1
|
Roh J, Jang JP, Oh T, Kim J, Lee B, Hong YS, Jang JH, Ko SK. Protective effect of hygrolansamycin C against corticosterone-induced toxicity and oxidative stress-mediated via autophagy and the MAPK signaling pathway. Pharmacol Rep 2024; 76:368-378. [PMID: 38498259 DOI: 10.1007/s43440-024-00572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Excessive stress, a major problem in modern societies, affects people of all ages worldwide. Corticosterone is one of the most abundant hormones secreted during stressful conditions and is associated with various dysfunctions in the body. In particular, we aimed to investigate the protective effects of hygrolansamycin C (HYGC) against corticosterone-induced cellular stress, a manifestation of excessive stress prevalent in contemporary societies. METHODS We isolated HYGC from Streptomyces sp. KCB17JA11 and subjected PC12 cells to corticosterone-induced stress. The effects of HYGC were assessed by measuring autophagy and the expression of mitogen-activated protein kinase (MAPK) phosphorylation-related genes. We used established cellular and molecular techniques to analyze protein levels and pathways. RESULTS HYGC effectively protected cells against corticosterone-induced injury. Specifically, it significantly reduced corticosterone-induced oxidative stress and inhibited the expression of autophagy-related proteins induced by corticosterone, which provided mechanistic insight into the protective effects of HYGC. At the signaling level, HYGC suppressed c-Jun N-terminal kinase and extracellular signal-regulated kinase phosphorylation and p38 activation. CONCLUSIONS HYGC is a promising candidate to counteract corticosterone-induced apoptosis and oxidative stress. Autophagy and MAPK pathway inhibition contribute to the protective effects of HYGC. Our findings highlight the potential of HYGC as a therapeutic agent for stress-related disorders and serve as a stepping stone for further exploration and development of stress management strategies.
Collapse
Affiliation(s)
- Jongtae Roh
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Jun-Pil Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea
| | - Taehoon Oh
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Korea
| | - Jihong Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Korea
| | - Byeongsan Lee
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Korea
| | - Young-Soo Hong
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Jae-Hyuk Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea.
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea.
| | - Sung-Kyun Ko
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Korea.
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea.
| |
Collapse
|
2
|
Kim S, Chaudhary PK, Kim S. Role of Prednisolone in Platelet Activation by Inhibiting TxA 2 Generation through the Regulation of cPLA 2 Phosphorylation. Animals (Basel) 2023; 13:ani13081299. [PMID: 37106862 PMCID: PMC10135208 DOI: 10.3390/ani13081299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Glucocorticoids have been commonly used in the treatment of inflammation and immune-mediated diseases in human beings and small animals such as cats and dogs. However, excessive use can lead to Cushing's syndrome along with several thrombotic and cardiovascular diseases. Although it is well-known that glucocorticoids exert a significant effect on coagulation, the effect of cortisol on platelet function is much less clear. Thus, we aimed to study the effects of prednisolone, one of the commonly used glucocorticoids, on the regulation of platelet function using murine platelets. We first evaluated the concentration-dependent effect of prednisolone on 2-MeSADP-induced platelet function and found that the 2-MeSADP-induced secondary wave of aggregation and dense granule secretion were completely inhibited from 500 nM prednisolone. Since 2-MeSADP-induced secretion and the resultant secondary wave of aggregation are mediated by TxA2 generation, this result suggested a role of prednisolone in platelet TxA2 generation. Consistently, prednisolone did not affect the 2-MeSADP-induced aggregation in aspirinated platelets, where the secondary wave of aggregation and secretion were blocked by eliminating the contribution of TxA2 generation by aspirin. In addition, thrombin-induced platelet aggregation and secretion were inhibited in the presence of prednisolone by inhibiting the positive-feedback effect of TxA2 generation on platelet function. Furthermore, prednisolone completely inhibited 2-MeSADP-induced TxA2 generation, confirming the role of prednisolone in TxA2 generation. Finally, Western blot analysis revealed that prednisolone significantly inhibited 2-MeSADP-induced cytosolic phospholipase A2 (cPLA2) and ERK phosphorylation in non-aspirinated platelets, while only cPLA2 phosphorylation, but not ERK phosphorylation, was significantly inhibited by prednisolone in aspirinated platelets. In conclusion, prednisolone affects platelet function by the inhibition of TxA2 generation through the regulation of cPLA2 phosphorylation, thereby shedding light on its clinical characterization and treatment efficacy in dogs with hypercortisolism in the future.
Collapse
Affiliation(s)
- Sanggu Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Preeti Kumari Chaudhary
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Soochong Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
3
|
Kokkinopoulou I, Diakoumi A, Moutsatsou P. Glucocorticoid Receptor Signaling in Diabetes. Int J Mol Sci 2021; 22:ijms222011173. [PMID: 34681832 PMCID: PMC8537243 DOI: 10.3390/ijms222011173] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022] Open
Abstract
Stress and depression increase the risk of Type 2 Diabetes (T2D) development. Evidence demonstrates that the Glucocorticoid (GC) negative feedback is impaired (GC resistance) in T2D patients resulting in Hypothalamic-Pituitary-Adrenal (HPA) axis hyperactivity and hypercortisolism. High GCs, in turn, activate multiple aspects of glucose homeostasis in peripheral tissues leading to hyperglycemia. Elucidation of the underlying molecular mechanisms revealed that Glucocorticoid Receptor (GR) mediates the GC-induced dysregulation of glucose production, uptake and insulin signaling in GC-sensitive peripheral tissues, such as liver, skeletal muscle, adipose tissue, and pancreas. In contrast to increased GR peripheral sensitivity, an impaired GR signaling in Peripheral Blood Mononuclear Cells (PBMCs) of T2D patients, associated with hyperglycemia, hyperlipidemia, and increased inflammation, has been shown. Given that GR changes in immune cells parallel those in brain, the above data implicate that a reduced brain GR function may be the biological link among stress, HPA hyperactivity, hypercortisolism and hyperglycemia. GR polymorphisms have also been associated with metabolic disturbances in T2D while dysregulation of micro-RNAs—known to target GR mRNA—has been described. Collectively, GR has a crucial role in T2D, acting in a cell-type and context-specific manner, leading to either GC sensitivity or GC resistance. Selective modulation of GR signaling in T2D therapy warrants further investigation.
Collapse
|
4
|
Lopes da Cunha P, Tintorelli R, Correa J, Budriesi P, Viola H. Behavioral tagging as a mechanism for aversive-memory formation under acute stress. Eur J Neurosci 2021; 55:2651-2665. [PMID: 33914357 DOI: 10.1111/ejn.15249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/01/2021] [Accepted: 04/15/2021] [Indexed: 02/03/2023]
Abstract
The behavioral tagging (BT) hypothesis postulates that a weak learning experience, which only induces short-term memory, may benefit from another event that provides plasticity-related proteins (PRPs) to establish a long-lasting memory. According to BT, the weak experience sets a transient learning tag at specific activated sites, and its temporal and spatial convergence with the PRPs allows the long-term memory (LTM) formation. In this work, rats were subjected to a weak inhibitory avoidance (IAw) training and we observed that acute stress (elevated platform, EP) experienced 1 hr before IAw promoted IA-LTM formation. This effect was dependent on glucocorticoid-receptor activity as well as protein synthesis in the dorsal hippocampus. However, the same stress has negative effects on IA-LTM formation when training is strong, probably by competing for necessary PRPs. Furthermore, our experiments showed that EP immediately after training did not impair the setting of the learning tag and even facilitated IA-LTM formation. These findings reveal different impacts of a given acute stressful experience on the formation of an aversive memory that could be explained by BT processes.
Collapse
Affiliation(s)
- Pamela Lopes da Cunha
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ramiro Tintorelli
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Correa
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Budriesi
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Haydee Viola
- Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular "Dr. Hector Maldonado" (FBMC), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
5
|
Ragozzino FJ, Arnold RA, Kowalski CW, Savenkova MI, Karatsoreos IN, Peters JH. Corticosterone inhibits vagal afferent glutamate release in the nucleus of the solitary tract via retrograde endocannabinoid signaling. Am J Physiol Cell Physiol 2020; 319:C1097-C1106. [PMID: 32966126 DOI: 10.1152/ajpcell.00190.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Circulating blood glucocorticoid levels are dynamic and responsive to stimuli that impact autonomic function. In the brain stem, vagal afferent terminals release the excitatory neurotransmitter glutamate to neurons in the nucleus of the solitary tract (NTS). Vagal afferents integrate direct visceral signals and circulating hormones with ongoing NTS activity to control autonomic function and behavior. Here, we investigated the effects of corticosterone (CORT) on glutamate signaling in the NTS using patch-clamp electrophysiology on brain stem slices containing the NTS and central afferent terminals from male C57BL/6 mice. We found that CORT rapidly decreased both action potential-evoked and spontaneous glutamate signaling. The effects of CORT were phenocopied by dexamethasone and blocked by mifepristone, consistent with glucocorticoid receptor (GR)-mediated signaling. While mRNA for GR was present in both the NTS and vagal afferent neurons, selective intracellular quenching of G protein signaling in postsynaptic NTS neurons eliminated the effects of CORT. We then investigated the contribution of retrograde endocannabinoid signaling, which has been reported to transduce nongenomic GR effects. Pharmacological or genetic elimination of the cannabinoid type 1 receptor signaling blocked CORT suppression of glutamate release. Together, our results detail a mechanism, whereby the NTS integrates endocrine CORT signals with fast neurotransmission to control autonomic reflex pathways.
