1
|
Wei S, Ma F, Feng S, Ha X. Integrating transcriptomics and proteomics to understand the molecular mechanisms underlying the pathogenesis of type 2 diabetes mellitus. Genomics 2024; 116:110964. [PMID: 39571829 DOI: 10.1016/j.ygeno.2024.110964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
The liver plays an important role in glucose regulation, and their dysfunction is closely associated with the development of type 2 diabetes mellitus (T2DM), and insulin resistance (IR) in hepatocyte mediate the pathogenesis of diabetes mellitus. In T2DM rats and their correlated control, we investigated various genes expression at transcriptional and translational level by utilizing transcriptomic using RNA sequencing (RNA-seq) and proteomics using isobaric tags for relative and absolute quantification (iTRAQ) to disclose potential candidates for Type 2 diabetes diagnosis and therapy. We found the lecithin retinol acyltransferase (Lrat) gene regulate hepatocyte IR in T2DM. Furthermore, BRL-3A cells, rat liver cells, worked as the IR model in vitro study. Hence, Lrat gene was overexpressed in BRL-3A cells to explore the role of Lrat gene in IR by measuring the cellular glucose consumption, TCHO, and LDL-C levels. Finally, we found that Lrat gene can improve the level of glycolipid metabolism in BRL-3A cells and reduce the degree of IR in BRL-3A cells. Therefore, further exploration of Lrat gene related molecular mechanism is meaningful.
Collapse
Affiliation(s)
- Shuyao Wei
- Department of Clinical Laboratory, The 940th Hospital of Joint Logistics Support force of Chinese People's Liberation Army, Lanzhou 730050, China; Department of Clinical Laboratory, Xuzhou Municipal First People's Hospital, Xuzhou 221009, China; Clinical Laboratory Diagnostics, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Stem Cells and Gene Drugs, Lanzhou 730050, China
| | - Feifei Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; Key Laboratory of Stem Cells and Gene Drugs, Lanzhou 730050, China
| | - Shanshan Feng
- Clinical Laboratory Diagnostics, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Stem Cells and Gene Drugs, Lanzhou 730050, China
| | - Xiaoqin Ha
- Department of Clinical Laboratory, The 940th Hospital of Joint Logistics Support force of Chinese People's Liberation Army, Lanzhou 730050, China; Clinical Laboratory Diagnostics, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Stem Cells and Gene Drugs, Lanzhou 730050, China.
| |
Collapse
|
2
|
Haga M, Iida K, Okada M. Positive and negative feedback regulation of the TGF-β1 explains two equilibrium states in skin aging. iScience 2024; 27:109708. [PMID: 38706856 PMCID: PMC11066433 DOI: 10.1016/j.isci.2024.109708] [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: 07/06/2023] [Revised: 02/05/2024] [Accepted: 04/06/2024] [Indexed: 05/07/2024] Open
Abstract
During aging, skin homeostasis is essential for maintaining appearance, as well as biological defense of the human body. In this study, we identified thrombospondin-1 (THBS1) and fibromodulin (FMOD) as positive and negative regulators, respectively, of the TGF-β1-SMAD4 axis in human skin aging, based on in vitro and in vivo omics analyses and mathematical modeling. Using transcriptomic and epigenetic analyses of senescent dermal fibroblasts, TGF-β1 was identified as the key upstream regulator. Bifurcation analysis revealed a binary high-/low-TGF-β1 switch, with THBS1 as the main controller. Computational simulation of the TGF-β1 signaling pathway indicated that THBS1 expression was sensitively regulated, whereas FMOD was regulated robustly. Results of sensitivity analysis and validation showed that inhibition of SMAD4 complex formation was a promising method to control THBS1 production and senescence. Therefore, this study demonstrated the potential of combining data-driven target discovery with mathematical approaches to determine the mechanisms underlying skin aging.
Collapse
Affiliation(s)
- Masatoshi Haga
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
- Basic Research Development Division, ROHTO Pharmaceutical Co., Ltd, Osaka 544-8666, Japan
| | - Keita Iida
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Liu S, Costa M. The role of NUPR1 in response to stress and cancer development. Toxicol Appl Pharmacol 2022; 454:116244. [PMID: 36116561 DOI: 10.1016/j.taap.2022.116244] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 10/31/2022]
Abstract
Stress contributes to the development of many human diseases, including cancer. Based on the source of stress, it can be divided into external stress, such as environmental carcinogens, chemicals, and radiation, and internal stress, like endoplasmic reticulum (ER) stress, hypoxia, and oxidative stress. Nuclear Protein 1 (NUPR1, p8 or Com-1) is a small, highly basic transcriptional regulator that participates in regulating a variety of cellular processes including DNA repair, ER stress, oxidative stress response, cell cycle, autophagy, apoptosis, ferroptosis and chromatin remodeling. A large number of studies have reported that NUPR1 expression can be stimulated rapidly in response to various stresses. Thus, NUPR1 is also known as a stress-response gene. Since the role of NUPR1 in breast cancer was identified in 1999, an increasing number of studies sought to reveal its function in cancer. High expression of NUPR1 has been identified in oral squamous cell carcinoma, breast cancer, lung cancer, multiple myeloma, liver cancer and renal cancer. In this review, we summarize current studies of NUPR1 in response to multiple external stressors and internal stressors, and its role in mediating stressors to cause different cell signaling responses. In addition, this review discusses the function of NUPR1 in carcinogenesis, tumorigenesis, metastasis, and cancer therapy. Thus, this review gives a comprehensive insight into the role of NUPR1 in mediating signals from stress to different cell responses, and this process plays a role in the development of cancer.
Collapse
Affiliation(s)
- Shan Liu
- Division of Environmental Medicine, Dept of Medicine, New York University School of Medicine, NY, USA.
| | - Max Costa
- Division of Environmental Medicine, Dept of Medicine, New York University School of Medicine, NY, USA.
| |
Collapse
|
4
|
Pham LT, Yamanaka K, Miyamoto Y, Waki H, Gouraud SSS. Estradiol-dependent gene expression profile in the amygdala of young ovariectomized spontaneously hypertensive rats. Physiol Genomics 2022; 54:99-114. [DOI: 10.1152/physiolgenomics.00082.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Estrogen plays a role in cardiovascular functions, emotional health, and energy homeostasis via estrogen receptors expressed in the brain. The comorbid relationship between rising blood pressure, a decline in mood and motivation, and body weight gain after menopause, when estrogen levels drop, suggests that the same brain area(s) contributes to protection from all of these postmenopausal disorders. The amygdala, a major limbic system nucleus known to express high estrogen receptor levels, is involved in the regulation of such physiological and psychological responses. We hypothesized that elevated estrogen levels contribute to premenopausal characteristics by activating specific genes and pathways in the amygdala. We examined the effect of 1-month estradiol treatment on the gene expression profile in the amygdala of ovariectomized young adult female spontaneously hypertensive rats. Estradiol substitution significantly decreased blood pressure, prevented body weight gain, and enhanced the voluntary physical activity of ovariectomized rats. In the amygdala of ovariectomized rats, estradiol treatment downregulated the expression of genes associated with estrogen signaling, cholinergic synapse, dopaminergic synapse, and long-term depression pathways. These findings indicate that the transcriptomic characteristics of the amygdala may be involved in estrogen-dependent regulation of blood pressure, physical activity motivation, and body weight control in young adult female spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Linh T Pham
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Ko Yamanaka
- Department of Physiology, Graduate School of Health and Sports Sciences, Juntendo University, Inzai, Chiba, Japan
| | | | - Hidefumi Waki
- Department of Physiolgy, Graduate School of Health and Sports Sciences, Juntendo University, Inzai, Chiba, Japan
| | - Sabine S. S. Gouraud
- College of Liberal Arts, Department of Natural Sciences, International Christian University, Tokyo, 東京都, Japan
| |
Collapse
|
5
|
Farmer C, Palin MF. Hyperprolactinemia using domperidone in prepubertal gilts: Effects on hormonal status, mammary development and mammary and pituitary gene expression. Domest Anim Endocrinol 2021; 76:106630. [PMID: 33979716 DOI: 10.1016/j.domaniend.2021.106630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/30/2021] [Accepted: 04/04/2021] [Indexed: 01/24/2023]
Abstract
Objectives of this experiment were to determine if the domperidone protocol previously used for gestating gilts can also lead to hyperprolactinemia in growing gilts, and to assess the effects of such a protocol on hormonal status, mammary development and gene expression in mammary and pituitary tissue of gilts at puberty. The impact on future lactation performance was also determined. At 75 ± 3 kg body weight (BW), gilts were divided between: 1) controls (CTL), receiving daily intramuscular (IM) injections of canola oil (1.1 mL) for 29 d (n = 41), and 2) treated (DOMP), receiving daily IM injections with 0.5 mg/kg BW of the dopamine receptor antagonist domperidone for 29 d (n = 40). In addition to that daily injection, treated gilts also received twice daily IM injections with 0.5 mg/kg BW of domperidone over the first 3 d of treatment. Fifteen gilts per treatment were sacrificed at 210 ± 5 d of age to collect mammary glands (for compositional analysis and gene expression) and the anterior pituitary (for gene expression). Remaining gilts were bred and allowed to farrow. Blood was sampled at the onset of treatment and on days 14 and 30. Gilts that farrowed were also blood sampled on days 3 and 20 of lactation. Blood was assayed for prolactin (PRL), leptin, insulin-like growth factor 1 (IGF-1), urea, free fatty acids and glucose. Concentrations of PRL increased after 14 d and 30 d of treatment (P < 0.01) and were lesser on day 3 of lactation in DOMP than CTL gilts (P < 0.01). At puberty, there were tendencies (P < 0.10) for total parenchymal protein and DNA to be greater in DOMP than CTL gilts. Treatment did not affect mRNA abundance of PRL or the long form of the PRL receptor genes in the pituitary gland at puberty but expression level of the dopamine receptor D2 and PRL genes was much lower in pubertal than late-pregnant gilts (P < 0.001). Furthermore, many genes related with PRL had a much greater expression level in late pregnancy than at puberty. On day 20 of lactation, CTL sows had greater concentrations of urea than DOMP sows (P < 0.01). The growth rate of litters was not affected by treatment nor was milk composition (P > 0.10). Even though PRL concentrations were increased with treatment, the absence of effect on mammary development was either due to timing relative to developmental stage, whereby treatment was initiated when gilts were too young, or was because all PRL receptors may have been saturated thereby preventing biological action of additional PRL.
