1
|
Alonge MM, Greville LJS, Ma X, Faure PA, Bentley GE. Acute restraint stress rapidly impacts reproductive neuroendocrinology and downstream gonad function in big brown bats (Eptesicus fuscus). J Exp Biol 2023; 226:jeb245592. [PMID: 37827114 PMCID: PMC10629485 DOI: 10.1242/jeb.245592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/23/2023] [Indexed: 10/14/2023]
Abstract
Animals face unpredictable challenges that require rapid, facultative physiological reactions to support survival but may compromise reproduction. Bats have a long-standing reputation for being highly sensitive to stressors, with sensitivity and resilience varying both within and among species, yet little is known about how stress affects the signaling that regulates reproductive physiology. Here, we provide the first description of the molecular response of the hypothalamic-pituitary-gonadal (HPG) axis of male big brown bats (Eptesicus fuscus) in response to short-term stress using a standardized restraint manipulation. This acute stressor was sufficient to upregulate plasma corticosterone and resulted in a rapid decrease in circulating testosterone. While we did not find differences in the mRNA expression of key steroidogenic enzymes (StAR, aromatase, 5-alpha reductase), seminiferous tubule diameter was reduced in stressed bats coupled with a 5-fold increase in glucocorticoid receptor (GR) mRNA expression in the testes. These changes, in part, may be mediated by RFamide-related peptide (RFRP) because fewer immunoreactive cell bodies were detected in the brains of stressed bats compared with controls - suggesting a possible increase in secretion - and increased RFRP expression locally in the gonads. The rapid sensitivity of the bat testes to stress may be connected to deleterious impacts on tissue health and function as supported by significant transcriptional upregulation of key pro-apoptotic signaling molecules (Bax, cytochrome c). Experiments like this broadly contribute to our understanding of the stronger ecological predictions regarding physiological responses of bats within the context of stress, which may impact decisions surrounding animal handling and conservation approaches.
Collapse
Affiliation(s)
- Mattina M. Alonge
- University of California, Berkeley, Department of Integrative Biology, Berkeley, CA 94720-3200, USA
| | - Lucas J. S. Greville
- McMaster University, Department of Psychology, Neuroscience & Behaviour, Hamilton, ON, Canada, L8S 4L8
- University of Waterloo, Department of Biology, Waterloo, ON, Canada, N2L 3G1
| | - Xuehao Ma
- University of California, Berkeley, Department of Integrative Biology, Berkeley, CA 94720-3200, USA
- Helen Wills Neuroscience Institute, Berkeley, CA 94720, USA
| | - Paul A. Faure
- McMaster University, Department of Psychology, Neuroscience & Behaviour, Hamilton, ON, Canada, L8S 4L8
| | - George E. Bentley
- University of California, Berkeley, Department of Integrative Biology, Berkeley, CA 94720-3200, USA
- Helen Wills Neuroscience Institute, Berkeley, CA 94720, USA
| |
Collapse
|
2
|
Bhaumik S, Lockett J, Cuffe J, Clifton VL. Glucocorticoids and Their Receptor Isoforms: Roles in Female Reproduction, Pregnancy, and Foetal Development. BIOLOGY 2023; 12:1104. [PMID: 37626990 PMCID: PMC10452123 DOI: 10.3390/biology12081104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023]
Abstract
Alterations in the hypothalamic-pituitary-adrenal (HPA) axis and associated changes in circulating levels of glucocorticoids are integral to an organism's response to stressful stimuli. Glucocorticoids acting via glucocorticoid receptors (GRs) play a role in fertility, reproduction, placental function, and foetal development. GRs are ubiquitously expressed throughout the female reproductive system and regulate normal reproductive function. Stress-induced glucocorticoids have been shown to inhibit reproduction and affect female gonadal function by suppressing the hypothalamic-pituitary-gonadal (HPG) axis at each level. Furthermore, during pregnancy, a mother's exposure to prenatal stress or external glucocorticoids can result in long-lasting alterations to the foetal HPA and neuroendocrine function. Several GR isoforms generated via alternative splicing or translation initiation from the GR gene have been identified in the mammalian ovary and uterus. The GR isoforms identified include the splice variants, GRα and GRβ, and GRγ and GR-P. Glucocorticoids can exert both stimulatory and inhibitory effects and both pro- and anti-inflammatory functions in the ovary, in vitro. In the placenta, thirteen GR isoforms have been identified in humans, guinea pigs, sheep, rats, and mice, indicating they are conserved across species and may be important in mediating a differential response to stress. Distinctive responses to glucocorticoids, differential birth outcomes in pregnancy complications, and sex-based variations in the response to stress could all potentially be dependent on a particular GR expression pattern. This comprehensive review provides an overview of the structure and function of the GR in relation to female fertility and reproduction and discusses the changes in the GR and glucocorticoid signalling during pregnancy. To generate this overview, an extensive non-systematic literature search was conducted across multiple databases, including PubMed, Web of Science, and Google Scholar, with a focus on original research articles, meta-analyses, and previous review papers addressing the subject. This review integrates the current understanding of GR variants and their roles in glucocorticoid signalling, reproduction, placental function, and foetal growth.
Collapse
Affiliation(s)
- Sreeparna Bhaumik
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
| | - Jack Lockett
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Brisbane 4102, Australia
| | - James Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane 4067, Australia;
| | - Vicki L. Clifton
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
| |
Collapse
|
3
|
Yang M, Ji Y, Yong T, Liu T, Yang S, Guo S, Meng F, Han X, Liang Q, Cao X, Huang L, Du X, Huang A, Kong F, Zeng X, Bu G. Corticosterone stage-dependently inhibits progesterone production presumably via impeding the cAMP-StAR cascade in granulosa cells of chicken preovulatory follicles. Poult Sci 2022; 102:102379. [PMID: 36608454 PMCID: PMC9829700 DOI: 10.1016/j.psj.2022.102379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Stress can suppress reproduction capacity in either wild or domestic animals, but the exact mechanism behind it, especially in terms of steroidogenesis, remains under-investigated so far. Considering the important roles of progesterone in avian breeding, we investigated the modulation of corticosterone on progesterone production in cultured granulosa cells of chicken follicles at different developmental stages. Using enzyme immunoassays, our study showed that corticosterone could only inhibit progesterone synthesis in granulosa cells from F5-6, F4, and F3 follicles, but not F2 and F1 follicles. Coincidentally, both quantitative real-time PCR and western blotting revealed that corticosterone could downregulate steroidogenic acute regulatory protein (StAR) expression, suggesting the importance of StAR in corticosterone-related actions. Using the dual-luciferase reporter system, we found that corticosterone can potentially enhance, rather than inhibit, the activity of StAR promoter. Of note, combining high-throughput transcriptomic analysis and quantitative real-time PCR, phosphodiesterase 10A (PDE10A), protein kinase cAMP-dependent type II regulatory subunit alpha (PRKAR2A) and cAMP responsive element modulator (CREM) were identified to exhibit the differential expression patterns consistent with cAMP blocking in granulosa cells from F5-6, F4, and F3, but not F2 and F1 follicles. Afterward, the expression profiles of these genes in granulosa cells of distinct developmental-stage follicles were examined by quantitative real-time PCR, in which all of them expressed correspondingly with progesterone levels of granulosa cells during development. Collectively, these findings indicate that corticosterone can stage-dependently inhibit progesterone production in granulosa cells of chicken preovulatory follicles, through impeding cAMP-induced StAR activity presumptively.
Collapse
Affiliation(s)
- Ming Yang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Yu Ji
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Tao Yong
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Tuoyuan Liu
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Shuai Yang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Shasha Guo
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Fengyan Meng
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Xingfa Han
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Qiuxia Liang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Xiaohan Cao
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Linyan Huang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Xiaogang Du
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Anqi Huang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Fanli Kong
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Xianyin Zeng
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China
| | - Guixian Bu
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China,Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an 625014, PR China,Corresponding author:
| |
Collapse
|
4
|
Xiao H, Xu Z, Zhu X, Wang J, Zheng Q, Zhang Q, Xu C, Tao W, Wang D. Cortisol safeguards oogenesis by promoting follicular cell survival. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1563-1577. [PMID: 35167018 DOI: 10.1007/s11427-021-2051-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
The role of glucocorticoids in oogenesis remains to be elucidated. cyp11c1 encodes the key enzyme involved in the synthesis of cortisol, the major glucocorticoid in teleosts. In our previous study, we mutated cyp11c1 in tilapia and analyzed its role in spermatogenesis. In this study, we analyzed its role in oogenesis. cyp11c1+/- XX tilapia showed normal ovarian morphology but poor egg quality, as indicated by the mortality of embryos before 3 d post fertilization, which could be partially rescued by the supplement of exogenous cortisol to the mother fish. Transcriptome analyses revealed reduced expression of maternal genes in the eggs of the cyp11c1+/- XX fish. The cyp11c1-/- females showed impaired vitellogenesis and arrested oogenesis due to significantly decreased serum cortisol. Further analyses revealed decreased serum E2 level and expression of amh, an important regulator of follicular cell development, and increased follicular cell apoptosis in the ovaries of cyp11c1-/- XX fish, which could be rescued by supplement of either exogenous cortisol or E2. Luciferase assays revealed a direct regulation of cortisol and E2 on amh transcription via GRs or ESRs. Taken together, our results demonstrate that cortisol safeguards oogenesis by promoting follicular cell survival probably via Amh signaling.