Collapse
Affiliation(s)
- Forrest J Ragozzino
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Rachel A Arnold
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Cody W Kowalski
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Marina I Savenkova
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington.,Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - James H Peters
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
6
|
Amrani Y, Panettieri RA, Ramos-Ramirez P, Schaafsma D, Kaczmarek K, Tliba O. Important lessons learned from studies on the pharmacology of glucocorticoids in human airway smooth muscle cells: Too much of a good thing may be a problem. Pharmacol Ther 2020; 213:107589. [PMID: 32473159 DOI: 10.1016/j.pharmthera.2020.107589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) are the treatment of choice for chronic inflammatory diseases such as asthma. Despite proven effective anti-inflammatory and immunosuppressive effects, long-term and/or systemic use of GCs can potentially induce adverse effects. Strikingly, some recent experimental evidence suggests that GCs may even exacerbate some disease outcomes. In asthma, airway smooth muscle (ASM) cells are among the targets of GC therapy and have emerged as key contributors not only to bronchoconstriction, but also to airway inflammation and remodeling, as implied by experimental and clinical evidence. We here will review the beneficial effects of GCs on ASM cells, emphasizing the differential nature of GC effects on pro-inflammatory genes and on other features associated with asthma pathogenesis. We will also summarize evidence describing how GCs can potentially promote pro-inflammatory and remodeling features in asthma with a specific focus on ASM cells. Finally, some of the possible solutions to overcome these unanticipated effects of GCs will be discussed.
Collapse
Affiliation(s)
- Yassine Amrani
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, Leicester Biomedical Research Center Respiratory, Leicester, UK
| | - Reynold A Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Patricia Ramos-Ramirez
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | | | - Klaudia Kaczmarek
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | - Omar Tliba
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA.
| |
Collapse
|
7
|
Cultured hippocampal neurons of dystrophic mdx mice respond differently from those of wild type mice to an acute treatment with corticosterone. Exp Cell Res 2020; 386:111715. [PMID: 31711918 DOI: 10.1016/j.yexcr.2019.111715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022]
Abstract
Duchenne muscular dystrophy is a lethal genetic disease characterised by progressive degeneration of skeletal muscles induced by deficiency of dystrophin, a cytoskeletal protein expressed in myocytes and in certain neuron populations. The severity of the neurological disorder varies in humans and animal models owing to dysfunction in numerous brain areas, including the hippocampus. Cyclic treatments with high-dose glucocorticoids remain a major pharmacological approach for treating the disease; however, elevated systemic levels of either stress-induced or exogenously administered anti-inflammatory molecules dramatically affect hippocampal activity. In this study, we analysed and compared the response of hippocampal neurons isolated from wild-type and dystrophic mdx mice to acute administration of corticosterone in vitro, without the influence of other glucocorticoid-regulated processes. Our results showed that in neurons of mdx mice, both the genomic and intracellular signalling-mediated responses to corticosterone were affected compared to those in wild-type animals, evoking the characteristic response to detrimental chronic glucocorticoid exposure. Responsiveness to glucocorticoids is, therefore, another function of hippocampal neurons possibly affected by deficiency of Dp427 since embryonic development. Knowing the pivotal role of hippocampus in stress hormone signalling, attention should be paid to the effects that prolonged glucocorticoid treatments may have on this and other brain areas of DMD patients.
Collapse
|
8
|
Koziol-White C, Johnstone TB, Corpuz ML, Cao G, Orfanos S, Parikh V, Deeney B, Tliba O, Ostrom RS, Dainty I, Panettieri RA. Budesonide enhances agonist-induced bronchodilation in human small airways by increasing cAMP production in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2019; 318:L345-L355. [PMID: 31747297 DOI: 10.1152/ajplung.00393.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The nongenomic mechanisms by which glucocorticoids modulate β2 agonist-induced-bronchodilation remain elusive. Our studies aimed to elucidate mechanisms mediating the beneficial effects of glucocorticoids on agonist-induced bronchodilation. Utilizing human precision-cut lung slices (hPCLS), we measured bronchodilation to formoterol, prostaglandin E2 (PGE2), cholera toxin (CTX), or forskolin in the presence and absence of budesonide. Using cultured human airway smooth muscle (HASM), intracellular cAMP was measured in live cells following exposure to formoterol, PGE2, or forskolin in the presence or absence of budesonide. We showed that simultaneous budesonide administration amplified formoterol-induced bronchodilation and attenuated agonist-induced phosphorylation of myosin light chain, a necessary signaling event mediating force generation. In parallel studies, cAMP levels were augmented by simultaneous exposure of HASM cells to formoterol and budesonide. Budesonide, fluticasone, and prednisone alone rapidly increased cAMP levels, but steroids alone had little effect on bronchodilation in hPCLS. Bronchodilation induced by PGE2, CTX, or forskolin was also augmented by simultaneous exposure to budesonide in hPCLS. Furthermore, HASM cells expressed membrane-bound glucocorticoid receptors that failed to translocate with glucocorticoid stimulation and that potentially mediated the rapid effects of steroids on β2 agonist-induced bronchodilation. Knockdown of glucocorticoid receptor-α had little effect on budesonide-induced and steroid-dependent augmentation of formoterol-induced cAMP generation in HASM. Collectively, these studies suggest that glucocorticoids amplify cAMP-dependent bronchodilation by directly increasing cAMP levels. These studies identify a molecular mechanism by which the combination of glucocorticoids and β2 agonists may augment bronchodilation in diseases such as asthma or chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Timothy B Johnstone
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| | - Maia L Corpuz
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Sarah Orfanos
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Vishal Parikh
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Brian Deeney
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Omar Tliba
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, New York
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| | - Ian Dainty
- Bioscience, Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
9
|
Johnstone WM, Honeycutt JL, Deck CA, Borski RJ. Nongenomic glucocorticoid effects and their mechanisms of action in vertebrates. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:51-96. [PMID: 31122395 DOI: 10.1016/bs.ircmb.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Glucocorticoids (GC) act on multiple organ systems to regulate a variety of physiological processes in vertebrates. Due to their immunosuppressive and anti-inflammatory actions, glucocorticoids are an attractive target for pharmaceutical development. Accordingly, they are one of the most widely prescribed classes of therapeutics. Through the classical mechanism of steroid action, glucocorticoids are thought to mainly affect gene transcription, both in a stimulatory and suppressive fashion, regulating de novo protein synthesis that subsequently leads to the physiological response. However, over the past three decades multiple lines of evidence demonstrate that glucocorticoids may work through rapid, nonclassical mechanisms that do not require alterations in gene transcription or translation. This review assimilates evidence across the vertebrate taxa on the diversity of nongenomic actions of glucocorticoids and the membrane-associated cellular mechanisms that may underlie rapid glucocorticoid responses to include potential binding sites characterized to date.
Collapse
Affiliation(s)
- William M Johnstone
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Jamie L Honeycutt
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Courtney A Deck
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Russell J Borski
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
10
|
Barfield ET, Gourley SL. Prefrontal cortical trkB, glucocorticoids, and their interactions in stress and developmental contexts. Neurosci Biobehav Rev 2018; 95:535-558. [PMID: 30477984 PMCID: PMC6392187 DOI: 10.1016/j.neubiorev.2018.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/14/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
The tropomyosin/tyrosine receptor kinase B (trkB) and glucocorticoid receptor (GR) regulate neuron structure and function and the hormonal stress response. Meanwhile, disruption of trkB and GR activity (e.g., by chronic stress) can perturb neuronal morphology in cortico-limbic regions implicated in stressor-related illnesses like depression. Further, several of the short- and long-term neurobehavioral consequences of stress depend on the developmental timing and context of stressor exposure. We review how the levels and activities of trkB and GR in the prefrontal cortex (PFC) change during development, interact, are modulated by stress, and are implicated in depression. We review evidence that trkB- and GR-mediated signaling events impact the density and morphology of dendritic spines, the primary sites of excitatory synapses in the brain, highlighting effects in adolescents when possible. Finally, we review the role of neurotrophin and glucocorticoid systems in stress-related metaplasticity. We argue that better understanding the long-term effects of developmental stressors on PFC trkB, GR, and related factors may yield insights into risk for chronic, remitting depression and related neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Elizabeth T Barfield
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| | - Shannon L Gourley
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Molecular and Systems Pharmacology Program, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
11
|
Panettieri RA, Schaafsma D, Amrani Y, Koziol-White C, Ostrom R, Tliba O. Non-genomic Effects of Glucocorticoids: An Updated View. Trends Pharmacol Sci 2018; 40:38-49. [PMID: 30497693 DOI: 10.1016/j.tips.2018.11.002] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/09/2018] [Accepted: 11/01/2018] [Indexed: 01/01/2023]
Abstract
Glucocorticoid (GC) anti-inflammatory effects generally require a prolonged onset of action and involve genomic processes. Because of the rapidity of some of the GC effects, however, the concept that non-genomic actions may contribute to GC mechanisms of action has arisen. While the mechanisms have not been completely elucidated, the non-genomic effects may play a role in the management of inflammatory diseases. For instance, we recently reported that GCs 'rapidly' enhanced the effects of bronchodilators, agents used in the treatment of allergic asthma. In this review article, we discuss (i) the non-genomic effects of GCs on pathways relevant to the pathogenesis of inflammatory diseases and (ii) the putative role of the membrane GC receptor. Since GC side effects are often considered to be generated through its genomic actions, understanding GC non-genomic effects will help design GCs with a better therapeutic index.