Collapse
Affiliation(s)
- C Farmer
- Agriculture and Agri-Food Canada, Sherbrooke R & D Centre, Sherbrooke, QC, J1M 0C8, Canada.
| | - M F Palin
- Agriculture and Agri-Food Canada, Sherbrooke R & D Centre, Sherbrooke, QC, J1M 0C8, Canada
| |
Collapse
|
6
|
Dean B, Gogos A. The impact of ovariectomy and chronic estrogen treatment on gene expression in the rat cortex: Implications for psychiatric disorders. Psychoneuroendocrinology 2021; 127:105192. [PMID: 33730612 DOI: 10.1016/j.psyneuen.2021.105192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 01/01/2023]
Abstract
Estrogens, via estrogen-mediated changes in CNS function, have been suggested to be beneficial in the treatment of several psychiatric disorders. Few studies have used transcriptomic technologies to determine the effect of estrogen on gene expression in the CNS. Thus, we aimed to examine the impact of ovariectomy (the removal of all ovarian hormones) and estrogen replacement on rat frontal cortical gene expression. We used the Agilent SurePrint G3 Gene Expression Rat Array to measure levels of RNA in intact (cycling) female rats and in ovariectomized rats that were, or were not, given 17β-estradiol in implants for 4 weeks. Compared to untreated ovariectomized rats, intact rats (effect of ovarian hormones; comparison 1) and rats receiving 17β-estradiol replacement (estrogen-specific effects; comparison 2) showed significant changes in cortical gene expression (58 and 36 genes, respectively). These changes in gene expression would be expected to affect pathways that regulate neurotransmitters, glutathione and sphingolipids; pathways known to be implicated in the pathophysiologies of psychiatric disorders. When we compared the levels of gene expression in the two comparisons that had a significance of p < 0.01 independent of magnitude of change, there was a strong correlation between fold changes in gene expression for 127 genes. We posit that this correlation is due to the level of expression of these genes being strongly influenced by both cycling and replacement estrogen. Further exploration of ovarian hormone- and estrogen-sensitive gene expression may provide new insight into the aetiology of aspects of psychiatric disorders that show sex differences.
Collapse
Affiliation(s)
- Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
7
|
Beletskiy A, Chesnokova E, Bal N. Insulin-Like Growth Factor 2 As a Possible Neuroprotective Agent and Memory Enhancer-Its Comparative Expression, Processing and Signaling in Mammalian CNS. Int J Mol Sci 2021; 22:ijms22041849. [PMID: 33673334 PMCID: PMC7918606 DOI: 10.3390/ijms22041849] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
A number of studies performed on rodents suggest that insulin-like growth factor 2 (IGF-2) or its analogs may possibly be used for treating some conditions like Alzheimer’s disease, Huntington’s disease, autistic spectrum disorders or aging-related cognitive impairment. Still, for translational research a comparative knowledge about the function of IGF-2 and related molecules in model organisms (rats and mice) and humans is necessary. There is a number of important differences in IGF-2 signaling between species. In the present review we emphasize species-specific patterns of IGF-2 expression in rodents, humans and some other mammals, using, among other sources, publicly available transcriptomic data. We provide a detailed description of Igf2 mRNA expression regulation and pre-pro-IGF-2 protein processing in different species. We also summarize the function of IGF-binding proteins. We describe three different receptors able to bind IGF-2 and discuss the role of IGF-2 signaling in learning and memory, as well as in neuroprotection. We hope that comprehensive understanding of similarities and differences in IGF-2 signaling between model organisms and humans will be useful for development of more effective medicines targeting IGF-2 receptors.
Collapse
|
8
|
Genazzani A, Panay N, Simoncini T, Depypere H, Mueck A, Egarter C, Biglia N, Fait T, Birkhaeuser M, Skouby SO, Brincat M, Goldstein S, Ruan X, Celis-Gonzales C, Palacios S. Purified and specific cytoplasmic pollen extract: a non-hormonal alternative for the treatment of menopausal symptoms. Gynecol Endocrinol 2020; 36:190-196. [PMID: 32019391 DOI: 10.1080/09513590.2020.1722994] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Research into non-hormonal, alternative therapies is necessary for women for whom menopausal hormone therapy is contraindicated or for women who do not wish to take hormones. This review focuses on one such non-hormonal option, namely, purified and specific cytoplasmic pollen extract, or PureCyTonin®. This extract has been evaluated in several preclinical and clinical studies, where it demonstrated its value as a safe and non-estrogenic alternative for menopause. This review presents the beneficial effects of PureCyTonin® in the treatment of menopausal symptoms (e.g. hot flushes) in healthy women, as well as in premenstrual syndrome. We discuss the mechanism of action of PureCyTonin®, an SSRI-'like' therapy. The lack of estrogenic effect demonstrated in preclinical studies suggests that PureCyTonin® may also be a suitable option for the management of menopausal symptoms in women with breast cancer.
Collapse
Affiliation(s)
- Andrea Genazzani
- Department of Gynecology and Obstetrics, University of Pisa, Pisa, Italy
| | - Nick Panay
- Queen Charlotte's & Chelsea and Westminster Hospitals and Imperial College, London, The International Menopause Society, London, UK
| | - Tommaso Simoncini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Herman Depypere
- Breast Clinic and Menopause Clinic, University Hospital, Gent, Belgium
| | - Alfred Mueck
- Department of Women's Health, University Hospitals of Tuebingen, Tuebingen, Germany
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Christian Egarter
- Department of Obstetrics and Gynecology, Division of Gynecologic Endocrinology and Reproductive Medicine, University of Vienna, Vienna, Austria
| | - Nicoletta Biglia
- Academic Division of Gynecology and Obstetrics, Mauriziano Hospital, University of Turin, Turin, Italy
| | - Tomas Fait
- Department of Obstetrics and Gynecology, Faculty Hospital Motol, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Birkhaeuser
- Department Obstetrics and Gynecology, Division of Gynecological Endocrinology and Reproductive Medicine, University of Bern, Bern, Switzerland
| | - Sven O Skouby
- Department of Gynecology and Obstetrics, Herlev University Hospital, Copenhagen, Denmark
| | - Mark Brincat
- Department of Obstetrics & Gynecology, Mater Dei Hospital, Msida, Malta
| | | | - Xiangyan Ruan
- Department of Women's Health, University Hospitals of Tuebingen, Tuebingen, Germany
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | | | | |
Collapse
|
9
|
Denley MCS, Gatford NJF, Sellers KJ, Srivastava DP. Estradiol and the Development of the Cerebral Cortex: An Unexpected Role? Front Neurosci 2018; 12:245. [PMID: 29887794 PMCID: PMC5981095 DOI: 10.3389/fnins.2018.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
The cerebral cortex undergoes rapid folding in an "inside-outside" manner during embryonic development resulting in the establishment of six discrete cortical layers. This unique cytoarchitecture occurs via the coordinated processes of neurogenesis and cell migration. In addition, these processes are fine-tuned by a number of extracellular cues, which exert their effects by regulating intracellular signaling pathways. Interestingly, multiple brain regions have been shown to develop in a sexually dimorphic manner. In many cases, estrogens have been demonstrated to play an integral role in mediating these sexual dimorphisms in both males and females. Indeed, 17β-estradiol, the main biologically active estrogen, plays a critical organizational role during early brain development and has been shown to be pivotal in the sexually dimorphic development and regulation of the neural circuitry underlying sex-typical and socio-aggressive behaviors in males and females. However, whether and how estrogens, and 17β-estradiol in particular, regulate the development of the cerebral cortex is less well understood. In this review, we outline the evidence that estrogens are not only present but are engaged and regulate molecular machinery required for the fine-tuning of processes central to the cortex. We discuss how estrogens are thought to regulate the function of key molecular players and signaling pathways involved in corticogenesis, and where possible, highlight if these processes are sexually dimorphic. Collectively, we hope this review highlights the need to consider how estrogens may influence the development of brain regions directly involved in the sex-typical and socio-aggressive behaviors as well as development of sexually dimorphic regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Nicholas J. F. Gatford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Katherine J. Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
10
|
Estrogen-induced neuroimmunomodulation as facilitator of and barrier to reproductive aging in brain and lymphoid organs. J Chem Neuroanat 2018; 95:6-12. [PMID: 29477446 DOI: 10.1016/j.jchemneu.2018.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/22/2018] [Accepted: 02/20/2018] [Indexed: 12/27/2022]
Abstract
Reproductive aging in females is marked by alterations in gonadal hormones, estrogen and progesterone, that facilitate cessation of reproductive cycles and onset of female-specific diseases such as autoimmune and neurodegenerative diseases, hormone-dependent cancers, and osteoporosis. Bidirectional communication between the three homeostatic systems, nervous system, endocrine system, and immune system, is essential for the maintenance of health and any dysfunction in the cross-talk promotes the development of diseases and cancer. The pleiotropic effects of estrogen on neural-immune interactions may promote either neuroprotection or inflammatory conditions depending on the site of action, dose and duration of treatment, type of estrogen receptors and its influence on intracellular signaling pathways, etc. Our studies involving treatment of early middle-aged female rats with low and high doses of estrogen and examining the brain areas, thymus, spleen, and lymph nodes revealed that estrogen-induced changes in neural-immune interactions are markedly affected in thymus followed by spleen and lymph nodes while it confers neuroprotection in the brain areas. These alterations are determined by antioxidant enzyme status, growth factors, intracellular signaling pathways involved in cell survival and inflammation, and metabolic enzymes and thus, may regulate the various stages in female reproductive aging. It is imperative that detailed longitudinal studies are carried out to understand the mechanisms of neuroendocrine-immune interactions in reproductive aging to facilitate healthy aging and for the development of better treatment strategies for female-specific diseases.
