1
|
Berisha B, Pfaffl MW, Thaqi G. Local Regulatory Changes of HSD11B1 and NR3C1 in the Follicular and Luteal Tissue During Experimentally Induced Ovulation in the Cow. Reprod Domest Anim 2024; 59:e14722. [PMID: 39295165 DOI: 10.1111/rda.14722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024]
Abstract
The objective of the study was to characterise the expression patterns of the two key components of cortisol action namely HSD11B1 (11-beta-hydroxysteroid dehydrogenase type 1) and NR3C1 (nuclear receptor subfamily 3, group C, member 1, also known as the glucocorticoid receptor) in superovulation induced bovine follicles during the periovulation and subsequent corpus luteum (CL) formation. Bovine ovaries containing preovulatory follicles or CL were timely defined during induced ovulation as follows: 0 h before GnRH (Gonadotropin-releasing hormone) application, and 4, 10, 20, 25 (follicles) and 60 h (early CL) after GnRH. The low mRNA expression of HSD11B1 and NR3C1 in the follicle group before the GnRH application increased significantly in the follicle group 20 h after GnRH and remained high afterward also in the early CL group. In contrast, the high NR3C1 mRNA decreased in follicles 25 h after GnRH (close to ovulation) and significantly increased again after ovulation (early CL). Our results indicated the involvement of HSD11B1 and NR3C1 as the two key components of cortisol action in the local mechanisms coordinating final follicle maturation, ovulation, follicular-luteal transition and CL development in the cow.
Collapse
Affiliation(s)
- Bajram Berisha
- Chair of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Munich, Germany
- Department of Animal Biotechnology, Faculty of Agriculture and Veterinary, University of Prishtina, Prishtina, Kosovo
- Academy of Sciences of Albania, Tirana, Albania
| | - Michael W Pfaffl
- Chair of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Granit Thaqi
- Chair of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Chilakamarthi U, Mahadik NS, Bhattacharyya T, Gangadhar PS, Giribabu L, Banerjee R. Glucocorticoid receptor mediated sensitization of colon cancer to photodynamic therapy induced cell death. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 251:112846. [PMID: 38237432 DOI: 10.1016/j.jphotobiol.2024.112846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/30/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024]
Abstract
Photodynamic therapy (PDT) is a clinically approved, non-invasive alternate cancer therapy. A synthetic glucocorticoid (GC), dexamethasone (Dex) has previously been demonstrated to sensitize cancer cells to chemotherapy. However, to the best of our knowledge, the sensitization effect of GCs on PDT has not yet been investigated. We hypothesized that glucocorticoid receptor (GR) targeting can selectively make cancer cells more sensitive to PDT treatment, as PDT induces hypoxia wherein GR-activity gets enhanced. In addition, Dex was reported to act against the PDT-induced cell survival pathways like HIF-1α, NRF2, NF-κB, STAT3 etc. Thus, both the treatments can complement each other and may result in increasing the effectiveness of combination therapy. Hence, in this study, we developed liposomal formulations of our previously reported PDT agent P-Nap, either alone (D1P-Nap) or in combination with Dex (D1XP-Nap) to elucidate the sensitization effect. Interestingly, our RT-PCR results in hypoxic conditions showed down-regulation of HIF-1α and over expression of GR-activated genes for glucose-6-phosphatase (G6Pase) and PEPCK enzymes, indicating prominent GR-transactivation. We also observed higher phototoxicity in CT26.WT cells treated with D1XP-Nap PDT under hypoxic conditions as compared to normoxic conditions. These effects were reversed when cells were pre-treated with RU486, a competitive inhibitor of GCs. Moreover, our in vivo findings of subcutaneous tumor model of Balb/C mice for colon cancer revealed a significant decrease in tumor volume as well as considerable enhancement in the survivability of PDT treated tumor-bearing mice when Dex was present in the formulation. A high Bax/Bcl-xL ratio, high p53 expression, enhanced E-cadherin expression and down-regulation of pro-tumorigenic transcription factors NF-κB and c-Myc were found in tumor lysates from mice treated with D1XP-Nap under PDT, indicating GR-mediated sensitization of the tumor to PDT-induced cell death and enhancement of life-span for tumor bearing mice.
Collapse
Affiliation(s)
- Ushasri Chilakamarthi
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Namita S Mahadik
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India
| | - Tithi Bhattacharyya
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India
| | - Palivela Siva Gangadhar
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India; Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Lingamallu Giribabu
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India; Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Rajkumar Banerjee
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India.
| |
Collapse
|
3
|
Baranova K, Nalivaeva N, Rybnikova E. Neuroadaptive Biochemical Mechanisms of Remote Ischemic Conditioning. Int J Mol Sci 2023; 24:17032. [PMID: 38069355 PMCID: PMC10707673 DOI: 10.3390/ijms242317032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
This review summarizes the currently known biochemical neuroadaptive mechanisms of remote ischemic conditioning. In particular, it focuses on the significance of the pro-adaptive effects of remote ischemic conditioning which allow for the prevention of the neurological and cognitive impairments associated with hippocampal dysregulation after brain damage. The neuroimmunohumoral pathway transmitting a conditioning stimulus, as well as the molecular basis of the early and delayed phases of neuroprotection, including anti-apoptotic, anti-oxidant, and anti-inflammatory components, are also outlined. Based on the close interplay between the effects of ischemia, especially those mediated by interaction of hypoxia-inducible factors (HIFs) and steroid hormones, the involvement of the hypothalamic-pituitary-adrenocortical system in remote ischemic conditioning is also discussed.
Collapse
Affiliation(s)
| | | | - Elena Rybnikova
- I. P. Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia; (K.B.); (N.N.)
| |
Collapse
|
4
|
Park HY, van Bruggen VLE, Peutz-Kootstra CJ, Ophelders DRMG, Jellema RK, Reutelingsperger CPM, Rutten BPF, Wolfs TGAM. Time Dependent Changes in the Ovine Neurovascular Unit; A Potential Neuroprotective Role of Annexin A1 in Neonatal Hypoxic-Ischemic Encephalopathy. Int J Mol Sci 2023; 24:ijms24065929. [PMID: 36983004 PMCID: PMC10054605 DOI: 10.3390/ijms24065929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Perinatal brain injury following hypoxia-ischemia (HI) is characterized by high mortality rates and long-term disabilities. Previously, we demonstrated that depletion of Annexin A1, an essential mediator in BBB integrity, was associated with a temporal loss of blood-brain barrier (BBB) integrity after HI. Since the molecular and cellular mechanisms mediating the impact of HI are not fully scrutinized, we aimed to gain mechanistic insight into the dynamics of essential BBB structures following global HI in relation to ANXA1 expression. Global HI was induced in instrumented preterm ovine fetuses by transient umbilical cord occlusion (UCO) or sham occlusion (control). BBB structures were assessed at 1, 3, or 7 days post-UCO by immunohistochemical analyses of ANXA1, laminin, collagen type IV, and PDGFRβ for pericytes. Our study revealed that within 24 h after HI, cerebrovascular ANXA1 was depleted, which was followed by depletion of laminin and collagen type IV 3 days after HI. Seven days post-HI, increased pericyte coverage, laminin and collagen type IV expression were detected, indicating vascular remodeling. Our data demonstrate novel mechanistic insights into the loss of BBB integrity after HI, and effective strategies to restore BBB integrity should potentially be applied within 48 h after HI. ANXA1 has great therapeutic potential to target HI-driven brain injury.
Collapse
Affiliation(s)
- Hyun Young Park
- Department of Pediatrics, School of Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Valéry L E van Bruggen
- Department of Pediatrics, School of Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | | | - Daan R M G Ophelders
- Department of Pediatrics, School of Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Reint K Jellema
- Department of Pediatrics, School of Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Pediatrics, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Chris P M Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Tim G A M Wolfs
- Department of Pediatrics, School of Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
5
|
Haftorn KL, Denault WRP, Lee Y, Page CM, Romanowska J, Lyle R, Næss ØE, Kristjansson D, Magnus PM, Håberg SE, Bohlin J, Jugessur A. Nucleated red blood cells explain most of the association between DNA methylation and gestational age. Commun Biol 2023; 6:224. [PMID: 36849614 PMCID: PMC9971030 DOI: 10.1038/s42003-023-04584-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Determining if specific cell type(s) are responsible for an association between DNA methylation (DNAm) and a given phenotype is important for understanding the biological mechanisms underlying the association. Our EWAS of gestational age (GA) in 953 newborns from the Norwegian MoBa study identified 13,660 CpGs significantly associated with GA (pBonferroni<0.05) after adjustment for cell type composition. When the CellDMC algorithm was applied to explore cell-type specific effects, 2,330 CpGs were significantly associated with GA, mostly in nucleated red blood cells [nRBCs; n = 2,030 (87%)]. Similar patterns were found in another dataset based on a different array and when applying an alternative algorithm to CellDMC called Tensor Composition Analysis (TCA). Our findings point to nRBCs as the main cell type driving the DNAm-GA association, implicating an epigenetic signature of erythropoiesis as a likely mechanism. They also explain the poor correlation observed between epigenetic age clocks for newborns and those for adults.
Collapse
Affiliation(s)
- Kristine L Haftorn
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway.
- Institute of Health and Society, University of Oslo, Oslo, Norway.
| | - William R P Denault
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Yunsung Lee
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Christian M Page
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Physical Health and Ageing, Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Julia Romanowska
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health and Primary Care, , University of Bergen, Bergen, Norway
| | - Robert Lyle
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Øyvind E Næss
- Institute of Health and Society, University of Oslo, Oslo, Norway
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Dana Kristjansson
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Oslo, Norway
| | - Per M Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Siri E Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jon Bohlin
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Division for Infection Control and Environmental Health, Department of Infectious Disease Epidemiology and Modelling, Norwegian Institute of Public Health, Oslo, Norway
| | - Astanand Jugessur
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health and Primary Care, , University of Bergen, Bergen, Norway
| |
Collapse
|
6
|
Inflammation in Pulmonary Hypertension and Edema Induced by Hypobaric Hypoxia Exposure. Int J Mol Sci 2022; 23:ijms232012656. [PMID: 36293512 PMCID: PMC9604159 DOI: 10.3390/ijms232012656] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/06/2022] Open
Abstract
Exposure to high altitudes generates a decrease in the partial pressure of oxygen, triggering a hypobaric hypoxic condition. This condition produces pathophysiologic alterations in an organism. In the lung, one of the principal responses to hypoxia is the development of hypoxic pulmonary vasoconstriction (HPV), which improves gas exchange. However, when HPV is exacerbated, it induces high-altitude pulmonary hypertension (HAPH). Another important illness in hypobaric hypoxia is high-altitude pulmonary edema (HAPE), which occurs under acute exposure. Several studies have shown that inflammatory processes are activated in high-altitude illnesses, highlighting the importance of the crosstalk between hypoxia and inflammation. The aim of this review is to determine the inflammatory pathways involved in hypobaric hypoxia, to investigate the key role of inflammation in lung pathologies, such as HAPH and HAPE, and to summarize different anti-inflammatory treatment approaches for these high-altitude illnesses. In conclusion, both HAPE and HAPH show an increase in inflammatory cell infiltration (macrophages and neutrophils), cytokine levels (IL-6, TNF-α and IL-1β), chemokine levels (MCP-1), and cell adhesion molecule levels (ICAM-1 and VCAM-1), and anti-inflammatory treatments (decreasing all inflammatory components mentioned above) seem to be promising mitigation strategies for treating lung pathologies associated with high-altitude exposure.