Collapse
Affiliation(s)
- Hesheng Xiao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Zhen Xu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xi Zhu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Jingrong Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qiaoyuan Zheng
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qingqing Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Chunmei Xu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Wenjing Tao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
5
|
Yang M, Jin C, Cheng X, Liu T, Ji Y, Meng F, Han X, Liang Q, Cao X, Huang L, Du X, Zeng X, Bu G. Corticosterone triggers anti-proliferative and apoptotic effects, and downregulates the ACVR1-SMAD1-ID3 cascade in chicken ovarian prehierarchical, but not preovulatory granulosa cells. Mol Cell Endocrinol 2022; 552:111675. [PMID: 35577112 DOI: 10.1016/j.mce.2022.111675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 10/18/2022]
Abstract
The coordinated proliferation and apoptosis of granulosa cells plays a critical role in follicular development. To identify the exact mechanisms of how stress-driven glucocorticoid production suppresses reproduction, granulosa cells were isolated from chicken follicles at different developmental stages and then treated with corticosterone. Using CCK-8, EDU and TUNEL assays, we showed that corticosterone could trigger both anti-proliferative and pro-apoptotic effects in granulosa cells from 6 to 8 mm follicles only, while depicting no influence on granulosa cells from any preovulatory follicles. High-throughput transcriptomic analysis identified 1362 transcripts showing differential expression profiles in granulosa cells from 6 to 8 mm follicles after corticosterone treatment. Interestingly, Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that 17 genes were enriched in the TGF-β signaling pathway, and 13 showed differential expression patterns consistent with corticosterone-induced effects. The differential expression profiles of these 13 genes were examined by quantitative real-time PCR in cultured chicken ovarian granulosa cells at diverse developmental stages following corticosterone challenge for a short (8 h) or long period (24 h). After 24 h of treatment, INHBB, FST, FMOD, NOG, ACVR1, SMAD1 and ID3 were the genes that responded consistently with corticosterone-induced proliferative and apoptotic events in all granulosa cells detected. However, only ACVR1, SMAD1 and ID3 could initiate coincident expression patterns after being treated for 8 h, suggesting their significance in corticosterone-mediated actions. Collectively, these findings indicate that corticosterone can inhibit proliferation and cause apoptosis in chicken ovarian prehierarchical, but not preovulatory granulosa cells, through impeding ACVR1-SMAD1-ID3 signaling presumptively.
Collapse
Affiliation(s)
- Ming Yang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Chenchen Jin
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xinyue Cheng
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Tuoyuan Liu
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Yu Ji
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Fengyan Meng
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xingfa Han
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Qiuxia Liang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xiaohan Cao
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Linyan Huang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xiaogang Du
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xianyin Zeng
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China.
| | - Guixian Bu
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China.
| |
Collapse
|
6
|
Zhidkova EM, Lylova ES, Savinkova AV, Mertsalov SA, Kirsanov KI, Belitsky GA, Yakubovskaya MG, Lesovaya EA. A Brief Overview of the Paradoxical Role of Glucocorticoids in Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2020; 14:1178223420974667. [PMID: 33424228 PMCID: PMC7755940 DOI: 10.1177/1178223420974667] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/21/2020] [Indexed: 11/15/2022]
Abstract
Glucocorticoids (GCs) are stress hormones that play multiple roles in the regulation of cancer cell differentiation, apoptosis, and proliferation. Some types of cancers, such as hematological malignancies, can be effectively treated by GCs, whereas the responses of epithelial cancers to GC treatment vary, even within cancer subtypes. In particular, GCs are frequently used as supporting treatment of breast cancer (BC) to protect against chemotherapy side effects. In the therapy of nonaggressive luminal subtypes of BC, GCs can have auxiliary antitumor effects due to their cytotoxic actions on cancer cells. However, GCs can promote BC progression, colonization of distant metastatic sites, and metastasis. The effects of GCs on cell proliferation vary with BC subtype and its molecular profile and are realized via the activation of glucocorticoid receptor (GR), a well-known transcriptional factor involved in the regulation of the expression of multiple genes, cell-cell adhesion, and cell migration and polarity. This review focuses on the roles of GC signaling in the adhesion, migration, and metastasis of BC cells. We discuss the molecular mechanisms of GC actions that lead to BC metastasis and propose alternative pharmacological uses of GCs for BC treatment.
Collapse
Affiliation(s)
- Ekaterina M Zhidkova
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Evgeniya S Lylova
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Alena V Savinkova
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | | | - Kirill I Kirsanov
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia.,Department of General Medical Practice, RUDN University, Moscow, Russia
| | - Gennady A Belitsky
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Marianna G Yakubovskaya
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Ekaterina A Lesovaya
- Department of Oncology, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia.,I.P. Pavlov Ryazan State Medical University, Ryazan, Russia
| |
Collapse
|
7
|
Brkic Z, Zivanovic A, Adzic M. Sex-specific Effects of Lipopolysaccharide on Hippocampal Mitochondrial Processes in Neuroinflammatory Model of Depression. Neuroscience 2020; 451:174-183. [PMID: 33039525 DOI: 10.1016/j.neuroscience.2020.09.059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/14/2020] [Accepted: 09/29/2020] [Indexed: 11/25/2022]
Abstract
Mitochondria play a significant role in pathogenesis of clinical depression and their function can be impaired by inflammation and alterations in hypothalamic-pituitary-adrenal axis. Sexual context is also a relevant factor in the incidence of mood disorders, and could have a strong influence during an immune challenge. Therefore, in this study we investigated whether the effects of seven-day lipopolysaccharide (LPS) treatment on glucocorticoid receptor (GR) could be associated with apoptosis and alterations in energy metabolism in hippocampus of female and male Wistar rats with depressive-like behavior. To that end, we measured the mitochondrial levels of GR and its phosphoisoforms pGR232 and pGR246 in hippocampus of female and male rats, as well as the mRNA levels of two GR-regulated mitochondrial genes, cyclooxygenase -1 and -3 (COX-1 and -3). We also measured alterations in the extrinsic and intrinsic apoptotic pathways in mitochondria and cytosol of hippocampus of these animals, and the levels of cleaved cytosolic poly [ADP-ribose] polymerase-1 (PARP-1) protein. We discovered that even though LPS treatment induced behavioral alterations and affected corticosterone levels and apoptosis in a similar manner in both sexes, it affected mitochondrial GR differently in males and females. Namely, the treatment decreased levels of mitochondrial GR and pGR232/pGR246 ratio only in females, and these alterations were followed by decreased mRNA levels of COX-1 and COX-3 only in this sex. The alterations in COX-1 and COX-3 mRNA levels could indicate impaired oxidative metabolism and diminished mitochondrial function in hippocampus of this sex.
Collapse
Affiliation(s)
- Zeljka Brkic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia.
| | - Ana Zivanovic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| |
Collapse
|
8
|
Mitochondrial signaling in inflammation-induced depressive behavior in female and male rats: The role of glucocorticoid receptor. Brain Res Bull 2019; 150:317-327. [PMID: 31251961 DOI: 10.1016/j.brainresbull.2019.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022]
Abstract
Mitochondrial dysfunction can result from the interplay between elevated inflammatory markers and alterations in hypothalamic-pituitary-adrenal (HPA) axis, and can contribute to pathogenesis of major depression. Therefore, in this study we investigated whether the effects of lipopolysaccharide (LPS) on glucocorticoid receptor (GR) could be associated with alterations in mitochondrial apoptotic signaling in the prefrontal cortex of male and female Wistar rats with depressive-like behavior. To that end, we measured LPS-induced alterations in the extrinsic and intrinsic apoptotic pathways in mitochondria and cytosol of PFC of female and male rats, as well as the levels of cleaved cytosolic PARP-1. We also measured the mitochondrial levels of GR and its phosphoisoforms pGR232 and pGR246, as well as the mRNA levels of two GR-regulated mitochondrial genes, COX-1 and COX-3. We discovered that although seven-day LPS treatment evoked depressive-like behavior and induced apoptosis in the PFC of both sexes, it affected apoptotic cascades in both sexes differently. In females the treatment initiated both intrinsic and extrinsic apoptotic cascade, while in males only intrinsic cascade was engaged. Alterations in intrinsic apoptotic pathway were more associated with GR alterations in males, where LPS treatment decreased levels of mitochondrial GR and increased pGR232/pGR246 ratio. Alterations in mitochondrial GR could be associated with changes in expression of genes involved in oxidative metabolism in the PFC of this sex, and could, in combination with elevated levels of BCL-2 and decreased levels of BAX detected in this cell fraction, mitigate the detrimental effect of LPS on mitochondria in male PFC.
Collapse
|
9
|
Hułas-Stasiak M, Dobrowolski P, Pawlikowska-Pawlęga B, Tomaszewska E, Muszyński S. The effects of dexamethasone administered during pregnancy on the postpartum spiny mouse ovary. PLoS One 2017; 12:e0183528. [PMID: 28827819 PMCID: PMC5565181 DOI: 10.1371/journal.pone.0183528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/04/2017] [Indexed: 01/30/2023] Open
Abstract
Excessive exposure to glucocorticoids can alter ovarian function by modulating oogenesis, folliculogenesis and steroidogenesis. The aim of the present study was to examine the effects of dexamethasone (DEX) administered during pregnancy on folliculogenesis and corpus luteum development in the postpartum spiny mouse ovary. DEX (125 μg kg-1 body weight per day) was applied to pregnant spiny mouse from day 20 of gestation to parturition. The obtained ovaries were fixed and used for immunohistochemistry and TEM analysis. The expression of proteins related to apoptosis (caspase-3, Bax, Bcl-2) and autophagy (Beclin1, Lamp1) as well as PCNA and GR receptors were evaluated by western-blot. In comparison with DEX-treated group a higher percentage of TUNEL positive granulosa and luteal cells was observed in the control group. These data were consistent with changes in caspase-3 and Bax expression, which increased in the control and decreased after DEX exposure. In turn, the proliferation index and PCNA expression were higher in the DEX-treated group. Moreover, the higher level of Beclin1, Lamp1, anti-apoptotic Bcl-2 protein and GR was observed in the DEX-treated females than in the control group. Beclin1 and Lamp1 were strongly expressed in luteal cells which exhibited an autophagic ultrastructure. Surprisingly, DEX augmented the number of ovarian follicles and corpora lutea, which resulted in a significant increase in ovarian weight. These findings suggest that DEX exerts anti-apoptotic action on granulosa layer and stimulates follicular maturation. Moreover, DEX induces autophagy in luteal cells promoting cell survival rather than cell death, which can prolong the corpus luteum life span.