Collapse
Affiliation(s)
- Reynold A Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson School of Medicine, New Brunswick, NJ, USA
| | | | - Yassine Amrani
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, Leicester Biomedical Research Center Respiratory, Leicester, UK
| | - Cynthia Koziol-White
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson School of Medicine, New Brunswick, NJ, USA
| | - Rennolds Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Omar Tliba
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA.
| |
Collapse
|
12
|
Bekhbat M, Rowson SA, Neigh GN. Checks and balances: The glucocorticoid receptor and NFĸB in good times and bad. Front Neuroendocrinol 2017; 46:15-31. [PMID: 28502781 PMCID: PMC5523465 DOI: 10.1016/j.yfrne.2017.05.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/21/2017] [Accepted: 05/09/2017] [Indexed: 01/23/2023]
Abstract
Mutual regulation and balance between the endocrine and immune systems facilitate an organism's stress response and are impaired following chronic stress or prolonged immune activation. Concurrent alterations in stress physiology and immunity are increasingly recognized as contributing factors to several stress-linked neuropsychiatric disorders including depression, anxiety, and post-traumatic stress disorder. Accumulating evidence suggests that impaired balance and crosstalk between the glucocorticoid receptor (GR) and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) - effectors of the stress and immune axes, respectively - may play a key role in mediating the harmful effects of chronic stress on mood and behavior. Here, we first review the molecular mechanisms of GR and NFκB interactions in health, then describe potential shifts in the GR-NFκB dynamics in chronic stress conditions within the context of brain circuitry relevant to neuropsychiatric diseases. Furthermore, we discuss developmental influences and sex differences in the regulation of these two transcription factors.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Emory University, Graduate Division of Biological Sciences, Neuroscience Graduate Program, United States
| | - Sydney A Rowson
- Emory University, Graduate Division of Biological Sciences, Molecular and Systems Pharmacology Graduate Studies Program, United States
| | - Gretchen N Neigh
- Virginia Commonwealth University, Department of Anatomy & Neurobiology, United States.
| |
Collapse
|
13
|
Yang S, Roselli F, Patchev AV, Yu S, Almeida OFX. Non-receptor-tyrosine kinases integrate fast glucocorticoid signaling in hippocampal neurons. J Biol Chem 2013; 288:23725-39. [PMID: 23818519 DOI: 10.1074/jbc.m113.470146] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite numerous descriptions of rapid effects of corticosterone on neuronal function, the intracellular mechanisms responsible for these changes remain elusive. The present comprehensive analysis reveals that signaling from a membrane-located G protein-coupled receptor activates PKC, Akt/PKB, and PKA, which subsequently trigger the phosphorylation of the tyrosine kinases Pyk2, Src, and Abl. These changes induce rapid cytoskeletal rearrangements (increased PSD-95 co-clustering) within the post-synaptic density; these events are accompanied by increased surface NMDA receptor expression, reflecting corticosterone-induced inhibition of NMDA receptor endocytosis. Notably, none of these signaling mechanisms require de novo protein synthesis. The observed up-regulation of ERK1/2 (downstream of NMDA receptor signaling) together with the fact that c-Abl integrates cytoplasmic and nuclear functions introduces a potential mechanism through which rapid signaling initiated at the plasma membrane may eventually determine the long term integrated response to corticosterone by impacting on the transcriptional machinery that is regulated by classical, nuclear mineralocorticoid, and glucocorticoid receptors.
Collapse
Affiliation(s)
- Silei Yang
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | | | | | | |
Collapse
|
14
|
Samarasinghe RA, Witchell SF, DeFranco DB. Cooperativity and complementarity: synergies in non-classical and classical glucocorticoid signaling. Cell Cycle 2012; 11:2819-27. [PMID: 22801547 DOI: 10.4161/cc.21018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids (GCs) are an ubiquitous class of steroid hormones that exert a wide array of physiological effects. Traditionally, GC action has been considered to primarily involve transcriptional effects following the binding of hormone to the glucocorticoid receptor (GR) and subsequent activation or repression of target genes. However, a number of findings suggest that cellular responses following GC exposure may be mediated by transcription-independent, or "non-classical," mechanisms. We have added to this growing body of work by recently uncovering a novel GC signaling pathway that operates through plasma membrane GRs to limit gap junction intercellular signaling and limit the proliferation of neural progenitor cells (NPCs). In this review, we highlight our current state of knowledge of non-classical GR signaling, in particular as it applies to neuronal function. Using NPCs as a cellular model, we speculate on the components of this non-classical pathway and the mechanisms whereby a number of cytoplasmic and nuclear signaling events may be integrated.
Collapse
Affiliation(s)
- Ranmal A Samarasinghe
- Department of Neuroscience, University of Pittsburgh, School of Medicine, and Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | | | | |
Collapse
|
15
|
Groeneweg FL, Karst H, de Kloet ER, Joëls M. Mineralocorticoid and glucocorticoid receptors at the neuronal membrane, regulators of nongenomic corticosteroid signalling. Mol Cell Endocrinol 2012; 350:299-309. [PMID: 21736918 DOI: 10.1016/j.mce.2011.06.020] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 06/15/2011] [Accepted: 06/20/2011] [Indexed: 02/06/2023]
Abstract
The balance between corticosteroid actions induced via activation of the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR) determines the brain's response to stress. While both receptors are best known for their delayed genomic role, it has become increasingly evident that they can also associate with the plasma membrane and act as mediators of rapid, nongenomic signalling. Nongenomic corticosteroid actions in the brain are required for the coordination of a rapid adaptive response to stress; membrane-associated MRs and GRs play a major role herein. However, many questions regarding the underlying mechanism are still unresolved. How do MR and GR translocate to the membrane and what are their downstream signalling partners? In this review we discuss these issues based on insights obtained from related receptors, most notably the estrogen receptor α.
Collapse
Affiliation(s)
- Femke L Groeneweg
- Department of Medical Pharmacology, Leiden Amsterdam Centre for Drug Research, Leiden University Medical Centre, Leiden University, Einsteinweg 55, Leiden, The Netherlands.
| | | | | | | |
Collapse
|
16
|
Nongenomic glucocorticoid receptor action regulates gap junction intercellular communication and neural progenitor cell proliferation. Proc Natl Acad Sci U S A 2011; 108:16657-62. [PMID: 21930911 DOI: 10.1073/pnas.1102821108] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) are used to treat pregnant women at risk for preterm delivery; however, prenatal exposure to GCs may trigger adverse neurological side effects due to reduced neural progenitor cell (NPC) proliferation. Whereas many established cell-cycle regulators impact NPC proliferation, other signaling molecules, such as the gap junction protein connexin-43 (Cx43), also influence proliferation. Gap junction intercellular communication (GJIC) is influenced by GCs in some cells, but such hormone effects have not been examined in coupled stem cells. We found that both continuous and transient exposure of embryonic day 14.5 mouse neurosphere cultures to dexamethasone (DEX) limits proliferation of coupled NPCs, which is manifested by both a reduction in S-phase progression and enhanced cell-cycle exit. A short (i.e., 1-h) DEX treatment also reduced GJIC as measured by live-cell fluorescence recovery after photobleaching, and altered the synchrony of spontaneous calcium transients in coupled NPCs. GC effects on GJIC in NPCs are transcription-independent and mediated through plasma membrane glucocorticoid receptors (GRs). This nongenomic pathway operates through lipid raft-associated GRs via a site-specific, MAPK-dependent phosphorylation of Cx43, which is linked to GR via caveolin-1 (Cav-1) and c-src. Cav-1 is essential for this nongenomic action of GR, as DEX effects on GJIC, Cx43 phosphorylation, and MAPK activation are not observed in Cav-1 knockout NPCs. As transient pharmacologic inhibition of GJIC triggers reduced S-phase progression but not enhanced cell-cycle exit, the nongenomic GR signaling pathway may operate via distinct downstream effectors to alter the proliferative capacity of NPCs.
Collapse
|
17
|
Abstract
Stress activation of the hypothalamic-pituitary-adrenal (HPA) axis culminates in increased circulating corticosteroid concentrations. Stress-induced corticosteroids exert diverse actions in multiple target tissues over a broad range of timescales, ranging from rapid actions, which are induced within seconds to minutes and gene transcription independent, to slow actions, which are delayed, long lasting, and transcription dependent. Rapid corticosteroid actions in the brain include, among others, a fast negative feedback mechanism responsible for shutting down the activated HPA axis centrally. We provide a brief review of the cellular mechanisms responsible for rapid corticosteroid actions in different brain structures of the rat, including the hypothalamus, hippocampus, amygdala, and in the anterior pituitary. We propose a model for the direct feedback inhibition of the HPA axis by glucocorticoids in the hypothalamus. According to this model, glucocorticoids activate membrane glucocorticoid receptors to induce endocannabinoid synthesis in the hypothalamic paraventricular nucleus (PVN) and retrograde cannabinoid type I receptor-mediated suppression of the excitatory synaptic drive to PVN neuroendocrine cells. Rapid corticosteroid actions in the hippocampus, amygdala, and pituitary are mediated by diverse cellular mechanisms and may also contribute to the rapid negative feedback regulation of the HPA neuroendocrine axis as well as to the stress regulation of emotional and spatial memory formation.
