering Hospital, San Giovanni Rotondo, Foggia I‑71013, Italy
| | - Tao Wu
- Department of Geriatrics, Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
| | - Antonio Greco
- Department of Medical Sciences, Geriatric Unit and Laboratory of Gerontology and Geriatrics, The Scientific Institute for Research and Health Care, Home for Relief of the Suffering Hospital, San Giovanni Rotondo, Foggia I‑71013, Italy
| | - Daniele Sancarlo
- Department of Medical Sciences, Geriatric Unit and Laboratory of Gerontology and Geriatrics, The Scientific Institute for Research and Health Care, Home for Relief of the Suffering Hospital, San Giovanni Rotondo, Foggia I‑71013, Italy
| | - Zhuowei Yu
- Department of Geriatrics, Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
23
|
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
|
24
|
Huffman J, Hoffmann C, Taylor GT. Integrating insulin-like growth factor 1 and sex hormones into neuroprotection: Implications for diabetes. World J Diabetes 2017; 8:45-55. [PMID: 28265342 PMCID: PMC5320748 DOI: 10.4239/wjd.v8.i2.45] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/24/2016] [Accepted: 11/22/2016] [Indexed: 02/05/2023] Open
Abstract
Brain integrity and cognitive aptitude are often impaired in patients with diabetes mellitus, presumably a result of the metabolic complications inherent to the disease. However, an increasing body of evidence has demonstrated the central role of insulin-like growth factor 1 (IGF1) and its relation to sex hormones in many neuroprotective processes. Both male and female patients with diabetes display abnormal IGF1 and sex-hormone levels but the comparison of these fluctuations is seldom a topic of interest. It is interesting to note that both IGF1 and sex hormones have the ability to regulate phosphoinositide 3-kinase-Akt and mitogen-activated protein kinases-extracellular signal-related kinase signaling cascades in animal and cell culture models of neuroprotection. Additionally, there is considerable evidence demonstrating the neuroprotective coupling of IGF1 and estrogen. Androgens have also been implicated in many neuroprotective processes that operate on similar signaling cascades as the estrogen-IGF1 relation. Yet, androgens have not been directly linked to the brain IGF1 system and neuroprotection. Despite the sex-specific variations in brain integrity and hormone levels observed in diabetic patients, the IGF1-sex hormone relation in neuroprotection has yet to be fully substantiated in experimental models of diabetes. Taken together, there is a clear need for the comprehensive analysis of sex differences on brain integrity of diabetic patients and the relationship between IGF1 and sex hormones that may influence brain-health outcomes. As such, this review will briefly outline the basic relation of diabetes and IGF1 and its role in neuroprotection. We will also consider the findings on sex hormones and diabetes as a basis for separately analyzing males and females to identify possible hormone-induced brain abnormalities. Finally, we will introduce the neuroprotective interplay of IGF1 and estrogen and how androgen-derived neuroprotection operates through similar signaling cascades. Future research on both neuroprotection and diabetes should include androgens into the interplay of IGF1 and sex hormones.
Collapse
|
25
|
Mendell AL, Creighton CE, Kalisch BE, MacLusky NJ. 5α-Androstane-3α,17β-Diol Inhibits Neurotoxicity in SH-SY5Y Human Neuroblastoma Cells and Mouse Primary Cortical Neurons. Endocrinology 2016; 157:4570-4578. [PMID: 27754784 DOI: 10.1210/en.2016-1508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Low free T levels in men are associated with age-related cognitive decline and increased risk for neurotoxicity, resulting in disease. The mechanisms underlying these observations remain poorly defined. Although rapid, androgen receptor-dependent activation of ERK has been postulated as a neurotrophic and neuroprotective mechanism, actions of T metabolites such as 5α-androstane-3α,17β-diol (3α-diol) may also be involved. We investigated the influence of 3α-diol on the induction of ERK phosphorylation in SH-SY5Y human female neuroblastoma cells and primary cortical neurons from male and female mice. In SH-SY5Y cells, ERK phosphorylation was induced by 10 nM DHT, epidermal growth factor, hydrogen peroxide (H2O2), and acetylcholine. The addition of 10 nM 3α-diol, which did not itself activate ERK, significantly inhibited ERK phosphorylation induced by DHT, epidermal growth factor, or H2O2, but not acetylcholine. In both SH-SY5Y cells and primary cortical neurons, prolonged ERK phosphorylation and caspase-3 cleavage resulting from amyloid β-peptide 1-42 (Aβ42) exposure were inhibited by cotreatment with 3α-diol. 3α-diol also reduced the loss in cellular viability induced by Aβ42 or H2O2 in SH-SY5Y cells. These data suggest that T-mediated neuroprotection may occur via two distinct but complementary mechanisms: an initial rapid activation of ERK phosphorylation, followed by modulation via 3α-diol of the potentially adverse consequences of prolonged ERK activation.
