1
|
Verouti S, Aeschlimann G, Wang Q, Del Olmo DA, Peyter AC, Menétrey S, Winter DV, Odermatt A, Pearce D, Hummler E, Vanderriele PE. Salt-sensitive hypertension in GR mutant rats is associated with altered plasma polyunsaturated fatty acid levels and aortic vascular reactivity. Pflugers Arch 2025; 477:37-53. [PMID: 39256246 PMCID: PMC11711871 DOI: 10.1007/s00424-024-03014-y] [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: 04/30/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024]
Abstract
In humans, glucocorticoid resistance is attributed to mutations in the glucocorticoid receptor (GR). Most of these mutations result in decreased ligand binding, transactivation, and/or translocation, albeit with normal protein abundances. However, there is no clear genotype‒phenotype relationship between the severity or age at disease presentation and the degree of functional loss of the receptor. Previously, we documented that a GR+/- rat line developed clinical features of glucocorticoid resistance, namely, hypercortisolemia, adrenal hyperplasia, and salt-sensitive hypertension. In this study, we analyzed the GR+/em4 rat model heterozygously mutant for the deletion of exon 3, which encompasses the second zinc finger, including the domains of DNA binding, dimerization, and nuclear localization signals. On a standard diet, mutant rats exhibited a trend toward increased corticosterone levels and a normal systolic blood pressure and heart rate but presented with adrenal hyperplasia. They exhibited increased adrenal soluble epoxide hydroxylase (sEH), favoring an increase in less active polyunsaturated fatty acids. Indeed, a significant increase in nonactive omega-3 and omega-6 polyunsaturated fatty acids, such as 5(6)-DiHETrE or 9(10)-DiHOME, was observed with advanced age (10 versus 5 weeks old) and following a switch to a high-salt diet accompanied by salt-sensitive hypertension. In thoracic aortas, a reduced soluble epoxide hydrolase (sEH) protein abundance resulted in altered vascular reactivity upon a standard diet, which was blunted upon a high-salt diet. In conclusion, mutations in the GR affecting the ligand-binding domain as well as the dimerization domain resulted in deregulated GR signaling, favoring salt-sensitive hypertension in the absence of obvious mineralocorticoid excess.
Collapse
Affiliation(s)
- S Verouti
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - G Aeschlimann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Q Wang
- Division of Nephrology and Hypertension, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - D Ancin Del Olmo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - A C Peyter
- Neonatal Research Laboratory, Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - S Menétrey
- Neonatal Research Laboratory, Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - D V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - A Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - D Pearce
- Department of Medicine and Cellular & Molecular Pharmacology, University of California, San Francisco, USA
| | - E Hummler
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland
| | - P E Vanderriele
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland.
| |
Collapse
|
2
|
Hage Z, Madeira MM, Koliatsis D, Tsirka SE. Convergence of endothelial dysfunction, inflammation and glucocorticoid resistance in depression-related cardiovascular diseases. BMC Immunol 2024; 25:61. [PMID: 39333855 PMCID: PMC11428380 DOI: 10.1186/s12865-024-00653-9] [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] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Major Depressive Disorder, or depression, has been extensively linked to dysregulated HPA axis function, chronic inflammation and cardiovascular diseases. While the former two have been studied in depth, the mechanistic connection between depression and cardiovascular disease is unclear. As major mediators of vascular homeostasis, vascular pathology and immune activity, endothelial cells represent an important player connecting the diseases. Exaggerated inflammation and glucocorticoid function are important topics to explore in the endothelial response to MDD. Glucocorticoid resistance in several cell types strongly promotes inflammatory signaling and results in worsened severity in many diseases. However, endothelial health and inflammation in chronic stress and depression are rarely considered from the perspective of glucocorticoid signaling and resistance. In this review, we aim to discuss (1) endothelial dysfunction in depression, (2) inflammation in depression, (3) general glucocorticoid resistance in depression and (4) endothelial glucocorticoid resistance in depression co-morbid inflammatory diseases. We will first describe vascular pathology, inflammation and glucocorticoid resistance separately in depression and then describe their potential interactions with one another in depression-relevant diseases. Lastly, we will hypothesize potential mechanisms by which glucocorticoid resistance in endothelial cells may contribute to vascular disease states in depressed people. Overall, endothelial-glucocorticoid signaling may play an important role in connecting depression and vascular pathology and warrants further study.
Collapse
Affiliation(s)
- Zachary Hage
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Miguel M Madeira
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Dimitris Koliatsis
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Stella E Tsirka
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
3
|
Tang LF, Xu A, Liu K. Pharmacogenomics-based individualized treatment of hypertension in preterm infants: A case report and review of the literature. World J Clin Cases 2023; 11:7440-7449. [PMID: 37969466 PMCID: PMC10643063 DOI: 10.12998/wjcc.v11.i30.7440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Neonatal hypertension is a rare but potentially serious condition that requires careful monitoring and treatment. Pharmacogenomics can help guide individualized drug therapy and improve outcomes. CASE SUMMARY We report a case of a preterm infant with multiple complications, including bronchopulmonary dysplasia (BPD), sepsis, intracranial hemorrhage, and hypertension. The infant was treated with various drugs, including dexamethasone and amlodipine. The infant was diagnosed with neonatal hypertension based on blood pressure measurements exceeding the 95th percentile for his age and sex. The possible causes of hypertension included dexamethasone, hydrochlorothiazide, spironolactone, and BPD. The infant was treated with oral amlodipine to lower his blood pressure. A pharmacogenomic test was performed to evaluate the genetic polymorphisms of ABCB1 and CYP3A5, which are involved in the metabolism and transport of dexamethasone and amlodipine. The infant's blood pressure was well controlled after the dose of amlodipine was reduced according to the pharmacogenomic results. The infant had a stable general condition and was discharged on the 100th d after birth. CONCLUSION This case illustrates the importance of regular blood pressure monitoring and etiological investigation in preterm infants with hypertension. Pharmacogenomics can provide useful information for individualized drug therapy and safety in this population.
Collapse
Affiliation(s)
- Lian-Fang Tang
- Department of Pediatrics, The First People’s Hospital of Yunnan Province, Kunming 650000, Yunnan Province, China
| | - Ao Xu
- Department of Pediatrics, The First People’s Hospital of Yunnan Province, Kunming 650000, Yunnan Province, China
| | - Kai Liu
- Pulmonary and Critical Care Medicine, Kunming Children’s Hospital, Kunming 650000, Yunnan Province, China
| |
Collapse
|
4
|
Role of glucocorticoid receptor mutations in hypertension and adrenal gland hyperplasia. Pflugers Arch 2022; 474:829-840. [PMID: 35732960 PMCID: PMC9217122 DOI: 10.1007/s00424-022-02715-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/03/2022]
Abstract
Hypertension is one of the leading causes of premature death in humans and exhibits a complex aetiology including environmental and genetic factors. Mutations within the glucocorticoid receptor (GR) can cause glucocorticoid resistance, which is characterized by several clinical features like hypercortisolism, hypokalaemia, adrenal hyperplasia and hypertension. Altered glucocorticoid receptor signalling further affects sodium and potassium homeostasis as well as blood pressure regulation and cell proliferation and differentiation that influence organ development and function. In salt-sensitive hypertension, excessive renal salt transport and sympathetic nervous system stimulation may occur simultaneously, and, thus, both the mineralocorticoid receptor (MR) and the GR-signalling may be implicated or even act interdependently. This review focuses on identified GR mutations in human primary generalized glucocorticoid resistance (PGGR) patients and their related clinical phenotype with specific emphasis on adrenal gland hyperplasia and hypertension. We compare these findings to mouse and rat mutants harbouring genetically engineered mutations to further dissect the cause and/or the consequence of clinical features which are common or different.