Collapse
Affiliation(s)
- Jeffrey G Tasker
- Department of Cell and Molecular Biology and Neuroscience Program, Tulane University, New Orleans, LA 70118, USA.
| | | |
Collapse
|
18
|
Tillis CC, Huang HW, Bi W, Pan S, Bruce SR, Alcorn JL. Glucocorticoid regulation of human pulmonary surfactant protein-B (SP-B) mRNA stability is independent of activated glucocorticoid receptor. Am J Physiol Lung Cell Mol Physiol 2011; 300:L940-50. [PMID: 21398497 DOI: 10.1152/ajplung.00420.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Adequate expression of surfactant protein-B (SP-B) is critical in the function of pulmonary surfactant to reduce alveolar surface tension. Expression of SP-B mRNA is restricted to specific lung-airway epithelial cells, and human SP-B mRNA stability is increased in the presence of the synthetic glucocorticoid dexamethasone (DEX). Although the mechanism of SP-B mRNA stabilization by DEX is unknown, studies suggest involvement of the glucocorticoid receptor (GR). We developed a dual-cistronic plasmid-based expression assay in which steady-state levels of SP-B mRNA, determined by Northern analysis, reproducibly reflect changes in SP-B mRNA stability. Using this assay, we found that steady-state levels of SP-B mRNA increased greater than twofold in transfected human-airway epithelial cells (A549) incubated with DEX (10(-7) M). DEX-mediated changes in SP-B mRNA levels required the presence of the SP-B mRNA 3'-untranslated region but did not require ongoing protein synthesis. The effect of DEX on SP-B mRNA levels was dose dependent, with maximal effect at 10(-7) M. DEX increased levels of SP-B mRNA in cells lacking GR, and the presence of the GR antagonist RU486 did not interfere with the effect of DEX. Surprisingly, other steroid hormones (progesterone, estradiol, and vitamin D; 10(-7) M) significantly increased SP-B mRNA levels, suggesting a common pathway of steroid hormone action on SP-B mRNA stability. These results indicate that the effect of DEX to increase SP-B mRNA stability is independent of activated GR and suggests that the mechanism is mediated by posttranscriptional or nongenomic effects of glucocorticoids.
Collapse
Affiliation(s)
- Ceá C Tillis
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Health Sciences Center at Houston, USA
| | | | | | | | | | | |
Collapse
|
19
|
Fragel-Madeira L, Meletti T, Mariante RM, Monteiro RQ, Einicker-Lamas M, Bernardo RR, Lopes AH, Linden R. Platelet activating factor blocks interkinetic nuclear migration in retinal progenitors through an arrest of the cell cycle at the S/G2 transition. PLoS One 2011; 6:e16058. [PMID: 21298035 PMCID: PMC3029264 DOI: 10.1371/journal.pone.0016058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 12/06/2010] [Indexed: 02/06/2023] Open
Abstract
Nuclear migration is regulated by the LIS1 protein, which is the regulatory subunit of platelet activating factor (PAF) acetyl-hydrolase, an enzyme complex that inactivates the lipid mediator PAF. Among other functions, PAF modulates cell proliferation, but its effects upon mechanisms of the cell cycle are unknown. Here we show that PAF inhibited interkinetic nuclear migration (IKNM) in retinal proliferating progenitors. The lipid did not, however, affect the velocity of nuclear migration in cells that escaped IKNM blockade. The effect depended on the PAF receptor, Erk and p38 pathways and Chk1. PAF induced no cell death, nor a reduction in nucleotide incorporation, which rules out an intra-S checkpoint. Notwithstanding, the expected increase in cyclin B1 content during G2-phase was prevented in the proliferating cells. We conclude that PAF blocks interkinetic nuclear migration in retinal progenitor cells through an unusual arrest of the cell cycle at the transition from S to G2 phases. These data suggest the operation, in the developing retina, of a checkpoint that monitors the transition from S to G2 phases of the cell cycle.
Collapse
Affiliation(s)
| | - Tamara Meletti
- Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael M. Mariante
- Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Q. Monteiro
- Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Robson R. Bernardo
- Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Angela H. Lopes
- Institute of Microbiology, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Linden
- Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
20
|
Riedemann T, Patchev AV, Cho K, Almeida OFX. Corticosteroids: way upstream. Mol Brain 2010; 3:2. [PMID: 20180948 PMCID: PMC2841592 DOI: 10.1186/1756-6606-3-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Accepted: 01/11/2010] [Indexed: 01/20/2023] Open
Abstract
Studies into the mechanisms of corticosteroid action continue to be a rich bed of research, spanning the fields of neuroscience and endocrinology through to immunology and metabolism. However, the vast literature generated, in particular with respect to corticosteroid actions in the brain, tends to be contentious, with some aspects suffering from loose definitions, poorly-defined models, and appropriate dissection kits. Here, rather than presenting a comprehensive review of the subject, we aim to present a critique of key concepts that have emerged over the years so as to stimulate new thoughts in the field by identifying apparent shortcomings. This article will draw on experience and knowledge derived from studies of the neural actions of other steroid hormones, in particular estrogens, not only because there are many parallels but also because 'learning from differences' can be a fruitful approach. The core purpose of this review is to consider the mechanisms through which corticosteroids might act rapidly to alter neural signaling.
Collapse
Affiliation(s)
- Therese Riedemann
- Max-Planck-Institute of Psychiatry, Kraepelin Str. 2-10, 80804 Munich, Germany
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK
| | - Alexandre V Patchev
- Max-Planck-Institute of Psychiatry, Kraepelin Str. 2-10, 80804 Munich, Germany
| | - Kwangwook Cho
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK
| | - Osborne FX Almeida
- Max-Planck-Institute of Psychiatry, Kraepelin Str. 2-10, 80804 Munich, Germany
| |
Collapse
|
21
|
González R, Ruiz-León Y, Gomendio M, Roldan ERS. The effect of glucocorticoids on ERK-1/2 phosphorylation during maturation of lamb oocytes and their subsequent fertilization and cleavage ability in vitro. Reprod Toxicol 2009; 29:198-205. [PMID: 19892010 DOI: 10.1016/j.reprotox.2009.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2008] [Revised: 10/15/2009] [Accepted: 10/22/2009] [Indexed: 10/20/2022]
Abstract
High levels of glucocorticoids may alter reproduction, but little is known about their direct actions on oocyte maturation, fertilization and subsequent development. Earlier work suggested negative effects of cortisol or dexamethasone on oocyte maturation but differences were noted between animal models. Both glucocorticoids reduce the p34(cdc2)-cyclin B1 complex but it is unknown if other signaling pathways important for meiosis progression are affected. In this study, using sheep oocytes as a model system, we assessed in vitro the effects of increasing concentration of glucocorticoids (0-250 microM) on oocyte maturation and underlying changes in the MAP kinase pathway, and the ability of oocytes to undergo fertilization and embryo development. Cortisol decreased oocyte maturation but only at the highest concentration, whereas dexamethasone had no effect. Fertilization and cleavage were not affected. On the other hand, both cortisol and dexamethasone inhibited ERK-1/2 activation in a concentration-dependent manner. It thus seems that oocytes can overcome deleterious effects of glucocorticoids during maturation despite the decrease in ERK-1/2 activity, but repercussions in vivo should be further explored.
Collapse
Affiliation(s)
- Raquel González
- Reproductive Ecology and Biology Group, Museo Nacional de Ciencias Naturales (CSIC), 28006 Madrid, Spain
| | | | | | | |
Collapse
|
22
|
Prager EM, Johnson LR. Stress at the synapse: signal transduction mechanisms of adrenal steroids at neuronal membranes. Sci Signal 2009; 2:re5. [PMID: 19724063 DOI: 10.1126/scisignal.286re5] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
As the key neuron-to-neuron interface, the synapse is involved in learning and memory, including traumatic memories during times of stress. However, the signal transduction mechanisms by which stress mediates its lasting effects on synapse transmission and on memory are not fully understood. A key component of the stress response is the increased secretion of adrenal steroids. Adrenal steroids (e.g., cortisol) bind to genomic mineralocorticoid and glucocorticoid receptors (gMRs and gGRs) in the cytosol. In addition, they may act through membrane receptors (mMRs and mGRs), and signal transduction through these receptors may allow for rapid modulation of synaptic transmission as well as modulation of membrane ion currents. mMRs increase synaptic and neuronal excitability; mechanisms include the facilitation of glutamate release through extracellular signal-regulated kinase signal transduction. In contrast, mGRs decrease synaptic and neuronal excitability by reducing calcium currents through N-methyl-D-aspartate receptors and voltage-gated calcium channels by way of protein kinase A- and G protein-dependent mechanisms. This body of functional data complements anatomical evidence localizing GRs to the postsynaptic membrane. Finally, accumulating data also suggest the possibility that mMRs and mGRs may show an inverted U-shaped dose response, whereby glutamatergic synaptic transmission is increased by low doses of corticosterone acting at mMRs and decreased by higher doses acting at mGRs. Thus, synaptic transmission is regulated by mMRs and mGRs, and part of the stress signaling response is a direct and bidirectional modulation of the synapse itself by adrenal steroids.