Collapse
|
7
|
Jinna N, Rida P, Smart M, LaBarge M, Jovanovic-Talisman T, Natarajan R, Seewaldt V. Adaptation to Hypoxia May Promote Therapeutic Resistance to Androgen Receptor Inhibition in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23168844. [PMID: 36012111 PMCID: PMC9408190 DOI: 10.3390/ijms23168844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) surpasses other BC subtypes as the most challenging to treat due to its lack of traditional BC biomarkers. Nearly 30% of TNBC patients express the androgen receptor (AR), and the blockade of androgen production and AR signaling have been the cornerstones of therapies for AR-positive TNBC. However, the majority of women are resistant to AR-targeted therapy, which is a major impediment to improving outcomes for the AR-positive TNBC subpopulation. The hypoxia signaling cascade is frequently activated in the tumor microenvironment in response to low oxygen levels; activation of the hypoxia signaling cascade allows tumors to survive despite hypoxia-mediated interference with cellular metabolism. The activation of hypoxia signaling networks in TNBC promotes resistance to most anticancer drugs including AR inhibitors. The activation of hypoxia network signaling occurs more frequently in TNBC compared to other BC subtypes. Herein, we examine the (1) interplay between hypoxia signaling networks and AR and (2) whether hypoxia and hypoxic stress adaptive pathways promote the emergence of resistance to therapies that target AR. We also pose the well-supported question, “Can the efficacy of androgen-/AR-targeted treatments be enhanced by co-targeting hypoxia?” By critically examining the evidence and the complex entwinement of these two oncogenic pathways, we argue that the simultaneous targeting of androgen biosynthesis/AR signaling and hypoxia may enhance the sensitivity of AR-positive TNBCs to AR-targeted treatments, derail the emergence of therapy resistance, and improve patient outcomes.
Collapse
Affiliation(s)
- Nikita Jinna
- Department of Population Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Max Smart
- Rowland Hall, Salt Lake City, UT 84102, USA
| | - Mark LaBarge
- Department of Population Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Victoria Seewaldt
- Department of Population Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Correspondence:
| |
Collapse
|
8
|
Burtscher J, Niedermeier M, Hüfner K, van den Burg E, Kopp M, Stoop R, Burtscher M, Gatterer H, Millet GP. The interplay of hypoxic and mental stress: Implications for anxiety and depressive disorders. Neurosci Biobehav Rev 2022; 138:104718. [PMID: 35661753 DOI: 10.1016/j.neubiorev.2022.104718] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Adequate oxygen supply is essential for the human brain to meet its high energy demands. Therefore, elaborate molecular and systemic mechanism are in place to enable adaptation to low oxygen availability. Anxiety and depressive disorders are characterized by alterations in brain oxygen metabolism and of its components, such as mitochondria or hypoxia inducible factor (HIF)-pathways. Conversely, sensitivity and tolerance to hypoxia may depend on parameters of mental stress and the severity of anxiety and depressive disorders. Here we discuss relevant mechanisms of adaptations to hypoxia, as well as their involvement in mental stress and the etiopathogenesis of anxiety and depressive disorders. We suggest that mechanisms of adaptations to hypoxia (including metabolic responses, inflammation, and the activation of chemosensitive brain regions) modulate and are modulated by stress-related pathways and associated psychiatric diseases. While severe chronic hypoxia or dysfunctional hypoxia adaptations can contribute to the pathogenesis of anxiety and depressive disorders, harnessing controlled responses to hypoxia to increase cellular and psychological resilience emerges as a novel treatment strategy for these diseases.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| | - Martin Niedermeier
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Clinic for Psychiatry II, Innsbruck Medical University, Innsbruck, Austria
| | - Erwin van den Burg
- Department of Psychiatry, Center of Psychiatric Neuroscience (CNP), University Hospital of Lausanne (CHUV), Prilly, Lausanne, Switzerland
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Ron Stoop
- Department of Psychiatry, Center of Psychiatric Neuroscience (CNP), University Hospital of Lausanne (CHUV), Prilly, Lausanne, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Hannes Gatterer
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Vanderhaeghen T, Timmermans S, Watts D, Paakinaho V, Eggermont M, Vandewalle J, Wallaeys C, Van Wyngene L, Van Looveren K, Nuyttens L, Dewaele S, Vanden Berghe J, Lemeire K, De Backer J, Dirkx L, Vanden Berghe W, Caljon G, Ghesquière B, De Bosscher K, Wielockx B, Palvimo JJ, Beyaert R, Libert C. Reprogramming of glucocorticoid receptor function by hypoxia. EMBO Rep 2022; 23:e53083. [PMID: 34699114 PMCID: PMC8728616 DOI: 10.15252/embr.202153083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 01/07/2023] Open
Abstract
Here, we investigate the impact of hypoxia on the hepatic response of glucocorticoid receptor (GR) to dexamethasone (DEX) in mice via RNA-sequencing. Hypoxia causes three types of reprogramming of GR: (i) much weaker induction of classical GR-responsive genes by DEX in hypoxia, (ii) a number of genes is induced by DEX specifically in hypoxia, and (iii) hypoxia induces a group of genes via activation of the hypothalamic-pituitary-adrenal (HPA) axis. Transcriptional profiles are reflected by changed GR DNA-binding as measured by ChIP sequencing. The HPA axis is induced by hypothalamic HIF1α and HIF2α activation and leads to GR-dependent lipolysis and ketogenesis. Acute inflammation, induced by lipopolysaccharide, is prevented by DEX in normoxia but not during hypoxia, and this is attributed to HPA axis activation by hypoxia. We unfold new physiological pathways that have consequences for patients suffering from GC resistance.
Collapse
|
10
|
Homeostatic Regulation of Glucocorticoid Receptor Activity by Hypoxia-Inducible Factor 1: From Physiology to Clinic. Cells 2021; 10:cells10123441. [PMID: 34943949 PMCID: PMC8699886 DOI: 10.3390/cells10123441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs) represent a well-known class of lipophilic steroid hormones biosynthesised, with a circadian rhythm, by the adrenal glands in humans and by the inter-renal tissue in teleost fish (e.g., zebrafish). GCs play a key role in the regulation of numerous physiological processes, including inflammation, glucose, lipid, protein metabolism and stress response. This is achieved through binding to their cognate receptor, GR, which functions as a ligand-activated transcription factor. Due to their potent anti-inflammatory and immune-suppressive action, synthetic GCs are broadly used for treating pathological disorders that are very often linked to hypoxia (e.g., rheumatoid arthritis, inflammatory, allergic, infectious, and autoimmune diseases, among others) as well as to prevent graft rejections and against immune system malignancies. However, due to the presence of adverse effects and GC resistance their therapeutic benefits are limited in patients chronically treated with steroids. For this reason, understanding how to fine-tune GR activity is crucial in the search for novel therapeutic strategies aimed at reducing GC-related side effects and effectively restoring homeostasis. Recent research has uncovered novel mechanisms that inhibit GR function, thereby causing glucocorticoid resistance, and has produced some surprising new findings. In this review we analyse these mechanisms and focus on the crosstalk between GR and HIF signalling. Indeed, its comprehension may provide new routes to develop novel therapeutic targets for effectively treating immune and inflammatory response and to simultaneously facilitate the development of innovative GCs with a better benefits-risk ratio.
Collapse
|
11
|
Xu K, Lu C, Ren X, Wang J, Xu P, Zhang Y. Overexpression of HIF-1α enhances the protective effect of mitophagy on steroid-induced osteocytes apoptosis. ENVIRONMENTAL TOXICOLOGY 2021; 36:2123-2137. [PMID: 34310007 DOI: 10.1002/tox.23327] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Glucocorticoid (GC; dexamethasone, DEX) -induced osteonecrosis of the femoral head (GIOFH) is a challenging orthopedic disease, and its underlying mechanism remains not clear. This study exposed murine long bone osteocyte-Y4 (MLO-Y4) cells to DEX below normoxic or hypoxic circumstances and found that cell autophagy have been reduced. At the same time, flow cytometry analysis showed increased apoptosis, which was more pronounced in hypoxic environments. Recent research also claimed that GC induces osteoporosis after osteocyte apoptosis, and subsequent microfractures lead to ischemia and hypoxia of the femoral head, resulted in GIOFH. Presently, we found that both mitophagy-related protein hypoxia-inducible factor-1α (HIF-1α) and BNIP3 were up-regulated in the hypoxic environment, and their expression was down-regulated when exposed to DEX. Besides, we demonstrated that overexpressing HIF-1α resisted DEX-induced apoptosis in a hypoxic environment. Here, we demonstrated that overexpression of HIF-1α, through its downstream marker BNIP3, reduced the suppression of DEX on mitophagy induced by hypoxia and protected bone cells from apoptosis. Also, these findings may provide a direction of the promising application for better GIOFH treatment shortly.