Collapse
|
5
|
Gu X, Ge L, Ren B, Fang Y, Li Y, Wang Y, Xu H. Glucocorticoids Promote Extracellular Matrix Component Remodeling by Activating YAP in Human Retinal Capillary Endothelial Cells. Front Cell Dev Biol 2022; 9:738341. [PMID: 34970541 PMCID: PMC8712730 DOI: 10.3389/fcell.2021.738341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Remodeling of extracellular matrix (ECM) components of endothelial cells is the main cause of retinal vascular basement membrane (BM) thickening, which leads to the initiation and perpetuation of microvasculopathy of diabetic retinopathy (DR). Excessive amounts of glucocorticoids (GCs) are related to the presence and severity of DR, however transcriptional effects of GCs on the biology of human retinal capillary endothelial cells (HRCECs) and its impacts on DR are still unclear. Here, we showed that GC (hydrocortisone) treatment induced ECM component [fibronectin (FN) and type IV collagen (Col IV)] expression and morphological changes in HRCECs via the glucocorticoid receptor (GR), which depended on the nuclear translocation of YAP coactivator. Mechanistically, GCs induced stress fiber formation in HRCECs, while blocking stress fiber formation inhibited GC-induced YAP nuclear translocation. Overexpression of FN, but not Col IV, activated YAP through the promotion of stress fiber formation via ECM-integrin signaling. Thus, a feedforward loop is established to sustain YAP activity. Using mRNA sequencing of HRCECs with overexpressed YAP or GC treatment, we found a similarity in Gene Ontology (GO) terms, differentially expressed genes (DEGs) and transcription factors (TFs) between the two RNA-seq datasets. In vivo, YAP was activated in retina vascular ECs of STZ-induced diabetic mice, and TF prediction analysis of published RNA-seq data of dermal vascular ECs from T2DM patients showed that GR and TEAD (the main transcription factor for YAP) were enriched. Together, GCs activate YAP and promote ECM component (FN and Col IV) remodeling in retinal capillary endothelial cells, and the underlying regulatory mechanism may provide new insights into the vascular BM thickening of the retina in the early pathogenesis of DR.
Collapse
Affiliation(s)
- Xianliang Gu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Lingling Ge
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Bangqi Ren
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yajie Fang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yijian Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yi Wang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| |
Collapse
|
6
|
Role of Endothelial Glucocorticoid Receptor in the Pathogenesis of Kidney Diseases. Int J Mol Sci 2021; 22:ijms222413295. [PMID: 34948091 PMCID: PMC8706765 DOI: 10.3390/ijms222413295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Accepted: 12/07/2021] [Indexed: 01/12/2023] Open
Abstract
Glucocorticoids, as multifunctional hormones, are widely used in the treatment of various diseases including nephrological disorders. They are known to affect immunological cells, effectively treating many autoimmune and inflammatory processes. Furthermore, there is a growing body of evidence demonstrating the potent role of glucocorticoids in non-immune cells such as podocytes. Moreover, novel data show additional pathways and processes affected by glucocorticoids, such as the Wnt pathway or autophagy. The endothelium is currently considered as a key organ in the regulation of numerous kidney functions such as glomerular filtration, vascular tone and the regulation of inflammation and coagulation. In this review, we analyse the literature concerning the effects of endothelial glucocorticoid receptor signalling on kidney function in health and disease, with special focus on hypertension, diabetic kidney disease, glomerulopathies and chronic kidney disease. Recent studies demonstrate the potential role of endothelial GR in the prevention of fibrosis of kidney tissue and cell metabolism through Wnt pathways, which could have a protective effect against disease progression. Another important aspect covered in this review is blood pressure regulation though GR and eNOS. We also briefly cover potential therapies that might affect the endothelial glucocorticoid receptor and its possible clinical implications, with special interest in selective or local GR stimulation and potential mitigation of GC treatment side effects.
Collapse
|
7
|
Dashti-Khavidaki S, Saidi R, Lu H. Current status of glucocorticoid usage in solid organ transplantation. World J Transplant 2021; 11:443-465. [PMID: 34868896 PMCID: PMC8603633 DOI: 10.5500/wjt.v11.i11.443] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/16/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids (GCs) have been the mainstay of immunosuppressive therapy in solid organ transplantation (SOT) for decades, due to their potent effects on innate immunity and tissue protective effects. However, some SOT centers are reluctant to administer GCs long-term because of the various related side effects. This review summarizes the advantages and disadvantages of GCs in SOT. PubMed and Scopus databases were searched from 2011 to April 2021 using search syntaxes covering “transplantation” and “glucocorticoids”. GCs are used in transplant recipients, transplant donors, and organ perfusate solution to improve transplant outcomes. In SOT recipients, GCs are administered as induction and maintenance immunosuppressive therapy. GCs are also the cornerstone to treat acute antibody- and T-cell-mediated rejections. Addition of GCs to organ perfusate solution and pretreatment of transplant donors with GCs are recommended by some guidelines and protocols, to reduce ischemia-reperfusion injury peri-transplant. GCs with low bioavailability and high potency for GC receptors, such as budesonide, nanoparticle-mediated targeted delivery of GCs to specific organs, and combination use of dexamethasone with inducers of immune-regulatory cells, are new methods of GC application in SOT patients to reduce side effects or induce immune-tolerance instead of immunosuppression. Various side effects involving different non-targeted organs/tissues, such as bone, cardiovascular, neuromuscular, skin and gastrointestinal tract, have been noted for GCs. There are also potential drug-drug interactions for GCs in SOT patients.