Collapse
Affiliation(s)
- Eric M Prager
- Center for the Study of Traumatic Stress, Department of Psychiatry and Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA
| | | |
Collapse
|
23
|
Reeves EKM, Gordish-Dressman H, Hoffman EP, Hathout Y. Proteomic profiling of glucocorticoid-exposed myogenic cells: Time series assessment of protein translocation and transcription of inactive mRNAs. Proteome Sci 2009; 7:26. [PMID: 19642986 PMCID: PMC2725035 DOI: 10.1186/1477-5956-7-26] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 07/30/2009] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Prednisone, one of the most highly prescribed drugs, has well characterized effects on gene transcription mediated by the glucocorticoid receptor. These effects are typically occurring on the scale of hours. Prednisone also has a number of non-transcriptional effects (occurring on minutes scale) on protein signaling, yet these are less well studied. We sought to expand the understanding of acute effects of prednisone action on cell signaling using a combination of SILAC strategy and subcellular fractionations from C2C12 myotubes. RESULTS De novo translation of proteins was inhibited in both SILAC labeled and unlabeled C2C12 myotubes. Unlabeled cells were exposed to prednisone while SILAC labeled cells remained untreated. After 0, 5, 15, and 30 minutes of prednisone exposure, labeled and unlabeled cells were mixed at 1:1 ratios and fractionated into cytosolic and nuclear fractions. A total of 534 proteins in the cytosol and 626 proteins in the nucleus were identified and quantitated, using 3 or more peptides per protein with peptide based probability < or = 0.001. We identified significant increases (1.7- to 3.1- fold) in cytoplasmic abundance of 11 ribosomal proteins within 5 minutes of exposure, all of which returned to baseline by 30 min. We hypothesized that these drug-induced acute changes in the subcellular localization of the cell's protein translational machinery could lead to altered translation of quiescent RNAs. To test this, de novo protein synthesis was assayed after 15 minutes of drug exposure. Quantitative fluorography identified 16 2D gel spots showing rapid changes in translation; five of these were identified by MS/MS (pyruvate kinase, annexin A6 isoform A and isoform B, nasopharyngeal epithelium specific protein 1, and isoform 2 of Replication factor C subunit 1), and all showed the 5' terminal oligopyrimidine motifs associated with mRNA sequestration to and from inactive mRNA pools. CONCLUSION We describe novel approaches of subcellular proteomic profiling and assessment of acute changes on a minute-based time scale. These data expand the current knowledge of acute, non-transcriptional activities of glucocorticoids, including changes in protein subcellular localization, altered translation of quiescent RNA pools, and PKC-mediated cytoskeleton remodeling.
Collapse
Affiliation(s)
- Erica K M Reeves
- Research Center for Genetic Medicine, Children's National Medical Center, NW, Washington, DC 20010, USA.
| | | | | | | |
Collapse
|
24
|
Menconi M, Gonnella P, Petkova V, Lecker S, Hasselgren PO. Dexamethasone and corticosterone induce similar, but not identical, muscle wasting responses in cultured L6 and C2C12 myotubes. J Cell Biochem 2009; 105:353-64. [PMID: 18615595 DOI: 10.1002/jcb.21833] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Dexamethasone-treated L6 (a rat cell line) and C2C12 (a mouse cell line) myotubes are frequently used as in vitro models of muscle wasting. We compared the effects of different concentrations of dexamethasone and corticosterone (the naturally occurring glucocorticoid in rodents) on protein breakdown rates, myotube size, and atrogin-1 and MuRF1 mRNA levels in the two cell lines. In addition, the expression of the glucocorticoid receptor (GR) and its role in glucocorticoid-induced metabolic changes were determined. Treatment with dexamethasone or corticosterone resulted in dose-dependent increases in protein degradation rates in both L6 and C2C12 myotubes accompanied by 25-30% reduction of myotube diameter. The same treatments increased atrogin-1 mRNA levels in L6 and C2C12 myotubes but, surprisingly, upregulated the expression of MuRF1 in L6 myotubes only. Both cell types expressed the GR and treatment with dexamethasone or corticosterone downregulated total cellular GR levels while increasing nuclear translocation of the GR in both L6 and C2C12 myotubes. The GR antagonist RU38486 inhibited the dexamethasone- and corticosterone-induced increases in atrogin-1 and MuRF1 expression in L6 myotubes but not in C2C12 myotubes. Interestingly, RU38486 exerted agonist effects in the C2C12, but not in the L6 myotubes. The present results suggest that muscle wasting-related responses to dexamethasone and corticosterone are similar, but not identical, in L6 and C2C12 myotubes. Most notably, the regulation by glucocorticoids of MuRF1 and the role of the GR may be different in the two cell lines. These differences need to be taken into account when cultured myotubes are used in future studies to further explore mechanisms of muscle wasting.
Collapse
Affiliation(s)
- Michael Menconi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | |
Collapse
|
25
|
Alexaki VI, Dermitzaki E, Charalampopoulos I, Kampa M, Nifli AP, Gravanis A, Margioris AN, Castanas E. Neuronal differentiation of PC12 cells abolishes the expression of membrane androgen receptors. Exp Cell Res 2006; 312:2745-56. [PMID: 16822503 DOI: 10.1016/j.yexcr.2006.04.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 04/01/2006] [Accepted: 04/06/2006] [Indexed: 11/29/2022]
Abstract
Sex steroids affect adrenal chromaffin cell function. In the present work, we have examined the expression and functional significance of membrane androgen receptor sites in normal rat adrenal chromaffin cells and in the PC12 rat pheochromocytoma cell line which can differentiate to either a neuronal or to an epithelial phenotype and expresses membrane estrogen receptor sites. Our data are as follows: (a) no cytosolic androgen receptors were found in both normal chromaffin and PC12 cells; (b) both types of chromaffin cells expressed high affinity membrane testosterone binding sites; (c) activation of these sites increased cytosolic Ca(2+), decreased catecholamine secretion and induced apoptosis; (d) NGF-induced neuronal differentiation of PC12 cells resulted in the suppression of the number of membrane testosterone sites. In conclusion, our data provide evidence for the existence of specific membrane testosterone receptors on adrenal chromaffin cells via which androgens, (some of them originating in the cortex) modulate their function. Neuronal differentiation of chromaffin cells results in a significant attenuation of these effects, via suppression of the expression of membrane androgen receptors suggesting, that the latter are specific for epithelioid chromaffin cells.
Collapse
Affiliation(s)
- Vassilia-Ismini Alexaki
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion 71003, Greece
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mulholland PJ, Self RL, Hensley AK, Little HJ, Littleton JM, Prendergast MA. A 24 h corticosterone exposure exacerbates excitotoxic insult in rat hippocampal slice cultures independently of glucocorticoid receptor activation or protein synthesis. Brain Res 2006; 1082:165-72. [PMID: 16510135 DOI: 10.1016/j.brainres.2006.01.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Revised: 01/18/2006] [Accepted: 01/18/2006] [Indexed: 11/26/2022]
Abstract
Elevations in circulating concentrations of glucocorticoids (GC) may increase the expression and/or sensitivity of ionotropic transmitter receptors in brain. For example, recent evidence suggests that acute and chronic GC exposure may alter the number and/or function of N-methyl-D-aspartate (NMDA)-type glutamate receptors, effects that may sensitize the brain to excitotoxic insults. The present studies examined the ability of short-term (24 h) corticosterone (CORT) exposure to potentiate NMDA-induced cytotoxicity in rat hippocampal slice cultures. Additional studies evaluated the role of mineralocorticoid (MR) and glucocorticoid receptor (GR) function, as well as de novo protein synthesis, in potentiation of toxicity by corticosterone exposure. Hippocampal slice cultures were exposed to NMDA (20 microM) for 24 h with cytotoxicity assessed by fluorescent detection of propidium iodide uptake. Exposure to NMDA caused significant propidium iodide uptake in each hippocampal region, while 24 h CORT (0.001-1 microM) exposure alone did not significantly increase propidium iodide uptake. Co-exposure of cultures to CORT and NMDA synergistically increased propidium iodide uptake in each hippocampal region, effects that were prevented by co-exposure to a non-toxic concentration of MK-801 (20 microM). In contrast, 24 h exposure with the MR antagonist spironolactone (1-10 microM), the GR antagonist RU-486 (1-10 microM), or the protein synthesis inhibitor cycloheximide (1 microM) failed to reduce the significant increase in propidium iodide uptake. These data suggest that relatively brief elevations in CORT levels may sensitize the hippocampus to injury independently of GC receptor activity and protein synthesis.