Collapse
Affiliation(s)
- Monika Hułas-Stasiak
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Lublin, Poland
| | - Piotr Dobrowolski
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Lublin, Poland
| | | | - Ewa Tomaszewska
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences, Lublin, Poland
| | - Siemowit Muszyński
- Department of Physics, Faculty of Production Engineering, University of Life Sciences, Lublin, Poland
| |
Collapse
|
10
|
Li Y, Zheng B, Tian H, Xu X, Sun Y, Mei Q, Lin X, Liu L. Yupingfeng Powder relieves the immune suppression induced by dexamethasone in mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 200:117-123. [PMID: 28161541 DOI: 10.1016/j.jep.2017.01.054] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/21/2016] [Accepted: 01/30/2017] [Indexed: 05/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yupingfeng Powder (YPF), a Chinese medical formula, is used traditionally for allergic diseases and characterized by reducing allergy relapse. In the present study, we attempted to investigate the effect of YPF on the immunity of mice and the possible mechanisms. MATERIALS AND METHODS An immunosuppressive mice model induced by Dexamethasone (Dex) was used. Blood samples, spleen and thymus were collected. Then, hematology parameters and organ weight were measured; Phenotypic analyses (CD4+/CD8+) of lymphocytes were performed using a flow cytometer; Phagocytosis of peritoneal macrophages were evaluated by particle tracers; Spleen lymphocytes were isolated, whose proliferation and apoptosis were assessed. NK cells' cytotoxicity was determined using the LDH release assay. RESULTS YPF could ameliorate weight loss and improve low thymus and spleen coefficients caused by Dex. Treatment with YPF made decreased lymphocytic activity of Dex-treated mice back to normal and inhibited Dex-induced apoptosis of lymphocytes. YPF increased the Dex caused low proportion of CD4+/CD8+, and upregulated Dex-reduced NK cells' activity. CONCLUSION The series of experiments demonstrated that YPF could exert immune regulation and enhance immunity of immunosuppressive mice through adjusting nonspecific and cellular immunity. The results would provide a basis for clinical application of YPF.
Collapse
Affiliation(s)
- Yuhua Li
- No. 422 Hospital of PLA, Zhanjiang 524005, Guangdong, PR China; Laboratory of Oncological Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Bin Zheng
- Department of Pharmacology, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China.
| | - Huajie Tian
- Department of Pharmacology, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China.
| | - Xiaotao Xu
- Department of Pharmacology, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China.
| | - Yang Sun
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi'an 710032, Shaanxi, PR China.
| | - Qibing Mei
- Department of Pharmacology, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi'an 710032, Shaanxi, PR China.
| | - Xiaotian Lin
- No. 422 Hospital of PLA, Zhanjiang 524005, Guangdong, PR China.
| | - Li Liu
- Department of Pharmacology, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China.
| |
Collapse
|
11
|
Tetsuka M, Takagi R, Ambo N, Myat TS, Zempo Y, Onuma A. Glucocorticoid metabolism in the bovine cumulus-oocyte complex matured in vitro. Reproduction 2015; 151:73-82. [PMID: 26519454 DOI: 10.1530/rep-15-0363] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/30/2015] [Indexed: 11/08/2022]
Abstract
Glucocorticoid action in target organs is regulated by relative activities of 11β-HSD type 1 (HSD11B1) that mainly converts cortisone to active cortisol and type 2 (HSD11B2) that inactivates cortisol to cortisone. HSD11Bs have been shown to be expressed in the ovary of various species. However, little is known about the expression and activity of HSD11Bs in the bovine cumulus-oocyte complex (COC). In the present study, we investigated the expression and activities of HSD11Bs in in vitro-matured (IVM) bovine COCs. Bovine COCs were matured in M199 supplemented with or without FSH and FCS. The expression of HSD11B1 and HSD11B2 was measured by using quantitative RT-PCR in denuded oocytes (DO) and cumulus cells (CC). Reductive and oxidative activities of HSD11Bs were determined by radiometric conversion assay using labeled cortisol, cortisone or dexamethasone in intact COCs, DO or CC in the presence or absence of 11-keto-progesterone (11kP), a selective inhibitor of HSD11B2. The presence of HSD11Bs in the oocyte was examined by immunofluorescence microscopy. Oocytes exclusively expressed HSD11B2 and its expression and activity were largely unchanged during IVM. CC, on the other hand, exclusively expressed HSD11B1 and its expression and activity were upregulated as IVM progressed. As a result, the net glucocorticoid metabolism shifted from inactivation to activation towards the end of IVM. These results indicate that the bovine COC is capable of modulating local glucocorticoid concentration and, by doing so, may create an environment that is favorable to ovulating oocyte for maturation, fertilization and subsequent development.
Collapse
Affiliation(s)
- Masafumi Tetsuka
- Department of Life Science and AgricultureObihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Ryo Takagi
- Department of Life Science and AgricultureObihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Nobuhiro Ambo
- Department of Life Science and AgricultureObihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Thet Su Myat
- Department of Life Science and AgricultureObihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Yuta Zempo
- Department of Life Science and AgricultureObihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Asuka Onuma
- Department of Life Science and AgricultureObihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
12
|
Sousa ML, Figueiredo F, Pinheiro C, Silva A, Malhão F, Rocha MJ, Rocha E, Urbatzka R. Morphological and molecular effects of cortisol and ACTH on zebrafish stage I and II follicles. Reproduction 2015; 150:429-36. [PMID: 26316549 DOI: 10.1530/rep-15-0271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/27/2015] [Indexed: 11/08/2022]
Abstract
Oogenesis in zebrafish (Danio rerio) is controlled by the hypothalamus-pituitary-gonadal axis and reproductive hormones. In addition, an interference of stress hormones is known with reproductive biology. In the presented work, we aimed to explore the hypothesis that cortisol (Cort) and ACTH may affect early oogenesis in zebrafish, given the presence of the specific receptors for glucocorticoids and ACTH in the zebrafish ovary. Follicles at stages I and II were exposed in vitro to 1 μM Cort and ACTH for 48 h, then ultrastructural and molecular effects were analyzed. The comet assay demonstrated increased tail moments for Cort and ACTH treatment indicative of DNA damage. The mRNA expression of apoptotic genes (bax, bcl-2) was not altered by both treatments, but Cort increased significantly the expression of the ACTH receptor (mc2r). Cort stimulated the presence of the endoplasmic reticulum, predominantly at stage II, while ACTH induced a strong vacuolization. Viability of oocytes was not affected by both treatments and fluorescent staining (monodansylcadaverine/acridine orange) indicated a reduced quantity of autophagosomes for ACTH, and lower presence of nucleic acids in ooplasm for Cort and ACTH. Concluding, different responses were observed for stress hormones on early stages of zebrafish oocytes, which suggest a role for both hormones in the stress-mediated adverse effects on female gametogenesis.
Collapse
Affiliation(s)
- Maria Lígia Sousa
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Francisco Figueiredo
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Pinheiro
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana Silva
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Fernanda Malhão
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Maria João Rocha
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Eduardo Rocha
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Ralph Urbatzka
- CIIMAR - Interdisciplinary Center of Marine and Environmental ResearchCIMAR Associated Laboratory, U. Porto - University of Porto, Rua dos Bragas 289, 4050-123 Porto, PortugalICBAS - Institute of Biomedical Sciences Abel SalazarUPorto - University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, PortugalISCS-N - Superior Institute of Health Sciences-NorthCESPU - Cooperative Higher Education, Polytechnic and University, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| |
Collapse
|
13
|
Heitmann RJ, Tobler KJ, Gillette L, Tercero J, Burney RO. Dexamethasone attenuates the embryotoxic effect of endometriotic peritoneal fluid in a murine model. J Assist Reprod Genet 2015. [PMID: 26198138 DOI: 10.1007/s10815-015-0516-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The in vitro fertilization (IVF) pregnancy rate of women with advanced stage endometriosis is nearly half that of the general population, suggesting incomplete targeting of the pathophysiology underlying endometriosis-associated infertility. Compelling evidence highlights inflammation as the etiologic link between endometriosis and infertility and a potential target for adjunctive treatment. The objective of this study was to examine the effect of dexamethasone on murine embryos exposed to human endometriotic peritoneal fluid (PF) using the established murine embryo assay model. METHODS PF was obtained from women with and without severe endometriosis. Murine embryos were harvested and randomly allocated to five groups of culture media conditions: (1) human tubal fluid (HTF), (2) HTF and 10 % PF from women without endometriosis, (3) HTF and 10 % PF from women with endometriosis (PF-E), (4) HTF with PF-E and 0.01 mcg/mL dexamethasone, and (5) HTF with PF-E and 0.1 mcg/mL dexamethasone. Embryos were cultured in standard conditions and evaluated for blastocyst development. RESULTS A total of 266 mouse embryos were cultured. Baseline blastulation rates were 63.6 %. The addition of peritoneal fluid from women with endometriosis decreased the blastocyst development rate to 38.9 % (P = 0.008). The addition of 0.1 mcg/mL of dexamethasone to the culture media restored the blastulation rate to near baseline levels (61.2 %; P = 0.019). CONCLUSIONS The results of our in vitro study demonstrate the capacity of dexamethasone to mitigate the deleterious impact of endometriotic PF on embryo development. If confirmed in vivo, dexamethasone may prove a useful adjunct for the treatment of endometriosis-associated infertility.