Collapse
Affiliation(s)
- Ke Xu
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, China
| | - Chao Lu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyu Ren
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, China
| | - Peng Xu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yingang Zhang
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Liang L, Gu X, Shen HJ, Shi YH, Li Y, Zhang J, Chen YY, Chen ZH, Ma JY, Li QY. Chronic Intermittent Hypoxia Reduces the Effects of Glucosteroid in Asthma via Activating the p38 MAPK Signaling Pathway. Front Physiol 2021; 12:703281. [PMID: 34512379 PMCID: PMC8430218 DOI: 10.3389/fphys.2021.703281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/03/2021] [Indexed: 11/14/2022] Open
Abstract
Aims Obstructive sleep apnea (OSA) is a risk factor for steroid-resistant (SR) asthma. However, the underlying mechanism is not well defined. This study aimed to investigate how chronic intermittent hypoxia (CIH), the main pathophysiology of OSA, influenced the effects of glucocorticoids (GCs) on asthma. Main Methods The effects of dexamethasone (Dex) were determined using the ovalbumin (OVA)-challenged mouse model of asthma and transforming growth factor (TGF)-β treated airway smooth muscle cells (ASMCs), with or without CIH. The p38 MAPK signaling pathway activity was then detected in the mouse (n = 6) and ASMCs models (n = 6), which were both treated with the p38 MAPK inhibitor SB239063. Key Findings Under CIH, mouse pulmonary resistance value, inflammatory cells in bronchoalveolar lavage fluid (BALF), and inflammation scores increased in OVA-challenged combined with CIH exposure mice compared with OVA-challenged mice (p < 0.05). These indicators were similarly raised in the OVA + CIH + Dex group compared with the OVA + Dex group (P < 0.05). CIH exposure enhanced the activation of the p38 MAPK pathway, oxidative stress injury, and the expression of NF-κB both in lung tissue and ASMCs, which were reversed by treatment with Dex and SB239063. In the in vitro study, treatment with Dex and SB239063 decreased ASMCs proliferation induced by TGF-β combined with CIH and suppressed activation of the p38 MAPK pathway, oxidative stress injury, and NF-κB nuclear transcription (p < 0.05). Significance These results indicated that CIH decreased GC sensitivity by activating the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Li Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Gu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Ji Shen
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Heng Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Li
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Yan Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen He Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Yun Ma
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Vanderhaeghen T, Beyaert R, Libert C. Bidirectional Crosstalk Between Hypoxia Inducible Factors and Glucocorticoid Signalling in Health and Disease. Front Immunol 2021; 12:684085. [PMID: 34149725 PMCID: PMC8211996 DOI: 10.3389/fimmu.2021.684085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoid-induced (GC) and hypoxia-induced transcriptional responses play an important role in tissue homeostasis and in the regulation of cellular responses to stress and inflammation. Evidence exists that there is an important crosstalk between both GC and hypoxia effects. Hypoxia is a pathophysiological condition to which cells respond quickly in order to prevent metabolic shutdown and death. The hypoxia inducible factors (HIFs) are the master regulators of oxygen homeostasis and are responsible for the ability of cells to cope with low oxygen levels. Maladaptive responses of HIFs contribute to a variety of pathological conditions including acute mountain sickness (AMS), inflammation and neonatal hypoxia-induced brain injury. Synthetic GCs which are analogous to the naturally occurring steroid hormones (cortisol in humans, corticosterone in rodents), have been used for decades as anti-inflammatory drugs for treating pathological conditions which are linked to hypoxia (i.e. asthma, ischemic injury). In this review, we investigate the crosstalk between the glucocorticoid receptor (GR), and HIFs. We discuss possible mechanisms by which GR and HIF influence one another, in vitro and in vivo, and the therapeutic effects of GCs on HIF-mediated diseases.
Collapse
Affiliation(s)
- Tineke Vanderhaeghen
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
Shackleton GL. Towards a biochemical approach to occupational stress management. Heliyon 2021; 7:e07175. [PMID: 34141933 PMCID: PMC8187824 DOI: 10.1016/j.heliyon.2021.e07175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/04/2020] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Given the immense and growing cost of occupational stress to society through lost productivity and the burden to healthcare systems, current best practices for detecting, managing and reducing stress in the workplace are clearly sub-optimal and substantially better methods are required. Subjective, self-reported psychology and psychiatry-based instruments are prone to biases whereas current objective, biology-based measures produce conflicting results and are far from reliable. A multivariate approach to occupational stress research is required that reflects the broad, coordinated, physiological response to demands placed on the body by exposure to diverse occupational stressors. A literature review was conducted to determine the extent of application of the emerging multivariate technology of metabolomics to occupational stress research. Of 170 articles meeting the search criteria, three were identified that specifically studied occupational stressors using metabolomics. A further ten studies were not specifically occupational or were of indirect or peripheral relevance. The occupational studies, although limited in number highlight the technological challenges associated with the application of metabolomics to investigate occupational stress. They also demonstrate the utility to evaluate stress more comprehensively than univariate biomarker studies. The potential of this multivariate approach to enhance our understanding of occupational stress has yet to be established. This will require more studies with broader analytical coverage of the metabolome, longitudinal sampling, combination with experience sampling methods and comparison with psychometric models of occupational stress. Progress will likely involve combining multi-omic data into a holistic, systems biology approach to detecting, managing and reducing occupational stress and optimizing workplace performance.
Collapse
|
15
|
Hasan F, Chiu Y, Shaw RM, Wang J, Yee C. Hypoxia acts as an environmental cue for the human tissue-resident memory T cell differentiation program. JCI Insight 2021; 6:138970. [PMID: 34027895 PMCID: PMC8262358 DOI: 10.1172/jci.insight.138970] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/07/2021] [Indexed: 12/31/2022] Open
Abstract
Tissue-resident memory T cells (TRM) provide frontline defense against infectious diseases and contribute to antitumor immunity; however, aside from the necessity of TGF-β, knowledge regarding TRM-inductive cues remains incomplete, particularly for human cells. Oxygen tension is an environmental cue that distinguishes peripheral tissues from the circulation, and here, we demonstrate that differentiation of human CD8+ T cells in the presence of hypoxia and TGF-β1 led to the development of a TRM phenotype, characterized by a greater than 5-fold increase in CD69+CD103+ cells expressing human TRM hallmarks and enrichment for endogenous human TRM gene signatures, including increased adhesion molecule expression and decreased expression of genes involved in recirculation. Hypoxia and TGF-β1 synergized to produce a significantly larger population of TRM phenotype cells than either condition alone, and comparison of these cells from the individual and combination conditions revealed distinct phenotypic and transcriptional profiles, indicating a programming response to milieu rather than a mere expansion. Our findings identify a likely previously unreported cue for the TRM differentiation program and can enable facile generation of human TRM phenotype cells in vitro for basic studies and translational applications such as adoptive cellular therapy.
Collapse
Affiliation(s)
- Farah Hasan
- Department of Melanoma Medical Oncology, University of Texas (UT) MD Anderson Cancer Center, Houston, Texas, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Yulun Chiu
- Department of Melanoma Medical Oncology, University of Texas (UT) MD Anderson Cancer Center, Houston, Texas, USA
| | - Rebecca M Shaw
- Department of Melanoma Medical Oncology, University of Texas (UT) MD Anderson Cancer Center, Houston, Texas, USA
| | - Junmei Wang
- Department of Melanoma Medical Oncology, University of Texas (UT) MD Anderson Cancer Center, Houston, Texas, USA
| | - Cassian Yee
- Department of Melanoma Medical Oncology, University of Texas (UT) MD Anderson Cancer Center, Houston, Texas, USA.,Department of Immunology, UT MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Yang N, Berry A, Sauer C, Baxter M, Donaldson IJ, Forbes K, Donn R, Matthews L, Ray D. Hypoxia regulates GR function through multiple mechanisms involving microRNAs 103 and 107. Mol Cell Endocrinol 2020; 518:111007. [PMID: 32871225 PMCID: PMC7646191 DOI: 10.1016/j.mce.2020.111007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 11/06/2022]
Abstract
Glucocorticoids (Gcs) potently inhibit inflammation, and regulate liver energy metabolism, often acting in a hypoxic environment. We now show hypoxic conditions open a specific GR cistrome, and prevent access of GR to part of the normoxic GR cistrome. Motif analysis identified enrichment of KLF4 binding sites beneath those peaks of GR binding exclusive to normoxia, implicating KLF4 as a pioneer, or co-factor under these conditions. Hypoxia reduced KLF4 expression, however, knockdown of KLF4 did not impair GR recruitment. KLF4 is a known target of microRNAs 103 and 107, both of which are induced by hypoxia. Expression of mimics to either microRNA103, or microRNA107 inhibited GR transactivation of normoxic target genes, thereby replicating the hypoxic effect. Therefore, studies in hypoxia reveal that microRNAs 103 and 107 are potent regulators of GR function. We have now identified a new pathway linking hypoxia through microRNAs 103 and 107 to regulation of GR function.
Collapse
Affiliation(s)
- Nan Yang
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX37LE, UK
| | - Andrew Berry
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Carolin Sauer
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX37LE, UK
| | - Matthew Baxter
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX37LE, UK
| | - Ian J Donaldson
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Karen Forbes
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Rachelle Donn
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Laura Matthews
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK.
| | - David Ray
- Faculty of Biology, Medicine, and Health, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX37LE, UK.
| |
Collapse
|
17
|
Marchi D, Santhakumar K, Markham E, Li N, Storbeck KH, Krone N, Cunliffe VT, van Eeden FJM. Bidirectional crosstalk between Hypoxia-Inducible Factor and glucocorticoid signalling in zebrafish larvae. PLoS Genet 2020; 16:e1008757. [PMID: 32379754 PMCID: PMC7237044 DOI: 10.1371/journal.pgen.1008757] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/19/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
In the last decades in vitro studies highlighted the potential for crosstalk between Hypoxia-Inducible Factor-(HIF) and glucocorticoid-(GC) signalling pathways. However, how this interplay precisely occurs in vivo is still debated. Here, we use zebrafish larvae (Danio rerio) to elucidate how and to what degree hypoxic signalling affects the endogenous glucocorticoid pathway and vice versa, in vivo. Firstly, our results demonstrate that in the presence of upregulated HIF signalling, both glucocorticoid receptor (Gr) responsiveness and endogenous cortisol levels are repressed in 5 days post fertilisation larvae. In addition, despite HIF activity being low at normoxia, our data show that it already impedes both glucocorticoid activity and levels. Secondly, we further analysed the in vivo contribution of glucocorticoids to HIF activity. Interestingly, our results show that both glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) play a key role in enhancing it. Finally, we found indications that glucocorticoids promote HIF signalling via multiple routes. Cumulatively, our findings allowed us to suggest a model for how this crosstalk occurs in vivo.
Collapse
Affiliation(s)
- Davide Marchi
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
- * E-mail: (DM); (FJMv)
| | - Kirankumar Santhakumar
- Department of Genetic Engineering, SRM Institute of Science and Technology Kattankulathur, India
| | - Eleanor Markham
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Nan Li
- The Bateson Centre & Department of Oncology and Metabolism, School of Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | - Nils Krone
- The Bateson Centre & Department of Oncology and Metabolism, School of Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Vincent T. Cunliffe
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Fredericus J. M. van Eeden
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
- * E-mail: (DM); (FJMv)
| |
Collapse
|
18
|
Merz T, Denoix N, Wigger D, Waller C, Wepler M, Vettorazzi S, Tuckermann J, Radermacher P, McCook O. The Role of Glucocorticoid Receptor and Oxytocin Receptor in the Septic Heart in a Clinically Relevant, Resuscitated Porcine Model With Underlying Atherosclerosis. Front Endocrinol (Lausanne) 2020; 11:299. [PMID: 32477273 PMCID: PMC7239997 DOI: 10.3389/fendo.2020.00299] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Abstract
The pathophysiology of sepsis-induced myocardial dysfunction is not resolved to date and comprises inflammation, barrier dysfunction and oxidative stress. Disease-associated reduction of tissue cystathionine-γ-lyase (CSE) expression, an endogenous H2S-producing enzyme, is associated with oxidative stress, barrier dysfunction and organ injury. CSE-mediated cardio-protection has been suggested to be related the upregulation of oxytocin receptor (OTR). CSE can also mediate glucocorticoid receptor (GR) signaling, which is important for normal heart function. A sepsis-related loss of cardiac CSE expression associated with impaired organ function has been reported previously. The aim of this current post hoc study was to investigate the role of cardiac GR and OTR after polymicrobial sepsis in a clinically relevant, resuscitated, atherosclerotic porcine model. Anesthetized and instrumented FBM (Familial Hypercholesterolemia Bretoncelles Meishan) pigs with high fat diet-induced atherosclerosis underwent poly-microbial septic shock (n = 8) or sham procedure (n = 5), and subsequently received intensive care therapy with fluid and noradrenaline administration for 24 h. Cardiac protein expression and mRNA levels were analyzed. Systemic troponin, a marker of cardiac injury, was significantly increased in septic animals in contrast to sham, whereas OTR and GR expression in septic hearts were reduced, along with a down-regulation of anti-inflammatory GR target genes and the antioxidant transcription factor NRF2. These results suggest a potential interplay between GR, CSE, and OTR in sepsis-mediated oxidative stress, inflammation and cardiac dysfunction.