Collapse
Affiliation(s)
- A L Mendell
- Department of Biomedical Sciences (A.L.M., C.E.C., B.E.K., N.J.M.), Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - C E Creighton
- Department of Biomedical Sciences (A.L.M., C.E.C., B.E.K., N.J.M.), Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - B E Kalisch
- Department of Biomedical Sciences (A.L.M., C.E.C., B.E.K., N.J.M.), Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Neil J MacLusky
- Department of Biomedical Sciences (A.L.M., C.E.C., B.E.K., N.J.M.), Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| |
Collapse
|
26
|
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]
|
27
|
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
|
28
|
Siddiqui AN, Siddiqui N, Khan RA, Kalam A, Jabir NR, Kamal MA, Firoz CK, Tabrez S. Neuroprotective Role of Steroidal Sex Hormones: An Overview. CNS Neurosci Ther 2016; 22:342-50. [PMID: 27012165 PMCID: PMC6492877 DOI: 10.1111/cns.12538] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/21/2016] [Accepted: 02/21/2016] [Indexed: 12/11/2022] Open
Abstract
Progesterone, estrogens, and testosterone are the well-known steroidal sex hormones, which have been reported to have "nonreproductive "effects in the brain, specifically in the neuroprotection and neurotrophy. In the last one decade, there has been a surge in the research on the role of these hormones in neuroprotection and their positive impact on different brain injuries. The said interest has been sparked by a desire to understand the action and mechanisms of these steroidal sex hormones throughout the body. The aim of this article was to highlight the potential outcome of the steroidal hormones, viz. progesterone, estrogens, and testosterone in terms of their role in neuroprotection and other brain injuries. Their possible mechanism of action at both genomic and nongenomic level will be also discussed. As far as our knowledge goes, we are for the first time reporting neuroprotective effect and possible mechanism of action of these hormones in a single article.
Collapse
Affiliation(s)
- Ali Nasir Siddiqui
- Department of Pharmaceutical Medicine, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Nahida Siddiqui
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Rashid Ali Khan
- Department of Pharmaceutical Medicine, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Abul Kalam
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Nasimudeen R Jabir
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Hebersham, NSW, Australia
| | | | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Caplan SL, Zheng B, Dawson-Scully K, White CA, West LM. Pseudopterosin A: Protection of Synaptic Function and Potential as a Neuromodulatory Agent. Mar Drugs 2016; 14:md14030055. [PMID: 26978375 PMCID: PMC4820309 DOI: 10.3390/md14030055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 02/28/2016] [Accepted: 03/04/2016] [Indexed: 01/07/2023] Open
Abstract
Natural products have provided an invaluable source of inspiration in the drug discovery pipeline. The oceans are a vast source of biological and chemical diversity. Recently, this untapped resource has been gaining attention in the search for novel structures and development of new classes of therapeutic agents. Pseudopterosins are group of marine diterpene glycosides that possess an array of potent biological activities in several therapeutic areas. Few studies have examined pseudopterosin effects during cellular stress and, to our knowledge, no studies have explored their ability to protect synaptic function. The present study probes pseudopterosin A (PsA) for its neuromodulatory properties during oxidative stress using the fruit fly, Drosophila melanogaster. We demonstrate that oxidative stress rapidly reduces neuronal activity, resulting in the loss of neurotransmission at a well-characterized invertebrate synapse. PsA mitigates this effect and promotes functional tolerance during oxidative stress by prolonging synaptic transmission in a mechanism that differs from scavenging activity. Furthermore, the distribution of PsA within mammalian biological tissues following single intravenous injection was investigated using a validated bioanalytical method. Comparable exposure of PsA in the mouse brain and plasma indicated good distribution of PsA in the brain, suggesting its potential as a novel neuromodulatory agent.
Collapse
Affiliation(s)
- Stacee Lee Caplan
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA.
| | - Bo Zheng
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, The University of Georgia, Athens, GA 30602, USA.
| | - Ken Dawson-Scully
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA.
| | - Catherine A White
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, The University of Georgia, Athens, GA 30602, USA.
| | - Lyndon M West
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, The University of Georgia, Athens, GA 30602, USA.
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA.
| |
Collapse
|
30
|
Shin MS, Chung KJ, Ko IG, Kim SH, Jin JJ, Kim SE, Lee JM, Ji ES, Kim TW, Cho HS, Kim CH, Cho YS, Kim CJ, Kim KH. Effects of surgical and chemical castration on spatial learning ability in relation to cell proliferation and apoptosis in hippocampus. Int Urol Nephrol 2016; 48:517-27. [PMID: 26781653 DOI: 10.1007/s11255-015-1200-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/23/2015] [Indexed: 11/28/2022]
Abstract
PURPOSE Chemical castration using luteinizing hormone-releasing hormone agonists and/or anti-androgens is an alternative to surgical castration. Goserelin and bicalutamide are representative drugs used for chemical castration. The effects of chemical castration on sexual functions are well documented; however, the possibility that chemical castration might induce undesirable effects on brain functions has been raised. We investigated the effects of chemical castration and surgical castration on spatial learning ability in relation to cell proliferation and apoptosis in hippocampus. METHODS Bilateral orchiectomy was performed for surgical castration, and chemical castration was induced by treatment with goserelin or bicalutamide for 28 days. To find out the effects of goserelin and bicalutamide with those of orchiectomy on the spatial learning ability, radial eight-arm maze test was performed. To find out the effects of goserelin and bicalutamide with those of orchiectomy in relation to cell proliferation and apoptosis in the hippocampus, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining, and immunohistochemistry for 5-bromo-2'-deoxyuridine, doublecortin, and caspase-3 were performed. Western blot for brain-derived neurotrophic factor, tyrosine kinase receptor B, Bax, and Bcl-2 in the hippocampus was also performed. RESULTS Orchiectomy caused deterioration of spatial learning ability with suppression of cell proliferation and enhancement of apoptosis in the hippocampus. However, treatment with goserelin and bicalutamide had no effect on spatial learning ability. Cell proliferation and apoptosis were not altered by treatment with goserelin and bicalutamide either. CONCLUSIONS Surgical castration causes deterioration of spatial learning ability, while chemical castration does not impair spatial learning ability. We should find out further mechanisms affect to the relationship between androgen level and neurogenesis and neuronal apoptosis.