Collapse
Affiliation(s)
- Ryan J Heitmann
- Department of Obstetrics and Gynecology, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA, 98431, USA
| | - Kyle J Tobler
- Department of Obstetrics and Gynecology, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA, 98431, USA
| | - Laurie Gillette
- Department of Clinical Investigation, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA, 98431, USA
| | - Juan Tercero
- Department of Clinical Investigation, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA, 98431, USA
| | - Richard O Burney
- Department of Obstetrics and Gynecology, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA, 98431, USA. .,Department of Clinical Investigation, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA, 98431, USA. .,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Madigan Healthcare Systems, ATTN: MCHJ-OG, 9040A Fitzsimmons Drive, Tacoma, WA, 98431-1100, USA.
| |
Collapse
|
14
|
Chakraborty P, Roy SK. Effect of azaline B on follicular development and functions in the hamster. Mol Cell Endocrinol 2015; 400:1-9. [PMID: 25462584 PMCID: PMC4274241 DOI: 10.1016/j.mce.2014.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/08/2023]
Abstract
The usefulness of azaline B, a GnRH antagonist, in suppressing gonadotropin secretion in the golden hamster was examined by examining follicular development, steroidogenesis and expression of steroidogenic enzymes. Serum levels of P and E declined significantly, while FSH or LH was undetectable in azaline B-treated hamsters. FSH significantly increased serum E levels, whereas LH upregulated serum P levels. The formation of antral follicles ceased in azaline-treated hamsters, but was reversed by FSH with or without LH supplement. FSH also activated the primordial follicle pool resulting in increased formation of primary and preantral follicles. Further, an increasing trend in the formation of preantral follicles in response to E or E + P, and the formation of antral follicles in response to E + P treatment was evident. The level of Cyp11a1 mRNA increased markedly in LH- or LH + FSH-treated hamsters, whereas FSH with or without LH upregulated Cyp17a1, Cyp19a1 and Fshr mRNA expression. E without or with P also upregulated ovarian Cyp19a1 mRNA expression. The expression of enzyme protein corroborated the mRNA data. In summary, azaline B is an efficient GnRH antagonist in the hamster, and will be useful in studying the selective effect of gonadotropins on ovarian functions without disrupting the physiological functions of other hormones in ovarian cells.
Collapse
Affiliation(s)
- Prabuddha Chakraborty
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shyamal K Roy
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
15
|
Stournaras C, Gravanis A, Margioris AN, Lang F. The actin cytoskeleton in rapid steroid hormone actions. Cytoskeleton (Hoboken) 2014; 71:285-93. [DOI: 10.1002/cm.21172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/20/2014] [Accepted: 02/26/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Christos Stournaras
- Department of Biochemistry; University of Crete Medical School; Heraklion Greece
- Department of Physiology; University of Tübingen; Tübingen Germany
| | - Achilles Gravanis
- Department of Pharmacology; University of Crete Medical School; Heraklion Greece
| | - Andrew N. Margioris
- Department of Clinical Chemistry; University of Crete Medical School; Heraklion Greece
| | - Florian Lang
- Department of Physiology; University of Tübingen; Tübingen Germany
| |
Collapse
|
16
|
Whirledge S, Cidlowski JA. A role for glucocorticoids in stress-impaired reproduction: beyond the hypothalamus and pituitary. Endocrinology 2013; 154:4450-68. [PMID: 24064362 PMCID: PMC3836069 DOI: 10.1210/en.2013-1652] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In addition to the well-characterized role of the sex steroid receptors in regulating fertility and reproduction, reproductive events are also mediated by the hypothalamic-pituitary-adrenal axis in response to an individual's environment. Glucocorticoid secretion in response to stress contributes to the well-characterized suppression of the hypothalamic-pituitary-gonadal axis through central actions in the hypothalamus and pituitary. However, both animal and in vitro studies indicate that other components of the reproductive system are also regulated by glucocorticoids. Furthermore, in the absence of stress, it appears that homeostatic glucocorticoid signaling plays a significant role in reproduction and fertility in all tissues comprising the hypothalamic-pituitary-gonadal axis. Indeed, as central regulators of the immune response, glucocorticoids are uniquely poised to integrate an individual's infectious, inflammatory, stress, nutritional, and metabolic status through glucocorticoid receptor signaling in target tissues. Endocrine signaling between tissues regulating the immune and stress response and those determining reproductive status provides an evolutionary advantage, facilitating the trade-off between reproductive investment and offspring fitness. This review focuses on the actions of glucocorticoids in tissues important for fertility and reproduction, highlighting recent studies that show glucocorticoid signaling plays a significant role throughout the hypothalamic-pituitary-gonadal axis and characterizing these effects as permissive or inhibitory in terms of facilitating reproductive success.
Collapse
Affiliation(s)
- Shannon Whirledge
- NIEHS/NIH, MD F3-07, P.O. Box 12233, Research Triangle Park, North Carolina 27709.
| | | |
Collapse
|
17
|
Hassanpour M, Joss J, Mohammad MG. Functional analyses of lymphocytes and granulocytes isolated from the thymus, spiral valve intestine, spleen, and kidney of juvenile Australian lungfish, Neoceratodus forsteri. FISH & SHELLFISH IMMUNOLOGY 2013; 35:107-114. [PMID: 23603235 DOI: 10.1016/j.fsi.2013.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 03/12/2013] [Accepted: 04/04/2013] [Indexed: 06/02/2023]
Abstract
Our current understanding of the lungfish immune system is limited. This study is characterizing the immune cells separated from primary and secondary immune organs of the Australian lungfish, Neoceratodus forsteri. Our functional studies utilized flow cytometry to study the immune cells extracted from the thymus, spiral valve intestine, spleen, and kidney. The different characteristics of lymphocytes and granulocytes were analyzed by utilization of viability, phagocytosis, oxidative burst, and apoptosis assays. Most of the nonviable intestinal cells were lymphocytes. Depending on the organ, 6-25% of the total population, predominantly granulocytes, underwent phagocytosis where the splenic cells were the most and intestinal cells the least phagocytic cells. Cells responded positively but differently to stimulation with phorbol myristate acetate (PMA) to produce radical oxygen species, an indication of their oxidative burst activity, which was mainly associated with granulocytes. Although cells were induced by dexamethasone to undergo apoptosis, such an induction did not follow a consistent pattern of dose of dexamethasone or incubation time between the different organs. In the absence of monoclonal antibodies against lungfish immune cells, these functional flow cytometric analyses aid our understanding on the functionality of immune cells.
Collapse
Affiliation(s)
- Masoud Hassanpour
- Department of Biological Sciences, Macquarie University, Sydney, New South Wales, Australia
| | | | | |
Collapse
|
18
|
Rusai K, Prokai A, Juanxing C, Meszaros K, Szalay B, Pásti K, Müller V, Heemann U, Lutz J, Tulassay T, Szabo A. Dexamethasone protects from renal ischemia/reperfusion injury: A possible association with SGK-1. ACTA ACUST UNITED AC 2013; 100:173-85. [DOI: 10.1556/aphysiol.100.2013.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
19
|
Giron-Michel J, Azzi S, Khawam K, Mortier E, Caignard A, Devocelle A, Ferrini S, Croce M, François H, Lecru L, Charpentier B, Chouaib S, Azzarone B, Eid P. Interleukin-15 plays a central role in human kidney physiology and cancer through the γc signaling pathway. PLoS One 2012; 7:e31624. [PMID: 22363690 PMCID: PMC3283658 DOI: 10.1371/journal.pone.0031624] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 01/16/2012] [Indexed: 12/16/2022] Open
Abstract
The ability of Interleukin-15 (IL-15) to activate many immune antitumor mechanisms renders the cytokine a good candidate for the therapy of solid tumors, particularly renal cell carcinoma. Although IL-15 is being currently used in clinical trials, the function of the cytokine on kidney's components has not been extensively studied; we thus investigated the role of IL-15 on normal and tumor renal epithelial cells. Herein, we analyzed the expression and the biological functions of IL-15 in normal renal proximal tubuli (RPTEC) and in their neoplastic counterparts, the renal clear cell carcinomas (RCC). This study shows that RPTEC express a functional heterotrimeric IL-15Rαβγc complex whose stimulation with physiologic concentrations of rhIL-15 is sufficient to inhibit epithelial mesenchymal transition (EMT) commitment preserving E-cadherin expression. Indeed, IL-15 is not only a survival factor for epithelial cells, but it can also preserve the renal epithelial phenotype through the γc-signaling pathway, demonstrating that the cytokine possess a wide range of action in epithelial homeostasis. In contrast, in RCC in vitro and in vivo studies reveal a defect in the expression of γc-receptor and JAK3 associated kinase, which strongly impacts IL-15 signaling. Indeed, in the absence of the γc/JAK3 couple we demonstrate the assembly of an unprecedented functional high affinity IL-15Rαβ heterodimer, that in response to physiologic concentrations of IL-15, triggers an unbalanced signal causing the down-regulation of the tumor suppressor gene E-cadherin, favoring RCC EMT process. Remarkably, the rescue of IL-15/γc-dependent signaling (STAT5), by co-transfecting γc and JAK3 in RCC, inhibits EMT reversion. In conclusion, these data highlight the central role of IL-15 and γc-receptor signaling in renal homeostasis through the control of E-cadherin expression and preservation of epithelial phenotype both in RPTEC (up-regulation) and RCC (down-regulation).
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Sandy Azzi
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Krystel Khawam
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Erwan Mortier
- INSERM UMRS 892, Institut de Recherche Thérapeutique de l'Université de Nantes (IRT UN), Nantes, France
| | - Anne Caignard
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris, France
| | - Aurore Devocelle
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Silvano Ferrini
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Michela Croce
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Hélène François
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Lola Lecru
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Bernard Charpentier
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Salem Chouaib
- INSERM UMR 753, Université de Paris-Sud, Institut Gustave Roussy (IGR), Villejuif, France
| | - Bruno Azzarone
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| | - Pierre Eid
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| |
Collapse
|
20
|
Jing Y, Qian Y, Ghandi M, He A, Borys MC, Pan SH, Li ZJ. A mechanistic study on the effect of dexamethasone in moderating cell death in Chinese Hamster Ovary cell cultures. Biotechnol Prog 2011; 28:490-6. [PMID: 22140034 DOI: 10.1002/btpr.747] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 09/30/2011] [Indexed: 11/08/2022]
Abstract
Dexamethasone (DEX) was previously shown (Jing et al., Biotechnol Bioeng. 2010;107:488-496) to play a dual role in increasing sialylation of recombinant glycoproteins produced by Chinese Hamster Ovary (CHO) cells. DEX addition increased sialic acid levels of a recombinant fusion protein through increased expression of α2,3-sialyltransferase and β1,4-galactosyltransferase, but also decreased the sialidase-mediated, extracellular degradation of sialic acid through slowing cell death at the end of the culture period. This study examines the underlying mechanism for this cytoprotective action by studying the transcriptional response of the CHO cell genome upon DEX treatment using DNA microarrays and gene ontology term analysis. Many of those genes showing a significant transcriptional response were associated with the regulation of programmed cell death. The gene with the highest change in expression level, as validated by Quantitative PCR assays with TaqMan® probes and confirmed by Western Blot analysis, was the antiapoptotic gene Tsc22d3, also referred to as GILZ (glucocorticoid-induced leucine zipper). The pathway by which DEX suppressed cell death towards the end of the culture period was also confirmed by showing involvement of glucocorticoid receptors and GILZ through studies using the glucocorticoid antagonist mifepristone (RU-486). These findings advance the understanding of the mechanism by which DEX suppresses cell death in CHO cells and provide a rationale for the application of glucocorticoids in CHO cell culture processes.