Collapse
Affiliation(s)
- Tamara Merz
- Ulm University Medical Center, Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm, Germany
- *Correspondence: Tamara Merz
| | - Nicole Denoix
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Daniela Wigger
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Martin Wepler
- Ulm University Medical Center, Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm, Germany
- Clinic for Anesthesia, Ulm University Medical Center, Ulm, Germany
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Peter Radermacher
- Ulm University Medical Center, Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm, Germany
| | - Oscar McCook
- Ulm University Medical Center, Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm, Germany
| |
Collapse
|
19
|
Somvanshi PR, Mellon SH, Flory JD, Abu-Amara D, Wolkowitz OM, Yehuda R, Jett M, Hood L, Marmar C, Doyle FJ. Mechanistic inferences on metabolic dysfunction in posttraumatic stress disorder from an integrated model and multiomic analysis: role of glucocorticoid receptor sensitivity. Am J Physiol Endocrinol Metab 2019; 317:E879-E898. [PMID: 31322414 PMCID: PMC6879860 DOI: 10.1152/ajpendo.00065.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/28/2019] [Accepted: 07/17/2019] [Indexed: 02/08/2023]
Abstract
Posttraumatic stress disorder (PTSD) is associated with neuroendocrine alterations and metabolic abnormalities; however, how metabolism is affected by neuroendocrine disturbances is unclear. The data from combat-exposed veterans with PTSD show increased glycolysis to lactate flux, reduced TCA cycle flux, impaired amino acid and lipid metabolism, insulin resistance, inflammation, and hypersensitive hypothalamic-pituitary-adrenal (HPA) axis. To analyze whether the co-occurrence of multiple metabolic abnormalities is independent or arises from an underlying regulatory defect, we employed a systems biological approach using an integrated mathematical model and multiomic analysis. The models for hepatic metabolism, HPA axis, inflammation, and regulatory signaling were integrated to perform metabolic control analysis (MCA) with respect to the observations from our clinical data. We combined the metabolomics, neuroendocrine, clinical laboratory, and cytokine data from combat-exposed veterans with and without PTSD to characterize the differences in regulatory effects. MCA revealed mechanistic association of the HPA axis and inflammation with metabolic dysfunction consistent with PTSD. This was supported by the data using correlational and causal analysis that revealed significant associations between cortisol suppression, high-sensitivity C-reactive protein, homeostatic model assessment of insulin resistance, γ-glutamyltransferase, hypoxanthine, and several metabolites. Causal mediation analysis indicates that the effects of enhanced glucocorticoid receptor sensitivity (GRS) on glycolytic pathway, gluconeogenic and branched-chain amino acids, triglycerides, and hepatic function are jointly mediated by inflammation, insulin resistance, oxidative stress, and energy deficit. Our analysis suggests that the interventions to normalize GRS and inflammation may help to manage features of metabolic dysfunction in PTSD.
Collapse
Affiliation(s)
- Pramod R Somvanshi
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Synthia H Mellon
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, California
| | - Janine D Flory
- Department of Psychiatry, James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Duna Abu-Amara
- Department of Psychiatry, New York Langone Medical School, New York, New York
| | - Owen M Wolkowitz
- Department of Psychiatry, University of California, San Francisco, California
| | - Rachel Yehuda
- Department of Psychiatry, James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marti Jett
- Integrative Systems Biology, US Army Medical Research and Materiel Command, US Army Center for Environmental Health Research, Fort Detrick, Frederick, Maryland
| | - Leroy Hood
- Institute for Systems Biology, Seattle, Washington
| | - Charles Marmar
- Department of Psychiatry, New York Langone Medical School, New York, New York
| | - Francis J Doyle
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
20
|
Lv J, Ma Q, Dasgupta C, Xu Z, Zhang L. Antenatal Hypoxia and Programming of Glucocorticoid Receptor Expression in the Adult Rat Heart. Front Physiol 2019; 10:323. [PMID: 31001129 PMCID: PMC6454194 DOI: 10.3389/fphys.2019.00323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/11/2019] [Indexed: 12/26/2022] Open
Abstract
Glucocorticoid receptor (GR) signaling is critical for development and function of the heart. Our previous study demonstrated that gestational hypoxia induced epigenetic repression of the GR gene in the developing heart. The present study aims to determine that the alterations of promoter methylation level and epigenetic repression of the GR gene in the developing heart in response to maternal hypoxia is sustained in adult offspring and potential gender differences in the programming of GR gene. Pregnant rats were treated with 10.5% O2 from gestational day 15 (E15) to 21 (E21). Hearts were isolated from 5-month-old male and female offspring with the developing stage being equivalent to 18-year-old human. GR mRNA and protein abundance was determined with real time qRT-PCR and Western blot. GR gene promoter methylation and binding of transcription factors were measured with methylated DNA immunoprecipitation (MeDIP) and Chromatin immunoprecipitation (ChIP). The results showed that antenatal hypoxia significantly decreased the expression of GR mRNA and protein in the hearts of adult male offspring, but not in females, which is ascribed to the differential changes of alternative exon1 mRNA variants of GR gene in male and female hearts in response to prenatal hypoxia. In addition, the downregulation of GR expression in the male heart was correlated with increased methylation levels of CpG dinucleotides in promoters of exon 14, 15, 16, 17, and 110, which resulted in a decrease in the binding of their transcription factors. Thus, the study reveals that antenatal hypoxia results in a reprogramming and long-term change in GR gene expression in the heart by hypermethylation of GR promoter in a sex-differential pattern, which provides a novel mechanism regarding the increased vulnerability of heart later in life with exposure of prenatal hypoxia.
Collapse
Affiliation(s)
- Juanxiu Lv
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Chiranjib Dasgupta
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
21
|
Concepcion KR, Zhang L. Corticosteroids and perinatal hypoxic-ischemic brain injury. Drug Discov Today 2018; 23:1718-1732. [PMID: 29778695 DOI: 10.1016/j.drudis.2018.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/13/2018] [Accepted: 05/11/2018] [Indexed: 01/15/2023]
Abstract
Perinatal hypoxic-ischemic (HI) brain injury is the major cause of neonatal mortality and severe long-term neurological morbidity. Yet, the effective therapeutic interventions currently available are extremely limited. Corticosteroids act on both mineralocorticoid (MR) and glucocorticoid (GR) receptors and modulate inflammation and apoptosis in the brain. Neuroinflammatory response to acute cerebral HI is a major contributor to the pathophysiology of perinatal brain injury. Here, we give an overview of current knowledge of corticosteroid-mediated modulations of inflammation and apoptosis in the neonatal brain, focusing on key regulatory cells of the innate and adaptive immune response. In addition, we provide new insights into targets of MR and GR in potential therapeutic strategies that could be beneficial for the treatment of infants with HI brain injury.
Collapse
Affiliation(s)
- Katherine R Concepcion
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
22
|
Ayroldi E, Cannarile L, Delfino DV, Riccardi C. A dual role for glucocorticoid-induced leucine zipper in glucocorticoid function: tumor growth promotion or suppression? Cell Death Dis 2018; 9:463. [PMID: 29695779 PMCID: PMC5916931 DOI: 10.1038/s41419-018-0558-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
Glucocorticoids (GCs), important therapeutic tools to treat inflammatory and immunosuppressive diseases, can also be used as part of cancer therapy. In oncology, GCs are used as anticancer drugs for lymphohematopoietic malignancies, while in solid neoplasms primarily to control the side effects of chemo/radiotherapy treatments. The molecular mechanisms underlying the effects of GCs are numerous and often overlapping, but not all have been elucidated. In normal, cancerous, and inflammatory tissues, the response to GCs differs based on the tissue type. The effects of GCs are dependent on several factors: the tumor type, the GC therapy being used, the expression level of the glucocorticoid receptor (GR), and the presence of any other stimuli such as signals from immune cells and the tumor microenvironment. Therefore, GCs may either promote or suppress tumor growth via different molecular mechanisms. Stress exposure results in dysregulation of the hypothalamic-pituitary-adrenal axis with increased levels of endogenous GCs that promote tumorigenesis, confirming the importance of GCs in tumor growth. Most of the effects of GCs are genomic and mediated by the modulation of GR gene transcription. Moreover, among the GR-induced genes, glucocorticoid-induced leucine zipper (GILZ), which was cloned and characterized primarily in our laboratory, mediates many GC anti-inflammatory effects. In this review, we analyzed the possible role for GILZ in the effects GCs have on tumors cells. We also suggest that GILZ, by affecting the immune system, tumor microenvironment, and directly cancer cell biology, has a tumor-promoting function. However, it may also induce apoptosis or decrease the proliferation of cancer cells, thus inhibiting tumor growth. The potential therapeutic implications of GILZ activity on tumor cells are discussed here.