Collapse
Affiliation(s)
- Mal-Soon Shin
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Kyung Jin Chung
- Department of Urology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, 405-760, Republic of Korea
| | - Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Sang-Hoon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Sung-Eun Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Jae-Min Lee
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Eun-Sang Ji
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Tae-Woon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Han-Sam Cho
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Chang Hee Kim
- Department of Urology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, 405-760, Republic of Korea
| | - Young-Sam Cho
- Department of Urology, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Jongro-gu, Seoul, 110-746, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Khae-Hawn Kim
- Department of Urology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, 405-760, Republic of Korea.
| |
Collapse
|
31
|
Ciocca G, Limoncin E, Carosa E, Di Sante S, Gravina GL, Mollaioli D, Gianfrilli D, Lenzi A, Jannini EA. Is Testosterone a Food for the Brain? Sex Med Rev 2016; 4:15-25. [PMID: 27872000 DOI: 10.1016/j.sxmr.2015.10.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/02/2015] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Testosterone is fundamental for psychological, sexological, cognitive, and reproductive aspects, and its lack or reduction largely impacts the quality of life in males and females. AIM Therefore, the aim of this review is to describe the role of testosterone in the neurophysiology of the brain and related aspects regarding the quality of general and sexual life. METHODS We listed and discussed the principal studies on the role of testosterone in the brain regarding sexual health, psychopathological conditions, and the elderly. The search strategies were composed by the insertion of specific terms in PubMed regarding the main studies from January 2000 to June 2015. MAIN OUTCOME MEASURES Using a psychoneuroendocrinologic perspective, we considered 4 main sections: brain and testosterone, sexuality and testosterone, psychopathology and testosterone, and cognitive impairment and testosterone. RESULTS Much evidence on the neuroendocrinology of testosterone regarding brain activity, sexual function, psychological health, and senescence was found. In any case, it is known that testosterone deficiency negatively impacts quality of life, first, but not exclusively, through a central effect. Moreover, testosterone and androgen receptors are differently expressed according to age and gender. This aspect contributes to gender differences and to the dimorphic physiological role of this hormone. CONCLUSION A universal role for testosterone can be recognized: low levels of testosterone are associated with mental disorders, sexual dysfunction, and cognitive impairment in both sexes. Hence, physicians should carefully assess testosterone levels, not only in the management of sexual dysfunctions but also when seeking to help patients with severe mental or organic diseases.
Collapse
Affiliation(s)
- Giacomo Ciocca
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Erika Limoncin
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Eleonora Carosa
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Stefania Di Sante
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni L Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Daniele Mollaioli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emmanuele A Jannini
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy.
| |
Collapse
|
32
|
CHEN JINGYU, HU RONG, GE HONGFEI, DUANMU WANGSHENG, LI YUHONG, XUE XINGSENG, HU SHENGLI, FENG HUA. G-protein-coupled receptor 30-mediated antiapoptotic effect of estrogen on spinal motor neurons following injury and its underlying mechanisms. Mol Med Rep 2015; 12:1733-1740. [PMID: 25872489 PMCID: PMC4464192 DOI: 10.3892/mmr.2015.3601] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 03/12/2015] [Indexed: 12/24/2022] Open
Abstract
Spinal cord injury (SCI) may result in severe dysfunction of motor neurons. G-protein-coupled receptor 30 (GPR30) expression in the motor neurons of the ventral horn of the spinal cord mediates neuroprotection through estrogen signaling. The present study explored the antiapoptotic effect of estrogen, mediated by GPR30 following SCI, and the mechanisms underlying this effect. Spinal motor neurons from rats were cultured in vitro in order to establish cell models of oxygen and glucose deprivation (OGD). The effects of estrogen, the estrogen agonist, G1, and the estrogen inhibitor, G15, on motor neurons were observed using MTT assays. The effects of E2, G1 and G15 on spinal motor neuron apoptosis following OGD, were detected using flow cytometry. The role of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) inhibitor, LY294002, was also determined using flow cytometry. Rat SCI models were established. E2, G1 and E2+LY294002 were administered in vivo. Motor function was scored at 3, 7, 14, 21 and 28 d following injury, using Basso-Beattie-Bresnahan (BBB) standards. Cell activity in the estrogen and G1 groups was higher than that in the solvent group, whereas cell activity in the E2+G15 group was lower than that in the E2 group (P<0.05). Following OGD, the proportion of apoptotic cells significantly increased (P<0.05). The proportion in the estrogen group was significantly lower than that in the solvent group, whereas the proportion of apoptotic cells in the E2+G15 and E2+LY294002 groups was higher than that in the E2 group (P<0.05). Treatment with E2 and G1 led to upregulation of P-Akt expression in normal cells and post-OGD cells. The BBB scores of rats in the E2 and G1 groups were higher than those in the placebo group (P<0.05). The BBB scores of the E2+LY294002 group were lower than those of the E2 group (P<0.05). Estrogen thus appears to exert a protective effect on spinal motor neurons following OGD, via GPR30. The PI3K/Akt pathway may be one of those involved in the estrogen‑related antiapoptotic effects mediated by GPR30.