Collapse
Affiliation(s)
- Ying Jing
- Biologics Process and Product Development, Technical Operations, Bristol-Myers Squibb Company, East Syracuse, NY 13057, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
D'Eliseo D, Pisu P, Romano C, Tubaro A, De Nunzio C, Morrone S, Santoni A, Stoppacciaro A, Velotti F. Granzyme B is expressed in urothelial carcinoma and promotes cancer cell invasion. Int J Cancer 2010; 127:1283-94. [PMID: 20027633 DOI: 10.1002/ijc.25135] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Granzyme B (GrB) is a serine proteinase known to be expressed by cytotoxic lymphocytes and to induce, in presence of perforin (Pf), apoptosis in target cells. Recently, GrB expression has been shown (often in absence of Pf) in nonlymphoid cells, but its function is not defined. In our study, we investigated GrB and Pf expression in bladder cancer cell lines and in urothelial carcinoma (UC) tissues by reverse transcription-polymerase chain reaction (RT-PCR), Western blot, ELISA, immunofluorescence and immunohistochemistry. We also assessed the function of GrB in UC cells; the in vitro function of GrB was examined by loss-of-function experiments. Our results revealed that GrB is expressed, in absence of Pf, in UC cells. Significant differences were found between GrB expression and both increasing pathological tumor spreading and high-grade vs. low-grade pTa tumors. Notably, GrB in UC tissues was concentrated at the cancer invasion front and was expressed in neoplastic cells undergoing epithelial-mesenchymal transition, a key event in carcinoma invasion. Indeed, GrB-positive cells also expressed Snail, N-cadherin or were negative for E-cadherin. GrB expressed in tumor cell lines was enzymatically active and capable of vitronectin cleavage, implying extracellular matrix (ECM) remodeling by GrB. Inhibition of GrB activity or Stealth RNA interference-mediated GrB gene silencing markedly suppressed bladder cancer cell invasion through matrigel. This data provides the first evidence for a role of GrB in promoting cancer cell invasion. Taken together, our findings suggest that GrB, via ECM degradation, contributes to the establishment of the UC invasive phenotype.
Collapse
Affiliation(s)
- Donatella D'Eliseo
- Department of Ecology and Sustainable Economic Development DECOS, Tuscia University, Viterbo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hoang KN, Dinh CT, Bas E, Chen S, Eshraghi AA, Van De Water TR. Dexamethasone treatment of naïve organ of Corti explants alters the expression pattern of apoptosis-related genes. Brain Res 2009; 1301:1-8. [DOI: 10.1016/j.brainres.2009.08.097] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/25/2009] [Accepted: 08/26/2009] [Indexed: 12/20/2022]
|
23
|
Kumar S, Allen DA, Kieswich JE, Patel NSA, Harwood S, Mazzon E, Cuzzocrea S, Raftery MJ, Thiemermann C, Yaqoob MM. Dexamethasone ameliorates renal ischemia-reperfusion injury. J Am Soc Nephrol 2009; 20:2412-25. [PMID: 19797168 DOI: 10.1681/asn.2008080868] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In the setting of renal ischemia-reperfusion injury (IRI), the effect and mechanism of action of glucocorticoids are not well understood. In rat renal IRI, a single dose of dexamethasone administered before ischemia, or at the onset of reperfusion, ameliorated biochemical and histologic acute kidney injury after 24 h. Dexamethasone upregulated Bcl-xL, downregulated ischemia-induced Bax, inhibited caspase-9 and caspase-3 activation, and reduced apoptosis and necrosis of proximal tubular cells. In addition, dexamethasone decreased the number of infiltrating neutrophils and ICAM-1. We observed the protective effect of dexamethasone in neutrophil-depleted mice, suggesting a neutrophil-independent mechanism. In vitro, dexamethasone protected human kidney proximal tubular (HK-2) cells during serum starvation and IRI-induced apoptosis, but inhibition of MEK 1/2 abolished its anti-apoptotic effects in these conditions. Dexamethasone stimulated rapid and transient phosphorylation of ERK 1/2, which required the presence of the glucocorticoid receptor and was independent of transcriptional activity. In summary, in the setting of renal ischemia-reperfusion injury, dexamethasone directly protects against kidney injury by a receptor-dependent, nongenomic mechanism.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, St. Bartholomew's, University of London, London EC1M 6BQ, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chang A, Eastwood H, Sly D, James D, Richardson R, O’Leary S. Factors influencing the efficacy of round window dexamethasone protection of residual hearing post-cochlear implant surgery. Hear Res 2009; 255:67-72. [DOI: 10.1016/j.heares.2009.05.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 05/01/2009] [Accepted: 05/18/2009] [Indexed: 10/20/2022]
|
25
|
Qian YH, Xiao Q, Chen H, Xu J. Dexamethasone inhibits camptothecin-induced apoptosis in C6-glioma via activation of Stat5/Bcl-xL pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:764-71. [PMID: 19339209 DOI: 10.1016/j.bbamcr.2009.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 01/02/2009] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
Abstract
Dexamethasone (DX) induces apoptosis resistance in most solid malignant tumors during co-treatment with chemotherapy agents, such as camptothecin (CAM). In this study, we investigated the mechanism by which DX reduces chemotherapy efficiency in C6-glioma. DX reduced CAM-increased DNA fragmentation and caspase-3 activation. The DX's protection was negated by RU486, an antagonist of glucocorticoid receptor (GR). DX itself increased anti-apoptotic gene, Bcl-xL expression, and its transcription factor, signaling transducer and activator of transcription 5 (Stat5), DNA binding activity and phospho-Stat5 expression. DX blocked the CAM-decreased Bcl-xL and phospho-Stat5 expression, and Stat5 binding activity. RU486 negated DX's actions. To determine whether Stat5 regulates Bcl-xL expression in CAM-induced cell death, C6-glioma was infected with an adenovirus containing a constitutively activated Stat5-GFP (Ad-Stat5ca). Overexpression of Stat5ca increased Bcl-xL and decreased CAM-induced cell death compared to control adenovirus infected cells; whereas Stat5 siRNA decreased DX-induced Bcl-xL and increased cell death. Phospho-Stat5 expression was observed in the nuclear extract by co-immunoprecipitation with an anti-GR antibody, indicating that Stat5 and GR were interactive and formed a complex in the nuclei. These results suggest that DX's prevention from CAM-induced apoptosis and RU486's antagonism of DX's protection may be through Stat5/Bcl-xL signal pathway regulated by a GR.
Collapse
Affiliation(s)
- Yi-Hua Qian
- Department of Human Anatomy and Histology-Embryology, School of Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | | | | | | |
Collapse
|
26
|
Herr I, Büchler MW, Mattern J. Glucocorticoid-mediated apoptosis resistance of solid tumors. Results Probl Cell Differ 2009; 49:191-218. [PMID: 19132324 DOI: 10.1007/400_2008_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
More than a quarter of a century ago, the phenomenon of glucocorticoid-induced apoptosis in the majority of hematological cells was first recognized. More recently, glucocorticoid-induced antiapoptotic signaling associated with apoptosis resistance towards cytotoxic therapy has been identified in cells of epithelial origin, most of malignant solid tumors and some other tissues. Despite these huge amounts of data demonstrating differential pro- and anti-apoptotic effects of glucocorticoids, the underlying mechanisms of cell type-specific glucocorticoid signaling are just beginning to be described. This review summarizes our present understanding of cell type-specific pro- and anti-apoptotic signaling induced by glucocorticoids. We shortly introduce mechanisms of glucocorticoid resistance of hematological cells. We highlight and discuss the emerging molecular evidence of a general induction of survival signaling in epithelial cells and carcinoma cells by glucocorticoids. We give a summary of our current knowledge of decreased proliferation rates in response to glucocorticoid pre- and combination treatment, which are suspicious to be involved not only in protection of normal tissues, but also in protection of solid tumors from cytotoxic effects of anticancer agents.