Collapse
Affiliation(s)
- Emira Ayroldi
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy.
| | - Lorenza Cannarile
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy
| | - Domenico V Delfino
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy
| |
Collapse
|
23
|
McHugh CI, Thipparthi MR, Lawhorn-Crews JM, Polin L, Gadgeel S, Akoury J, Mangner TJ, Douglas KA, Li J, Ratnam M, Shields AF. Using Radiolabeled 3'-Deoxy-3'- 18F-Fluorothymidine with PET to Monitor the Effect of Dexamethasone on Non-Small Cell Lung Cancer. J Nucl Med 2018; 59:1544-1550. [PMID: 29674424 DOI: 10.2967/jnumed.117.207258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/09/2018] [Indexed: 01/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer mortality in the United States, and pemetrexed-based therapies are regularly used to treat nonsquamous NSCLC. Despite widespread use, pemetrexed has a modest effect on progression-free survival, with varying efficacy between individuals. Recent work has indicated that dexamethasone, given to prevent pemetrexed toxicity, is able to protect a subset of NSCLC cells from pemetrexed cytotoxicity by temporarily suppressing the S phase of the cell cycle. Therefore, dexamethasone might block treatment efficacy in a subpopulation of patients and might be contributing to the variable response to pemetrexed. Methods: Differences in retention of the experimental PET tracer 3'-deoxy-3'-fluorothymidine (FLT) were used to monitor S-phase suppression by dexamethasone in NSCLC cell models, animals with tumor xenografts, and patients with advanced cancer. Results: Significant reductions in tracer uptake were observed after 24 h of dexamethasone treatment in NSCLC cell lines and xenograft models expressing high levels of glucocorticoid receptor α, coincident with pemetrexed resistance visualized by attenuation of the flare effect associated with pemetrexed activity. Two of 4 patients imaged in a pilot study with 18F-FLT PET after dexamethasone treatment demonstrated reductions in tracer uptake from baseline, with a variable response between individual tumor lesions. Conclusion: 18F-FLT PET represents a useful method for the noninvasive monitoring of dexamethasone-mediated S-phase suppression in NSCLC and might provide a way to individualize chemotherapy in patients receiving pemetrexed-based regimens.
Collapse
Affiliation(s)
| | | | - Jawana M Lawhorn-Crews
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Lisa Polin
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Shirish Gadgeel
- Department of Oncology, University of Michigan Health System, Ann Arbor, Michigan
| | - Janice Akoury
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | | | - Kirk A Douglas
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Jing Li
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Manohar Ratnam
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Anthony F Shields
- Wayne State University School of Medicine, Detroit, Michigan .,Karmanos Cancer Institute, Detroit, Michigan; and
| |
Collapse
|
24
|
Nguyen V, Sabeur K, Maltepe E, Ameri K, Bayraktar O, Rowitch DH. Sonic Hedgehog Agonist Protects Against Complex Neonatal Cerebellar Injury. CEREBELLUM (LONDON, ENGLAND) 2018; 17:213-227. [PMID: 29134361 PMCID: PMC5849674 DOI: 10.1007/s12311-017-0895-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cerebellum undergoes rapid growth during the third trimester and is vulnerable to injury and deficient growth in infants born prematurely. Factors associated with preterm cerebellar hypoplasia include chronic lung disease and postnatal glucocorticoid administration. We modeled chronic hypoxemia and glucocorticoid administration in neonatal mice to study whole cerebellar and cell type-specific effects of dual exposure. Chronic neonatal hypoxia resulted in permanent cerebellar hypoplasia. This was compounded by administration of prednisolone as shown by greater volume loss and Purkinje cell death. In the setting of hypoxia and prednisolone, administration of a small molecule Smoothened-Hedgehog agonist (SAG) preserved cerebellar volume and protected against Purkinje cell death. Such protective effects were observed even when SAG was given as a one-time dose after dual insult. To model complex injury and determine cell type-specific roles for the hypoxia inducible factor (HIF) pathway, we performed conditional knockout of von Hippel Lindau (VHL) to hyperactivate HIF1α in cerebellar granule neuron precursors (CGNP) or Purkinje cells. Surprisingly, HIF activation in either cell type resulted in no cerebellar deficit. However, in mice administered prednisolone, HIF overactivation in CGNPs resulted in significant cerebellar hypoplasia, whereas HIF overactivation in Purkinje cells caused cell death. Together, these findings indicate that HIF primes both cell types for injury via glucocorticoids, and that hypoxia/HIF + postnatal glucocorticoid administration act on distinct cellular pathways to cause cerebellar injury. They further suggest that SAG is neuroprotective in the setting of complex neonatal cerebellar injury.
Collapse
Affiliation(s)
- Vien Nguyen
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Khalida Sabeur
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Emin Maltepe
- Division of Neonatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Kurosh Ameri
- Department of Cardiology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Omer Bayraktar
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
- Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, Cambridge University, Cambridge, UK
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Division of Neonatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, Cambridge University, Cambridge, UK.
| |
Collapse
|
25
|
Phenylethanolamine N-methyltransferase gene expression in PC12 cells exposed to intermittent hypoxia. Neurosci Lett 2018; 666:169-174. [DOI: 10.1016/j.neulet.2017.12.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/23/2017] [Accepted: 12/26/2017] [Indexed: 11/22/2022]
|
26
|
de Theije CC, Schols AMWJ, Lamers WH, Ceelen JJM, van Gorp RH, Hermans JJR, Köhler SE, Langen RCJ. Glucocorticoid Receptor Signaling Impairs Protein Turnover Regulation in Hypoxia-Induced Muscle Atrophy in Male Mice. Endocrinology 2018; 159:519-534. [PMID: 29069356 DOI: 10.1210/en.2017-00603] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/18/2017] [Indexed: 02/04/2023]
Abstract
Hypoxemia may contribute to muscle wasting in conditions such as chronic obstructive pulmonary disease. Muscle wasting develops when muscle proteolysis exceeds protein synthesis. Hypoxia induces skeletal muscle atrophy in mice, which can in part be attributed to reduced food intake. We hypothesized that hypoxia elevates circulating corticosterone concentrations by reduced food intake and enhances glucocorticoid receptor (GR) signaling in muscle, which causes elevated protein degradation signaling and dysregulates protein synthesis signaling during hypoxia-induced muscle atrophy. Muscle-specific GR knockout and control mice were subjected to normoxia, normobaric hypoxia (8% oxygen), or pair-feeding to the hypoxia group for 4 days. Plasma corticosterone and muscle GR signaling increased after hypoxia and pair-feeding. GR deficiency prevented muscle atrophy by pair-feeding but not by hypoxia. GR deficiency differentially affected activation of ubiquitin 26S-proteasome and autophagy proteolytic systems by pair-feeding and hypoxia. Reduced food intake suppressed mammalian target of rapamycin complex 1 (mTORC1) activity under normoxic but not hypoxic conditions, and this retained mTORC1 activity was mediated by GR. We conclude that GR signaling is required for muscle atrophy and increased expression of proteolysis-associated genes induced by decreased food intake under normoxic conditions. Under hypoxic conditions, muscle atrophy and elevated gene expression of the ubiquitin proteasomal system-associated E3 ligases Murf1 and Atrogin-1 are mostly independent of GR signaling. Furthermore, impaired inhibition of mTORC1 activity is GR-dependent in hypoxia-induced muscle atrophy.
Collapse
MESH Headings
- Animals
- Autophagy
- Cell Size
- Corticosterone/blood
- Corticosterone/metabolism
- Crosses, Genetic
- Gene Expression Regulation, Enzymologic
- Glucocorticoids/metabolism
- Hypoxia/blood
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle Fibers, Fast-Twitch/enzymology
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/pathology
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/etiology
- Proteasome Endopeptidase Complex/metabolism
- Proteolysis
- Random Allocation
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Chiel C de Theije
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Wouter H Lamers
- Department of Anatomy and Embryology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Judith J M Ceelen
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Rick H van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - J J Rob Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - S Elonore Köhler
- Department of Anatomy and Embryology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Ramon C J Langen
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
27
|
Baranova KA, Rybnikova EA, Samoilov MO. The dynamics of HIF-1α expression in the rat brain at different stages of experimental posttraumatic stress disorder and its correction with moderate hypoxia. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417020027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Siebenmann C, Robach P, Lundby C. Regulation of blood volume in lowlanders exposed to high altitude. J Appl Physiol (1985) 2017; 123:957-966. [PMID: 28572493 DOI: 10.1152/japplphysiol.00118.2017] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/21/2022] Open
Abstract
Humans ascending to high altitude (HA) experience a reduction in arterial oxyhemoglobin saturation and, as a result, arterial O2 content ([Formula: see text]). As HA exposure extends, this reduction in [Formula: see text] is counteracted by an increase in arterial hemoglobin concentration. Initially, hemoconcentration is exclusively related to a reduction in plasma volume (PV), whereas after several weeks a progressive expansion in total red blood cell volume (RCV) contributes, although often to a modest extent. Since the decrease in PV is more rapid and usually more pronounced than the expansion in RCV, at least during the first weeks of exposure, a reduction in circulating blood volume is common at HA. Although the regulation of hematological responses to HA has been investigated for decades, it remains incompletely understood. This is not only related to the large number of mechanisms that could be involved and the complexity of their interplay but also to the difficulty of conducting comprehensive experiments in the often secluded HA environment. In this review, we present our understanding of the kinetics, the mechanisms and the physiological relevance of the HA-induced reduction in PV and expansion in RCV.
Collapse
Affiliation(s)
- Christoph Siebenmann
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; and
| | - Paul Robach
- National School for Mountain Sports, Site of the National School for Skiing and Mountaineering (ENSA), Chamonix, France
| | - Carsten Lundby
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; and
| |
Collapse
|
29
|
Taylor CT, Doherty G, Fallon PG, Cummins EP. Hypoxia-dependent regulation of inflammatory pathways in immune cells. J Clin Invest 2016; 126:3716-3724. [PMID: 27454299 DOI: 10.1172/jci84433] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Uncontrolled inflammation underpins a diverse range of diseases where effective therapy remains an unmet clinical need. Hypoxia is a prominent feature of the inflammatory microenvironment that regulates key transcription factors including HIF and NF-κB in both innate and adaptive immune cells. In turn, altered activity of the pathways controlled by these factors can affect the course of inflammation through the regulation of immune cell development and function. In this review, we will discuss these pathways and the oxygen sensors that confer hypoxic sensitivity in immune cells. Furthermore, we will describe how hypoxia-dependent pathways contribute to immunity and discuss their potential as therapeutic targets in inflammatory and infectious disease.
Collapse
|
30
|
Simko V, Takacova M, Debreova M, Laposova K, Ondriskova-Panisova E, Pastorekova S, Csaderova L, Pastorek J. Dexamethasone downregulates expression of carbonic anhydrase IX via HIF-1α and NF-κB-dependent mechanisms. Int J Oncol 2016; 49:1277-88. [PMID: 27431580 PMCID: PMC5021256 DOI: 10.3892/ijo.2016.3621] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/21/2016] [Indexed: 12/31/2022] Open
Abstract
Dexamethasone is a synthetic glucocorticoid frequently used to suppress side-effects of anticancer chemotherapy. In the present study, we showed that dexamethasone treatment leads to concentration-dependent downregulation of cancer-associated marker, carbonic anhydrase IX (CA IX), at the level of promoter activity, mRNA and protein expression in 2D and 3D cancer cell models. The effect of dexamethasone on CA IX expression under hypoxic conditions is predominantly mediated by impaired transcriptional activity and decreased protein level of the main hypoxic transcription factor HIF-1α. In addition, CA9 downregulation can be caused by protein-protein interactions between activated glucocorticoid receptors, major effectors of glucocorticoid action, and transcription factors that trigger CA9 transcription (e.g. AP-1). Moreover, we identified a potential NF-κB binding site in the CA9 promoter and propose the involvement of NF-κB in the dexamethasone-mediated inhibition of CA9 transcription. As high level of CA IX is often linked to aggressive tumor behavior, poor prognosis and chemo- and radiotherapy resistance, uncovering its reduction after dexa-methasone treatment and implication of additional regulatory mechanisms can be relevant for the CA IX-related clinical applications.