Collapse
Affiliation(s)
- JINGYU CHEN
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - RONG HU
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - HONGFEI GE
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - WANGSHENG DUANMU
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - YUHONG LI
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - XINGSENG XUE
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - SHENGLI HU
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - HUA FENG
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
33
|
Gürer B, Kertmen H, Kasim E, Yilmaz ER, Kanat BH, Sargon MF, Arikok AT, Ergüder BI, Sekerci Z. Neuroprotective effects of testosterone on ischemia/reperfusion injury of the rabbit spinal cord. Injury 2015; 46:240-248. [PMID: 25467821 DOI: 10.1016/j.injury.2014.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/25/2014] [Accepted: 11/03/2014] [Indexed: 02/02/2023]
Abstract
AIM Previous studies demonstrated the neuroprotective effects of testosterone, but no previous study has examined the neuroprotective effects of testosterone on spinal cord ischemia/reperfusion injury. The purpose of this study was to evaluate whether testosterone could protect the spinal cord from ischemia/reperfusion injury. METHODS Rabbits were randomised into four groups of eight animals as follows: group 1 (control), group 2 (ischemia), group 3 (methylprednisolone) and group 4 (testosterone). In the control group only a laparotomy was performed. In all other groups, the spinal cord ischemia model was created by the occlusion of the aorta just caudal to the renal artery. Levels of malondialdehyde and catalase were analysed, as were the activities of caspase-3, myeloperoxidase, and xanthine oxidase. Histopathological and ultrastructural evaluations were performed. Neurological evaluation was performed with the Tarlov scoring system. RESULTS After ischemia-reperfusion injury, increases were found in caspase-3 activity, myeloperoxidase activity, malondialdehyde levels, and xanthine oxidase activity. In contrast, decreases in catalase levels were observed. After the administration of testosterone, decreases were observed in caspase-3 activity, myeloperoxidase activity, malondialdehyde levels, and xanthine oxidase activity, whereas catalase levels increased. Furthermore, testosterone treatment showed improved results concerning histopathological scores, ultrastructural score and Tarlov scores. CONCLUSIONS Our results revealed for the first time that testosterone exhibits meaningful neuroprotective activity following ischemia-reperfusion injury of the spinal cord.
Collapse
Affiliation(s)
- Bora Gürer
- Ministry of Health, Fatih Sultan Mehmet Education and Research Hospital, Neurosurgey Clinic, Istanbul, Turkey.
| | - Hayri Kertmen
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| | - Emin Kasim
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| | - Erdal Resit Yilmaz
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| | - Burhan Hakan Kanat
- Ministry of Health, Elazig Education and Research Hospital, General Surgery Clinic, Elazig, Turkey
| | - Mustafa Fevzi Sargon
- Hacettepe University, Faculty of Medicine, Department of Anatomy, Ankara, Turkey
| | - Ata Türker Arikok
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Department of Pathology, Ankara, Turkey
| | - Berrin Imge Ergüder
- Ankara University, Faculty of Medicine, Department of Biochemistry, Ankara, Turkey
| | - Zeki Sekerci
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| |
Collapse
|
34
|
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
|
35
|
Abstract
Background Neuronal cells are vulnerable to many stresses that can cause apoptosis. Reprogramming of fibroblasts into induced neural stem cells (iNSCs) is a potentially unlimited source of neurons. Discovering agents that can provide neuronal protection against these apoptotic stimuli is important for developing therapeutic strategies for various brain diseases. Material/Methods We investigated the therapeutic effects of iNSCs against apoptosis activator II (AAII)-induced apoptosis of cortical neuronal cells. Apoptosis was confirmed by double immunocytochemistry with NeuN and 4′,6-diamidino-2-phenylindole using terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP-biotin nick-end labeling. We performed Western blot analyses for activated caspase-3, Bcl-2, phosphorylated Akt, and phosphorylated extracellular signal-regulated protein kinase (ERK). The level of vascular endothelial growth factor (VEGF) was analyzed using enzyme-linked immunosorbent assays (P<0.05). Results Cortical neuronal cells cultured with iNSCs had fewer apoptotic cells than those cultured without iNSCs. We found that cells cultured with iNSCs had a significantly lower caspase-3 level and a significantly higher Bcl-2 level than cells cultured without iNSCs. Cells cultured with iNSCs had higher VEGF levels than cells cultured without iNSCs. The levels of phosphorylated Akt and phosphorylated ERK were significantly higher in cells cultured with iNSCs than in cells cultured without iNSCs. Conclusions Our findings suggest that iNSCs activate Akt and ERK, which are associated with the inhibition of neuronal apoptosis. Thus, treatment with iNSCs may help reduce neuronal loss in brain disease. Further studies aimed at proving this hypothesis might help establish therapeutic agents that can prevent neuronal cell death and help cure neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Neurosurgery, College of Medicine, Korea University, Seoul, Korea
| | - Jangbo Lee
- Department of Neurosurgery, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
36
|
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
|
37
|
Protective effects of testosterone on presynaptic terminals against oligomeric β-amyloid peptide in primary culture of hippocampal neurons. BIOMED RESEARCH INTERNATIONAL 2014; 2014:103906. [PMID: 25045655 PMCID: PMC4086619 DOI: 10.1155/2014/103906] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022]
Abstract
Increasing lines of evidence support that testosterone may have neuroprotective effects. While observational studies reported an association between higher bioavailable testosterone or brain testosterone levels and reduced risk of Alzheimer's disease (AD), there is limited understanding of the underlying neuroprotective mechanisms. Previous studies demonstrated that testosterone could alleviate neurotoxicity induced by β-amyloid (Aβ), but these findings mainly focused on neuronal apoptosis. Since synaptic dysfunction and degeneration are early events during the pathogenesis of AD, we aim to investigate the effects of testosterone on oligomeric Aβ-induced synaptic changes. Our data suggested that exposure of primary cultured hippocampal neurons to oligomeric Aβ could reduce the length of neurites and decrease the expression of presynaptic proteins including synaptophysin, synaptotagmin, and synapsin-1. Aβ also disrupted synaptic vesicle recycling and protein folding machinery. Testosterone preserved the integrity of neurites and the expression of presynaptic proteins. It also attenuated Aβ-induced impairment of synaptic exocytosis. By using letrozole as an aromatase antagonist, we further demonstrated that the effects of testosterone on exocytosis were unlikely to be mediated through the estrogen receptor pathway. Furthermore, we showed that testosterone could attenuate Aβ-induced reduction of HSP70, which suggests a novel mechanism that links testosterone and its protective function on Aβ-induced synaptic damage. Taken together, our data provide further evidence on the beneficial effects of testosterone, which may be useful for future drug development for AD.