Collapse
Affiliation(s)
- Ingrid Herr
- Department of Surgery, University of Heidelberg, Germany.
| | | | | |
Collapse
|
27
|
Dickinson RE, Myers M, Duncan WC. Novel regulated expression of the SLIT/ROBO pathway in the ovary: possible role during luteolysis in women. Endocrinology 2008; 149:5024-34. [PMID: 18566128 DOI: 10.1210/en.2008-0204] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The human corpus luteum (CL) undergoes luteolysis, associated with marked tissue and vascular remodeling, unless conception occurs and the gland is rescued by human chorionic gonadotropin (hCG). In Drosophila the Slit gene product, a secreted glycoprotein, acts as a ligand for the roundabout (robo) transmembrane receptor. Together they influence the guidance and migration of neuronal and nonneuronal cells. In vertebrates three Slit (Slit1, Slit2, Slit3) and four Robo (Robo1, Robo2, Robo3/Rig-1, Robo4/Magic Robo) genes have been identified. ROBO1, SLIT2, and SLIT3 are also inactivated in human cancers and may regulate apoptosis and metastasis. Because processes such as apoptosis and tissue remodeling occur during the regression of the CL, the aim of this study was to investigate the expression, regulation, and effects of the SLIT and ROBO genes in human luteal cells. Immunohistochemistry and RT-PCR revealed that SLIT2, SLIT3, ROBO1, and ROBO2 are expressed in luteal steroidogenic cells and fibroblast-like cells of the human CL. Furthermore, using real-time quantitative PCR, expression of SLIT2, SLIT3, and ROBO2 was maximal in the late-luteal phase and significantly reduced after luteal rescue in vivo with exogenous hCG (P<0.05). Additionally, hCG significantly inhibited SLIT2, SLIT3, and ROBO2 expression in cultured luteinized granulosa cells (P<0.05). Blocking SLIT-ROBO activity increased migration and significantly decreased levels of apoptosis in primary cultures of luteal cells (P<0.05). Overall, these results suggest the SLIT/ROBO pathway could play an important role in luteolysis in women.
Collapse
Affiliation(s)
- Rachel E Dickinson
- Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, Department of Reproductive and Developmental Sciences, The Queen's Medical Research Institute, Edinburgh, Scotland, United Kingdom.
| | | | | |
Collapse
|
28
|
O'Leary S, Lloyd ML, Shellam GR, Robertson SA. Immunization with recombinant murine cytomegalovirus expressing murine zona pellucida 3 causes permanent infertility in BALB/c mice due to follicle depletion and ovulation failure. Biol Reprod 2008; 79:849-60. [PMID: 18667753 DOI: 10.1095/biolreprod.108.067884] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Zona pellucida (ZP) glycoproteins are promising candidate antigens for use in immunocontraceptive vaccines because of their crucial role in mammalian fertilization. A single intraperitoneal immunization with recombinant murine cytomegalovirus engineered to express murine ZP3 (rMCMV-mZP3) induces permanent infertility with no evident systemic illness in female BALB/c mice. To investigate the mechanisms underpinning reproductive failure elicited by rMCMV-mZP3, ovarian parameters and reproductive function were evaluated at time points spanning 10 days to 5 wk after virus inoculation. Fertility was substantially impaired by 14 days after inoculation with rMCMV-mZP3 and was fully ablated by 21 days. Pregnancies established after inoculation but before complete infertility showed no adverse effects on fetal viability assessed at Day 17.5 post coitum (pc). Infertile mice retained estrous cycling activity and remained receptive to mating; however, at Day 3.5 pc there were fewer developing embryos and corpora lutea, plasma progesterone content was reduced, and there was no evidence of excess unfertilized oocytes. Consistent with this, profound ovarian pathology was evident from 10 days after rMCMV-mZP3 inoculation, with a decline first in mature ovarian follicles and then in immature ovarian follicles and with diminished expression of genes regulating follicle development, including Nobox, Gdf9, and Gja1 (connexin43). Follicle loss was associated with mild focal oophoritis and with recruitment of inflammatory leukocytes, predominantly CD4(+) and CD8(+) T cells evident from 10 days after virus inoculation. These data indicate that vaccination with rMCMV-mZP3 causes permanent infertility in BALB/c mice principally due to induction of ovarian autoimmune pathology leading to progressive oocyte depletion and eventual ovulation failure.
Collapse
Affiliation(s)
- Sean O'Leary
- Research Centre for Reproductive Health, Discipline of Obstetrics and Gynaecology, University of Adelaide, Adelaide, South Australia, Australia 5005
| | | | | | | |
Collapse
|
29
|
Haller J, Mikics E, Makara GB. The effects of non-genomic glucocorticoid mechanisms on bodily functions and the central neural system. A critical evaluation of findings. Front Neuroendocrinol 2008; 29:273-91. [PMID: 18054070 DOI: 10.1016/j.yfrne.2007.10.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 10/02/2007] [Accepted: 10/17/2007] [Indexed: 11/15/2022]
Abstract
Mounting evidence suggests that--beyond the well-known genomic effects--glucocorticoids affect cell function via non-genomic mechanisms. Such mechanisms operate in many major systems and organs including the cardiovascular, immune, endocrine and nervous systems, smooth and skeletal muscles, liver, and fat cells. Non-genomic effects are exerted by direct actions on membrane lipids (affecting membrane fluidity), membrane proteins (e.g. ion channels and neurotransmitter receptors), and cytoplasmic proteins (e.g. MAPKs, phospholipases, protein kinases, etc.). These actions are mediated by the glucocorticoids per se or by the proteins dissociated from the liganded glucocorticoid receptor complex. The MR and GR also activate non-genomic mechanisms in certain cases. Some effects of glucocorticoids are shared by a variety of steroids, whereas others are more selective. Moreover, "ultra-selective" effects-mediated by certain glucocorticoids only-were also shown. Disparate findings suggest that non-genomic mechanisms also show "demand-specificity", i.e. require the coincidence of two or more processes. Some of the non-genomic mechanisms activated by glucocorticoids are therapeutically relevant; moreover, the "non-genomic specificity" of certain glucocorticoids raises the possibility of therapeutic applications. Despite the large body of evidence, however, the non-genomic mechanisms of glucocorticoids are still poorly understood. Criteria for differentiating genomic and non-genomic mechanisms are often loosely applied; interactions between various mechanisms are unknown, and non-genomic mechanism-specific pharmacological (potentially therapeutic) agents are lacking. Nevertheless, the discovery of non-genomic mechanisms is a major breakthrough in stress research, and further insights into these mechanisms may open novel approaches for the therapy of various diseases.
Collapse
Affiliation(s)
- József Haller
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest 1083, Hungary
| | | | | |
Collapse
|
30
|
Abstract
Gap junctions that allow the direct communication between cytoplasmic compartments of neighboring cells are present in a variety of tissues and organs and play pivotal roles in a wide range of physiological processes. In the ovary, gap junctions consist mainly of connexin (Cx) 43 and Cx37, and their indispensable role in regulating folliculogenesis and oogenesis is well established. The ovarian Cx43 is regulated by gonadotropins at the transcriptional, translational and post-translational levels whereas the regulation of the ovarian Cx37 is yet unknown. In addition to their involvement in normal ovarian functions, gap junction proteins, particularly Cx43, seem to act as cancer suppressors. A summary of our present knowledge regarding gap junctional communication (GJC) and the ovarian gap junction proteins in normally developing ovaries and under pathological conditions is presented in this review.
Collapse
Affiliation(s)
- Eran Gershon
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | |
Collapse
|
31
|
Myers M, Lamont MC, van den Driesche S, Mary N, Thong KJ, Hillier SG, Duncan WC. Role of luteal glucocorticoid metabolism during maternal recognition of pregnancy in women. Endocrinology 2007; 148:5769-79. [PMID: 17872369 DOI: 10.1210/en.2007-0742] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The human corpus luteum (hCL) is an active, transient, and dynamic endocrine gland. It will experience extensive tissue and vascular remodeling followed by 1) demise of the whole gland without any apparent scarring or 2) maintenance of structural and functional integrity dependent on conceptus-derived human chorionic gonadotropin (hCG). Because cortisol has well-characterized roles in tissue remodeling and repair, we hypothesized that it may have a role in controlling luteal dissolution during luteolysis and would be locally produced toward the end of the luteal cycle. Glucocorticoid-metabolizing enzymes [11beta-hydroxysteroid dehydrogenase (11betaHSD) types 1 and 2] and the glucocorticoid receptor (GR) were assessed in hCL and cultures of luteinized granulosa cells (LGC) using immunofluorescence and quantitative RT-PCR. Furthermore, the effect of cortisol on steroidogenic cell survival and fibroblast-like cell activity was explored in vitro. The hCL expressed 11betaHSD isoenzymes in LGC and nuclear GR in several cell types. hCG up-regulated the expression and activity of 11betaHSD type 1 (P < 0.05) and down-regulated type 2 enzyme (P < 0.05) in vitro and tended to do the same in vivo. Cortisol increased the survival of LGC treated with RU486 (P < 0.05) and suppressed the activity of a proteolytic enzyme associated with luteolysis in fibroblast-like cells (P < 0.05). Our results suggest that, rather than during luteolysis, it is luteal rescue with hCG that is associated with increased local cortisol generation by 11betaHSD type 1. Locally generated cortisol may therefore act on the hCL through GR to have a luteotropic role in the regulation of luteal tissue remodeling during maternal recognition of pregnancy.
Collapse
Affiliation(s)
- Michelle Myers
- Obstetrics and Gynaecology, The Queen's Medical Research Institute Centre for Reproductive Biology, 47 Little France Crescent, Edinburgh, Scotland, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
32
|
James DP, Eastwood H, Richardson RT, O’Leary SJ. Effects of Round Window Dexamethasone on Residual Hearing in a Guinea Pig Model of Cochlear Implantation. Audiol Neurootol 2007; 13:86-96. [DOI: 10.1159/000111780] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 07/27/2007] [Indexed: 12/20/2022] Open
|
33
|
Sivertson KL, Seeds MC, Long DL, Peachman KK, Bass DA. The differential effect of dexamethasone on granulocyte apoptosis involves stabilization of Mcl-1L in neutrophils but not in eosinophils. Cell Immunol 2007; 246:34-45. [PMID: 17573055 PMCID: PMC2213750 DOI: 10.1016/j.cellimm.2007.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 05/03/2007] [Accepted: 05/04/2007] [Indexed: 10/23/2022]
Abstract
In the absence of activation signals, circulating human neutrophils and eosinophils undergo spontaneous apoptosis. The glucocorticoid dexamethasone (Dex) accelerates apoptosis in inflammatory cells such as eosinophils, but uniquely delays neutrophil apoptosis. Corresponding to the opposite effects of Dex on granulocyte apoptosis, we demonstrate that in neutrophils and eosinophils Dex oppositely affects expression of the anti-apoptotic Bcl-2 family protein Mcl-1L. Mcl-1L expression declines over time in vitro; however, Dex maintains Mcl-1L expression in neutrophils. In contrast, Dex accelerates Mcl-1L protein loss in eosinophils. Neither Mcl-1S, a pro-apoptotic splice variant, nor Bax were affected. Dex treatment in the presence of a translation inhibitor stabilized existing Mcl-1L protein in neutrophils, while Mcl-1L stability in eosinophils was unaffected. Accordingly, delay of neutrophil apoptosis by Dex was prevented by antisense Mcl-1L siRNA. Our findings suggest that regulation of Mcl-1L degradation plays an important role in the opposite effects of Dex on granulocyte apoptosis.