Collapse
Affiliation(s)
- Simin Dashti-Khavidaki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155, Iran
| | - Reza Saidi
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| |
Collapse
|
8
|
Hendrickx JO, De Moudt S, Van Dam D, De Deyn PP, Fransen P, De Meyer GRY. Altered stress hormone levels affect in vivo vascular function in the hAPP23 +/- overexpressing mouse model of Alzheimer's disease. Am J Physiol Heart Circ Physiol 2021; 321:H905-H919. [PMID: 34506227 DOI: 10.1152/ajpheart.00254.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/02/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) has long been considered a brain-specific dementia syndrome. However, in recent decades, the occurrence of cardiovascular (CV) disease in the progression of AD has been confirmed by increasing epidemiological evidence. In this study, we conducted an in-depth cardiovascular characterization of a humanized amyloid precursor protein (APP) overexpressing mouse model (hAPP23+/-), which overexpresses the Swedish mutation (KM670/671NL). At the age of 6 mo, hAPP23+/- mice had a lower survival, lower body weight, and increased corticosterone and VMA levels compared with C57BL/6 littermates. Systolic blood pressure was increased in hAPP23+/- animals compared with C57BL/6 littermates, but diastolic blood pressure was not statistically different. Pulse pressure remained unchanged but abdominal and carotid pulse-wave velocity (aPWV and cPWV) were increased in hAPP23+/- compared with C57BL/6 mice. Echocardiography showed no differences in systolic or diastolic cardiac function. Ex vivo evaluation of vascular function showed decreased adreno receptor dependent vasoconstriction of hAPP23+/- aortic segments, although the isobaric biomechanics of the aortic wall were similar to C57BL/6 aortic segments. In conclusion, hAPP23+/- mice exhibited high serum corticosterone levels, elevated systolic blood pressure, and increased arterial stiffness in vivo. However, ex vivo aortic stiffness of hAPP23+/- aortic segments was not changed and vascular reactivity to α1-adrenoceptor stimulation was attenuated. These findings highlight the need for more frequent assessment of circulating stress hormone levels and PWV measurements in daily clinical practice for people at risk of AD.NEW & NOTEWORTHY We showed that male amyloid precursor protein (APP) transgenic mice have higher circulating stress hormone levels. As a result, higher systolic blood pressure and pulse-wave velocity were measured in vivo in addition to a smaller α-adrenergic receptor-dependent contraction upon ex vivo stimulation with phenylephrine. Our findings highlight the need for more frequent assessment of circulating stress hormone levels and PWV measurements in daily clinical practice for people at risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Jhana O Hendrickx
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Paul Fransen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
9
|
Chen F, Hao L, Zhu S, Yang X, Shi W, Zheng K, Wang T, Chen H. Potential Adverse Effects of Dexamethasone Therapy on COVID-19 Patients: Review and Recommendations. Infect Dis Ther 2021; 10:1907-1931. [PMID: 34296386 PMCID: PMC8298044 DOI: 10.1007/s40121-021-00500-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the context of the coronavirus disease 2019 (COVID-19) pandemic, the global healthcare community has raced to find effective therapeutic agents against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To date, dexamethasone is the first and an important therapeutic to significantly reduce the risk of death in COVID-19 patients with severe disease. Due to powerful anti-inflammatory and immunosuppressive effects, dexamethasone could attenuate SARS-CoV-2-induced uncontrolled cytokine storm, severe acute respiratory distress syndrome and lung injury. Nevertheless, dexamethasone treatment is a double-edged sword, as numerous studies have revealed that it has significant adverse impacts later in life. In this article, we reviewed the literature regarding the adverse effects of dexamethasone administration on different organ systems as well as related disease pathogenesis in an attempt to clarify the potential harms that may arise in COVID-19 patients receiving dexamethasone treatment. Overall, taking the threat of COVID19 pandemic into account, we think it is necessary to apply dexamethasone as a pharmaceutical therapy in critical patients. However, its adverse side effects cannot be ignored. Our review will help medical professionals in the prognosis and follow-up of patients treated with dexamethasone. In addition, given that a considerable amount of uncertainty, confusion and even controversy still exist, further studies and more clinical trials are urgently needed to improve our understanding of the parameters and the effects of dexamethasone on patients with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Fei Chen
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China.
| | - Lanting Hao
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Shiheng Zhu
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Xinyuan Yang
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Wenhao Shi
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Kai Zheng
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Tenger Wang
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Huiran Chen
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| |
Collapse
|
10
|
Liu B, Zhou H, Zhang T, Gao X, Tao B, Xing H, Zhuang Z, Dardik A, Kyriakides TR, Goodwin JE. Loss of endothelial glucocorticoid receptor promotes angiogenesis via upregulation of Wnt/β-catenin pathway. Angiogenesis 2021; 24:631-645. [PMID: 33650028 PMCID: PMC8292305 DOI: 10.1007/s10456-021-09773-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Objective The glucocorticoid receptor (GR) is a member of the nuclear receptor family that controls key biological processes in the cardiovascular system and has recently been shown to modulate Wnt signaling in endothelial cells. Wnt/β-catenin signaling has been demonstrated to be crucial in the process of angiogenesis. In the current study, we studied whether GR could regulate angiogenesis via the Wnt/β-catenin pathway. Approach and Resultsa Key components of the Wnt/β-catenin pathway were evaluated using quantitative PCR and Western blot in the presence or absence of GR. Enhanced angiogenesis was found in GR deficiency in vitro and confirmed with cell viability assays, proliferation assays and tube formation assays. Consistent with these in vitro findings, endothelial cell-specific GR loss GR in vivo promoted angiogenesis in both a hind limb ischemia model and sponge implantation assay. Results were further verified in a novel mouse model lacking endothelial LRP5/6, a key receptor in canonical Wnt signaling, and showed substantially suppressed angiogenesis using these same in vitro and in vivo assays. To further investigate the mechanism of GR regulation of Wnt signaling, autophagy flux was investigated in endothelial cells by visualizing auto phagolysosomes as well as by assessing P62 degradation and LC3B conversion. Results indicated that potentiated autophagy flux participated in GR-Wnt regulation. Conclusions Lack of endothelial GR triggers autophagy flux, leads to activation of Wnt/β-catenin signaling and promotes angiogenesis. There may also be a synergistic interaction between autophagy and Wnt/β-catenin signaling. Supplementary Information The online version of this article (10.1007/s10456-021-09773-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bing Liu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Han Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Tiening Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xixiang Gao
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University and Institute of Vascular Surgery, Capital Medical University, Beijing, China
| | - Bo Tao
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Hao Xing
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Zhenwu Zhuang
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, CT, 06510-3221, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT, 06516, USA
| | - Themis R Kyriakides
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Pathology, Yale University, New Haven, CT, 06510, USA
| | - Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
11
|
You Y, Tan W, Guo Y, Luo M, Shang FF, Xia Y, Luo S. Progesterone promotes endothelial nitric oxide synthase expression through enhancing nuclear progesterone receptor-SP-1 formation. Am J Physiol Heart Circ Physiol 2020; 319:H341-H348. [PMID: 32618512 DOI: 10.1152/ajpheart.00206.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Progesterone exerts antihypertensive actions partially by modulating endothelial nitric oxide synthase (eNOS) activity. Here, we aimed to investigate the effects and mechanisms of progesterone on eNOS expression. First, human umbilical vein endothelial cells (HUVECs) were exposed to progesterone and then the eNOS transcription factor specificity protein-1 (SP-1) and progesterone receptor (PRA/B) expression were assessed by Western blotting and qRT-PCR. The interaction between SP-1 and PRA/B was next determined through coimmunoprecipitation assay. The chromatin immunoprecipitation assay and luciferase assay were used to investigate the relationship of PRA/B, SP-1, and eNOS promoter. At last, rats were intraperitoneally injected with progesterone receptor antagonist RU-486, and then the expression of eNOS and vasodilation function in thoracic aorta and mesenteric artery were measured. The results showed that progesterone could increase eNOS expression in HUVECs. Further study showed that progesterone increased PRA-SP-1 complex formation and facilitated PRA/B and SP-1 binding to eNOS promoter. Mutating SP-1 or PR-binding motif on eNOS promoter abolished the effect of progesterone on eNOS gene transcription. We also observed that progesterone receptor antagonist RU-486 reduced eNOS expression and impaired vasodilation in rats. Those results suggest that progesterone modulates eNOS expression through promoting PRA-SP-1 complex formation, and progesterone antagonist attenuates eNOS expression, leading to the loss of vascular relaxation.NEW & NOTEWORTHY Progesterone directly upregulated endothelial nitric oxide synthase (eNOS) expression in human endothelial cells. Progesterone augmented eNOS promoter activity through a progesterone receptor A- and specificity protein-1-dependent manner. Antagonism of the progesterone receptor reduced eNOS expression and impaired vasodilation in rats.