Collapse
|
11
|
Antidepressant-Like Effect of Lipid Extract of Channa striatus in Postpartum Model of Depression in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:1469209. [PMID: 29317891 PMCID: PMC5727658 DOI: 10.1155/2017/1469209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/15/2017] [Indexed: 12/16/2022]
Abstract
Postpartum depression affects 15% of women. Channa striatus, a freshwater fish, is consumed in local Malay population as a rejuvenating diet during postpartum period. This study evaluated the antidepressant-like effect of lipid extract of C. striatus fillet and its mechanism of action in female Sprague-Dawley rats in postpartum model of depression. The rats were ovariectomized and treated with high dose of progesterone and estradiol benzoate for 23 days to have hormone-simulated pregnancy. The day 24 and afterwards were considered as the postpartum period. During the postpartum period, lipid extract was administered at 125, 250, and 500 mg/kg through intraperitoneal route for 15 days. Fluoxetine (10 mg/kg) was used as the positive control. On postpartum day 15, the animals were tested in forced swimming test (FST) and open field test (OFT) followed by biochemical analysis. Withdrawal of hormone administration during the postpartum period induced depressive-like behavior in FST. Administration of lipid extract reversed that depressive-like behavior at 125, 250, and 500 mg/kg in FST. In OFT, it decreased the exploratory activity. The mechanism of the antidepressant-like effect may be mediated through the decrease in plasma corticosterone, increase in plasma oxytocin, and decrease in nuclear factor-kappa B in prefrontal cortex of rats.
Collapse
|
12
|
DiCarlo LM, Vied C, Nowakowski RS. The stability of the transcriptome during the estrous cycle in four regions of the mouse brain. J Comp Neurol 2017; 525:3360-3387. [PMID: 28685836 DOI: 10.1002/cne.24282] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 01/21/2023]
Abstract
We analyzed the transcriptome of the C57BL/6J mouse hypothalamus, hippocampus, neocortex, and cerebellum to determine estrous cycle-specific changes in these four brain regions. We found almost 16,000 genes are present in one or more of the brain areas but only 210 genes, ∼1.3%, are significantly changed as a result of the estrous cycle. The hippocampus has the largest number of differentially expressed genes (DEGs) (82), followed by the neocortex (76), hypothalamus (63), and cerebellum (26). Most of these DEGs (186/210) are differentially expressed in only one of the four brain regions. A key finding is the unique expression pattern of growth hormone (Gh) and prolactin (Prl). Gh and Prl are the only DEGs to be expressed during only one stage of the estrous cycle (metestrus). To gain insight into the function of the DEGs, we examined gene ontology and phenotype enrichment and found significant enrichment for genes associated with myelination, hormone stimulus, and abnormal hormone levels. Additionally, 61 of the 210 DEGs are known to change in response to estrogen in the brain. 50 of the 210 genes differentially expressed as a result of the estrous cycle are related to myelin and oligodendrocytes and 12 of the 63 DEGs in the hypothalamus are oligodendrocyte- and myelin-specific genes. This transcriptomic analysis reveals that gene expression in the female mouse brain is remarkably stable during the estrous cycle and demonstrates that the genes that do fluctuate are functionally related.
Collapse
Affiliation(s)
- Lisa M DiCarlo
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| | - Cynthia Vied
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida.,Translational Science Laboratory, Florida State University College of Medicine, Tallahassee, Florida
| | - Richard S Nowakowski
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| |
Collapse
|
13
|
Sárvári M, Kalló I, Hrabovszky E, Solymosi N, Rodolosse A, Liposits Z. Long-Term Estrogen Receptor Beta Agonist Treatment Modifies the Hippocampal Transcriptome in Middle-Aged Ovariectomized Rats. Front Cell Neurosci 2016; 10:149. [PMID: 27375434 PMCID: PMC4901073 DOI: 10.3389/fncel.2016.00149] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/27/2016] [Indexed: 11/13/2022] Open
Abstract
Estradiol (E2) robustly activates transcription of a broad array of genes in the hippocampal formation of middle-aged ovariectomized rats via estrogen receptors (ERα, ERβ, and G protein-coupled ER). Selective ERβ agonists also influence hippocampal functions, although their downstream molecular targets and mechanisms are not known. In this study, we explored the effects of long-term treatment with ERβ agonist diarylpropionitrile (DPN, 0.05 mg/kg/day, sc.) on the hippocampal transcriptome in ovariectomized, middle-aged (13 month) rats. Isolated hippocampal formations were analyzed by Affymetrix oligonucleotide microarray and quantitative real-time PCR. Four hundred ninety-seven genes fulfilled the absolute fold change higher than 2 (FC > 2) selection criterion. Among them 370 genes were activated. Pathway analysis identified terms including glutamatergic and cholinergic synapse, RNA transport, endocytosis, thyroid hormone signaling, RNA degradation, retrograde endocannabinoid signaling, and mRNA surveillance. PCR studies showed transcriptional regulation of 58 genes encoding growth factors (Igf2, Igfb2, Igf1r, Fgf1, Mdk, Ntf3, Bdnf), transcription factors (Otx2, Msx1), potassium channels (Kcne2), neuropeptides (Cck, Pdyn), peptide receptors (Crhr2, Oprm1, Gnrhr, Galr2, Sstr1, Sstr3), neurotransmitter receptors (Htr1a, Htr2c, Htr2a, Gria2, Gria3, Grm5, Gabra1, Chrm5, Adrb1), and vesicular neurotransmitter transporters (Slc32a1, Slc17a7). Protein-protein interaction analysis revealed networking of clusters associated with the regulation of growth/troph factor signaling, transcription, translation, neurotransmitter and neurohormone signaling mechanisms and potassium channels. Collectively, the results reveal the contribution of ERβ-mediated processes to the regulation of transcription, translation, neurogenesis, neuromodulation, and neuroprotection in the hippocampal formation of ovariectomized, middle-aged rats and elucidate regulatory channels responsible for DPN-altered functional patterns. These findings support the notion that selective activation of ERβ may be a viable approach for treating the neural symptoms of E2 deficiency in menopause.