Collapse
|
38
|
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
|
39
|
Jayaraman A, Pike CJ. Differential effects of synthetic progestagens on neuron survival and estrogen neuroprotection in cultured neurons. Mol Cell Endocrinol 2014; 384:52-60. [PMID: 24424444 PMCID: PMC3954450 DOI: 10.1016/j.mce.2014.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 11/22/2013] [Accepted: 01/03/2014] [Indexed: 11/19/2022]
Abstract
Progesterone and other progestagens are used in combination with estrogens for clinical purposes, including contraception and postmenopausal hormone therapy. Progesterone and estrogens have interactive effects in brain, however interactions between synthetic progestagens and 17β-estradiol (E2) in neurons are not well understood. In this study, we investigated the effects of seven clinically relevant progestagens on estrogen receptor (ER) mRNA expression, E2-induced neuroprotection, and E2-induced BDNF mRNA expression. We found that medroxyprogesterone acetate decreased both ERα and ERβ expression and blocked E2-mediated neuroprotection and BDNF expression. Conversely, levonorgestrel and nesterone increased ERα and or ERβ expression, were neuroprotective, and failed to attenuate E2-mediated increases in neuron survival and BDNF expression. Other progestagens tested, including norethindrone, norethindrone acetate, norethynodrel, and norgestimate, had variable effects on the measured endpoints. Our results demonstrate a range of qualitatively different actions of progestagens in cultured neurons, suggesting significant variability in the neural effects of clinically utilized progestagens.
Collapse
Affiliation(s)
- Anusha Jayaraman
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
40
|
Panizzon MS, Hauger R, Xian H, Vuoksimaa E, Spoon KM, Mendoza SP, Jacobson KC, Vasilopoulos T, Rana BK, McKenzie R, McCaffery JM, Lyons MJ, Kremen WS, Franz CE. Interaction of APOE genotype and testosterone on episodic memory in middle-aged men. Neurobiol Aging 2013; 35:1778.e1-8. [PMID: 24444806 DOI: 10.1016/j.neurobiolaging.2013.12.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/19/2013] [Accepted: 12/21/2013] [Indexed: 01/12/2023]
Abstract
Age-related changes in testosterone are believed to be a key component of the processes that contribute to cognitive aging in men. The APOE-ε4 allele may interact with testosterone and moderate the hormone's association with cognition. The goals of the present study were to examine the degree to which free testosterone is associated with episodic memory in a community-based sample of middle-aged men, and examine the potential interaction between free testosterone and the APOE-ε4 allele. Data were used from 717 participants in the Vietnam Era Twin Study of Aging. Average age was 55.4 years (standard deviation = 2.5). Significant positive associations were observed between free testosterone level and verbal episodic memory, as well as a significant interaction between free testosterone and APOE-ε4 status. In ε4 carriers free testosterone was positively associated with verbal episodic memory performance (story recall), whereas no association was observed in ε4 noncarriers. Results support the hypothesis that APOE-ε4 status increases susceptibility to other risk factors, such as low testosterone, which may ultimately contribute to cognitive decline or dementia.
Collapse
Affiliation(s)
- Matthew S Panizzon
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA.