Collapse
Affiliation(s)
- Yuehua You
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wanying Tan
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minghao Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei-Fei Shang
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yong Xia
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
- Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio
| | - Suxin Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Kračun D, Klop M, Knirsch A, Petry A, Kanchev I, Chalupsky K, Wolf CM, Görlach A. NADPH oxidases and HIF1 promote cardiac dysfunction and pulmonary hypertension in response to glucocorticoid excess. Redox Biol 2020; 34:101536. [PMID: 32413743 PMCID: PMC7226895 DOI: 10.1016/j.redox.2020.101536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular side effects are frequent problems accompanying systemic glucocorticoid therapy, although the underlying mechanisms are not fully resolved. Reactive oxygen species (ROS) have been shown to promote various cardiovascular diseases although the link between glucocorticoid and ROS signaling has been controversial. As the family of NADPH oxidases has been identified as important source of ROS in the cardiovascular system we investigated the role of NADPH oxidases in response to the synthetic glucocorticoid dexamethasone in the cardiovascular system in vitro and in vivo in mice lacking functional NADPH oxidases due to a mutation in the gene coding for the essential NADPH oxidase subunit p22phox. We show that dexamethasone induced NADPH oxidase-dependent ROS generation, leading to vascular proliferation and angiogenesis due to activation of the transcription factor hypoxia-inducible factor-1 (HIF1). Chronic treatment of mice with low doses of dexamethasone resulted in the development of systemic hypertension, cardiac hypertrophy and left ventricular dysfunction, as well as in pulmonary hypertension and pulmonary vascular remodeling. In contrast, mice deficient in p22phox-dependent NADPH oxidases were protected against these cardiovascular side effects. Mechanistically, dexamethasone failed to upregulate HIF1α levels in these mice, while vascular HIF1α deficiency prevented pulmonary vascular remodeling. Thus, p22phox-dependent NADPH oxidases and activation of the HIF pathway are critical elements in dexamethasone-induced cardiovascular pathologies and might provide interesting targets to limit cardiovascular side effects in patients on chronic glucocorticoid therapy.
Collapse
Affiliation(s)
- Damir Kračun
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Mathieu Klop
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Anna Knirsch
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Ivan Kanchev
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Karel Chalupsky
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany; Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Cordula M Wolf
- Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
13
|
Soto-Piña AE, Franklin C, Rani CSS, Fernandez E, Cardoso-Peña E, Benítez-Arciniega AD, Gottlieb H, Hinojosa-Laborde C, Strong R. Dexamethasone Causes Hypertension in Rats Even Under Chemical Blockade of Peripheral Sympathetic Nerves. Front Neurosci 2019; 13:1305. [PMID: 31866814 PMCID: PMC6909820 DOI: 10.3389/fnins.2019.01305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/21/2019] [Indexed: 11/19/2022] Open
Abstract
Synthetic glucocorticoids (GCs) are widely used to treat inflammatory conditions. However, chronic use of GCs can lead to hypertension. The cause of this undesired side effect remains unclear. Previously, we developed an in vivo rat model to study the mechanisms underlying hypertension induced by the chronic administration of the potent synthetic GC, dexamethasone (DEX) and found that the catecholamine biosynthetic pathway plays an important role. In the current study, we used this model to investigate the role of the adrenal medulla, renal nerves, and other peripheral sympathetic nerves in DEX-induced hypertension. After 5 days of baseline telemetric recording of mean arterial pressure (MAP) and heart rate (HR), rats were subjected to one of the following treatments: renal denervation (RDNX), adrenal medullectomy (ADMX), 6-hydroxydopamine (6-OHDA, 20 mg/kg, i.p.) to induce chemical sympathectomy, or a combination of ADMX and 6-OHDA. On day 11, the animals received vehicle (VEH) or DEX in drinking water for 7 days, with the latter causing an increase in MAP in control animals. ADMX and RDNX by themselves exacerbated the pressor effect of DEX. In the chemical sympathectomy group, DEX still caused a rise in MAP but the response was lower (ΔMAP of 6-OHDA/DEX < VEH/DEX, p = 0.039). However, when ΔMAP was normalized to day 10, 6-OHDA + DEX did not show any difference from VEH + DEX, certainly not an increase as observed in DEX + ADMX or RDNX groups. This indicates that sympathetic nerves do not modulate the pressor effect of DEX. TH mRNA levels increased in the adrenal medulla in both VEH/DEX (p = 0.009) and 6-OHDA/DEX (p = 0.031) groups. In the 6-OHDA group, DEX also increased plasma levels of norepinephrine (NE) (p = 0.016). Our results suggest that the activation of catecholamine synthetic pathway could be involved in the pressor response to DEX in animals even under chemical sympathectomy with 6-OHDA.
Collapse
Affiliation(s)
| | - Cynthia Franklin
- Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX, United States
| | - C S Sheela Rani
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Elizabeth Fernandez
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Elías Cardoso-Peña
- Unidad de Medicina Familiar 220, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | | - Helmut Gottlieb
- Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX, United States
| | - Carmen Hinojosa-Laborde
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Randy Strong
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, United States
| |
Collapse
|
14
|
Liu B, Zhang TN, Knight JK, Goodwin JE. The Glucocorticoid Receptor in Cardiovascular Health and Disease. Cells 2019; 8:cells8101227. [PMID: 31601045 PMCID: PMC6829609 DOI: 10.3390/cells8101227] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
The glucocorticoid receptor is a member of the nuclear receptor family that controls many distinct gene networks, governing various aspects of development, metabolism, inflammation, and the stress response, as well as other key biological processes in the cardiovascular system. Recently, research in both animal models and humans has begun to unravel the profound complexity of glucocorticoid signaling and convincingly demonstrates that the glucocorticoid receptor has direct effects on the heart and vessels in vivo and in vitro. This research has contributed directly to improving therapeutic strategies in human disease. The glucocorticoid receptor is activated either by the endogenous steroid hormone cortisol or by exogenous glucocorticoids and acts within the cardiovascular system via both genomic and non-genomic pathways. Polymorphisms of the glucocorticoid receptor are also reported to influence the progress and prognosis of cardiovascular disease. In this review, we provide an update on glucocorticoid signaling and highlight the critical role of this signaling in both physiological and pathological conditions of the cardiovascular system. With increasing in-depth understanding of glucocorticoid signaling, the future is promising for the development of targeted glucocorticoid treatments and improved clinical outcomes.
Collapse
Affiliation(s)
- Bing Liu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Tie-Ning Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jessica K Knight
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
15
|
Rosiglitazone, a Ligand to PPAR γ, Improves Blood Pressure and Vascular Function through Renin-Angiotensin System Regulation. PPAR Res 2019; 2019:1371758. [PMID: 30863432 PMCID: PMC6378057 DOI: 10.1155/2019/1371758] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 01/20/2019] [Indexed: 12/16/2022] Open
Abstract
Rosiglitazone (RGZ), a peroxisome proliferator-activated receptor gamma (PPARγ) ligand, has been reported to act as insulin sensitizer and exert cardiovascular actions. In this work, we hypothesized that RGZ exerts a PPARγ–dependent regulation of blood pressure through modulation of angiotensin-converting enzyme (ACE)-type 2 (ACE2)/angiotensin-(1-7)/angiotensin II type-2 receptor (AT2R) axis in an experimental model of high blood pressure. We carried on experiments in normotensive (Sham) and aortic coarctation (AoCo)-induced hypertensive male Wistar rats. Both sham and AoCo rats were treated 7 days with vehicle (V), RGZ (5 mg/kg/day), or RGZ+BADGE (120 mg/kg/day) post-coarctation. We measured blood pressure and vascular reactivity on aortic rings, as well as the expression of renin-angiotensin system (RAS) proteins. We found that RGZ treatment in AoCo group decreases blood pressure values and improves vascular response to acetylcholine, both parameters dependent on PPARγ-stimulation. RGZ lowered serum angiotensin II (AngII) but increased Ang-(1-7) levels. It also decreased 8-hydroxy-2′-deoxyguanosine (8-OH-2dG), malondialdehyde (MDA), and improved the antioxidant capacity. Regarding protein expression of RAS, RGZ decreases ACE and angiotensin II type 1 receptor (AT1R) and improved ACE2, AT2R, and Mas receptor in AoCo rats. Additionally, an in silico analysis revealed that 5′UTR regions of RAS and PPARγ share motifs with a transcriptional regulatory role. We conclude that RGZ lowers blood pressure values by increasing the expression of RAS axis proteins ACE2 and AT2R, decreasing the levels of AngII and increasing levels of Ang-(1-7) in a PPARγ-dependent manner. The in silico analysis is a valuable tool to predict the interaction between PPARγ and RAS.