Collapse
Affiliation(s)
- Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary; Faculty of Information Technology and Bionics, Pázmány Péter Catholic UniversityBudapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Norbert Solymosi
- Faculty of Veterinary Science, Szent István University Budapest, Hungary
| | - Annie Rodolosse
- Functional Genomics Core, Institute for Research in Biomedicine Barcelona, Spain
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary; Faculty of Information Technology and Bionics, Pázmány Péter Catholic UniversityBudapest, Hungary
| |
Collapse
|
14
|
Chong HP, Cordeaux Y, Ranjan YS, Richardson S, Liquet B, Smith GCS, Charnock‐Jones DS. Age-related changes in murine myometrial transcript profile are mediated by exposure to the female sex hormones. Aging Cell 2016; 15:177-80. [PMID: 26490259 PMCID: PMC4717263 DOI: 10.1111/acel.12406] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2015] [Indexed: 01/12/2023] Open
Abstract
In humans, the risk of operative first delivery increases linearly with maternal age. We previously hypothesized that prolonged, cyclical, prepregnancy exposure to estrogen and progesterone contributes to uterine aging. Here, we test this hypothesis. Myometrium was obtained from four groups of virgin mice: (i) 10- to 12-week- and 28- to 30-week-old mice; (ii) 10- to 12-week- and 38- to 40-week-old mice; (iii) 38-week-old mice that had an ovariectomy or sham operation early in life; (iv) 38-week-old mice that had been treated with progesterone or vehicle containing implants from 8 to 36 weeks. Transcript profiling was carried out using Affymetrix Gene ST 1.1 arrays, and data were normalized. We identified 60 differentially regulated transcripts associated with advancing age (group 1). We validated these changes in group 2 (P for overlap = 5.8 × 10(-46) ). Early ovariectomy prevented the age-related changes in myometrial transcript profile. Similarly, progesterone-mediated long-term ovarian suppression prevented the age-related changes in myometrial transcript profile. Interferon regulatory factor 7 (Irf7) mRNA was regulated by age and hormonal exposure, and was identified as a predicted regulator of the other differentially expressed transcripts by both promoter sequence and canonical pathway activation analysis (P = 8.47 × 10(-5) and P < 10(-10) , respectively). Immunohistochemistry demonstrated IRF7 in both mouse and human myometrium. We conclude the following: (i) Myometrial aging in mice is associated with reproducible changes in transcript profile; (ii) these changes can be prevented by interventions which inhibit cyclical changes in the female sex hormones; and (iii) IRF7 may be an important regulator of myometrial function and aging.
Collapse
Affiliation(s)
- Hsu P. Chong
- Department of Obstetrics and Gynaecology & NIHR Biomedical Research Centre University of Cambridge The Rosie Hospital Robinson Way Cambridge CB2 0SW UK
| | - Yolande Cordeaux
- Department of Obstetrics and Gynaecology & NIHR Biomedical Research Centre University of Cambridge The Rosie Hospital Robinson Way Cambridge CB2 0SW UK
| | - Yorain Sri Ranjan
- Department of Obstetrics and Gynaecology & NIHR Biomedical Research Centre University of Cambridge The Rosie Hospital Robinson Way Cambridge CB2 0SW UK
| | - Sylvia Richardson
- MRC Biostatistics Unit Cambridge Institute of Public Health Cambridge CB2 0SR UK
| | - Benoit Liquet
- MRC Biostatistics Unit Cambridge Institute of Public Health Cambridge CB2 0SR UK
| | - Gordon C. S. Smith
- Department of Obstetrics and Gynaecology & NIHR Biomedical Research Centre University of Cambridge The Rosie Hospital Robinson Way Cambridge CB2 0SW UK
| | - David Stephen Charnock‐Jones
- Department of Obstetrics and Gynaecology & NIHR Biomedical Research Centre University of Cambridge The Rosie Hospital Robinson Way Cambridge CB2 0SW UK
| |
Collapse
|
15
|
Duclot F, Kabbaj M. The estrous cycle surpasses sex differences in regulating the transcriptome in the rat medial prefrontal cortex and reveals an underlying role of early growth response 1. Genome Biol 2015; 16:256. [PMID: 26628058 PMCID: PMC4667491 DOI: 10.1186/s13059-015-0815-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/27/2015] [Indexed: 01/22/2023] Open
Abstract
Background Males and females differ in cognitive functions and emotional processing, which in part have been associated with baseline sex differences in gene expression in the medial prefrontal cortex. Nevertheless, a growing body of evidence suggests that sex differences in medial prefrontal cortex-dependent cognitive functions are attenuated by hormonal fluctuations within the menstrual cycle. Despite known genomic effects of ovarian hormones, the interaction of the estrous cycle with sex differences in gene expression in the medial prefrontal cortex remains unclear and warrants further investigations. Results We undertake a large-scale characterization of sex differences and their interaction with the estrous cycle in the adult medial prefrontal cortex transcriptome and report that females with high and low ovarian hormone levels exhibited a partly opposed sexually biased transcriptome. The extent of regulation within females vastly exceeds sex differences, and supports a multi-level reorganization of synaptic function across the estrous cycle. Genome-wide analysis of the transcription factor early growth response 1 binding highlights its role in controlling the synapse-related genes varying within females. Conclusions We uncover a critical influence of the estrous cycle on the adult rat medial prefrontal cortex transcriptome resulting in partly opposite sex differences in proestrus when compared to diestrus females, and we discovered a direct role for Early Growth Response 1 in this opposite regulation. In addition to illustrating the importance of accounting for the estrous cycle in females, our data set the ground for a better understanding of the female specificities in cognition and emotional processing. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0815-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Florian Duclot
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 W Call Street, Tallahassee, FL, 32306, USA. .,Program in Neuroscience, College of Medicine, Florida State University, 1115 W Call Street, Tallahassee, FL, 32306, USA.
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 W Call Street, Tallahassee, FL, 32306, USA. .,Program in Neuroscience, College of Medicine, Florida State University, 1115 W Call Street, Tallahassee, FL, 32306, USA.
| |
Collapse
|
16
|
DeMambro VE, Le PT, Guntur AR, Maridas DE, Canalis E, Nagano K, Baron R, Clemmons DR, Rosen CJ. Igfbp2 Deletion in Ovariectomized Mice Enhances Energy Expenditure but Accelerates Bone Loss. Endocrinology 2015; 156:4129-40. [PMID: 26230658 PMCID: PMC4606757 DOI: 10.1210/en.2014-1452] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Previously, we reported sexually dimorphic bone mass and body composition phenotypes in Igfbp2(-/-) mice (-/-), where male mice exhibited decreased bone and increased fat mass, whereas female mice displayed increased bone but no changes in fat mass. To investigate the interaction between IGF-binding protein (IGFBP)-2 and estrogen, we subjected Igfbp2 -/- and +/+ female mice to ovariectomy (OVX) or sham surgery at 8 weeks of age. At 20 weeks of age, mice underwent metabolic cage analysis and insulin tolerance tests before killing. At harvest, femurs were collected for microcomputed tomography, serum for protein levels, brown adipose tissue (BAT) and inguinal white adipose tissue (IWAT) adipose depots for histology, gene expression, and mitochondrial respiration analysis of whole tissue. In +/+ mice, serum IGFBP-2 dropped 30% with OVX. In the absence of IGFBP-2, OVX had no effect on preformed BAT; however, there was significant "browning" of the IWAT depot coinciding with less weight gain, increased insulin sensitivity, lower intraabdominal fat, and increased bone loss due to higher resorption and lower formation. Likewise, after OVX, energy expenditure, physical activity and BAT mitochondrial respiration were decreased less in the OVX-/- compared with OVX+/+. Mitochondrial respiration of IWAT was reduced in OVX+/+ yet remained unchanged in OVX-/- mice. These changes were associated with significant increases in Fgf21 and Foxc2 expression, 2 proteins known for their insulin sensitizing and browning of WAT effects. We conclude that estrogen deficiency has a profound effect on body and bone composition in the absence of IGFBP-2 and may be related to changes in fibroblast growth factor 21.
Collapse
Affiliation(s)
- Victoria E DeMambro
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Phuong T Le
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Anyonya R Guntur
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - David E Maridas
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Ernesto Canalis
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Kenichi Nagano
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Roland Baron
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - David R Clemmons
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Clifford J Rosen
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| |
Collapse
|
17
|
Abstract
UNLABELLED Parkinson's disease (PD) is characterized by the progressive loss of select neuronal populations, but the prodeath genes mediating the neurodegenerative processes remain to be fully elucidated. Trib3 (tribbles pseudokinase 3) is a stress-induced gene with proapoptotic activity that was previously described as highly activated at the transcriptional level in a 6-hydroxydopamine (6-OHDA) cellular model of PD. Here, we report that Trib3 immunostaining is elevated in dopaminergic neurons of the substantia nigra pars compacta (SNpc) of human PD patients. Trib3 protein is also upregulated in cellular models of PD, including neuronal PC12 cells and rat dopaminergic ventral midbrain neurons treated with 6-OHDA, 1-methyl-4-phenylpyridinium (MPP+), or α-synuclein fibrils (αSYN). In the toxin models, Trib3 induction is substantially mediated by the transcription factors CHOP and ATF4. Trib3 overexpression is sufficient to promote neuronal death; conversely, Trib3 knockdown protects neuronal PC12 cells as well as ventral midbrain dopaminergic neurons from 6-OHDA, MPP+, or αSYN. Mechanism studies revealed that Trib3 physically interacts with Parkin, a prosurvival protein whose loss of function is associated with PD. Elevated Trib3 reduces Parkin expression in cultured cells; and in the SNpc of PD patients, Parkin levels are reduced in a subset of dopaminergic neurons expressing high levels of Trib3. Loss of Parkin at least partially mediates the prodeath actions of Trib3 in that Parkin knockdown in cellular PD models abolishes the protective effect of Trib3 downregulation. Together, these findings identify Trib3 and its regulatory pathways as potential targets to suppress the progression of neuron death and degeneration in PD. SIGNIFICANCE STATEMENT Parkinson's disease (PD) is the most common neurodegenerative movement disorder. Current treatments ameliorate symptoms, but not the underlying neuronal death. Understanding the core neurodegenerative processes in PD is a prerequisite for identifying new therapeutic targets and, ultimately, curing this disease. Here, we describe a novel pathway involving the proapoptotic protein Trib3 in neuronal death associated with PD. These findings are supported by data from multiple cellular models of PD and by immunostaining of postmortem PD brains. Upstream, Trib3 is induced by the transcription factors ATF4 and CHOP; and downstream, Trib3 interferes with the PD-associated prosurvival protein Parkin to mediate death. These findings establish this new pathway as a potential and promising therapeutic target for treatment of PD.