| | - Richard Hauger
- Department of Psychiatry, University of California, San Diego, CA, USA; VA San Diego Healthcare System, CA, USA; Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Hong Xian
- Department of Biostatistics, St. Louis University, College for Public Health & Social Justice, St. Louis, MO, USA; Research Service, St. Louis Veterans Affairs Medical Center, St. Louis, MO
| | - Eero Vuoksimaa
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Kelly M Spoon
- Computational Science Research Center, San Diego State University, San Diego, CA, USA
| | - Sally P Mendoza
- Department of Psychology, University of California, Davis, CA, USA
| | | | | | - Brinda K Rana
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA
| | - Ruth McKenzie
- Department of Psychology, Boston University, Boston, MA, USA
| | - Jeanne M McCaffery
- Department of Psychiatry and Human Behavior, The Miriam Hospital and Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | - Michael J Lyons
- Department of Psychology, Boston University, Boston, MA, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA; VA San Diego Healthcare System, CA, USA; Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Carol E Franz
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA
| |
Collapse
|
41
|
Birnie M, Morrison R, Camara R, Strauss KI. Temporal changes of cytochrome P450 (Cyp) and eicosanoid-related gene expression in the rat brain after traumatic brain injury. BMC Genomics 2013; 14:303. [PMID: 23642095 PMCID: PMC3658912 DOI: 10.1186/1471-2164-14-303] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 04/16/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) induces arachidonic acid (ArA) release from cell membranes. ArA metabolites form a class of over 50 bioactive eicosanoids that can induce both adaptive and/or maladaptive brain responses. The dynamic metabolism of ArA to eicosanoids, and how they affect the injured brain, is poorly understood due to their diverse activities, trace levels, and short half-lives. The eicosanoids produced in the brain postinjury depend upon the enzymes present locally at any given time. Eicosanoids are synthesized by heme-containing enzymes, including cyclooxygenases, lipoxygenases, and arachidonate monoxygenases. The latter comprise a subset of the cytochrome P450 "Cyp" gene family that metabolize fatty acids, steroids, as well as endogenous and exogenous toxicants. However, for many of these genes neither baseline neuroanatomical nor injury-related temporal expression have been studied in the brain.In a rat model of parietal cortex TBI, Cyp and eicosanoid-related mRNA levels were determined at 6 h, 24 h, 3d, and 7d postinjury in parietal cortex and hippocampus, where dynamic changes in eicosanoids have been observed. Quantitative real-time polymerase chain reaction with low density arrays were used to assay 62 rat Cyps, 37 of which metabolize ArA or other unsaturated fatty acids; 16 eicosanoid-related enzymes that metabolize ArA or its metabolites; 8 eicosanoid receptors; 5 other inflammatory- and recovery-related genes, plus 2 mouse Cyps as negative controls and 3 highly expressed "housekeeping" genes. RESULTS Sixteen arachidonate monoxygenases, 17 eicosanoid-related genes, and 12 other Cyps were regulated in the brain postinjury (p < 0.05, Tukey HSD). Discrete tissue levels and distinct postinjury temporal patterns of gene expression were observed in hippocampus and parietal cortex. CONCLUSIONS The results suggest complex regulation of ArA and other lipid metabolism after TBI. Due to the temporal nature of brain injury-induced Cyp gene induction, manipulation of each gene (or its products) at a given time after TBI will be required to assess their contributions to secondary injury and/or recovery. Moreover, a better understanding of brain region localization and cell type-specific expression may be necessary to deduce the role of these eicosanoid-related genes in the healthy and injured brain.
Collapse
Affiliation(s)
- Matthew Birnie
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
| | - Ryan Morrison
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
| | - Ramatoulie Camara
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
| | - Kenneth I Strauss
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
- Present Address: Michigan State University College of Human Medicine, 333 Bostwick Ave NE, 49503 Grand Rapids, MI, USA
| |
Collapse
|
42
|
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: 164] [Impact Index Per Article: 13.7] [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
|
43
|
Filová B, Ostatníková D, Celec P, Hodosy J. The effect of testosterone on the formation of brain structures. Cells Tissues Organs 2013; 197:169-77. [PMID: 23306974 DOI: 10.1159/000345567] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2012] [Indexed: 11/19/2022] Open
Abstract
It has been confirmed in several studies that testosterone can significantly affect brain development. Following metabolism of this hormone by 5α-reductase to dihydrotestosterone, testosterone may act via androgen receptors, or after conversion by aromatase to estradiol, it may act via estrogen receptors. The parts of the brain which are changed under the influence of sex hormones are known as sexually dimorphic nuclei, especially in the preoptic area of the hypothalamus. Nevertheless, evidence suggests that testosterone also influences the structure of the hippocampus, specifically CA1 and CA3 areas of the hippocampus, as well as the amygdala. These brain areas are designed to convert information from short-term into long-term memory. In this review, we summarize the effects of testosterone on the organization of brain structures with respect to spatial cognitive abilities in small rodents.
Collapse
Affiliation(s)
- Barbora Filová
- Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | | | | | | |
Collapse
|
44
|
Farivar TN, Najafipour R, Johari P. Nano - drug Delivery of Apoptosis Activator 2 to AGS Cells by Liposomes Conjugated with Anti-TROP2 Antibody. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2012. [PMID: 23181231 PMCID: PMC3503378 DOI: 10.4103/1947-2714.103319] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background: Gastric cancer is the second most common causes of cancer related death in the world and is responsible for two third of cancer related death in the developing countries. survival rate surgery is low and radiation therapy and chemotherapy as alternatives ways for treatment of gastric cancer are not very promising. Thus there is an urgent need for introducing novel treatment procedures and promising new anti-canceric drugs. Aim: In this study we used pre-prepared liposomes and after necessary manipulations ,these modified liposomes were used for delivery of apoptosis activator 2 to gastric adenocarcinoma cell line (AGS). Materials and Methods: we used pre-prepared liposomes and after necessary manipulations, these modified liposomes were used for delivery of apoptosis activator 2 to AGS cells and induced apoptosis was evaluated by related apoptotic DNA ladder, TUNNEL and Cell Death experiments. Results: Evaluation of apoptosis by Apoptotic DNA Ladder in liposome treated and untreated AGS cells by DNA laddering and fragmentation, TUNEL and Cell Death Detection confirmed that treatment of AGS cell lines with apoptosis activator 2 loaded liposomes which targeted cell surface TROP2 antigen in cancer cells significantly increased apoptosis in these cells. Conclusion: Nano drug delivery of apoptosis activator 2 to human gastric adenocarcinoma cell line with liposomes targeted TROP2 antigen is a possible way for smart killing of human gastric adenocarcinoma cells.