Collapse
Affiliation(s)
- Patrick J Mulholland
- Department of Psychology, 012-I Kastle Hall, University of Kentucky, Lexington, 40506-0044, USA
| | | | | | | | | | | |
Collapse
|
27
|
Khan MM, Hadman M, De Sevilla LM, Mahesh VB, Buccafusco J, Hill WD, Brann DW. Cloning, distribution, and colocalization of MNAR/PELP1 with glucocorticoid receptors in primate and nonprimate brain. Neuroendocrinology 2006; 84:317-29. [PMID: 17142998 DOI: 10.1159/000097746] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Accepted: 10/12/2006] [Indexed: 11/19/2022]
Abstract
MNAR/PELP1 (see text) is a newly identified scaffold protein/coactivator initially thought to modulate nongenomic and genomic actions of the estrogen receptor; however, it has been recently shown to interact with multiple steroid receptors, including androgen and glucocorticoid receptors. In the present study, we cloned the monkey MNAR/PELP1 gene, deduced its domain structure, examined its localization pattern and colocalization with glucocorticoid receptor in monkey brain, and determined its subcellular localization. PCR-based cloning of MNAR/PELP1 from monkey brain produced a transcript of approximately 3.4 kb which showed high homology to the human and rat MNAR/PELP1 gene. Domain analysis showed that all the key steroid-receptor-interacting (LXXLL) domains, SH3-interacting (PXXP) domains and several C-terminal glutamic-acid-rich clusters, as well as various kinase domains are conserved in the monkey MNAR/PELP1 gene. Anatomical mapping of MNAR/PELP1 immunoreactivity in several regions of the monkey brain showed a similar pattern of MNAR/PELP1 distribution as previously observed in rat and mouse brains. MNAR/PELP1 also showed an absolute colocalization with glucocorticoid receptors in both primate and nonprimate brain, including those regions of the brain, where other steroid receptors are not significantly expressed, such as hippocampus, striatum, and thalamus - suggesting that MNAR/PELP1 may modulate glucocorticoid actions in the brain. Finally, ultrastructural electron microscopic studies showed that MNAR/PELP1-reactive gold particles are located within nucleus, cytoplasm, dendritic/synaptic terminals, and astrocytic processes. As a whole, the studies demonstrate that MNAR/PELP1 is expressed and colocalizes with glucocorticoid receptors in monkey and rat brains and may have multiple cellular functions based on its subcellular localizations.
Collapse
Affiliation(s)
- Mohammad M Khan
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta GA 30912, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Yang H, Menconi MJ, Wei W, Petkova V, Hasselgren PO. Dexamethasone upregulates the expression of the nuclear cofactor p300 and its interaction with C/EBPbeta in cultured myotubes. J Cell Biochem 2005; 94:1058-67. [PMID: 15669015 DOI: 10.1002/jcb.20371] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Muscle wasting during sepsis and other catabolic conditions is, at least in part, mediated by glucocorticoids and is associated with upregulated transcription of multiple genes in the ubiquitin-proteasome proteolytic pathway. In addition to transcription factors, nuclear cofactors, including p300, regulate gene transcription. We tested the hypothesis that glucocorticoids upregulate the expression of p300 in muscle cells. Treatment of cultured L6 myotubes, a rat skeletal muscle cell line, with dexamethasone resulted in a dose- and time-dependent increase in p300 protein and mRNA levels. Surprisingly, the effect of dexamethasone on p300 levels was not inhibited by the glucocorticoid receptor (GR) antagonist RU38486 and RU38486 exerted an agonist effect on p300, increasing its expression. Co-immunoprecipitation showed that treatment of the myotubes with dexamethasone resulted in protein-protein interaction between p300 and C/EBPbeta, but not C/EBPdelta. The present results suggest that glucocorticoids upregulate the expression of p300 and its interaction with C/EBPbeta in skeletal muscle. Increased expression and activity of p300 may be involved in the regulation of gene transcription in glucocorticoid-dependent muscle wasting.
Collapse
Affiliation(s)
- Hongmei Yang
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
29
|
Xiao L, Qi A, Chen Y. Cultured embryonic hippocampal neurons deficient in glucocorticoid (GC) receptor: a novel model for studying nongenomic effects of GC in the neural system. Endocrinology 2005; 146:4036-41. [PMID: 15961565 DOI: 10.1210/en.2004-1652] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glucocorticoid (GC) acts through both genomic and nongenomic mechanisms. It affects the structure and function of the central nervous system, especially the hippocampus. Here we report an in vitro culture system that can yield embryonic hippocampal neurons deficient in the expression of GC receptor as demonstrated by immunoblotting, immunocytochemistry, and RT-PCR. Owing to this unique feature, those neuron preparations can serve as an ideal model for studying the nongenomic actions of GC on neural cells. In this study, we found that the Erk1/2, c-Jun N-terminal kinase (JNK), and p38 MAPKs were activated in these neurons by BSA-conjugated corticosterone within 15 min of treatment. This activation was not blocked by RU38486, spironolactone, or cycloheximide. Therefore, it is concluded that the activation of MAPKs observed here was due to the nongenomic action of GC. Furthermore, a 24-h incubation with corticosterone at concentrations ranged from 10(-11)-10(-5) M did not have an effect on the viability of GC receptor-deficient neurons.
Collapse
Affiliation(s)
- Lin Xiao
- Institute of Neuroscience, Department of Neurobiology, Second Military Medical University, 800 XiangYin Road, Shanghai 200433, People's Republic of China
| | | | | |
Collapse
|
30
|
Yang H, Mammen J, Wei W, Menconi M, Evenson A, Fareed M, Petkova V, Hasselgren PO. Expression and activity of C/EBPbeta and delta are upregulated by dexamethasone in skeletal muscle. J Cell Physiol 2005; 204:219-26. [PMID: 15669083 DOI: 10.1002/jcp.20278] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The influence of glucocorticoids on the expression and activity of the transcription factors CCAAT/enhancer binding protein (C/EBP)beta and delta in skeletal muscle was examined by treating rats or cultured L6 myotubes with dexamethasone. Treatment of rats with 10 mg/kg of dexamethasone resulted in increased C/EBPbeta and delta DNA binding activity in the extensor digitorum longus muscle as determined by electrophoretic mobility shift assay (EMSA) and supershift analysis. A similar response was noticed in dexamethasone-treated myotubes. In other experiments, myocytes were transfected with a plasmid containing a promoter construct consisting of multiple C/EBP binding elements upstream of a luciferase reporter gene. Treatment of these cells with dexamethasone resulted in a fourfold increase in luciferase activity, suggesting that glucocorticoids increase C/EBP-dependent gene activation in muscle cells. In addition, dexamethasone upregulated the protein and gene expression of C/EBPbeta and delta in the myotubes in a time- and dose-dependent fashion as determined by Western blotting and real-time PCR, respectively. The results suggest that glucocorticoids increase C/EBPbeta and delta activity and expression through a direct effect in skeletal muscle.
Collapse
Affiliation(s)
- Hongmei Yang
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Qi AQ, Qiu J, Xiao L, Chen YZ. Rapid activation of JNK and p38 by glucocorticoids in primary cultured hippocampal cells. J Neurosci Res 2005; 80:510-7. [PMID: 15846779 DOI: 10.1002/jnr.20491] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Rapid activation of JNK and p38 and their translocation to the cell nucleus by glucocorticoids, corticosterone (Cort), and bovine serum-conjugated corticosterone (Cort-BSA) were studied in primary cultured hippocampal cells by using immunoblotting and immunofluorescence confocal microscopy. The rapid activation occurred 5 min after stimulation and was maintained at plateau for as long as 2-4 hr; i.e., the response persisted for 2 hr after washing out the 15-min application of Cort-BSA. The activation occurred at a minimal concentration of 10(-9) M for Cort and 10(-8) M for Cort-BSA. GDPbetaS blocked the activation, but RU38486, a nuclear glucocorticoid receptor antagonist, could not block the activation, indicating the involvement of the membrane-delineated receptor in this reaction. The protein kinase C (PKC) inhibitor Go6976 blocked the response, whereas the protein kinase A inhibitor H89 could not, implying the involvement of PKC in the intracellular signal transduction pathway. The nongenomic nature of the responses and the transduction pathway and the significance of persistent action and biological significance are discussed.
Collapse
Affiliation(s)
- Ai-Qun Qi
- Department of Physiology, Second Military Medical University, Shanghai, China
| | | | | | | |
Collapse
|
32
|
Yang CH, Huang CC, Hsu KS. Behavioral stress modifies hippocampal synaptic plasticity through corticosterone-induced sustained extracellular signal-regulated kinase/mitogen-activated protein kinase activation. J Neurosci 2005; 24:11029-34. [PMID: 15590919 PMCID: PMC6730281 DOI: 10.1523/jneurosci.3968-04.2004] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The induction of hippocampal long-term synaptic plasticity is exquisitely sensitive to behavioral stress, but the underlying mechanisms are still unclear. We report here that hippocampal slices prepared from adult rats that had experienced unpredictable and inescapable restraint tail-shock stress showed marked impairments of long-term potentiation (LTP) in the CA1 region. The same stress promoted the induction of long-term depression (LTD). These effects were prevented when the animals were given the glucocorticoid receptor antagonist 11beta, 17beta-11[4-(dimethylamino)phenyl]-17-hydroxy-17-(1-propynyl)-estra-4-9-dien-3-one before the stress. Immunoblotting analyses revealed that stress induced a profound and prolonged extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK1/2 MAPK) hyperphosphorylation through small GTPase Ras, Raf-1, and MAPK kinase 1/2 (MEK1/2). Furthermore, the stress effects were obviated by the intrahippocampal injection of specific inhibitors of MEK1/2 (U0126), protein kinase C (bisindolylmaleimide I), tyrosine kinase (K252a), and BDNF antisense oligonucleotides. These results suggest that the effects of stress on LTP and LTD originate from the corticosterone-induced sustained activation of ERK1/2-coupled signaling cascades.