Collapse
|
18
|
Stankiewicz AM, Goscik J, Dyr W, Juszczak GR, Ryglewicz D, Swiergiel AH, Wieczorek M, Stefanski R. Novel candidate genes for alcoholism--transcriptomic analysis of prefrontal medial cortex, hippocampus and nucleus accumbens of Warsaw alcohol-preferring and non-preferring rats. Pharmacol Biochem Behav 2015; 139:27-38. [PMID: 26455281 DOI: 10.1016/j.pbb.2015.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/06/2015] [Accepted: 10/06/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Animal models provide opportunity to study neurobiological aspects of human alcoholism. Changes in gene expression have been implicated in mediating brain functions, including reward system and addiction. The current study aimed to identify genes that may underlie differential ethanol preference in Warsaw High Preferring (WHP) and Warsaw Low Preferring (WLP) rats. METHODS Microarray analysis comparing gene expression in nucleus accumbens (NAc), hippocampus (HP) and medial prefrontal cortex (mPFC) was performed in male WHP and WLP rats bred for differences in ethanol preference. RESULTS Differential and stable between biological repeats expression of 345, 254 and 129 transcripts in NAc, HP and mPFC was detected. Identified genes and processes included known mediators of ethanol response (Mx2, Fam111a, Itpr1, Gabra4, Agtr1a, LTP/LTD, renin-angiotensin signaling pathway), toxicity (Sult1c2a, Ces1, inflammatory response), as well as genes involved in regulation of important addiction-related brain systems such as dopamine, tachykinin or acetylcholine (Gng7, Tac4, Slc5a7). CONCLUSIONS The identified candidate genes may underlie differential ethanol preference in an animal model of alcoholism. COMMENT Names of genes are written in italics, while names of proteins are written in standard font. Names of human genes/proteins are written in all capital letters. Names of rodent genes/proteins are written in capital letter followed by small letters.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Animal Behaviour, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Joanna Goscik
- Software Department, Faculty of Computer Science, Bialystok University of Technology, 15-351 Bialystok, Poland
| | - Wanda Dyr
- Department of Pharmacology and Physiology of the Nervous System, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Grzegorz R Juszczak
- Department of Animal Behaviour, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Danuta Ryglewicz
- First Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Artur H Swiergiel
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland; Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA71130, USA.
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Roman Stefanski
- Department of Pharmacology and Physiology of the Nervous System, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| |
Collapse
|
19
|
Gogos A, Sbisa AM, Sun J, Gibbons A, Udawela M, Dean B. A Role for Estrogen in Schizophrenia: Clinical and Preclinical Findings. Int J Endocrinol 2015; 2015:615356. [PMID: 26491441 PMCID: PMC4600562 DOI: 10.1155/2015/615356] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/21/2015] [Accepted: 08/23/2015] [Indexed: 12/30/2022] Open
Abstract
Gender differences in schizophrenia have been extensively researched and it is being increasingly accepted that gonadal steroids are strongly attributed to this phenomenon. Of the various hormones implicated, the estrogen hypothesis has been the most widely researched one and it postulates that estrogen exerts a protective effect by buffering females against the development and severity of the illness. In this review, we comprehensively analyse studies that have investigated the effects of estrogen, in particular 17β-estradiol, in clinical, animal, and molecular research with relevance to schizophrenia. Specifically, we discuss the current evidence on estrogen dysfunction in schizophrenia patients and review the clinical findings on the use of estradiol as an adjunctive treatment in schizophrenia patients. Preclinical research that has used animal models and molecular probes to investigate estradiol's underlying protective mechanisms is also substantially discussed, with particular focus on estradiol's impact on the major neurotransmitter systems implicated in schizophrenia, namely, the dopamine, serotonin, and glutamate systems.
Collapse
Affiliation(s)
- Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Alyssa M. Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia
| | - Jeehae Sun
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia
| | - Andrew Gibbons
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Madhara Udawela
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
20
|
Royall DR, Bishnoi RJ, Palmer RF. Serum IGF-BP2 strongly moderates age's effect on cognition: a MIMIC analysis. Neurobiol Aging 2015; 36:2232-2240. [PMID: 26043140 DOI: 10.1016/j.neurobiolaging.2015.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 03/30/2015] [Accepted: 04/04/2015] [Indexed: 01/13/2023]
Abstract
We have used structural equation models to explicitly distinguish functional status and therefore "dementia-relevant" variance in cognitive task performance (i.e., "δ" for dementia). Our approach is modular and can be directed to other targets. In this analysis, we construct a δ ortholog representing the "cognitive correlates of age" (cAGE). cAGE largely mediates age's effects on dementia severity, as rated by the Clinical Dementia Rating Scale Sum of boxes and has an area under the receiver operating curve = 0.96 for the diagnosis of Alzheimer's Disease versus controls. We then test cAGE's association with serum insulin-like growth factor binding protein 2 (IGF-BP2), which has previously been associated with age-related cognitive changes in animals, and with cortical atrophy in older humans. IGF-BP2's adverse effects on cognition are largely mediated through cAGE, independent of education, ethnicity, gender, depression ratings, serum homocysteine levels, hemoglobin A1c, and apolipoprotein e4 status. This suggests that age-specific cognitive decline may be moderated by IGF-BP2 and that modulation of that protein's function(s) might ameliorate age-specific cognitive impairments.
Collapse
Affiliation(s)
- Donald R Royall
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, TX, USA; Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Department of Family and Community Medicine, University of Texas Health Science Center, San Antonio, TX, USA; South Texas Veterans' Health System Audie L. Murphy Division GRECC, San Antonio, TX, USA.
| | - Ram J Bishnoi
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Raymond F Palmer
- Department of Family and Community Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
21
|
Sárvári M, Kalló I, Hrabovszky E, Solymosi N, Rodolosse A, Vastagh C, Auer H, Liposits Z. Hippocampal Gene Expression Is Highly Responsive to Estradiol Replacement in Middle-Aged Female Rats. Endocrinology 2015; 156:2632-45. [PMID: 25924104 DOI: 10.1210/en.2015-1109] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the hippocampus, estrogens are powerful modulators of neurotransmission, synaptic plasticity and neurogenesis. In women, menopause is associated with increased risk of memory disturbances, which can be attenuated by timely estrogen therapy. In animal models of menopause, 17β-estradiol (E2) replacement improves hippocampus-dependent spatial memory. Here, we explored the effect of E2 replacement on hippocampal gene expression in a rat menopause model. Middle-aged ovariectomized female rats were treated continuously for 29 days with E2, and then, the hippocampal transcriptome was investigated with Affymetrix expression arrays. Microarray data were analyzed by Bioconductor packages and web-based softwares, and verified with quantitative PCR. At standard fold change selection criterion, 156 genes responded to E2. All alterations but 4 were transcriptional activation. Robust activation (fold change > 10) occurred in the case of transthyretin, klotho, claudin 2, prolactin receptor, ectodin, coagulation factor V, Igf2, Igfbp2, and sodium/sulfate symporter. Classification of the 156 genes revealed major groups, including signaling (35 genes), metabolism (31 genes), extracellular matrix (17 genes), and transcription (16 genes). We selected 33 genes for further studies, and all changes were confirmed by real-time PCR. The results suggest that E2 promotes retinoid, growth factor, homeoprotein, neurohormone, and neurotransmitter signaling, changes metabolism, extracellular matrix composition, and transcription, and induces protective mechanisms via genomic effects. We propose that these mechanisms contribute to effects of E2 on neurogenesis, neural plasticity, and memory functions. Our findings provide further support for the rationale to develop safe estrogen receptor ligands for the maintenance of cognitive performance in postmenopausal women.
Collapse
Affiliation(s)
- Miklós Sárvári
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Erik Hrabovszky
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Norbert Solymosi
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Annie Rodolosse
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Csaba Vastagh
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Herbert Auer
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology (M.S., I.K., E.H., C.V., Z.L.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Faculty of Information Technology and Bionics (I.K., Z.L.), Pázmány Péter Catholic University, 1083 Budapest, Hungary; Faculty of Veterinary Science (N.S.), Szent István University, 1078 Budapest, Hungary; Functional Genomics Core (A.R.), Institute for Research in Biomedicine, 08028 Barcelona, Spain; and Functional Genomics Consulting (H.A.), 08780 Palleja, Spain
| |
Collapse
|
22
|
Pickard A, McCance DJ. IGF-Binding Protein 2 - Oncogene or Tumor Suppressor? Front Endocrinol (Lausanne) 2015; 6:25. [PMID: 25774149 PMCID: PMC4343188 DOI: 10.3389/fendo.2015.00025] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/13/2015] [Indexed: 01/08/2023] Open
Abstract
The role of insulin-like growth factor binding protein 2 (IGFBP2) in cancer is unclear. In general, IGFBP2 is considered to be oncogenic and its expression is often observed to be elevated in cancer. However, there are a number of conflicting reports in vitro and in vivo where IGFBP2 acts in a tumor suppressor manner. In this mini-review, we discuss the factors influencing the variation in IGFBP2 expression in cancer and our interpretation of these findings.