Collapse
|
45
|
Schwarz ER, Phan A, Willix RD. Andropause and the development of cardiovascular disease presentation-more than an epi-phenomenon. J Geriatr Cardiol 2012; 8:35-43. [PMID: 22783283 PMCID: PMC3390065 DOI: 10.3724/sp.j.1263.2011.00035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 03/14/2011] [Accepted: 03/21/2011] [Indexed: 01/11/2023] Open
Abstract
Andropause refers to a generalized decline of male hormones, including testosterone and dehydroepiandrosterone in middle-aged and aging men. This decline in hormones has been associated with changes such as depression, loss of libido, sexual dysfunction, and changes in body composition. Aging has been associated with an abundance of concomitant diseases, in particular cardiovascular diseases, and although andropause is correlated to aging, a causal relationship between reduction of androgens and the development of chronic diseases such as atherosclerosis and heart failure has not been convincingly established yet. On the other hand, increasing data has emerged that revealed the effects of low levels of androgens on cardiovascular disease progression. As an example, low levels of testosterone have been linked to a higher incidence of coronary artery disease. Whether hormone replacement therapy that is used for andropausal men to alleviate symptoms of "male menopause" can halt progression of cardiovascular disease, remains controversially discussed, primarily due to the lack of well-designed, randomized controlled trials. At least for symptom improvement, the use of androgen replacement therapy in andropausal men may be clinically indicated, and with the appropriate supervision and follow up may prove to be beneficial with regard to preservation of the integrity of cardiovascular health at higher ages.
Collapse
Affiliation(s)
- Ernst R Schwarz
- Cedars Sinai Heart Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, Suite 6215, Los Angeles, CA 90048, USA; Cenegenics Medical Institute, 851 Rampart Blvd., Las Vegas, NV 89145, USA
| | | | | |
Collapse
|
46
|
Mild exercise increases dihydrotestosterone in hippocampus providing evidence for androgenic mediation of neurogenesis. Proc Natl Acad Sci U S A 2012; 109:13100-5. [PMID: 22807478 DOI: 10.1073/pnas.1210023109] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mild exercise activates hippocampal neurons through the glutamatergic pathway and also promotes adult hippocampal neurogenesis (AHN). We hypothesized that such exercise could enhance local androgen synthesis and cause AHN because hippocampal steroid synthesis is facilitated by activated neurons via N-methyl-D-aspartate receptors. Here we addressed this question using a mild-intense treadmill running model that has been shown to be a potent AHN stimulator. A mass-spectrometric analysis demonstrated that hippocampal dihydrotestosterone increased significantly, whereas testosterone levels did not increase significantly after 2 wk of treadmill running in both orchidectomized (ORX) and sham castrated (Sham) male rats. Furthermore, analysis of mRNA expression for the two isoforms of 5α-reductases (srd5a1, srd5a2) and for androgen receptor (AR) revealed that both increased in the hippocampus after exercise, even in ORX rats. All rats were injected twice with 5'-bromo-2'deoxyuridine (50 mg/kg body weight, i.p.) on the day before training. Mild exercise significantly increased AHN in both ORX and Sham rats. Moreover, the increase of doublecortin or 5'-bromo-2'deoxyuridine/NeuN-positive cells in ORX rats was blocked by s.c. flutamide, an AR antagonist. It was also found that application of an estrogen receptor antagonist, tamoxifen, did not suppress exercise-induced AHN. These results support the hypothesis that, in male animals, mild exercise enhances hippocampal synthesis of dihydrotestosterone and increases AHN via androgenenic mediation.
Collapse
|
47
|
Kohtz AS, Frye CA. Dissociating behavioral, autonomic, and neuroendocrine effects of androgen steroids in animal models. Methods Mol Biol 2012; 829:397-431. [PMID: 22231829 DOI: 10.1007/978-1-61779-458-2_26] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Developments in behavioral assessment, autonomic and/or baseline reactivity, psychopharmacology, and genetics, have contributed significantly to the assessment of performance-enhancing drugs in animal models. Particular classes of steroid hormones: androgenic steroids are of interest. Anecdotally, the performance enhancing effects of androgens are attributed to anabolic events. However, there is a discrepancy between anecdotal evidence and investigative data. While some androgen steroids may promote muscle growth (myogenesis), effects of androgens on performance enhancement are not always seen. Indeed, some effects of androgens on performance may be attributable to their psychological and cardiovascular effects. As such, we consider androgen effects in terms of their behavioral, autonomic, and neuroendocrine components. Techniques are discussed in this chapter, some of which are well established, while others have been more recently developed to study androgen action. Androgens may be considered for their positive impact, negative consequence, or psychotropic properties. Thus, this review aims to elucidate some of the effects and/or mechanisms of androgens on behavioral, autonomic, and/or neuroendocrine assessment that may underlie their controversial performance enhancing effects.