Collapse
|
16
|
Balkaya M, Bacak G. Effects of L-NAME, DEXA and L-NAME+DEXA on Systemic Blood Pressure of Hypertensive Pregnant and Non-Pregnant Wistar albino Rats. ACTA ACUST UNITED AC 2018. [DOI: 10.30704/http-www-jivs-net.460833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
17
|
Kho J, Tian X, Wong WT, Bertin T, Jiang MM, Chen S, Jin Z, Shchelochkov OA, Burrage LC, Reddy AK, Jiang H, Abo-Zahrah R, Ma S, Zhang P, Bissig KD, Kim JJ, Devaraj S, Rodney GG, Erez A, Bryan NS, Nagamani SC, Lee BH. Argininosuccinate Lyase Deficiency Causes an Endothelial-Dependent Form of Hypertension. Am J Hum Genet 2018; 103:276-287. [PMID: 30075114 DOI: 10.1016/j.ajhg.2018.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
Primary hypertension is a major risk factor for ischemic heart disease, stroke, and chronic kidney disease. Insights obtained from the study of rare Mendelian forms of hypertension have been invaluable in elucidating the mechanisms causing primary hypertension and development of antihypertensive therapies. Endothelial cells play a key role in the regulation of blood pressure; however, a Mendelian form of hypertension that is primarily due to endothelial dysfunction has not yet been described. Here, we show that the urea cycle disorder, argininosuccinate lyase deficiency (ASLD), can manifest as a Mendelian form of endothelial-dependent hypertension. Using data from a human clinical study, a mouse model with endothelial-specific deletion of argininosuccinate lyase (Asl), and in vitro studies in human aortic endothelial cells and induced pluripotent stem cell-derived endothelial cells from individuals with ASLD, we show that loss of ASL in endothelial cells leads to endothelial-dependent vascular dysfunction with reduced nitric oxide (NO) production, increased oxidative stress, and impaired angiogenesis. Our findings show that ASLD is a unique model for studying NO-dependent endothelial dysfunction in human hypertension.
Collapse
|
18
|
Ivy JR, Evans LC, Moorhouse R, Richardson RV, Al-Dujaili EAS, Flatman PW, Kenyon CJ, Chapman KE, Bailey MA. Renal and Blood Pressure Response to a High-Salt Diet in Mice With Reduced Global Expression of the Glucocorticoid Receptor. Front Physiol 2018; 9:848. [PMID: 30038578 PMCID: PMC6046455 DOI: 10.3389/fphys.2018.00848] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/14/2018] [Indexed: 01/02/2023] Open
Abstract
Salt-sensitive hypertension is common in glucocorticoid excess. Glucocorticoid resistance also presents with hypercortisolemia and hypertension but the relationship between salt intake and blood pressure (BP) is not well defined. GRβgeo/+ mice have global glucocorticoid receptor (GR) haploinsufficiency and increased BP. Here we examined the effect of high salt diet on BP, salt excretion and renal blood flow in GRβgeo/+mice. Basal BP was ∼10 mmHg higher in male GRβgeo/+ mice than in GR+/+ littermates. This modest increase was amplified by ∼10 mmHg following a high-salt diet in GRβgeo/+ mice. High salt reduced urinary aldosterone excretion but increased renal mineralocorticoid receptor expression in both genotypes. Corticosterone, and to a lesser extent deoxycorticosterone, excretion was increased in GRβgeo/+ mice following a high-salt challenge, consistent with enhanced 24 h production. GR+/+ mice increased fractional sodium excretion and reduced renal vascular resistance during the high salt challenge, retaining neutral sodium balance. In contrast, sodium excretion and renal vascular resistance did not adapt to high salt in GRβgeo/+ mice, resulting in transient sodium retention and sustained hypertension. With high-salt diet, Slc12a3 and Scnn1a mRNAs were higher in GRβgeo/+ than controls, and this was reflected in an exaggerated natriuretic response to thiazide and benzamil, inhibitors of NCC and ENaC, respectively. Reduction in GR expression causes salt-sensitivity and an adaptive failure of the renal vasculature and tubule, most likely reflecting sustained mineralocorticoid receptor activation. This provides a mechanistic basis to understand the hypertension associated with loss-of-function polymorphisms in GR in the context of habitually high salt intake.
Collapse
Affiliation(s)
- Jessica R Ivy
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Louise C Evans
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rebecca Moorhouse
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel V Richardson
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emad A S Al-Dujaili
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Peter W Flatman
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher J Kenyon
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Karen E Chapman
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew A Bailey
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
19
|
Whirledge S, DeFranco DB. Glucocorticoid Signaling in Health and Disease: Insights From Tissue-Specific GR Knockout Mice. Endocrinology 2018; 159:46-64. [PMID: 29029225 PMCID: PMC5761604 DOI: 10.1210/en.2017-00728] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are adrenally produced hormones critically involved in development, general physiology, and control of inflammation. Since their discovery, glucocorticoids have been widely used to treat a variety of inflammatory conditions. However, high doses or prolonged use leads to a number of side effects throughout the body, which preclude their clinical utility. The primary actions of glucocorticoids are mediated by the glucocorticoid receptor (GR), a transcription factor that regulates many complex signaling pathways. Although GR is nearly ubiquitous throughout the body, glucocorticoids exhibit cell- and tissue-specific effects. For example, glucocorticoids stimulate glucose production in the liver, reduce glucose uptake in the skeletal muscle, and decrease insulin secretion from the pancreatic β-cells. Mouse models represent an important approach to understanding the dynamic functions of GR signaling in normal physiology, disease, and resistance. In the absence of a viable GR null model, gene-targeting techniques utilizing promoter-driven recombination have provided an opportunity to characterize the tissue-specific actions of GR. The aim of the present review is to describe the organ systems in which GR has been conditionally deleted and summarize the functions ascribed to glucocorticoid action in those tissues.
Collapse
Affiliation(s)
- Shannon Whirledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut 06520
| | - Donald B. DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
20
|
Abstract
The metabolic syndrome describes a clustering of risk factors—visceral obesity, dyslipidaemia, insulin resistance, and salt-sensitive hypertension—that increases mortality related to cardiovascular disease, type 2 diabetes, cancer, and non-alcoholic fatty liver disease. The prevalence of these concurrent comorbidities is ~ 25–30% worldwide, and metabolic syndrome therefore presents a significant global public health burden. Evidence from clinical and preclinical studies indicates that glucocorticoid excess is a key causal feature of metabolic syndrome. This is not increased systemic in circulating cortisol, rather increased bioavailability of active glucocorticoids within tissues. This review examines the role of covert glucocorticoid excess on the hypertension of the metabolic syndrome. Here, the role of the 11β-hydroxysteroid dehydrogenase enzymes, which exert intracrine and paracrine control over glucocorticoid signalling, is examined. 11βHSD1 amplifies glucocorticoid action in cells and contributes to hypertension through direct and indirect effects on the kidney and vasculature. The deactivation of glucocorticoid by 11βHSD2 controls ligand access to glucocorticoid and mineralocorticoid receptors: loss of function promotes salt retention and hypertension. As for hypertension in general, high blood pressure in the metabolic syndrome reflects a complex interaction between multiple systems. The clear association between high dietary salt, glucocorticoid production, and metabolic disorders has major relevance for human health and warrants systematic evaluation.