Collapse
Affiliation(s)
- Adam Pickard
- Centre for Cancer Research and Cell Biology, Queen’s University, Belfast, UK
- *Correspondence: Adam Pickard, Centre for Cancer Research and Cell Biology, Queen’s University Belfast, 97 Lisburn Road, Belfast BT7 9BL, UK e-mail:
| | - Dennis J. McCance
- Centre for Cancer Research and Cell Biology, Queen’s University, Belfast, UK
| |
Collapse
|
23
|
Humphreys GI, Ziegler YS, Nardulli AM. 17β-estradiol modulates gene expression in the female mouse cerebral cortex. PLoS One 2014; 9:e111975. [PMID: 25372139 PMCID: PMC4221195 DOI: 10.1371/journal.pone.0111975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/10/2014] [Indexed: 01/13/2023] Open
Abstract
17β-estradiol (E2) plays critical roles in a number of target tissues including the mammary gland, reproductive tract, bone, and brain. Although it is clear that E2 reduces inflammation and ischemia-induced damage in the cerebral cortex, the molecular mechanisms mediating the effects of E2 in this brain region are lacking. Thus, we examined the cortical transcriptome using a mouse model system. Female adult mice were ovariectomized and implanted with silastic tubing containing oil or E2. After 7 days, the cerebral cortices were dissected and RNA was isolated and analyzed using RNA-sequencing. Analysis of the transcriptomes of control and E2-treated animals revealed that E2 treatment significantly altered the transcript levels of 88 genes. These genes were associated with long term synaptic potentiation, myelination, phosphoprotein phosphatase activity, mitogen activated protein kinase, and phosphatidylinositol 3-kinase signaling. E2 also altered the expression of genes linked to lipid synthesis and metabolism, vasoconstriction and vasodilation, cell-cell communication, and histone modification. These results demonstrate the far-reaching and diverse effects of E2 in the cerebral cortex and provide valuable insight to begin to understand cortical processes that may fluctuate in a dynamic hormonal environment.
Collapse
Affiliation(s)
- Gwendolyn I. Humphreys
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yvonne S. Ziegler
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Ann M. Nardulli
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
24
|
Peng Y, Jiang B, Wu H, Dai R, Tan L. Effects of genistein on neuronal apoptosis, and expression of Bcl-2 and Bax proteins in the hippocampus of ovariectomized rats. Neural Regen Res 2014; 7:2874-81. [PMID: 25317139 PMCID: PMC4190945 DOI: 10.3969/j.issn.1673-5374.2012.36.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 08/01/2012] [Indexed: 11/18/2022] Open
Abstract
Genistein is one of several isoflavones that has a structure similar to 17β-estradiol, has a strong antioxidant effect, and a high affinity to estrogen receptors. At 15 weeks after ovariectomy, the expression of Bcl-2 in the hippocampus of rats decreased and Bax expression increased, with an obvious upregulation of apoptosis. However, intraperitoneal injection of genistein or 17β-estradiol for 15 consecutive weeks from the second day after operation upregulated Bcl-2 protein expression, downregulated Bax protein expression, and attenuated hippocampal neuron apoptosis. Our experimental findings indicate that long-term intervention with genistein can lead to a decrease in apoptosis in hippocampal neurons following ovariectomy, upregulate the expression of Bcl-2, and downregulate the expression of Bax. In addition, genistein and 17β-estradiol play equal anti-apoptotic and neuroprotective roles.
Collapse
Affiliation(s)
- Yun Peng
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Bo Jiang
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Huiling Wu
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Ruchun Dai
- Institute of Endocrinology and Metabolism, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Liming Tan
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
25
|
Sárvári M, Deli L, Kocsis P, Márk L, Maász G, Hrabovszky E, Kalló I, Gajári D, Vastagh C, Sümegi B, Tihanyi K, Liposits Z. Estradiol and isotype-selective estrogen receptor agonists modulate the mesocortical dopaminergic system in gonadectomized female rats. Brain Res 2014; 1583:1-11. [DOI: 10.1016/j.brainres.2014.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023]
|
26
|
Ascenzi P, Gustincich S, Marino M. Mammalian nerve globins in search of functions. IUBMB Life 2014; 66:268-76. [DOI: 10.1002/iub.1267] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/02/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Paolo Ascenzi
- Interdepartmental Laboratory of Electron Microscopy; University Roma Tre; Roma Italy
| | | | - Maria Marino
- Department of Science; University Roma Tre; Roma Italy
| |
Collapse
|
27
|
Ovariectomy and subsequent treatment with estrogen receptor agonists tune the innate immune system of the hippocampus in middle-aged female rats. PLoS One 2014; 9:e88540. [PMID: 24551115 PMCID: PMC3923802 DOI: 10.1371/journal.pone.0088540] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/07/2014] [Indexed: 01/23/2023] Open
Abstract
The innate immune system including microglia has a major contribution to maintenance of the physiological functions of the hippocampus by permanent monitoring of the neural milieu and elimination of tissue-damaging threats. The hippocampus is vulnerable to age-related changes ranging from gene expression to network connectivity. The risk of hippocampal deterioration increases with the decline of gonadal hormone supply. To explore the impact of hormone milieu on the function of the innate immune system in middle-aged female rats, we compared mRNA expression in the hippocampus after gonadal hormone withdrawal, with or without subsequent estrogen replacement using estradiol and isotype-selective estrogen receptor (ER) agonists. Targeted profiling assessed the status of the innate immune system (macrophage-associated receptors, complement, inhibitory neuronal ligands), local estradiol synthesis (P450 aromatase) and estrogen reception (ER). Results established upregulation of macrophage-associated (Cd45, Iba1, Cd68, Cd11b, Cd18, Fcgr1a, Fcgr2b) and complement (C3, factor B, properdin) genes in response to ovariectomy. Ovariectomy upregulated Cd22 and downregulated semaphorin3A (Sema3a) expression, indicating altered neuronal regulation of microglia. Ovariectomy also led to downregulation of aromatase and upregulation of ERα gene. Of note, analogous changes were observed in the hippocampus of postmenopausal women. In ovariectomized rats, estradiol replacement attenuated Iba1, Cd11b, Fcgr1a, C3, increased mannose receptor Mrc1, Cd163 and reversed Sema3a expression. In contrast, reduced expression of aromatase was not reversed by estradiol. While the effects of ERα agonist closely resembled those of estradiol, ERβ agonist was also capable of attenuating the expression of several macrophage-associated and complement genes. These data together indicate that the innate immune system of the aging hippocampus is highly responsive to the gonadal hormone milieu. In ovariectomized female rats, estradiol replacement exerts potent immunomodulatory effects including attenuation of microglia sensitization, initiation of M2-like activation and modulation of complement expression by targeting hippocampal neurons and glial cells through ERα and ERβ.
Collapse
|
28
|
Hoeflich A, Wirthgen E, David R, Classen CF, Spitschak M, Brenmoehl J. Control of IGFBP-2 Expression by Steroids and Peptide Hormones in Vertebrates. Front Endocrinol (Lausanne) 2014; 5:43. [PMID: 24778626 PMCID: PMC3985015 DOI: 10.3389/fendo.2014.00043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/20/2014] [Indexed: 12/03/2022] Open
Abstract
IGFBP-2 (1) has been described as a brain tumor oncogene (2) and is widely expressed in cancers from different origins (3-8). IGFBP-2 alone cannot cause malignant transformation, yet progression of brain tumors to higher grade (9) and also has been provided as a protective element in earlier stages of multistage colon carcinogenesis (10). Therefore, it is crucial to understand the factors that determine expression patterns of IGFBP-2 under normal and malignant conditions. The present review provides a comprehensive update of known factors that have an impact on expression of IGFBP-2.