Collapse
Affiliation(s)
- Kelly L. Sivertson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michael C. Seeds
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- *Corresponding author: Michael C. Seeds, PhD, Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1054. Tel: 336-716-9811; Fax: 336-716-1214; E-mail:
| | - David L. Long
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kristina K. Peachman
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - David A. Bass
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Section on Pulmonary and Critical Care Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
34
|
|
35
|
Herr I, Gassler N, Friess H, Büchler MW. Regulation of differential pro- and anti-apoptotic signaling by glucocorticoids. Apoptosis 2007; 12:271-291. [PMID: 17191112 DOI: 10.1007/s10495-006-0624-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
More than a quarter of a century ago, the phenomenon of glucocorticoid-induced apoptosis in the majority of hematological cells was first recognized. More recently, glucocorticoid-induced antiapoptotic signaling associated with apoptosis resistance has been identified in cells of epithelial origin, most of malignant solid tumors and some other tissues. Despite these huge amount of data demonstrating differential pro- and anti-apoptotic effects of glucocorticoids, the underlying mechanisms of cell type specific glucocorticoid signaling are just beginning to be described. This review summarizes our present understanding of cell type-specific pro- and anti-apoptotic signaling induced by glucocorticoids. In the first section we give a summary and update of known glucocorticoid-induced pathways mediating apoptosis in hematological cells. We shortly introduce mechanisms of glucocorticoid resistance of hematological cells. We highlight and discuss the emerging molecular evidence of a general induction of survival signaling in epithelial cells and carcinoma cells by glucocorticoids. We provide a model for glucocorticoid-induced resistance in cells growing in a tissue formation. Thus, attachment to the extracellular matrix and cell-cell contacts typical for e.g. epithelial and tumor cells may be crucially involved in switching the balance of several interacting pathways to survival upon treatment with glucocorticoids.
Collapse
Affiliation(s)
- Ingrid Herr
- Department of Surgery, University of Heidelberg, Heidelberg, Germany.
| | | | | | | |
Collapse
|
36
|
Gabai G, Mollo A, Marinelli L, Badan M, Bono G. Endocrine and Ovarian Responses to Prolonged Adrenal Stimulation at the Time of Induced Corpus Luteum Regression. Reprod Domest Anim 2006; 41:485-93. [PMID: 17107506 DOI: 10.1111/j.1439-0531.2006.00697.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The endocrine and ovarian responses to prolonged adrenal stimulation at the time of corpus luteum (CL) regression were studied in non-lactating non-pregnant Friesian cows. Cows were synchronized with two cloprostenol (PG) injections 11 days apart (second PG referred as time 0). Experiment 1 was carried out on five animals in two phases with a resting period in between. Between -48 and 84 h, animals received 12 injections of either saline (CTR) or adrenocorticotrophic hormone (ACTH) agonist (Synacthen; SYN) every 12 h. Cortisol (C), progesterone (P4), oestradiol (E2) and LH were analysed in the blood samples collected every 8-12 h between days -3 and 4. Pulsatile LH release was studied 4 h before and 4 h after naloxone administration beginning at 96 h. Experiment 2 was carried out on four cows in a cross-over experimental design (two phases, with a resting period in between). Treatments were performed by administering either saline (CTR) or Synacthen (SYN) every 12 h between -36 and 24 h. The concentrations of C, P4 and E2 were measured in blood plasma every 4-12 h from days -3 to 3, then every day from days 5 to 9. In both experiments, ovaries were examined by ultrasonography every 1-3 days. ACTH administration induced a significant increase (p < 0.001) of plasma C lasting for 7 days (experiment 1), and for 3-4 days (experiment 2). Plasma C returned to baseline levels within 6 days (expt 1) or 36 h (expt 2) after treatment interruption. During the SYN phase, LH pre-ovulatory surge was not detectable. During the CTR phase, naloxone administration induced a significant increase (p < 0.05) of average LH concentrations that was not evident during the SYN phase. The dominant follicle development was retarded and mean plasma E2 concentrations were significantly lower during the SYN phase (p < 0.01). Luteolysis was completed within 2 days. However, P4 decline between 0 and 4 h was slower (p < 0.01) during the SYN phase. Our results indicate that, under prolonged adrenal stimulation, follicular development is delayed and LH release is impaired, which are independent of CL function.
Collapse
Affiliation(s)
- G Gabai
- Dipartimento di Scienze Sperimentali Veterinarie, Università di Padova, Legnaro (PD), Italy.
| | | | | | | | | |
Collapse
|
37
|
Hernandez-Pigeon H, Jean C, Charruyer A, Haure MJ, Titeux M, Tonasso L, Quillet-Mary A, Baudouin C, Charveron M, Laurent G. Human keratinocytes acquire cellular cytotoxicity under UV-B irradiation. Implication of granzyme B and perforin. J Biol Chem 2006; 281:13525-13532. [PMID: 16524880 DOI: 10.1074/jbc.m512694200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ultraviolet (UV) radiation from the sun is widely considered as a major cause of human skin photoaging and skin cancer. Granzyme B (GrB) and perforin (PFN) are two proteins contained in granules and implicated in one of the mechanisms by which cytotoxic lymphocytes and natural killer cells exert their cytotoxicity against virus-infected, alloreactive, or transformed cells. The distribution of GrB and PFN in the skin has received little attention. However, Berthou and co-workers (Berthou, C., Michel, L., Soulie, A., Jean-Louis, F., Flageul, B., Dubertret, L., Sigaux, F., Zhang, Y., and Sasportes, M. (1997) J. Immunol. 159, 5293-5300) described that, whereas freshly isolated epidermal cells did not express GrB or PFN, keratinocyte growth to confluence was associated with GrB and PFN mRNA and protein synthesis. In this work, we have investigated the possible role of UV-B on GrB and PFN expression in keratinocytes. We found that UV-B induces GrB and PFN expression in these cells through redox-, epidermal growth factor receptor-, and mitogen-activated protein kinase-dependent signaling. Furthermore, under UV irradiation, keratinocytes acquire a significant cytotoxicity, which is GrB and PFN dependent, toward a variety of cellular targets including transformed T-lymphocytes, melanocytes, and keratinocytes. This phenomenon may have important functional consequences in the regulation of skin inflammatory response and in the emergence of cancer skin.
Collapse
Affiliation(s)
- Hélène Hernandez-Pigeon
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France.
| | - Christine Jean
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France
| | - Alexandra Charruyer
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France
| | - Marie-José Haure
- CERPER, Institut de Recherche Pierre Fabre, Laboratoire de Biologie Cellulaire Cutanée, Toulouse, France
| | - Matthias Titeux
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France
| | - Laure Tonasso
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France
| | - Anne Quillet-Mary
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France
| | - Caroline Baudouin
- CERPER, Institut de Recherche Pierre Fabre, Laboratoire de Biologie Cellulaire Cutanée, Toulouse, France
| | - Marie Charveron
- CERPER, Institut de Recherche Pierre Fabre, Laboratoire de Biologie Cellulaire Cutanée, Toulouse, France
| | - Guy Laurent
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr. Baylac, Centre Hospitalier Universitaire Purpan, BP 3028, 31024 Toulouse cedex 3, France; Service d'Hématologie, Centre Hospitalier Universitaire Purpan, 31059 Toulouse, France
| |
Collapse
|
38
|
Guilloton F, de Thonel A, Jean C, Demur C, Mansat-De Mas V, Laurent G, Quillet-Mary A. TNFα stimulates NKG2D-mediated lytic activity of acute myeloid leukemic cells. Leukemia 2005; 19:2206-14. [PMID: 16239914 DOI: 10.1038/sj.leu.2403952] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mechanism by which leukemic cells interfere with normal hematopoiesis remains unclear. We show here that, whereas the leukemic KG1a cells are naturally devoid from cellular cytotoxicity, once activated by TNFalpha, they display cytolytic activity toward various cellular targets including CFU-GM. This mechanism is dependent on stimulation of the granzyme B/perforin system. In addition, KG1a cells expressed the NKG2D receptor and its signal-transducing adaptator DAP 10, which were functional as confirmed by redirected lysis experiments. Interestingly, flow cytometry analysis of 20 samples of patients with acute myeloid leukemia (AML) (FAB M0-M5) revealed the expression of NKG2D (40%) and other natural cytotoxicity receptors (40% for NKp30, 74% for NKp44, 39% for NKp46) by a pool >15% of leukemic cells. Furthermore, CD34+ hematopoietic progenitors undergoing granulomonocytic differentiation expressed NKG2D ligands. Altogether, we propose a model in which, upon stimulation by TNFalpha, leukemic cells may exert cytotoxicity against myeloid progenitors. This finding may have important clinical implications in the context of diseases characterized by TNFalpha accumulation, such as AML or myelodisplasic syndromes.