Collapse
Affiliation(s)
- Veronika Simko
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Martina Takacova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Michaela Debreova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Katarina Laposova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Elena Ondriskova-Panisova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Silvia Pastorekova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Lucia Csaderova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jaromir Pastorek
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| |
Collapse
|
31
|
Orgeig S, Morrison JL, Daniels CB. Evolution, Development, and Function of the Pulmonary Surfactant System in Normal and Perturbed Environments. Compr Physiol 2015; 6:363-422. [PMID: 26756637 DOI: 10.1002/cphy.c150003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Surfactant lipids and proteins form a surface active film at the air-liquid interface of internal gas exchange organs, including swim bladders and lungs. The system is uniquely positioned to meet both the physical challenges associated with a dynamically changing internal air-liquid interface, and the environmental challenges associated with the foreign pathogens and particles to which the internal surface is exposed. Lungs range from simple, transparent, bag-like units to complex, multilobed, compartmentalized structures. Despite this anatomical variability, the surfactant system is remarkably conserved. Here, we discuss the evolutionary origin of the surfactant system, which likely predates lungs. We describe the evolution of surfactant structure and function in invertebrates and vertebrates. We focus on changes in lipid and protein composition and surfactant function from its antiadhesive and innate immune to its alveolar stability and structural integrity functions. We discuss the biochemical, hormonal, autonomic, and mechanical factors that regulate normal surfactant secretion in mature animals. We present an analysis of the ontogeny of surfactant development among the vertebrates and the contribution of different regulatory mechanisms that control this development. We also discuss environmental (oxygen), hormonal and biochemical (glucocorticoids and glucose) and pollutant (maternal smoking, alcohol, and common "recreational" drugs) effects that impact surfactant development. On the adult surfactant system, we focus on environmental variables including temperature, pressure, and hypoxia that have shaped its evolution and we discuss the resultant biochemical, biophysical, and cellular adaptations. Finally, we discuss the effect of major modern gaseous and particulate pollutants on the lung and surfactant system.
Collapse
Affiliation(s)
- Sandra Orgeig
- School of Pharmacy & Medical Sciences and Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Janna L Morrison
- School of Pharmacy & Medical Sciences and Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Christopher B Daniels
- School of Pharmacy & Medical Sciences and Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| |
Collapse
|
32
|
Zou W, Yang S, Zhang T, Sun H, Wang Y, Xue H, Zhou D. Hypoxia enhances glucocorticoid-induced apoptosis and cell cycle arrest via the PI3K/Akt signaling pathway in osteoblastic cells. J Bone Miner Metab 2015; 33:615-24. [PMID: 25230819 DOI: 10.1007/s00774-014-0627-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 08/18/2014] [Indexed: 12/12/2022]
Abstract
Although osteonecrosis of the femoral head is a known primary limitation of long-term or high-dose clinical administration of glucocorticoids, the mechanisms underlying this side effect remain unclear. Hypoxia is an important biological state under numerous pathological conditions. In this study, we investigated glucocorticoid-induced osteonecrosis under hypoxic conditions in the MC3T3-E1 osteoblast cell line using a cell cytotoxicity assay, flow cytometry, and western blotting. 6α-Methylprednisolone sodium succinate (MPSL) more effectively induced apoptosis and G0/G1 arrest of MC3T3-E1 osteoblasts under hypoxic conditions than under normoxic conditions. Correspondingly, MPSL more effectively upregulated cellular levels of cleaved caspase 3, p53, and its target p21, and downregulated cyclin D1 levels in hypoxia. Moreover, overexpression of Akt abrogated the MPSL activation of p53, p21, and cleaved caspase 3 and the attenuation of cyclin D1 expression and rescued osteoblasts from MPSL-induced cell cycle arrest and apoptosis, indicating that phosphatidylinositol 3-kinase (PI3K)/Akt signaling might play an essential role in MPSL-induced inhibition of osteoblasts. Furthermore, the suppression of PI3K/Akt signaling and upregualtion of cellular p85α monomer levels by MPSL were more pronounced under hypoxic conditions than under normoxic conditions. Finally, we found that the enhancement of the effects of MPSL under hypoxic conditions was attributed to hypoxia-upregulated glucocorticoid receptor activity. In conclusion, our results demonstrate that MPSL, a synthetic glucocorticoid receptor agonist, promotes the level of p85α and inhibits PI3K/Akt signaling to induce apoptosis and cell cycle arrest in osteoblasts, and that this effect is enhanced under hypoxic conditions.
Collapse
Affiliation(s)
- Wanjing Zou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Shu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Tie Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Haimei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yuying Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hong Xue
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Deshan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
33
|
Swenson ER. Pharmacology of acute mountain sickness: old drugs and newer thinking. J Appl Physiol (1985) 2015; 120:204-15. [PMID: 26294748 DOI: 10.1152/japplphysiol.00443.2015] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/12/2015] [Indexed: 01/09/2023] Open
Abstract
Pharmacotherapy in acute mountain sickness (AMS) for the past half century has largely rested on the use of carbonic anhydrase (CA) inhibitors, such as acetazolamide, and corticosteroids, such as dexamethasone. The benefits of CA inhibitors are thought to arise from their known ventilatory stimulation and resultant greater arterial oxygenation from inhibition of renal CA and generation of a mild metabolic acidosis. The benefits of corticosteroids include their broad-based anti-inflammatory and anti-edemagenic effects. What has emerged from more recent work is the strong likelihood that drugs in both classes act on other pathways and signaling beyond their classical actions to prevent and treat AMS. For the CA inhibitors, these include reduction in aquaporin-mediated transmembrane water transport, anti-oxidant actions, vasodilation, and anti-inflammatory effects. In the case of corticosteroids, these include protection against increases in vascular endothelial and blood-brain barrier permeability, suppression of inflammatory cytokines and reactive oxygen species production, and sympatholysis. The loci of action of both classes of drug include the brain, but may also involve the lung as revealed by benefits that arise with selective administration to the lungs by inhalation. Greater understanding of their pluripotent actions and sites of action in AMS may help guide development of better drugs with more selective action and fewer side effects.
Collapse
Affiliation(s)
- Erik R Swenson
- Veterans Affairs Puget Sound Health Care System, Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle
| |
Collapse
|
34
|
Zhang C, Qiang Q, Jiang Y, Hu L, Ding X, Lu Y, Hu G. Effects of hypoxia inducible factor-1α on apoptotic inhibition and glucocorticoid receptor downregulation by dexamethasone in AtT-20 cells. BMC Endocr Disord 2015; 15:24. [PMID: 26002039 PMCID: PMC4464719 DOI: 10.1186/s12902-015-0017-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/20/2015] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Hypoxia inducible factor-1α (HIF-1α) is the central transcriptional regulator of hypoxic responses during the progression of pituitary adenomas. Although previous immunohistochemical studies revealed that HIF-1α is expressed in adreno-cortico-tropic-hormone (ACTH) pituitary adenomas, the role of HIF-1α remains unclear. METHODS AtT-20 cells were incubated under hypoxic conditions (1 % O2) for 12 h. HIF-1α mRNA and protein expression levels were measured by real-time PCR and western blotting, respectively. BrdU was used to determine the effects of hypoxia on cell viability. AtT-20 cells were transfected with siRNA targeting HIF-1α, followed by hypoxia (1 % O2) for 12 h. Apoptosis was determined by annexin V-FITC flow cytometry and Tdt-mediated dUTP nick end-labelling (TUNEL) assay. In addition, we examined interactions between HIF-1α, glucocorticoid receptor (GR), and dexamethasone under both normoxic and hypoxic conditions. RESULTS Hypoxia triggered the time-dependent proliferation of AtT-20 cells in association with increased HIF-1α mRNA and protein levels. However, the viability of AtT-20 cells decreased greatly when they were first transfected with HIF-1α-siRNA and then exposed to hypoxia. According to flow cytometry (annexin V-FITC and PI staining) and TUNEL analyses, a greater percentage of cells were apoptotic when transfected with HIF-1α siRNA and subsequently cultured under hypoxic conditions compared to those in the normoxia and mock groups. After AtT-20 cells were cultured in 1 % O2 and then treated with dexamethasone, HIF-1α levels significantly increased or decreased in normoxic or hypoxic conditions, respectively. Dexamethasone suppressed GR expression to a higher degree in hypoxic than normoxic conditions. Downregulation of GR by dexamethasone was greatly prevented in cells that were transfected with HIF-1α siRNA. CONCLUSIONS These findings strongly suggest that HIF-1α exerts an antiapoptotic role and participates in the downregulation of GR by dexamethasone in hypoxic AtT-20 cells.
Collapse
Affiliation(s)
- Chenran Zhang
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Feng-Yang Road, Shanghai, 200003, China.
| | - Qiang Qiang
- Department of Neurology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| | - Ying Jiang
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Feng-Yang Road, Shanghai, 200003, China.
| | - Liuhua Hu
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Xuehua Ding
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Feng-Yang Road, Shanghai, 200003, China.
| | - Yicheng Lu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Feng-Yang Road, Shanghai, 200003, China.
| | - Guohan Hu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Feng-Yang Road, Shanghai, 200003, China.
| |
Collapse
|
35
|
de Theije CC, Langen RCJ, Lamers WH, Gosker HR, Schols AMWJ, Köhler SE. Differential sensitivity of oxidative and glycolytic muscles to hypoxia-induced muscle atrophy. J Appl Physiol (1985) 2014; 118:200-11. [PMID: 25429096 DOI: 10.1152/japplphysiol.00624.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hypoxia as a consequence of acute and chronic respiratory disease has been associated with muscle atrophy. This study investigated the sensitivity of oxidative and glycolytic muscles to hypoxia-induced muscle atrophy. Male mice were exposed to 8% normobaric oxygen for up to 21 days. Oxidative soleus and glycolytic extensor digitorum longus (EDL) muscles were isolated, weighed, and assayed for expression profiles of the ubiquitin-proteasome system (UPS), the autophagy-lysosome pathway (ALP), and glucocorticoid receptor (GR) and hypoxia-inducible factor-1α (HIF1α) signaling. Fiber-type composition and the capillary network were investigated. Hypoxia-induced muscle atrophy was more prominent in the EDL than the soleus muscle. Although increased expression of HIF1α target genes showed that both muscle types sensed hypoxia, their adaptive responses differed. Atrophy consistently involved a hypoxia-specific effect (i.e., not attributable to a hypoxia-mediated reduction of food intake) in the EDL only. Hypoxia-specific activation of the UPS and ALP and increased expression of the glucocorticoid receptor (Gr) and its target genes were also mainly observed in the EDL. In the soleus, stimulation of gene expression of those pathways could be mimicked to a large extent by food restriction alone. Hypoxia increased the number of capillary contacts per fiber cross-sectional area in both muscles. In the EDL, this was due to type II fiber atrophy, whereas in the soleus the absolute number of capillary contacts increased. These responses represent two distinct modes to improve oxygen supply to muscle fibers, but may aggravate muscle atrophy in chronic obstructive pulmonary disease patients who have a predominance of type II fibers.