Collapse
Affiliation(s)
- Matthew A Bailey
- The British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
21
|
Loss of the podocyte glucocorticoid receptor exacerbates proteinuria after injury. Sci Rep 2017; 7:9833. [PMID: 28852159 PMCID: PMC5575043 DOI: 10.1038/s41598-017-10490-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Nephrotic syndrome is a common disorder in adults and children whose etiology is largely unknown. Glucocorticoids remain the mainstay of therapy in most cases, though their mechanism of action remains poorly understood. Emerging evidence suggests that immunomodulatory therapies used in nephrotic syndrome directly target the podocytes. To study how steroids directly affect the podocytes in the treatment of proteinuria, we created a mouse model with podocyte-specific deletion of the glucocorticoid receptor. The podocyte-specific glucocorticoid receptor (GR) knockout mice had similar renal function and protein excretion compared to wild type. However, after glomerular injury induced by either LPS or nephrotoxic serum, the podocyte GR knockout mice demonstrated worsened proteinuria compared to wild type. Ultrastructural examination of podocytes confirmed more robust foot process effacement in the knockout animals. Expression of several key slit diaphragm protein was down regulated in pGR KO mice. Primary podocytes isolated from wild type and podocyte GR knockout mice showed similar actin stress fiber staining patterns in unstimulated conditions. Yet, when exposed to LPS, GR knockout podocytes demonstrated fewer stress fibers and impaired migration compared to wild type podocytes. We conclude that the podocyte glucocorticoid receptor is important for limiting proteinuria in settings of podocyte injury.
Collapse
|
22
|
Puzserova A, Bernatova I. Blood pressure regulation in stress: focus on nitric oxide-dependent mechanisms. Physiol Res 2017; 65:S309-S342. [PMID: 27775419 DOI: 10.33549/physiolres.933442] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Stress is considered a risk factor associated with the development of various civilization diseases including cardiovascular diseases, malignant tumors and mental disorders. Research investigating mechanisms involved in stress-induced hypertension have attracted much attention of physicians and researchers, however, there are still ambiguous results concerning a causal relationship between stress and long-term elevation of blood pressure (BP). Several studies have observed that mechanisms involved in the development of stress-induced hypertension include increased activity of sympathetic nervous system (SNS), glucocorticoid (GC) overload and altered endothelial function including decreased nitric oxide (NO) bioavailability. Nitric oxide is well known neurotransmitter, neuromodulator and vasodilator involved in regulation of neuroendocrine mechanisms and cardiovascular responses to stressors. Thus NO plays a crucial role in the regulation of the stress systems and thereby in the BP regulation in stress. Elevated NO synthesis, especially in the initial phase of stress, may be considered a stress-limiting mechanism, facilitating the recovery from stress to the resting levels via attenuation of both GC release and SNS activity as well as by increased NO-dependent vasorelaxation. On the other hand, reduced levels of NO were observed in the later phases of stress and in subjects with genetic predisposition to hypertension, irrespectively, in which reduced NO bioavailability may account for disruption of NO-mediated BP regulatory mechanisms and accentuated SNS and GC effects. This review summarizes current knowledge on the role of stress in development of hypertension with a special focus on the interactions among NO and other biological systems affecting blood pressure and vascular function.
Collapse
Affiliation(s)
- A Puzserova
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | |
Collapse
|
23
|
Liern M, Codianni P, Vallejo G. [Comparative study of the conventional scheme and prolonged treatment with steroids on primary steroid-sensitive nephrotic syndrome in children]. BOLETIN MEDICO DEL HOSPITAL INFANTIL DE MEXICO 2016; 73:309-317. [PMID: 29384123 DOI: 10.1016/j.bmhimx.2016.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/06/2016] [Accepted: 07/25/2016] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND In the steroid-sensitive nephrotic syndrome (SSNS) the prolonged treatment with steroids could decrease the frequency of relapses. We conducted a comparative study of prolonged steroid scheme and the usual treatment of primary SSNS to assess: the number of patients with relapses, mean time to treatment initiation, to remission and to first relapse, total number of relapses, total cumulative dose of steroids, and the steroid toxicity. METHODS Patients were divided into two groups: group A (27 patients) received 16-β-methylprednisolone for 12 weeks, reducing the steroid until week 24. Group B (29 patients) received 16-β-methylprednisolone for 12 weeks and placebo until week 24. RESULTS Cumulative incidence rate of relapse (person/years) for group A was of 36/100 and 66/100 for group B (p=0.04). Average elapsed time to first relapse was of 114 days for group A and of 75 days to for group B (p=0.01). The difference in time for initial response to treatment and up to achieve remission between both groups was not significant. Total cumulative relapses were 9 for group A and 17 for group B (p=0.04). Total patients with relapses were 3 for group A and 7 for group B (p=0.17). Cumulative average dose per patient was 5,243mg/m2 for group A and 4,306mg/m2 for group B (p=0.3), and serum cortisol was 14μg/dl for group A and 16μg/dl for group B (p=0.4). There were no steroid toxicity differences between groups. CONCLUSIONS The duration of the treatment had an impact on the number of relapses without increasing steroid toxicity.
Collapse
Affiliation(s)
- Miguel Liern
- Servicio de Nefrología, Hospital General de Niños Dr. Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina.
| | - Paola Codianni
- Servicio de Nefrología, Hospital General de Niños Dr. Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| | - Graciela Vallejo
- Servicio de Nefrología, Hospital General de Niños Dr. Ricardo Gutiérrez, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
24
|
Chang HY, Tain YL. Postnatal dexamethasone-induced programmed hypertension is related to the regulation of melatonin and its receptors. Steroids 2016; 108:1-6. [PMID: 26921678 DOI: 10.1016/j.steroids.2016.02.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 02/18/2016] [Accepted: 02/20/2016] [Indexed: 01/08/2023]
Abstract
Adulthood hypertension can be programmed by glucocorticoid exposure in early life. We found that maternal melatonin therapy prevents postnatal dexamethasone (DEX)-induced programmed hypertension. Melatonin acts through specific receptors, including MT1 and MT2 membrane receptors, and retinoid related orphan nuclear receptors of the RZR/ROR family. Thus we tested whether postnatal DEX-induced hypertension is related to changes of melatonin receptors in the kidney and heart, which was preserved by maternal melatonin therapy. Male neonates were assigned to four groups (n=6-8/group): control, DEX, control+melatonin (MEL), and DEX+MEL. Male rat pups were injected i.p. with DEX on d 1 (0.5 mg/kg BW), d 2 (0.3 mg/kg BW), and d 3 (0.1 mg/kg BW) after birth. Melatonin was administered in drinking water (0.01%) during the lactation period. We found DEX group developed hypertension at 16 weeks of age, which melatonin therapy prevented. Postnatal DEX treatment increased mRNA expression of MT1 and MT2, while decreased RORα and RZRβ in the kidney. These changes were prevented by melatonin therapy. Postnatal DEX decreased protein level of MT2 in the kidney, which was attenuated by melatonin therapy. Renal protein level of RORα was higher in DEX+MEL group compared to control and DEX group. Renal melatonin level was higher in the MEL and DEX+MEL groups compared to control. We concluded that melatonin therapy has long-term protection on postnatal DEX-induced programmed hypertension, which is associated with regulation on melatonin receptors in the kidney. Our findings would offer potential therapeutic approaches to prevent programmed hypertension in premature baby receiving glucocorticoids.