Collapse
Affiliation(s)
- Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- *Correspondence: Andreas Hoeflich, Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, Dummerstorf 18196, Germany e-mail:
| | | | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock, Germany
| | | | - Marion Spitschak
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Julia Brenmoehl
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
29
|
Balgobin S, Montoya TI, Shi H, Acevedo JF, Keller PW, Riegel M, Wai CY, Word RA. Estrogen alters remodeling of the vaginal wall after surgical injury in guinea pigs. Biol Reprod 2013; 89:138. [PMID: 24174572 DOI: 10.1095/biolreprod.113.112367] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Loss of pelvic organ support (i.e., pelvic organ prolapse) is common in menopausal women. Surgical reconstruction of pelvic organ prolapse is plagued with high failure rates. The objective of this study was to determine the effects of estrogen on biomechanical properties, lysyl oxidase (LOX), collagen content, and histomorphology of the vagina with or without surgical injury. Nulliparous ovariectomized guinea pigs were treated systemically with either 50 μg/kg/day estradiol (E2,) or vehicle. After 2 wk, vaginal surgery was performed, and animals were treated with either beta-aminopropionitrile (BAPN, an irreversible LOX inhibitor), or vehicle to determine the role of LOX in recovery of the vaginal wall from injury with or without E2. Estradiol resulted in (i) significant growth, increased smooth muscle, and increased thickness of the vagina, (ii) increased distensibility without compromise of maximal force at failure, and (iii) increased total and cross-linked collagen. In the absence of E2, BAPN resulted in decreased collagen and vaginal wall strength in the area of the injury. In contrast, in E2-treated animals, increased distensibility, maximal forces, and total collagen were maintained despite BAPN. Interestingly, LOX mRNA was induced dramatically (9.5-fold) in the injured vagina with or without E2 at 4 days. By 21 days, however, LOX levels declined to near baseline in E2-deprived animals. LOX mRNA levels remained strikingly elevated (12-fold) at 21 days in the estrogenized vagina. The results suggest that prolonged E2 induced increases in LOX, and collagen cross-links may act to sustain a matrix environment that optimizes long-term surgical wound healing in the vagina.
Collapse
Affiliation(s)
- Sunil Balgobin
- Departments of Obstetrics and Gynecology and Animal Resource Center, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Willmes DM, Birkenfeld AL. The Role of INDY in Metabolic Regulation. Comput Struct Biotechnol J 2013; 6:e201303020. [PMID: 24688728 PMCID: PMC3962103 DOI: 10.5936/csbj.201303020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/02/2013] [Accepted: 12/02/2013] [Indexed: 01/20/2023] Open
Abstract
Reduced expression of the Indy (I'm Not Dead Yet) gene in D. melanogaster and C. elegans extends longevity. Indy and its mammalian homolog mINDY (Slc13a5, NaCT) are transporters of TCA cycle intermediates, mainly handling the uptake of citrate via the plasma membrane into the cytosol. Deletion of mINDY in mice leads to significant metabolic changes akin to caloric restriction, likely caused by reducing the effects of mINDY-imported citrate on fatty acid and cholesterol synthesis, glucose metabolism and ß-oxidation. This review will provide an overview on different mammalian SLC1 3 family members with a focus on mINDY (SLCl3A5) in glucose and energy metabolism and will highlight the role of mINDY as a putative therapeutic target for the treatment of obesity, non-alcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- Diana M Willmes
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| | - Andreas L Birkenfeld
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| |
Collapse
|
31
|
El Khoudary SR, Shields KJ, Chen HY, Matthews KA. Menopause, complement, and hemostatic markers in women at midlife: the Study of Women's Health Across the Nation. Atherosclerosis 2013; 231:54-8. [PMID: 24125410 PMCID: PMC3844281 DOI: 10.1016/j.atherosclerosis.2013.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/27/2013] [Accepted: 08/29/2013] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To evaluate whether higher circulating levels of complement proteins C3 and C4 are associated with menopausal status and with hemostatic/thrombus formation markers (circulating factor VII (factor VIIc), fibrinogen, plasminogen activator inhibitor-1 (PAI-1) and tissue plasminogen activator antigen (tPA-ag)) in a sample of midlife women. METHODS AND RESULTS A total of 100 women (50 late peri-/postmenopausal and 50 pre-/early peri- menopausal women) from the Study of Women's Health Across the Nation (SWAN) Pittsburgh site were included in the present analysis. Factor VIIc and PAI-1 were log transformed. Linear regression was used for analysis. The mean age of the study participants was 50.5 ± 2.6 years with 73% were Caucasian and 27% were African American. C3 but not C4 was significantly higher in postmenopausal women compared to premenopausal women (P value = 0.03), adjusting for age, race and BMI. In final model (adjusting for age, race, BMI and menopausal status), C3 was associated with higher levels of log PAI-1 (P value = 0.0009) and tPA-ag (P value = 0.0003), while C4 was associated with higher levels of log factor VIIc (P value = 0.04) and fibrinogen (P value = 0.005). CONCLUSIONS These data suggest that C3 and C4 may be related to blood clots via their associations with hemostatic markers and that C3 is related to menopausal status. Complement proteins C3 and C4 could be possible pathways by which postmenopausal women are at higher risk of atherosclerosis and cardiovascular related events. It is important to replicate these findings in a larger sample size.
Collapse
Affiliation(s)
| | - Kelly J. Shields
- West Penn Allegheny Health System’s research program, Allegheny General Hospital Lupus Center of Excellence, PA
| | | | | |
Collapse
|
32
|
Sodium-coupled dicarboxylate and citrate transporters from the SLC13 family. Pflugers Arch 2013; 466:119-30. [PMID: 24114175 DOI: 10.1007/s00424-013-1369-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/19/2013] [Accepted: 09/23/2013] [Indexed: 12/30/2022]
Abstract
The SLC13 family in humans and other mammals consists of sodium-coupled transporters for anionic substrates: three transporters for dicarboxylates/citrate and two transporters for sulfate. This review will focus on the di- and tricarboxylate transporters: NaDC1 (SLC13A2), NaDC3 (SLC13A3), and NaCT (SLC13A5). The substrates of these transporters are metabolic intermediates of the citric acid cycle, including citrate, succinate, and α-ketoglutarate, which can exert signaling effects through specific receptors or can affect metabolic enzymes directly. The SLC13 transporters are important for regulating plasma, urinary and tissue levels of these metabolites. NaDC1, primarily found on the apical membranes of renal proximal tubule and small intestinal cells, is involved in regulating urinary levels of citrate and plays a role in kidney stone development. NaDC3 has a wider tissue distribution and high substrate affinity compared with NaDC1. NaDC3 participates in drug and xenobiotic excretion through interactions with organic anion transporters. NaCT is primarily a citrate transporter located in the liver and brain, and its activity may regulate metabolic processes. The recent crystal structure of the Vibrio cholerae homolog, VcINDY, provides a new framework for understanding the mechanism of transport in this family. This review summarizes current knowledge of the structure, function, and regulation of the di- and tricarboxylate transporters of the SLC13 family.
Collapse
|
33
|
Meitzen J, Perry AN, Westenbroek C, Hedges VL, Becker JB, Mermelstein PG. Enhanced striatal β1-adrenergic receptor expression following hormone loss in adulthood is programmed by both early sexual differentiation and puberty: a study of humans and rats. Endocrinology 2013; 154:1820-31. [PMID: 23533220 PMCID: PMC3628022 DOI: 10.1210/en.2012-2131] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
After reproductive senescence or gonadectomy, changes occur in neural gene expression, ultimately altering brain function. The endocrine mechanisms underlying these changes in gene expression beyond immediate hormone loss are poorly understood. To investigate this, we measured changes in gene expression the dorsal striatum, where 17β-estradiol modulates catecholamine signaling. In human caudate, quantitative PCR determined a significant elevation in β1-adrenergic receptor (β1AR) expression in menopausal females when compared with similarly aged males. No differences were detected in β2-adrenergic and D1- and D2-dopamine receptor expression. Consistent with humans, adult ovariectomized female rats exhibited a similar increase in β1AR expression when compared with gonadectomized males. No sex difference in β1AR expression was detected between intact adults, prepubertal juveniles, or adults gonadectomized before puberty, indicating the necessity of pubertal development and adult ovariectomy. Additionally, increased β1AR expression in adult ovariectomized females was not observed if animals were masculinized/defeminized with testosterone injections as neonates. To generate a model system for assessing functional impact, increased β1AR expression was induced in female-derived cultured striatal neurons via exposure to and then removal of hormone-containing serum. Increased β1AR action on cAMP formation, cAMP response element-binding protein phosphorylation and gene expression was observed. This up-regulation of β1AR action was eliminated with 17β-estradiol addition to the media, directly implicating this hormone as a regulator of β1AR expression. Beyond having implications for the known sex differences in striatal function and pathologies, these data collectively demonstrate that critical periods early in life and at puberty program adult gene responsiveness to hormone loss after gonadectomy and potentially reproductive senescence.