Collapse
Affiliation(s)
- Chih-Hao Yang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | | | | |
Collapse
|
33
|
Revest JM, Di Blasi F, Kitchener P, Rougé-Pont F, Desmedt A, Turiault M, Tronche F, Piazza PV. The MAPK pathway and Egr-1 mediate stress-related behavioral effects of glucocorticoids. Nat Neurosci 2005; 8:664-72. [PMID: 15834420 DOI: 10.1038/nn1441] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Accepted: 03/28/2005] [Indexed: 11/08/2022]
Abstract
Many of the behavioral consequences of stress are mediated by the activation of the glucocorticoid receptor by stress-induced high levels of glucocorticoid hormones. To explore the molecular mechanisms of these effects, we combined in vivo and in vitro approaches. We analyzed mice carrying a brain-specific mutation (GR(NesCre)) in the glucocorticoid receptor gene (GR, also called Nr3c1) and cell lines that either express endogenous glucocorticoid receptor or carry a constitutively active form of the receptor (DeltaGR) that can be transiently induced. In the hippocampus of the wild-type [corrected] mice after stress, as well as in the cell lines, activation of glucocorticoid receptors greatly increased the expression and enzymatic activity of proteins in the MAPK signaling pathway and led to an increase in the levels of both Egr-1 mRNA and protein. In parallel, inhibition of the MAPK pathway within the hippocampus abolished the increase in contextual fear conditioning induced by glucocorticoids. The present results provide a molecular mechanism for the stress-related effects of glucocorticoids on fear memories.
Collapse
Affiliation(s)
- Jean-Michel Revest
- INSERM U588, Laboratoire de Physiopathologie du Comportement, Bordeaux Institute for Neurosciences, University Victor Segalen-Bordeaux 2, Domaine de Carreire, 146 Rue Léo Saignat, 33077 Bordeaux Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Szewczyk NJ, Jacobson LA. Signal-transduction networks and the regulation of muscle protein degradation. Int J Biochem Cell Biol 2005; 37:1997-2011. [PMID: 16125109 DOI: 10.1016/j.biocel.2005.02.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Revised: 01/18/2005] [Accepted: 02/22/2005] [Indexed: 02/05/2023]
Abstract
Protein degradation in muscle functions in maintaining normal physiological homeostasis and adapting to new homeostatic states, and is required for muscle wasting or atrophy in various pathological states. The interplay between protein synthesis and degradation to maintain homeostasis is complex and responds to a variety of autocrine and intercellular signals from neuronal inputs, hormones, cytokines, growth factors and other regulatory molecules. The intracellular events that connect extracellular signals to the molecular control of protein degradation are incompletely understood, but likely involve interacting signal-transduction networks rather than isolated pathways. We review some examples of signal-transduction systems that regulate protein degradation, including effectors of proteolysis inducing factor (PIF), insulin and insulin-like growth factor (IGF) and their receptors, and fibroblast growth factor (FGF) and its receptors.
Collapse
Affiliation(s)
- Nathaniel J Szewczyk
- Department of Biological Sciences, University of Pittsburgh, 304 Langley Hall, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
35
|
López-Toledano MA, Redondo C, Lobo MVT, Reimers D, Herranz AS, Paíno CL, Bazán E. Tyrosine hydroxylase induction by basic fibroblast growth factor and cyclic AMP analogs in striatal neural stem cells: role of ERK1/ERK2 mitogen-activated protein kinase and protein kinase C. J Histochem Cytochem 2004; 52:1177-89. [PMID: 15314085 DOI: 10.1369/jhc.3a6244.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neural stem cells (NSC) with self-renewal and multilineage potential are considered good candidates for cell replacement of damaged nervous tissue. In vitro experimental conditions can differentiate these cells into specific neuronal phenotypes. In the present study, we describe the combined effect of basic fibroblast growth factor (bFGF) and dibutyryladenosine 3',5'-cyclic monophosphate (dbcAMP) on the differentiation of fetal rat striatal NSC into tyrosine hydroxylase-positive cells. Tyrosine hydroxylase induction was accompanied by the activation of ERK1/ERK2 mitogen-activated protein kinase and was inhibited by the ERK1/ERK2 pathway blocker PD98059, suggesting that ERK activation may be important for this process. In addition, protein kinase C (PKC) was shown to be required for tyrosine hydroxylase protein expression. The inhibition of PKC by staurosporin, as well as its downregulation, decreased the ability of bFGF+dbcAMP to generate tyrosine hydroxylase-positive cells. Moreover, the PKC activator phorbol 12-myristate 13-acetate (PMA) together with bFGF and dbcAMP led to a significant increase in phospho-ERK1/ERK2 levels, and the percentage of beta-tubulin III-positive cells that expressed tyrosine hydroxylase increased by 3.5-fold. PMA also promoted the phosphorylation of the cyclic AMP response element binding protein that might contribute to the increase in tyrosine hydroxylase-positive cells observed in bFGF+dbcAMP+PMA-treated cultures. From these results, we conclude that the manipulation in vitro of NSC from rat fetal striatum with bFGF, cyclic AMP analogs, and PKC activators promotes the generation of tyrosine hydroxylase-positive neurons.
Collapse
Affiliation(s)
- Miguel A López-Toledano
- Servicio de Neurobiologia-Investigación, Hospital Ramón y Cajal, Carretera de Colmenar Viejo, Km 9.1, 28034 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
36
|
Baccan GC, Oliveira RDR, Mantovani B. Stress and immunological phagocytosis: possible nongenomic action of corticosterone. Life Sci 2004; 75:1357-68. [PMID: 15234193 DOI: 10.1016/j.lfs.2004.02.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2003] [Accepted: 02/26/2004] [Indexed: 10/26/2022]
Abstract
Some immunological responses triggered by stress can be mediated by corticosterone activity through cytosolic receptors regulating gene expression. There are, however some reports on the possibility of a nongenomic effect of this hormone to explain phenomena observed in a few minutes. We have found that macrophages from mice subjected to 10 min of cold stress (at -15 degrees C) showed a lower phagocytic capacity mediated by Fcgamma-receptors than cells from control animals. Treating mice with glucocorticoid antagonist RU 486 did not block the decrease in phagocytic capacity. This inhibitory effect on phagocytosis was also observed by experiments in vitro with corticosterone in the concentration found in serum after stress, and could not be prevented by RU 486, actinomicyn D or cycloheximide. These results indicate that corticosterone could affect phagocytosis by macrophages through a nongenomic mechanism, and may have physiological implications.
Collapse
Affiliation(s)
- Gyselle C Baccan
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | | | | |
Collapse
|
37
|
Rossol-Haseroth K, Zhou Q, Braun S, Boldyreff B, Falkenstein E, Wehling M, Lösel RM. Mineralocorticoid receptor antagonists do not block rapid ERK activation by aldosterone. Biochem Biophys Res Commun 2004; 318:281-8. [PMID: 15110785 DOI: 10.1016/j.bbrc.2004.04.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2004] [Indexed: 11/21/2022]
Abstract
Aldosterone can elicit rapid nongenomic effects both in vivo and in vitro, often mediated by signal transduction cascades. However, it is not understood how these rapid effects are initiated. In this study we show that aldosterone leads to rapid activation of mitogen activated protein kinases ERK1/2 in the cortical collecting duct cell line M-1. Inhibitors of transcription and translation could not block this activation, which suggests an extranuclear (nongenomic) mechanism. Although it is known that M-1 cells do not contain a transcriptionally functional MR, it is not known whether a closely related protein still could mediate the effects, or an unrelated nonclassic receptor. To test this hypothesis, the effects of four classical mineralocorticoid receptor antagonists were studied. None of the compounds could block the response to aldosterone. Altogether, the data suggest that rapid aldosterone effects in M-1 cells are initiated by a receptor different from the classical mineralocorticoid receptor.
Collapse
Affiliation(s)
- Karin Rossol-Haseroth
- Department of Clinical Pharmacology, Faculty of Clinical Medicine Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Ishida-Takahashi R, Uotani S, Abe T, Degawa-Yamauchi M, Fukushima T, Fujita N, Sakamaki H, Yamasaki H, Yamaguchi Y, Eguchi K. Rapid Inhibition of Leptin Signaling by Glucocorticoids in Vitro and in Vivo. J Biol Chem 2004; 279:19658-64. [PMID: 14993217 DOI: 10.1074/jbc.m310864200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Elevated secretion of glucocorticoids (GCs) or hypersensitivity to GCs has a permissive effect on the development of obesity and leads to abnormalities of body fat distribution. Recent studies demonstrated GCs act as antagonists of leptin in rodents. However, little is known about the interaction between GCs and leptin signaling. In the present study, we investigated the effects of GCs on leptin action in vitro and in vivo. GCs rapidly inhibited the leptin-induced STAT3 phosphorylation in a dose- and time-dependent manner, as assayed by Western blotting using anti-phosphospecific-STAT3 in human hepatoma cell lines (Huh7) transiently expressing long form leptin receptor. GCs also inhibited the leptin-induced JAK2 tyrosine phosphorylation but unaltered the specific binding of (125)I-leptin to the cells. Parallel experiments, however, demonstrated that the inhibitory effects of GCs were not observed in either IL-6- or LIF-induced STAT3 phosphorylation. Furthermore, we examined the feeding behavior and hypothalamic leptin signaling following intracerebroventricular (icv) infusion of GCs prior to icv leptin infusion in Sprague-Dawley rats. The food intake after 24 h of icv leptin injection increased 3-fold in GCs-treated animals. In addition, central infusion of GCs resulted in a marked reduction of hypothalamic STAT3 phosphorylation in response to icv infusion of leptin. To clarify the molecular mechanism by which GCs rapidly reduce leptin-induced JAK/STAT signaling, we examined the intracellular signal transduction pathway potentially mediated by GCs. PD98059, a specific MEK inhibitor, blocked the inhibitory effects of GCs on leptin-induced JAK/STAT activation in Huh7 cells. These results suggest GCs antagonize leptin action by a rapid inhibition of the leptin-induced JAK/STAT pathway partly via MAPK cascade.