Collapse
Affiliation(s)
- Hsin-Yu Chang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - You-Lin Tain
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
25
|
Effect of Formulation and Processing Variables on Dexamethasone Entrapment and Release of Niosomes. J CLUST SCI 2015. [DOI: 10.1007/s10876-015-0908-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
26
|
d'Emmanuele di Villa Bianca R, Mitidieri E, Donnarumma E, Tramontano T, Brancaleone V, Cirino G, Bucci M, Sorrentino R. Hydrogen sulfide is involved in dexamethasone-induced hypertension in rat. Nitric Oxide 2015; 46:80-6. [DOI: 10.1016/j.niox.2014.11.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/14/2014] [Accepted: 11/21/2014] [Indexed: 11/25/2022]
|
27
|
Goodwin JE, Zhang X, Rotllan N, Feng Y, Zhou H, Fernández-Hernando C, Yu J, Sessa WC. Endothelial glucocorticoid receptor suppresses atherogenesis--brief report. Arterioscler Thromb Vasc Biol 2015; 35:779-782. [PMID: 25810297 DOI: 10.1161/atvbaha.114.304525] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The purpose of this study was to determine the role of the endothelial glucocorticoid receptor in the pathogenesis of atherosclerosis. APPROACH AND RESULTS Control mice and mice lacking the endothelial glucocorticoid receptor were bred onto an Apoe knockout background and subjected to high-fat diet feeding for 12 weeks. Assessment of body weight and total cholesterol and triglycerides before and after the diet revealed no differences between the 2 groups of mice. However, mice lacking the endothelial glucocorticoid receptor developed more severe atherosclerotic lesions in the aorta, brachiocephalic artery, and aortic sinus, as well as a heightened inflammatory milieu as evidenced by increased macrophage recruitment in the lesions. CONCLUSIONS These data suggest that the endothelial glucocorticoid receptor is important for tonic inhibition of inflammation and limitation of atherosclerosis progression in this model.
Collapse
Affiliation(s)
- Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, CT
| | - Noemi Rotllan
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT
| | - Yan Feng
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Han Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Carlos Fernández-Hernando
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT
| | - Jun Yu
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, CT
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
28
|
Animal Models of Altered Glucocorticoid Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [DOI: 10.1007/978-1-4939-2895-8_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Goodwin JE. Glucocorticoids and the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [DOI: 10.1007/978-1-4939-2895-8_13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
Ji P, Hu H, Yang X, Wei X, Zhu C, Liu J, Feng Y, Yang F, Okanurak K, Li N, Zeng X, Zheng H, Wu Z, Lv Z. AcCystatin, an immunoregulatory molecule from Angiostrongylus cantonensis, ameliorates the asthmatic response in an aluminium hydroxide/ovalbumin-induced rat model of asthma. Parasitol Res 2014; 114:613-24. [PMID: 25399816 DOI: 10.1007/s00436-014-4223-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 12/19/2022]
Abstract
Epidemiological surveys have demonstrated that helminth infections are negatively related to atopic diseases, including asthma. Defining and characterising specific helminth molecules that have excellent immunomodulatory capacities as potential therapeutics for the treatment or prophylaxis of allergic manifestations are of great interest. AcCystatin, a cystatin protease inhibitor of Angiostrongylus cantonensis, is a homologue of other nematode cystatins with immunoregulatory properties. Here, we aim to determine the effects of AcCystatin on an ovalbumin/aluminium hydroxide (OVA/Al[OH]3)-induced rat model of asthma. Wistar rats were randomly divided into four groups, including a control group, an OVA/Al[OH]3-induced asthma group, a group receiving AcCystatin immunisation prior to OVA/Al[OH]3-induced asthma and a group receiving AcCystatin treatment after OVA/Al[OH]3-induced asthma. The numbers of eosinophils, basophils, neutrophils, lymphocytes and monocytes in the peripheral blood and of eosinophils in the bronchoalveolar lavage fluid (BALF) were counted for each animal. The expression levels of the cytokines interferon-γ, interleukin (IL) 4, IL-5, IL-6, IL-10, IL17A and tumour necrosis factor receptor-α in BALF, of OVA-specific immunoglobulin E in BALF and serum and of the chemokines eotaxin-1, eotaxin-2, eotaxin-3, MCP-1 and MCP-3 in lung tissue were measured. In addition, the degree of peribronchial and perivascular inflammation and the intensity of goblet cell metaplasia were qualitatively evaluated. The sensitised/challenged rats developed an extensive cell inflammatory response of the airways. AcCystatin administration significantly reduced the cellular infiltrate in the perivascular and peribronchial lung tissues and reduced both goblet mucous production and eosinophil infiltration. The rats that were treated with AcCystatin before or after sensitisation with OVA showed significant decreases in eotaxin-1, eotaxin-3 and MCP-1 expression in the lung tissue. The production of IL-4, IL-5, IL-6 and IL-17A and of OVA-specific IgE antibodies was also significantly reduced in AcCystatin-treated rats compared with untreated asthmatic rats. The AcCystatin treatment was associated with a significant increase in IL-10 levels. Our present findings provide the first demonstration that AcCystatin is an effective agent in the prevention and treatment of the airway inflammation associated with asthma.
Collapse
Affiliation(s)
- Pengyu Ji
- Zhongshan School of Medicine, Sun Yat-sen University, 74 2nd Zhongshan Road, Guangzhou, 510080, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Goodwin JE, Feng Y, Velazquez H, Zhou H, Sessa WC. Loss of the endothelial glucocorticoid receptor prevents the therapeutic protection afforded by dexamethasone after LPS. PLoS One 2014; 9:e108126. [PMID: 25299055 PMCID: PMC4191990 DOI: 10.1371/journal.pone.0108126] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/25/2014] [Indexed: 12/26/2022] Open
Abstract
Glucocorticoids are normally regarded as anti-inflammatory therapy for a wide variety of conditions and have been used with some success in treating sepsis and sepsis-like syndromes. We previously demonstrated that mice lacking the glucocorticoid receptor in the endothelium (GR EC KO mice) are extremely sensitive to low-dose LPS and demonstrate prolonged activation and up regulation of NF-κB. In this study we pre-treated these GR EC KO mice with dexamethasone and assessed their response to an identical dose of LPS. Surprisingly, the GR EC KO mice fared even worse than when given LPS alone demonstrating increased mortality, increased levels of the inflammatory cytokines TNF-α and IL-6 and increased nitric oxide release after the dexamethasone pre-treatment. As expected, control animals pre-treated with dexamethasone showed improvement in all parameters assayed. Mechanistically we demonstrate that GR EC KO mice show increased iNOS production and NF-κB activation despite treatment with dexamethasone.