Collapse
Affiliation(s)
- John Meitzen
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Bergeron M, Clémençon B, Hediger M, Markovich D. SLC13 family of Na+-coupled di- and tri-carboxylate/sulfate transporters. Mol Aspects Med 2013; 34:299-312. [DOI: 10.1016/j.mam.2012.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 11/16/2012] [Indexed: 12/22/2022]
|
35
|
Sárvári M, Hrabovszky E, Kalló I, Solymosi N, Likó I, Berchtold N, Cotman C, Liposits Z. Menopause leads to elevated expression of macrophage-associated genes in the aging frontal cortex: rat and human studies identify strikingly similar changes. J Neuroinflammation 2012. [PMID: 23206327 PMCID: PMC3558453 DOI: 10.1186/1742-2094-9-264] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background The intricate interactions between the immune, endocrine and central nervous systems shape the innate immune response of the brain. We have previously shown that estradiol suppresses expression of immune genes in the frontal cortex of middle-aged ovariectomized rats, but not in young ones reflecting elevated expression of these genes in middle-aged, ovarian hormone deficient animals. Here, we explored the impact of menopause on the microglia phenotype capitalizing on the differential expression of macrophage-associated genes in quiescent and activated microglia. Methods We selected twenty-three genes encoding phagocytic and recognition receptors expressed primarily in microglia, and eleven proinflammatory genes and followed their expression in the rat frontal cortex by real-time PCR. We used young, middle-aged and middle-aged ovariectomized rats to reveal age- and ovariectomy-related alterations. We analyzed the expression of the same set of genes in the postcentral and superior frontal gyrus of pre- and postmenopausal women using raw microarray data from our previous study. Results Ovariectomy caused up-regulation of four classic microglia reactivity marker genes including Cd11b, Cd18, Cd45 and Cd86. The change was reversible since estradiol attenuated transcriptional activation of the four marker genes. Expression of genes encoding phagocytic and toll-like receptors such as Cd11b, Cd18, C3, Cd32, Msr2 and Tlr4 increased, whereas scavenger receptor Cd36 decreased following ovariectomy. Ovarian hormone deprivation altered the expression of major components of estrogen and neuronal inhibitory signaling which are involved in the control of microglia reactivity. Strikingly similar changes took place in the postcentral and superior frontal gyrus of postmenopausal women. Conclusions Based on the overlapping results of rat and human studies we propose that the microglia phenotype shifts from the resting toward the reactive state which can be characterized by up-regulation of CD11b, CD14, CD18, CD45, CD74, CD86, TLR4, down-regulation of CD36 and unchanged CD40 expression. As a result of this shift, microglial cells have lower threshold for subsequent activation in the forebrain of postmenopausal women.
Collapse
Affiliation(s)
- Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony Utca 43, Budapest 1083, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The current male bias in biomedical research should be eliminated. The large sex differences in incidence and progression of diseases mean that sex-biased factors are an untapped source of factors that protect from disease. Greater understanding will come from intensified study of the "sexome," which is the sum of sex-biased effects on gene networks and cell systems. The global search for sites and mechanisms of sex-specific regulation in diverse tissues will provide unanticipated insights into physiological regulation and targets for novel therapies.
Collapse
Affiliation(s)
- Arthur P Arnold
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California 90095-7239, USA.
| | | |
Collapse
|
37
|
Farkas I, Sárvári M, Aller M, Okada N, Okada H, Likó I, Liposits Z. Estrogen receptor alpha and beta differentially mediate C5aR agonist evoked Ca2+-influx in neurons through L-type voltage-gated Ca2+ channels. Neurochem Int 2012; 60:631-9. [DOI: 10.1016/j.neuint.2012.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/19/2011] [Accepted: 02/21/2012] [Indexed: 01/19/2023]
|
38
|
Ota M, Fujii T, Nemoto K, Tatsumi M, Moriguchi Y, Hashimoto R, Sato N, Iwata N, Kunugi H. A polymorphism of the ABCA1 gene confers susceptibility to schizophrenia and related brain changes. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1877-83. [PMID: 21839797 DOI: 10.1016/j.pnpbp.2011.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/14/2011] [Accepted: 07/26/2011] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The ATP-binding cassette transporter A1 (ABCA1) mediates cellular cholesterol efflux through the transfer of cholesterol from the inner to the outer layer of the cell membrane and regulates extracellular cholesterol levels in the central nervous system. Several lines of evidence have indicated lipid and myelin abnormalities in schizophrenia. METHOD Initially, we examined the possible association of the polymorphisms of the ABCA1 gene (ABCA1) with susceptibility to schizophrenia in 506 patients with schizophrenia (DSM-IV) and 941 controls. The observed association was then subject to a replication analysis in an independent sample of 511 patients and 539 controls. We further examined the possible effect of the risk allele on gray matter volume assessed with magnetic resonance imaging (MRI) in 86 patients with schizophrenia (49 males) and 139 healthy controls (47 males). RESULTS In the initial association study, the 1587 K allele (rs2230808) was significantly more common in male patients with schizophrenia than in male controls. Although such a significant difference was not observed in the second sample alone, the increased frequency of the 1587 K allele in male patients remained to be significant in the combined male sample of 556 patients and 594 controls. Male schizophrenia patients carrying the 1587 K allele had a smaller amount of gray matter volume than those who did not carry the allele. CONCLUSION Our data suggest a male-specific association of the 1587 K allele of ABCA1 with susceptibility to schizophrenia and smaller gray matter volume in schizophrenia.
Collapse
Affiliation(s)
- Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sárvári M, Hrabovszky E, Kalló I, Solymosi N, Tóth K, Likó I, Széles J, Mahó S, Molnár B, Liposits Z. Estrogens regulate neuroinflammatory genes via estrogen receptors α and β in the frontal cortex of middle-aged female rats. J Neuroinflammation 2011; 8:82. [PMID: 21774811 PMCID: PMC3161870 DOI: 10.1186/1742-2094-8-82] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 07/20/2011] [Indexed: 12/18/2022] Open
Abstract
Background Estrogens exert anti-inflammatory and neuroprotective effects in the brain mainly via estrogen receptors α (ERα) and β (ERβ). These receptors are members of the nuclear receptor superfamily of ligand-dependent transcription factors. This study was aimed at the elucidation of the effects of ERα and ERβ agonists on the expression of neuroinflammatory genes in the frontal cortex of aging female rats. Methods To identify estrogen-responsive immunity/inflammation genes, we treated middle-aged, ovariectomized rats with 17β-estradiol (E2), ERα agonist 16α-lactone-estradiol (16α-LE2) and ERβ agonist diarylpropionitrile (DPN), or vehicle by Alzet minipump delivery for 29 days. Then we compared the transcriptomes of the frontal cortex of estrogen-deprived versus ER agonist-treated animals using Affymetrix Rat230 2.0 expression arrays and TaqMan-based quantitative real-time PCR. Microarray and PCR data were evaluated by using Bioconductor packages and the RealTime StatMiner software, respectively. Results Microarray analysis revealed the transcriptional regulation of 21 immunity/inflammation genes by 16α-LE2. The subsequent comparative real-time PCR study analyzed the isotype specific effects of ER agonists on neuroinflammatory genes of primarily glial origin. E2 regulated the expression of sixteen genes, including down-regulation of complement C3 and C4b, Ccl2, Tgfb1, macrophage expressed gene Mpeg1, RT1-Aw2, Cx3cr1, Fcgr2b, Cd11b, Tlr4 and Tlr9, and up-regulation of defensin Np4 and RatNP-3b, IgG-2a, Il6 and ER gene Esr1. Similar to E2, both 16α-LE2 and DPN evoked up-regulation of defensins, IgG-2a and Il6, and down-regulation of C3 and its receptor Cd11b, Ccl2, RT1-Aw2 and Fcgr2b. Conclusions These findings provide evidence that E2, 16α-LE2 and DPN modulate the expression of neuroinflammatory genes in the frontal cortex of middle-aged female rats via both ERα and ERβ. We propose that ERβ is a promising target to suppress regulatory functions of glial cells in the E2-deprived female brain and in various neuroinflammatory diseases.
Collapse
Affiliation(s)
- Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony utca 43, Budapest, 1083, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Buechel HM, Popovic J, Searcy JL, Porter NM, Thibault O, Blalock EM. Deep sleep and parietal cortex gene expression changes are related to cognitive deficits with age. PLoS One 2011; 6:e18387. [PMID: 21483696 PMCID: PMC3070733 DOI: 10.1371/journal.pone.0018387] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 03/06/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Age-related cognitive deficits negatively affect quality of life and can presage serious neurodegenerative disorders. Despite sleep disruption's well-recognized negative influence on cognition, and its prevalence with age, surprisingly few studies have tested sleep's relationship to cognitive aging. METHODOLOGY We measured sleep stages in young adult and aged F344 rats during inactive (enhanced sleep) and active (enhanced wake) periods. Animals were behaviorally characterized on the Morris water maze and gene expression profiles of their parietal cortices were taken. PRINCIPAL FINDINGS Water maze performance was impaired, and inactive period deep sleep was decreased with age. However, increased deep sleep during the active period was most strongly correlated to maze performance. Transcriptional profiles were strongly associated with behavior and age, and were validated against prior studies. Bioinformatic analysis revealed increased translation and decreased myelin/neuronal pathways. CONCLUSIONS The F344 rat appears to serve as a reasonable model for some common sleep architecture and cognitive changes seen with age in humans, including the cognitively disrupting influence of active period deep sleep. Microarray analysis suggests that the processes engaged by this sleep are consistent with its function. Thus, active period deep sleep appears temporally misaligned but mechanistically intact, leading to the following: first, aged brain tissue appears capable of generating the slow waves necessary for deep sleep, albeit at a weaker intensity than in young. Second, this activity, presented during the active period, seems disruptive rather than beneficial to cognition. Third, this active period deep sleep may be a cognitively pathologic attempt to recover age-related loss of inactive period deep sleep. Finally, therapeutic strategies aimed at reducing active period deep sleep (e.g., by promoting active period wakefulness and/or inactive period deep sleep) may be highly relevant to cognitive function in the aging community.
Collapse
Affiliation(s)
- Heather M. Buechel
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jelena Popovic
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - James L. Searcy
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Nada M. Porter
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Olivier Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Eric M. Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|