Collapse
Affiliation(s)
- C C de Theije
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands; Department of Anatomy and Embryology, Maastricht University Medical Center+, Maastricht, The Netherlands; and
| | - R C J Langen
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands; Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - W H Lamers
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands; Department of Anatomy and Embryology, Maastricht University Medical Center+, Maastricht, The Netherlands; and
| | - H R Gosker
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands; Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - A M W J Schols
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands; Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - S E Köhler
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands; Department of Anatomy and Embryology, Maastricht University Medical Center+, Maastricht, The Netherlands; and
| |
Collapse
|
36
|
Lim W, Park C, Shim MK, Lee YH, Lee YM, Lee Y. Glucocorticoids suppress hypoxia-induced COX-2 and hypoxia inducible factor-1α expression through the induction of glucocorticoid-induced leucine zipper. Br J Pharmacol 2014; 171:735-45. [PMID: 24172143 DOI: 10.1111/bph.12491] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 10/18/2013] [Accepted: 10/22/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE The COX-2/PGE2 pathway in hypoxic cancer cells has important implications for stimulation of inflammation and tumourigenesis. However, the mechanism by which glucocorticoid receptors (GRs) inhibit COX-2 during hypoxia has not been elucidated. Hence, we explored the mechanisms underlying glucocorticoid-mediated inhibition of hypoxia-induced COX-2 in human distal lung epithelial A549 cells. EXPERIMENTAL APPROACH The expressions of COX-2 and glucocorticoid-induced leucine zipper (GILZ) in A549 cells were determined by Western blot and/or quantitative real time-PCR respectively. The anti-invasive effect of GILZ on A549 cells was evaluated using the matrigel invasion assay. KEY RESULTS The hypoxia-induced increase in COX-2 protein and mRNA levels and promoter activity were suppressed by dexamethasone, and this effect of dexamethasone was antagonized by the GR antagonist RU486. Overexpression of GILZ in A549 cells also inhibited hypoxia-induced COX-2 expression levels and knockdown of GILZ reduced the glucocorticoid-mediated inhibition of hypoxia-induced COX-2 expression, indicating that the inhibitory effects of dexamethasone on hypoxia-induced COX-2 are mediated by GILZ. GILZ suppressed the expression of hypoxia inducible factor (HIF)-1α at the protein level and affected its signalling pathway. Hypoxia-induced cell invasion was also dramatically reduced by GILZ expression. CONCLUSION AND IMPLICATIONS Dexamethasone-induced upregulation of GILZ not only inhibits the hypoxic-evoked induction of COX-2 expression and cell invasion but further blocks the HIF-1 pathway by destabilizing HIF-1α expression. Taken together, these findings suggest that the suppression of hypoxia-induced COX-2 by glucocorticoids is mediated by GILZ. Hence, GILZ is a potential key therapeutic target for suppression of inflammation under hypoxia.
Collapse
Affiliation(s)
- Wonchung Lim
- Department of Bioscience and Biotechnology, College of Life Science, Institute of Biotechnology, Sejong University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
37
|
Sau S, Agarwalla P, Mukherjee S, Bag I, Sreedhar B, Pal-Bhadra M, Patra CR, Banerjee R. Cancer cell-selective promoter recognition accompanies antitumor effect by glucocorticoid receptor-targeted gold nanoparticle. NANOSCALE 2014; 6:6745-54. [PMID: 24824564 DOI: 10.1039/c4nr00974f] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Nanoparticles, such as gold nanoparticles (GNP), upon convenient modifications perform multi tasks catering to many biomedical applications. However, GNP or any other type of nanoparticles is yet to achieve the feat of intracellular regulation of endogenous genes of choice such as through manipulation of a gene-promoter in a chromosome. As for gene modulation and delivery, GNP (or other nanoparticles) showed only limited gene therapy potential, which relied on the delivery of 'exogenous' genes invoking gene knockdown or replacement. Practically, there are no instances for the nanoparticle-mediated promoter regulation of 'endogenous' genes, more so, as a cancer selective phenomenon. In this regard, we report the development of a simple, easily modifiable GNP-formulation, which promoted/up-regulated the expression of a specific category of 'endogenous' genes, the glucocorticoid responsive genes. This genetic up-regulation was induced in only cancer cells by modified GNP-mediated transcriptional activation of its cytoplasmic receptor, glucocorticoid receptor (GR). Normal cells and their GR remained primarily unperturbed by this GNP-formulation. The most potent gene up-regulating GNP-formulation down-regulated a cancer-specific proliferative signal, phospho-Akt in cancer cells, which accompanied retardation of tumor growth in the murine melanoma model. We show that GR-targeted GNPs may find potential use in the targeting and modulation of genetic information in cancer towards developing novel anticancer therapeutics.
Collapse
Affiliation(s)
- Samaresh Sau
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Tarnaka, Uppal Road, Hyderabad 500007, India.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Involvement of transcriptional factor induced by hypoxia in the neuronal mechanisms of adaptation to psychoemotional and hypoxic stress. ACTA ACUST UNITED AC 2014. [DOI: 10.15407/fz59.06.088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
39
|
Inhibition of protein translation as a mechanism of acidotic pH protection against ischaemic injury through inhibition of CREB mediated tRNA synthetase expression. Exp Cell Res 2013; 319:3116-27. [DOI: 10.1016/j.yexcr.2013.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/22/2013] [Accepted: 07/18/2013] [Indexed: 12/11/2022]
|
40
|
Chen JC, Cai HY, Wang Y, Ma YY, Song LN, Yin LJ, Cao DM, Diao F, Li YD, Lu J. Up-regulation of stomatin expression by hypoxia and glucocorticoid stabilizes membrane-associated actin in alveolar epithelial cells. J Cell Mol Med 2013; 17:863-72. [PMID: 23672602 PMCID: PMC3822891 DOI: 10.1111/jcmm.12069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 03/24/2013] [Indexed: 11/27/2022] Open
Abstract
Stomatin is an important lipid raft-associated protein which interacts with membrane proteins and plays a role in the membrane organization. However, it is unknown whether it is involved in the response to hypoxia and glucocorticoid (GC) in alveolar epithelial cells (AEC). In this study we found that hypoxia and dexamethasone (dex), a synthetic GC not only up-regulated the expression of stomatin alone, but also imposed additive effect on the expression of stomatin in A549 cells, primary AEC and lung of rats. Then we investigated whether hypoxia and dex transcriptionally up-regulated the expression of stomatin by reporter gene assay, and found that dex, but not hypoxia could increase the activity of a stomatin promoter-driven reporter gene. Further deletion and mutational studies demonstrated that a GC response element (GRE) within the promoter region mainly contributed to the induction of stomatin by dex. Moreover, we found that hypoxia exposure did not affect membrane-associated actin, but decreased actin in cytoplasm in A549 cells. Inhibiting stomatin expression by stomatin siRNA significantly decreased dense of peripheral actin ring in hypoxia or dex treated A549 cells. Taken all together, these data indicated that dex and/or hypoxia significantly up-regulated the expression of stomatin in vivo and in vitro, which could stabilize membrane-associated actin in AEC. We suppose that the up-regulation of stomatin by hypoxia and dex may enhance the barrier function of alveolar epithelia and mediate the adaptive role of GC to hypoxia.
Collapse
Affiliation(s)
- Ji-Cheng Chen
- Department of Pathophysiology, The Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Liu C, Croft QPP, Kalidhar S, Brooks JT, Herigstad M, Smith TG, Dorrington KL, Robbins PA. Dexamethasone mimics aspects of physiological acclimatization to 8 hours of hypoxia but suppresses plasma erythropoietin. J Appl Physiol (1985) 2013; 114:948-56. [PMID: 23393065 PMCID: PMC3633439 DOI: 10.1152/japplphysiol.01414.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/31/2013] [Indexed: 11/22/2022] Open
Abstract
Dexamethasone ameliorates the severity of acute mountain sickness (AMS) but it is unknown whether it obtunds normal physiological responses to hypoxia. We studied whether dexamethasone enhanced or inhibited the ventilatory, cardiovascular, and pulmonary vascular responses to sustained (8 h) hypoxia. Eight healthy volunteers were studied, each on four separate occasions, permitting four different protocols. These were: dexamethasone (20 mg orally) beginning 2 h before a control period of 8 h of air breathing; dexamethasone with 8 h of isocapnic hypoxia (end-tidal Po(2) = 50 Torr); placebo with 8 h of air breathing; and placebo with 8 h of isocapnic hypoxia. Before and after each protocol, the following were determined under both euoxic and hypoxic conditions: ventilation; pulmonary artery pressure (estimated using echocardiography to assess maximum tricuspid pressure difference); heart rate; and cardiac output. Plasma concentrations of erythropoietin (EPO) were also determined. Dexamethasone had no early (2-h) effect on any variable. Both dexamethasone and 8 h of hypoxia increased euoxic values of ventilation, pulmonary artery pressure, and heart rate, together with the ventilatory sensitivity to acute hypoxia. These effects were independent and additive. Eight hours of hypoxia, but not dexamethasone, increased the sensitivity of pulmonary artery pressure to acute hypoxia. Dexamethasone, but not 8 h of hypoxia, increased both cardiac output and systemic arterial pressure. Dexamethasone abolished the rise in EPO induced by 8 h of hypoxia. In summary, dexamethasone enhances ventilatory acclimatization to hypoxia. Thus, dexamethasone in AMS may improve oxygenation and thereby indirectly lower pulmonary artery pressure.