Collapse
Affiliation(s)
- Ryoko Ishida-Takahashi
- Division of Immunology, Endocrinology, and Metabolism, Department of Medical and Dental Sciences, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Da Silva CA, Kassel O, Lebouquin R, Lacroix EJ, Frossard N. Paradoxical early glucocorticoid induction of stem cell factor (SCF) expression in inflammatory conditions. Br J Pharmacol 2003; 141:75-84. [PMID: 14662725 PMCID: PMC1574180 DOI: 10.1038/sj.bjp.0705598] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Stem cell factor (SCF) is a major growth factor for mast cells, promoting their differentiation and chemotaxis. Its expression is regulated by glucocorticoids in inflammatory conditions, showing an early increased protein expression, before the expected anti-inflammatory decrease (Da Silva et al., Br. J. Pharmacol. 2002:135,1634). 2. We here evaluated the early kinetic of SCF expression regulated by interleukin (IL)-1beta, budesonide and the combination of both in human lung fibroblasts in culture. 3. Budesonide potentiated the IL-1beta-enhanced expression of SCF mRNA (+103%) and protein (+98%) very shortly after treatment (at 30 min and 1 h, respectively). A gentle downregulation followed. This potentiating effect of budesonide was related to increased SCF mRNA stability and SCF gene transcription. 4. Deletion of a kappaB-like site that we identified in the first intron of the SCF gene, in a luciferase reporter system, abolished the potentiation by budesonide, as well as the effect of IL-1beta alone, as compared to the wild-type construction activity. 5. All budesonide-induced effects were glucocorticoid-receptor dependent, since they were reproduced by dexamethasone and blocked by RU486. 6. IL-1beta+budesonide did not affect the relative expression of the soluble and membrane-bound forms of SCF. 7. In conclusion, our results clearly show that glucocorticoids act very early to adversely increase the expression of SCF mRNA and protein in the inflammatory conditions created by IL-1beta, and that this effect involves increased mRNA stability and increased gene expression through activation of the NF-kappaB-like responsive element.
Collapse
MESH Headings
- Budesonide/antagonists & inhibitors
- Budesonide/pharmacology
- Cells, Cultured
- DNA, Complementary/drug effects
- DNA, Complementary/genetics
- Down-Regulation
- Drug Synergism
- Fibroblasts/drug effects
- Fibroblasts/physiology
- Glucocorticoids/antagonists & inhibitors
- Glucocorticoids/pharmacology
- Glucocorticoids/physiology
- Humans
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/physiopathology
- Interleukin-1/metabolism
- Interleukin-1/pharmacology
- Lung/cytology
- Mifepristone/pharmacology
- NF-kappa B/physiology
- Plasmids/drug effects
- Plasmids/genetics
- RNA Stability/drug effects
- RNA Stability/genetics
- RNA, Messenger/chemical synthesis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Glucocorticoid/antagonists & inhibitors
- Receptors, Glucocorticoid/physiology
- Stem Cell Factor/drug effects
- Stem Cell Factor/genetics
- Stem Cell Factor/metabolism
- Tissue Engineering/methods
- Transcription, Genetic/drug effects
- Transcription, Genetic/genetics
- Transfection/methods
Collapse
|
40
|
Abstract
The circadian rhythms are daily oscillations in various biological processes that are regulated by an endogenous clock. Disruption of these rhythms has been associated with cancer in humans. One of the cellular processes that is regulated by circadian rhythm is cell proliferation, which often shows asynchrony between normal and malignant tissues. This asynchrony highlights the importance of the circadian clock in tumour suppression in vivo and is one of the theoretical foundations for cancer chronotherapy. Investigation of the mechanisms by which the circadian clock controls cell proliferation and other cellular functions might lead to new therapeutic targets.
Collapse
Affiliation(s)
- Loning Fu
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | | |
Collapse
|
41
|
Takahashi T, Kimoto T, Tanabe N, Hattori TA, Yasumatsu N, Kawato S. Corticosterone acutely prolonged N-methyl-d-aspartate receptor-mediated Ca2+ elevation in cultured rat hippocampal neurons. J Neurochem 2002; 83:1441-51. [PMID: 12472898 DOI: 10.1046/j.1471-4159.2002.01251.x] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This work reports the first demonstration that corticosterone (CORT) has a rapid and transient effect on NMDA receptor-mediated Ca2+ signaling in cultured rat hippocampal neurons. Using single cell Ca2+ imaging, CORT and agonists of glucocorticoid receptors were observed to modulate the NMDA receptor-mediated Ca2+ signals in a completely different fashion from pregnenolone sulfate. In the absence of steroids, 100 micro m NMDA induced a transient Ca2+ signal that lasted for 30-70 s in 86.1% of the neurons prepared from postnatal rats (3-5 days old). After pre-treatment with 0.1-100 micro m CORT for 10-20 min, NMDA induced extremely prolonged Ca2+ elevation. This prolonged Ca2+ elevation was terminated by the application of MK-801 and followed by washing out of CORT. The proportion of CORT-modulated neurons within the NMDA-responsive cells increased from 25.1 to 95.5% when the concentration of CORT was raised from 0.1 to 50 micro m. Substitution of BSA-conjugated CORT produced essentially the same results. When hippocampal neurons were preincubated with 10 micro m cortisol and 1 micro m dexamethasone for 20 min, a very prolonged Ca2+ elevation was also observed upon NMDA stimulation. The CORT-prolonged Ca2+ elevation caused a long-lasting depolarization of the mitochondrial membrane, as observed with rhodamine 123. In contrast, incubation with 100 micro m pregnenolone sulfate did not considerably alter the time duration of NMDA-induced transient Ca2+ elevation, but caused a significant increase in the peak amplitude of Ca2+ elevation in hippocampal neurons. These results imply that high levels of CORT induce a rapid and non-genomic prolongation of NMDA receptor-mediated Ca2+ elevation, probably via putative membrane surface receptors for CORT in the hippocampal neurons.
Collapse
Affiliation(s)
- Taiki Takahashi
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo at Komaba, Meguro, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Cato ACB, Nestl A, Mink S. Rapid actions of steroid receptors in cellular signaling pathways. SCIENCE'S STKE : SIGNAL TRANSDUCTION KNOWLEDGE ENVIRONMENT 2002; 2002:re9. [PMID: 12084906 DOI: 10.1126/stke.2002.138.re9] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Steroid hormones regulate cellular processes by binding to intracellular receptors that, in turn, interact with discrete nucleotide sequences to alter gene expression. Because most steroid receptors in target cells are located in the cytoplasm, they need to get into the nucleus to alter gene expression. This process typically takes at least 30 to 60 minutes. In contrast, other regulatory actions of steroid hormones are manifested within seconds to a few minutes. These time periods are far too rapid to be due to changes at the genomic level and are therefore termed nongenomic or rapid actions, to distinguish them from the classical steroid hormone action of regulation of gene expression. The rapid effects of steroid hormones are manifold, ranging from activation of mitogen-activated protein kinases (MAPKs), adenylyl cyclase (AC), protein kinase C (PKC), and heterotrimeric guanosine triphosphate-binding proteins (G proteins). In some cases, these rapid actions of steroids are mediated through the classical steroid receptor that can also function as a ligand-activated transcription factor, whereas in other instances the evidence suggests that these rapid actions do not involve the classical steroid receptors. One candidate target for the nonclassical receptor-mediated effects are G protein-coupled receptors (GPCRs), which activate several signal transduction pathways. One characteristic of responses that are not mediated by the classical steroid receptors is insensitivity to steroid antagonists, which has contributed to the notion that a new class of steroid receptors may be responsible for part of the rapid action of steroids. Evidence suggests that the classical steroid receptors can be localized at the plasma membrane, where they may trigger a chain of reactions previously attributed only to growth factors. Identification of interaction domains on the classical steroid receptors involved in the rapid effects, and separation of this function from the genomic action of these receptors, should pave the way to a better understanding of the rapid action of steroid hormones.
Collapse
Affiliation(s)
- Andrew C B Cato
- Forschungszentrum Karlsruhe, Institute of Toxicology and Genetics, Post Office Box 3640, D-76021 Karlsruhe, Germany.
| | | | | |
Collapse
|
44
|
Chen YZ, Qiu J. Possible genomic consequence of nongenomic action of glucocorticoids in neural cells. NEWS IN PHYSIOLOGICAL SCIENCES : AN INTERNATIONAL JOURNAL OF PHYSIOLOGY PRODUCED JOINTLY BY THE INTERNATIONAL UNION OF PHYSIOLOGICAL SCIENCES AND THE AMERICAN PHYSIOLOGICAL SOCIETY 2001; 16:292-6. [PMID: 11719608 DOI: 10.1152/physiologyonline.2001.16.6.292] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The nongenomic, rapid effects of glucocorticoid activate multiple intracellular transduction pathways. This review proposes a possible genomic consequence of the nongenomic action of steroids. The genomic actions of hormonal steroids may be twofold: classic genomic and nongenomically induced genomic.
Collapse
Affiliation(s)
- Y Z Chen
- Institute of Neuroscience, Department of Neurobiology, Second Military Medical University, Shanghai 200433, China
| | | |
Collapse
|