Collapse
Affiliation(s)
- F Guilloton
- INSERMU563/CPTP, Pavillon Lefebvre Bât B, Dpt G DELSOL, Equipe G LAURENT, CHU Purpan, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
39
|
Wada T, Pippin JW, Marshall CB, Griffin SV, Shankland SJ. Dexamethasone Prevents Podocyte Apoptosis Induced by Puromycin Aminonucleoside: Role of p53 and Bcl-2–Related Family Proteins. J Am Soc Nephrol 2005; 16:2615-25. [PMID: 15987750 DOI: 10.1681/asn.2005020142] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Nephrotic-range proteinuria is due to glomerular diseases characterized by podocyte injury. Glucocorticoids are the standard of care for most forms of nephrotic syndrome. However, the precise mechanisms underlying the beneficial effects of glucocorticoids on podocytes, beyond its general immunosuppressive and anti-inflammatory effects, are still unknown. This study tested the hypothesis that the synthetic glucocorticoid dexamethasone directly reduces podocyte apoptosis. Growth-restricted immortalized mouse podocytes in culture were exposed to puromycin aminonucleoside (PA) to induce apoptosis. Our results showed that dexamethasone significantly reduced PA-induced apoptosis by 2.81-fold. Dexamethasone also rescued podocyte viability when exposed to PA. PA-induced apoptosis was associated with increased p53 expression, which was completely blocked by dexamethasone. Furthermore, the inhibition of p53 by the p53 inhibitor pifithrin-alpha protected against PA-induced apoptosis. Dexamethasone also lowered the increase in the proapoptotic Bax, which was increased by PA, and increased expression of the antiapoptotic Bcl-xL protein. Moreover, the decrease in p53 by dexamethasone was associated with increased Bcl-xL levels. Podocyte apoptosis induced by PA was caspase-3 independent but was associated with the translocation of apoptosis-inducing factor (AIF) from the cytoplasm to nuclei. AIF translocation was inhibited by dexamethasone. These results show that PA-induced podocyte apoptosis is p53 dependent and associated with changes in Bcl-2-related proteins and AIF translocation. The protective effects of dexamethasone on PA-induced apoptosis were associated with decreasing p53, increasing Bcl-xL, and inhibition of AIF translocation. These novel findings provide new insights into the beneficial effects of corticosteroids on podocytes directly, independent of its immunosuppressive effects.
Collapse
Affiliation(s)
- Takehiko Wada
- Division of Nephrology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
40
|
Burton JL, Madsen SA, Chang LC, Weber PSD, Buckham KR, van Dorp R, Hickey MC, Earley B. Gene expression signatures in neutrophils exposed to glucocorticoids: A new paradigm to help explain “neutrophil dysfunction” in parturient dairy cows. Vet Immunol Immunopathol 2005; 105:197-219. [PMID: 15808301 DOI: 10.1016/j.vetimm.2005.02.012] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Neutrophils are the first line of immunity against most pathogens that infect cattle. These normally short-lived white blood cells develop from myeloid-lineage cells in bone marrow. Upon maturation, bone marrow neutrophils are released into the circulation where they marginate on inflamed blood vessel endothelial cells and migrate through them into the area of infection. Once migrated, neutrophils do not reenter the circulation, but rather, perform their bactericidal functions and die by apoptosis in the tissue. The cytokine and hormonal milieu of the blood and extracellular tissue fluid can influence neutrophil development and immunity-related activities, but the molecular basis of these phenotypic changes and physiological benefits or drawbacks of them are poorly understood. In the current paper, we review new gene expression information that resulted from two of our functional genomics studies designed to evaluate effects of glucocorticoid hormones on bovine neutrophils. This work provides one model to describe complex changes that occur in neutrophils as the cells respond to glucocorticoids, which might act to alter the cells' functional priorities and tip the delicate balance between health and disease during stress, including at parturition. A bovine immunobiology microarray and real time RT-PCR were used to study blood neutrophils collected during the natural surge of endogenous glucocorticoid (cortisol) in parturient dairy cows and bone marrow neutrophils collected from glucocorticoid (dexamethasone)-treated dairy steers. The gene expression signatures we observed led us to perform additional phenotyping of the neutrophils and correlation analyses, which together painted a picture suggesting that glucocorticoids have key roles in modulating neutrophil development, life span, and tissue defense functions during parturition and hormone therapy. Based on these observations, we postulate that glucocorticoids orchestrate adaptive changes in the entire neutrophil system that support increased cell numbers and longevity in blood and heightened remodeling activity in tissues, while at the same time decreasing some important antimicrobial defense activities of the cells. Thus, our functional genomics studies have enabled us to elucidate multiple consequences of neutrophil exposure to glucocorticoids, highlighting a probable role for this interaction in the induction of parturition and partly explaining why some parturient dairy cows may experience heightened incidence and severity of inflammatory diseases like mastitis.
Collapse
Affiliation(s)
- Jeanne L Burton
- Immunogenetics Laboratory, Department of Animal Science, Michigan State University, 1205E Anthony Hall, East Lansing, MI 48824, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ngezahayo A, Altmann B, Steffens M, Kolb HA. Gap Junction Coupling and Apoptosis in GFSHR-17 Granulosa Cells. J Membr Biol 2005; 204:137-44. [PMID: 16245036 DOI: 10.1007/s00232-005-0756-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2005] [Revised: 06/10/2005] [Indexed: 11/30/2022]
Abstract
Recently, we found that intracellular washout of cGMP induces gap junction uncoupling and proposed a link between gap junction uncoupling and stimulation of apoptotic reactions in GFSHR-17 granulosa cells. In the present report we show that an inhibitor of guanylyl cyclase, ODQ, reduces gap junction coupling and promotes apoptotic reactions such as chromatin condensation and DNA strand breaks. To analyze whether gap junction uncoupling and induction of apoptotic reactions are related, the cells were treated with heptanol and 18 beta-GA, two known gap junction uncouplers. Gap junction coupling of GFSHR-17 cells could be restored if the incubation time with the gap junction uncouplers was less than 10 min. A prolonged incubation time irreversibly suppressed gap junction coupling and caused chromatin condensation as well as DNA degradation. The promotion of apoptotic reactions by heptanol or 18 beta-GA was not observed in cells with low gap junction coupling like HeLa cells, indicating that the observed genotoxic reactions are not caused by unspecific effects of gap junction uncouplers. Additionally, it was observed that heptanol or 18 beta-GA did not induce a sustained rise of [Ca(2+)](i). The effects of gap junction uncouplers could not be suppressed by the presence of 8-Br-cGMP. It is discussed that irreversible gap junction uncoupling can be mediated by cGMP-dependent as well as cGMP-independent pathways and in turn could lead to stimulation of apoptotic reactions in granulosa cells.
Collapse
Affiliation(s)
- A Ngezahayo
- Institute of Biophysics, University Hannover, Herrenhäuserstr. 2, D-30419 Hannover, Germany.
| | | | | | | |
Collapse
|
42
|
Wu W, Pew T, Zou M, Pang D, Conzen SD. Glucocorticoid receptor-induced MAPK phosphatase-1 (MPK-1) expression inhibits paclitaxel-associated MAPK activation and contributes to breast cancer cell survival. J Biol Chem 2004; 280:4117-24. [PMID: 15590693 DOI: 10.1074/jbc.m411200200] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucocorticoid receptor (GR) activation has recently been shown to inhibit apoptosis in breast epithelial cells. We have previously described a group of genes that is rapidly up-regulated in these cells following dexamethasone (Dex) treatment. In an effort to dissect the mechanisms of GR-mediated breast epithelial cell survival, we now examine the molecular events downstream of GR activation. Here we show that GR activation leads to both the rapid induction of MAPK phosphatase-1 (MKP-1) mRNA and its sustained expression. Induction of the MKP-1 protein in the MCF10A-Myc and MDA-MB-231 breast epithelial cell lines was also seen. Paclitaxel treatment resulted in MAPK activation and apoptosis of MDA-MB-231 breast cancer cells, and both processes were inhibited by Dex pretreatment. Furthermore, induction of MKP-1 correlated with the inhibition of extracellular signal-regulated kinase (ERK1/2) and c-Jun N-terminal kinase (JNK) activity, whereas p38 activity was minimally affected. Blocking Dex-induced MKP-1 induction using small interfering RNA increased ERK1/2 and JNK phosphorylation and decreased cell survival. ERK1/2 and JNK inactivation was associated with Ets-like transcription factor-1 (ELK-1) dephosphorylation. To explore the gene expression changes that occur downstream of ELK-1 dephosphorylation, we used a combination of temporal gene expression data and promoter element analyses. This approach revealed a previously unrecognized transcriptional target of ELK-1, the human tissue plasminogen activator (tPA). We verified the predicted ELK-1--> tPA transcriptional regulatory relationship using a luciferase reporter assay. We conclude that GR-mediated MAPK inactivation contributes to cell survival and that the potential transcriptional targets of this inhibition can be identified from large scale gene array analysis.
Collapse
MESH Headings
- Amino Acid Motifs
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis
- Blotting, Northern
- Blotting, Western
- Breast/metabolism
- Cell Cycle Proteins/metabolism
- Cell Cycle Proteins/physiology
- Cell Line, Tumor
- Cell Survival
- DNA-Binding Proteins/biosynthesis
- Down-Regulation
- Dual Specificity Phosphatase 1
- Enzyme Activation
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Humans
- Immediate-Early Proteins/metabolism
- Immediate-Early Proteins/physiology
- JNK Mitogen-Activated Protein Kinases/biosynthesis
- JNK Mitogen-Activated Protein Kinases/metabolism
- Luciferases/metabolism
- MAP Kinase Kinase 4
- MAP Kinase Signaling System
- Mitogen-Activated Protein Kinase 1/biosynthesis
- Mitogen-Activated Protein Kinase 3/biosynthesis
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Mitogen-Activated Protein Kinases/metabolism
- Models, Biological
- Oligonucleotide Array Sequence Analysis
- Paclitaxel/pharmacology
- Phosphoprotein Phosphatases/metabolism
- Phosphoprotein Phosphatases/physiology
- Phosphorylation
- Protein Binding
- Protein Phosphatase 1
- Protein Tyrosine Phosphatases/metabolism
- Protein Tyrosine Phosphatases/physiology
- Proto-Oncogene Proteins/biosynthesis
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Receptors, Glucocorticoid/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
- Tissue Plasminogen Activator/metabolism
- Transcription Factors/biosynthesis
- Transcription, Genetic
- Transfection
- ets-Domain Protein Elk-1
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine and the Committee on Cancer Biology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | |
Collapse
|