Collapse
Affiliation(s)
- Amy S Kohtz
- Department of Psychology, The University at Albany-SUNY, Albany, NY, USA
| | | |
Collapse
|
48
|
Zuloaga DG, Carbone DL, Quihuis A, Hiroi R, Chong DL, Handa RJ. Perinatal dexamethasone-induced alterations in apoptosis within the hippocampus and paraventricular nucleus of the hypothalamus are influenced by age and sex. J Neurosci Res 2012; 90:1403-12. [PMID: 22388926 DOI: 10.1002/jnr.23026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 12/06/2011] [Accepted: 12/08/2011] [Indexed: 01/18/2023]
Abstract
Exposure to high levels of glucocorticoids (GCs) during development leads to long-term changes in hypothalamic-pituitary-adrenal (HPA) axis regulation, although little is known about the neural mechanisms that underlie these alterations. In this study, we investigated the effects of late gestational (days 18-22) or postnatal (days 4-6) administration of the GC receptor agonist dexamethasone (DEX) on an apoptosis marker in two brain regions critical to HPA axis regulation, the hippocampus and the hypothalamic paraventricular nucleus (PVN). One day after the final DEX injection, male and female rats were sacrificed, and brains were processed for immunohistochemical detection of cleaved caspase-3, an apoptotic cell death indicator. DEX increased cleaved caspase-3 immunoreactivity in the CA1 hippocampal region of both sexes following prenatal but not postnatal treatment. Prenatal DEX also increased caspase-3 immunoreactivity in the CA3 region, an elevation that tended to be greater in females. In contrast, postnatal DEX resulted in a much smaller, albeit significant, induction in CA3 caspase-3 compared with prenatal treatment. Quantitative real-time PCR analysis revealed that prenatal but not postnatal DEX-induced hippocampal cleaved caspase-3 correlated with elevated mRNA of the proapoptotic gene Bad. Few caspase-3-ir cells were identified within the PVN regardless of treatment age, although postnatal but not prenatal DEX increased this number. However, the region immediately surrounding the PVN (peri-PVN) showed significant increases in caspase-3-ir cells following pre- and postnatal DEX. Together these findings indicate that developmental GC exposure increases apoptosis in HPAaxis-associated brain regions in an age- and sex-dependent manner.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Paus T, Bernard M, Chakravarty MM, Davey Smith G, Gillis J, Lourdusamy A, Melka MG, Leonard G, Pavlidis P, Perron M, Pike GB, Richer L, Schumann G, Timpson N, Toro R, Veillette S, Pausova Z. KCTD8 gene and brain growth in adverse intrauterine environment: a genome-wide association study. ACTA ACUST UNITED AC 2011; 22:2634-42. [PMID: 22156575 DOI: 10.1093/cercor/bhr350] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The most dramatic growth of the human brain occurs in utero and during the first 2 years of postnatal life. Genesis of the cerebral cortex involves cell proliferation, migration, and apoptosis, all of which may be influenced by prenatal environment. Here, we show that variation in KCTD8 (potassium channel tetramerization domain 8) is associated with brain size in female adolescents (rs716890, P = 5.40 × 10(-09)). Furthermore, we found that the KCTD8 locus interacts with prenatal exposure to maternal cigarette smoking vis-à-vis cortical area and cortical folding: In exposed girls only, the KCTD8 locus explains up to 21% of variance. Using head circumference as a proxy of brain size at 7 years of age, we have replicated this gene-environment interaction in an independent sample. We speculate that KCTD8 might modulate adverse effects of smoking during pregnancy on brain development via apoptosis triggered by low intracellular levels of potassium, possibly reducing the number of progenitor cells.
Collapse
Affiliation(s)
- Tomás Paus
- Rotman Research Institute, University of Toronto, Toronto, Ontario M6A 2E1, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sigwalt AR, Budde H, Helmich I, Glaser V, Ghisoni K, Lanza S, Cadore EL, Lhullier FLR, de Bem AF, Hohl A, de Matos FJ, de Oliveira PA, Prediger RD, Guglielmo LGA, Latini A. Molecular aspects involved in swimming exercise training reducing anhedonia in a rat model of depression. Neuroscience 2011; 192:661-74. [PMID: 21712072 DOI: 10.1016/j.neuroscience.2011.05.075] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 04/19/2011] [Accepted: 05/28/2011] [Indexed: 12/19/2022]
Abstract
Patients suffering from depression frequently display hyperactivity of the hypothalamic-pituitary-adrenal axis (HPA) resulting in elevated cortisol levels. One main symptom of this condition is anhedonia. There is evidence that exercise training can be used as a rehabilitative intervention in the treatment of depressive disorders. In this scenario, the aim of the present study was to assess the effect of an aerobic exercise training protocol on the depressive-like behavior, anhedonia, induced by repeated dexamethasone administration. The study was carried out on adult male Wistar rats randomly divided into four groups: the "control group" (C), "exercise group" (E), "dexamethasone group" (D) and the "dexamethasone plus exercise group" (DE). The exercise training consisted of swimming (1 h/d, 5 d/wk) for 3 weeks, with an overload of 5% of the rat body weight. Every day rats were injected with either dexamethasone (D/DE) or saline solution (C/E). Proper positive controls, using fluoxetine, were run in parallel. Decreased blood corticosterone levels, reduced adrenal cholesterol synthesis and adrenal weight (HPA disruption), reduced preference for sucrose consumption and increased immobility time (depressive-like behavior), marked hippocampal DNA oxidation, increased IL-10 and total brain-derived neurotrophic factor (BDNF; pro-plus mature-forms) and a severe loss of body mass characterized the dexamethasone-treated animals. Besides increasing testosterone blood concentrations, the swim training protected depressive rats from the anhedonic state, following the same profile as fluoxetine, and also from the dexamethasone-induced impaired neurochemistry. The data indicate that physical exercise could be a useful tool in preventing and treating depressive disorders.
Collapse
Affiliation(s)
- A R Sigwalt
- Laboratório de Bioenergética e Estresse Oxidativo, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|