Collapse
Affiliation(s)
- Chun Liu
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
42
|
RSUME enhances glucocorticoid receptor SUMOylation and transcriptional activity. Mol Cell Biol 2013; 33:2116-27. [PMID: 23508108 DOI: 10.1128/mcb.01470-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Glucocorticoid receptor (GR) activity is modulated by posttranslational modifications, including phosphorylation, ubiquitination, and SUMOylation. The GR has three SUMOylation sites: lysine 297 (K297) and K313 in the N-terminal domain (NTD) and K721 within the ligand-binding domain. SUMOylation of the NTD sites mediates the negative effect of the synergy control motifs of GR on promoters with closely spaced GR binding sites. There is scarce evidence on the role of SUMO conjugation to K721 and its impact on GR transcriptional activity. We have previously shown that RSUME (RWD-containing SUMOylation enhancer) increases protein SUMOylation. We now demonstrate that RSUME interacts with the GR and increases its SUMOylation. RSUME regulates GR transcriptional activity and the expression of its endogenous target genes, FKBP51 and S100P. RSUME uncovers a positive role for the third SUMOylation site, K721, on GR-mediated transcription, demonstrating that GR SUMOylation acts positively in the presence of a SUMOylation enhancer. Both mutation of K721 and small interfering RNA-mediated RSUME knockdown diminish GRIP1 coactivator activity. RSUME, whose expression is induced under stress conditions, is a key factor in heat shock-induced GR SUMOylation. These results show that inhibitory and stimulatory SUMO sites are present in the GR and at higher SUMOylation levels the stimulatory one becomes dominant.
Collapse
|
43
|
Baitharu I, Deep SN, Jain V, Prasad D, Ilavazhagan G. Inhibition of glucocorticoid receptors ameliorates hypobaric hypoxia induced memory impairment in rat. Behav Brain Res 2013; 240:76-86. [DOI: 10.1016/j.bbr.2012.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/05/2012] [Accepted: 11/07/2012] [Indexed: 01/03/2023]
|
44
|
Abstract
Adverse environments during the fetal and neonatal development period may permanently program physiology and metabolism, and lead to increased risk of diseases in later life. Programming of the hypothalamic-pituitary-adrenal (HPA) axis is one of the key mechanisms that contribute to altered metabolism and response to stress. Programming of the HPA axis often involves epigenetic modification of the glucocorticoid receptor (GR) gene promoter, which influences tissue-specific GR expression patterns and response to stimuli. This review summarizes the current state of research on the HPA axis and programming of health and disease in the adult, focusing on the epigenetic regulation of GR gene expression patterns in response to fetal and neonatal stress. Aberrant GR gene expression patterns in the developing brain may have a significant negative impact on protection of the immature brain against hypoxic-ischemic encephalopathy in the critical period of development during and immediately after birth.
Collapse
|
45
|
Yang N, Ray DW, Matthews LC. Current concepts in glucocorticoid resistance. Steroids 2012; 77:1041-9. [PMID: 22728894 DOI: 10.1016/j.steroids.2012.05.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/24/2012] [Accepted: 05/29/2012] [Indexed: 01/04/2023]
Abstract
Glucocorticoids (GCs) are the most potent anti-inflammatory agents known. A major factor limiting their clinical use is the wide variation in responsiveness to therapy. The high doses of GC required for less responsive patients means a high risk of developing very serious side effects. Variation in sensitivity between individuals can be due to a number of factors. Congenital, generalized GC resistance is very rare, and is due to mutations in the glucocorticoid receptor (GR) gene, the receptor that mediates the cellular effects of GC. A more common problem is acquired GC resistance. This localized, disease-associated GC resistance is a serious therapeutic concern and limits therapeutic response in patients with chronic inflammatory disease. It is now believed that localized resistance can be attributed to changes in the cellular microenvironment, as a consequence of chronic inflammation. Multiple factors have been identified, including alterations in both GR-dependent and -independent signaling downstream of cytokine action, oxidative stress, hypoxia and serum derived factors. The underlying mechanisms are now being elucidated, and are discussed here. Attempts to augment tissue GC sensitivity are predicted to permit safe and effective use of low-dose GC therapy in inflammatory disease.
Collapse
Affiliation(s)
- Nan Yang
- Endocrine Sciences Research Group, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
46
|
Lee YJ, Lee JS, Kang EH, Lee YK, Kim SY, Song YW, Koo KH. Vascular endothelial growth factor polymorphisms in patients with steroid-induced femoral head osteonecrosis. J Orthop Res 2012; 30:21-7. [PMID: 21710604 DOI: 10.1002/jor.21492] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 06/09/2011] [Indexed: 02/04/2023]
Abstract
To investigate an association between steroid-induced femoral head osteonecrosis (FHON) and functional vascular endothelial growth factor (VEGF) gene (-2578A/C, -1154A/G, -634C/G, and +405C/G) polymorphisms polymerase chain reaction-restriction fragment length polymorphism genotyping was performed in 160 patients (86 idiopathic FHON and 74 steroid-induced FHON) and 160 gender- and age-matched controls. The steroid-induced subgroup had a significantly lower prevalence of -1154A allele (7.4% vs. 18.1%, odds ratio (OR) = 0.363) and genotype carrying -1154A (14.9% vs. 32.5%, OR = 0.333 in a recessive model) than controls. In a dominant model, the frequency of genotype carrying +405G (74.3% vs. 84.4%, OR = 0.492) was significantly lower in steroid-induced FHON than in controls. The distribution of haplotypes was significantly different between controls and FHON patients (p = 0.00011). Especially, when haplotypes were classified into high (CGCG and AAGG) or low (CGGC and AGGC) VEGF inducing haplotypes, patients with steroid-induced FHON had a significantly lower prevalence of high inducing haplotypes (7.4% vs. 15.9%, OR = 0.424) and a significantly higher prevalence of low inducing haplotypes (4.7% vs. 0.6%, OR = 7.894) than controls. Low inducing VEGF haplotypes may confer an increased risk and high inducing haplotypes have a protective effect for the development of steroid-induced FHON in Korea.
Collapse
Affiliation(s)
- Yun Jong Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, South Korea
| | | | | | | | | | | | | |
Collapse
|
47
|
Michailidou Z, Turban S, Miller E, Zou X, Schrader J, Ratcliffe PJ, Hadoke PWF, Walker BR, Iredale JP, Morton NM, Seckl JR. Increased angiogenesis protects against adipose hypoxia and fibrosis in metabolic disease-resistant 11β-hydroxysteroid dehydrogenase type 1 (HSD1)-deficient mice. J Biol Chem 2011; 287:4188-97. [PMID: 22158867 PMCID: PMC3281676 DOI: 10.1074/jbc.m111.259325] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In obesity, rapidly expanding adipose tissue becomes hypoxic, precipitating inflammation, fibrosis, and insulin resistance. Compensatory angiogenesis may prevent these events. Mice lacking the intracellular glucocorticoid-amplifying enzyme 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1−/−) have “healthier” adipose tissue distribution and resist metabolic disease with diet-induced obesity. Here we show that adipose tissues of 11βHSD1−/− mice exhibit attenuated hypoxia, induction of hypoxia-inducible factor (HIF-1α) activation of the TGF-β/Smad3/α-smooth muscle actin (α-SMA) signaling pathway, and fibrogenesis despite similar fat accretion with diet-induced obesity. Moreover, augmented 11βHSD1−/− adipose tissue angiogenesis is associated with enhanced peroxisome proliferator-activated receptor γ (PPARγ)-inducible expression of the potent angiogenic factors VEGF-A, apelin, and angiopoietin-like protein 4. Improved adipose angiogenesis and reduced fibrosis provide a novel mechanism whereby suppression of intracellular glucocorticoid regeneration promotes safer fat expansion with weight gain.
Collapse
Affiliation(s)
- Zoi Michailidou
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wang Y, Ma YY, Song XL, Cai HY, Chen JC, Song LN, Yang R, Lu J. Upregulations of glucocorticoid-induced leucine zipper by hypoxia and glucocorticoid inhibit proinflammatory cytokines under hypoxic conditions in macrophages. THE JOURNAL OF IMMUNOLOGY 2011; 188:222-9. [PMID: 22124125 DOI: 10.4049/jimmunol.1002958] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hypoxia and inflammation often develop concurrently in numerous diseases, and the influence of hypoxia on natural evolution of inflammatory responses is widely accepted. Glucocorticoid-induced leucine zipper (GILZ) is thought to be an important mediator of anti-inflammatory and immune-suppressive actions of glucocorticoid (GC). However, whether GILZ is involved in hypoxic response is still unclear. In this study, we investigated the effects of hypoxic exposure and/or the administration of dexamethasone (Dex), a synthetic GC on GILZ expression both in vitro and in vivo, and further explored the relationship between GILZ and proinflammatory cytokines IL-1β, IL-6, and TNF-α under normoxic and hypoxic conditions. We found that hypoxia not only remarkably upregulated the expression of GILZ, but also significantly enhanced Dex-induced expression of GILZ in macrophages and the spleen of rats. ERK activity is found involved in the upregulation of GILZ induced by hypoxia. Inhibiting the expression of GILZ in RAW264.7 cells using specific GILZ small interfering RNA led to a significant increase in mRNA production and protein secretion of IL-1β and IL-6 in hypoxia and abrogated the inhibitory effect of Dex on expression of IL-1β and IL-6 in hypoxia. We also found that adrenal hormones played pivotal roles in upregulation of GILZ expression in vivo. Altogether, data presented in this study suggest that GILZ has an important role not only in adjusting adaptive responses to hypoxia by negatively regulating the activation of macrophages and the expression of proinflammatory cytokines, but also in mediating the anti-inflammatory action of GC under hypoxic conditions.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathophysiology, Second Military Medical University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Hassan-Smith Z, Cooper MS. Overview of the endocrine response to critical illness: how to measure it and when to treat. Best Pract Res Clin Endocrinol Metab 2011; 25:705-17. [PMID: 21925072 DOI: 10.1016/j.beem.2011.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The assessment and manipulation of the endocrine system in patients with critical illness is one of the most complex and controversial areas in endocrinology. Severe acute illness causes dramatic changes in most endocrine systems. This can lead to considerable difficulty in recognising pre-existing endocrine disorders in severely ill patients. Critical care itself might also induce types of endocrine dysfunction not seen outside the critical care unit. It is important to clarify whether or not such endocrine dysfunction occurs. Where it does occur it is also important to determine whether endocrine intervention is useful in improving outcome. There is also the issue of whether endocrine manipulation in critically ill patients without endocrine dysfunction could benefit from endocrine intervention, e.g. to improve haemodynamics or reverse a catabolic state. This review will discuss some of these contentious issues. It will highlight how endocrine assessment of a patient with critical illness differs from that in other types of patient. It will emphasise the added need to place the biochemical assessment and its interpretation in the context of the patients underlying condition.
Collapse
Affiliation(s)
- Zaki Hassan-Smith
- Centre for Endocrinology, Diabetes and Metabolism, 2nd Floor Institute for Biomedical Research, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
| | | |
Collapse
|
50
|
du Souich P, Fradette C. The effect and clinical consequences of hypoxia on cytochrome P450, membrane carrier proteins activity and expression. Expert Opin Drug Metab Toxicol 2011; 7:1083-100. [DOI: 10.1517/17425255.2011.586630] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|