Collapse
Affiliation(s)
- Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| | - Yan Feng
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Heino Velazquez
- Department of Internal Medicine, Veterans Affairs Hospital, West Haven, Connecticut, United States of America
| | - Han Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - William C. Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
32
|
Urbańska J, Karewicz A, Nowakowska M. Polymeric delivery systems for dexamethasone. Life Sci 2013; 96:1-6. [PMID: 24373835 DOI: 10.1016/j.lfs.2013.12.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/25/2013] [Accepted: 12/13/2013] [Indexed: 12/28/2022]
Abstract
Glucocorticoids (GCs) are broadly used in the treatment of inflammation and in suppressing hyperactivity of the immune system expressed in allergies, asthma, autoimmune diseases and sepsis. They are pleiotropic in nature, showing a wide range of diverse effects, including those which are harmful for the organism. Dexamethasone (DEX) is one of the most frequently used GCs and is considered as one of the safest. Still serious side-effects have been observed for this drug, mostly due to its hydrophobicity and low bioavailability. The potentially promising polymeric carrier systems to deliver DEX effectively are revised.
Collapse
Affiliation(s)
- Justyna Urbańska
- Faculty of Chemistry, Jagiellonian University, 30-060 Kraków, Ingardena 3, Poland
| | - Anna Karewicz
- Faculty of Chemistry, Jagiellonian University, 30-060 Kraków, Ingardena 3, Poland.
| | - Maria Nowakowska
- Faculty of Chemistry, Jagiellonian University, 30-060 Kraków, Ingardena 3, Poland
| |
Collapse
|
33
|
Prykhozhij SV, Marsico A, Meijsing SH. Zebrafish Expression Ontology of Gene Sets (ZEOGS): a tool to analyze enrichment of zebrafish anatomical terms in large gene sets. Zebrafish 2013; 10:303-15. [PMID: 23656298 DOI: 10.1089/zeb.2012.0865] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The zebrafish (Danio rerio) is an established model organism for developmental and biomedical research. It is frequently used for high-throughput functional genomics experiments, such as genome-wide gene expression measurements, to systematically analyze molecular mechanisms. However, the use of whole embryos or larvae in such experiments leads to a loss of the spatial information. To address this problem, we have developed a tool called Zebrafish Expression Ontology of Gene Sets (ZEOGS) to assess the enrichment of anatomical terms in large gene sets. ZEOGS uses gene expression pattern data from several sources: first, in situ hybridization experiments from the Zebrafish Model Organism Database (ZFIN); second, it uses the Zebrafish Anatomical Ontology, a controlled vocabulary that describes connected anatomical structures; and third, the available connections between expression patterns and anatomical terms contained in ZFIN. Upon input of a gene set, ZEOGS determines which anatomical structures are overrepresented in the input gene set. ZEOGS allows one for the first time to look at groups of genes and to describe them in terms of shared anatomical structures. To establish ZEOGS, we first tested it on random gene selections and on two public microarray datasets with known tissue-specific gene expression changes. These tests showed that ZEOGS could reliably identify the tissues affected, whereas only very few enriched terms to none were found in the random gene sets. Next we applied ZEOGS to microarray datasets of 24 and 72 h postfertilization zebrafish embryos treated with beclomethasone, a potent glucocorticoid. This analysis resulted in the identification of several anatomical terms related to glucocorticoid-responsive tissues, some of which were stage-specific. Our studies highlight the ability of ZEOGS to extract spatial information from datasets derived from whole embryos, indicating that ZEOGS could be a useful tool to automatically analyze gene expression pattern features of any large zebrafish gene set.
Collapse
|
34
|
Lee SR, Kim HK, Song IS, Youm J, Dizon LA, Jeong SH, Ko TH, Heo HJ, Ko KS, Rhee BD, Kim N, Han J. Glucocorticoids and their receptors: insights into specific roles in mitochondria. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 112:44-54. [PMID: 23603102 DOI: 10.1016/j.pbiomolbio.2013.04.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/27/2022]
Abstract
Glucocorticoids (GCs) affect most physiological systems and are the most frequently used drugs for multiple disorders and organ transplantation. GC functions depend on a balance between circulating GC and cytoplasmic glucocorticoid receptor II (GR). Mitochondria individually enclose circular, double-stranded DNA that is expressed and replicated in response to nuclear-encoded factors imported from the cytoplasm. Fine-tuning and response to cellular demands should be coordinately regulated by the nucleus and mitochondria; thus mitochondrial-nuclear interaction is vital to optimal mitochondrial function. Elucidation of the direct and indirect effects of steroids, including GCs, on mitochondria is an important and emerging field of research. Mitochondria may also be under GC control because GRs are present in mitochondria, and glucocorticoid response elements (GREs) reside in the mitochondrial genome. Therefore, mitochondrial gene expression can be regulated by GCs via at least two different mechanisms: direct action on mitochondrial DNA and oxidative phosphorylation (OXPHOS) genes, or by an indirect effect through interaction with nuclear genes. In this review, we outline possible mechanisms of regulation of mitochondrial genes in response to GCs in view of translocation of the GR into mitochondria and the possible regulation of OXPHOS genes by GREs in the mitochondrial genome.
Collapse
Affiliation(s)
- Sung-Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, 633-165 Gaegeum-Dong, Busanjin-Gu, 613-735 Busan, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kurakula K, Hamers AAJ, de Waard V, de Vries CJM. Nuclear Receptors in atherosclerosis: a superfamily with many 'Goodfellas'. Mol Cell Endocrinol 2013; 368:71-84. [PMID: 22664910 DOI: 10.1016/j.mce.2012.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 01/07/2023]
Abstract
Nuclear Receptors form a superfamily of 48 transcription factors that exhibit a plethora of functions in steroid hormone signaling, regulation of metabolism, circadian rhythm and cellular differentiation. In this review, we describe our current knowledge on the role of Nuclear Receptors in atherosclerosis, which is a multifactorial disease of the vessel wall. Various cell types are involved in this chronic inflammatory pathology in which multiple cellular processes and numerous genes are dysregulated. Systemic risk factors for atherosclerosis are among others adverse blood lipid profiles, enhanced circulating cytokine levels, as well as increased blood pressure. Since many Nuclear Receptors modulate lipid profiles or regulate blood pressure they indirectly affect atherosclerosis. In the present review, we focus on the functional involvement of Nuclear Receptors within the atherosclerotic vessel wall, more specifically on their modulation of cellular functions in endothelial cells, smooth muscle cells and macrophages. Collectively, this overview shows that most of the Nuclear Receptors are athero-protective in atherosclerotic lesions.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
36
|
Endothelial glucocorticoid receptor is required for protection against sepsis. Proc Natl Acad Sci U S A 2012; 110:306-11. [PMID: 23248291 DOI: 10.1073/pnas.1210200110] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The glucocorticoid receptor (GR) is ubiquitously expressed on nearly all cell types, but tissue-specific deletion of this receptor can produce dramatic whole organism phenotypes. In this study we investigated the role of the endothelial GR in sepsis in vivo and in vitro. Mice with an endothelial-specific GR deletion and controls were treated with 12.5 mg/kg LPS and phenotyped. Mice lacking GR showed significantly increased mortality, more hemodynamic instability, higher nitric oxide levels, and higher levels of the inflammatory cytokines, tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) compared with controls. There were no differences in rates of apoptosis or macrophage recruitment between the two groups. Both endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) expression were increased after LPS challenge in mice with endothelial GR deficiency, and aminoguanidine, a specific iNOS inhibitor in mice was able to rescue hemodynamic collapse in these animals. In vitro, human umbilical vein cells (HUVECs) subjected to GR knockdown by siRNA showed increased expression of eNOS at baseline that persisted after treatment with LPS. Both eNOS and iNOS mRNA was increased by qPCR. In HUVECs lacking GR, NF-κB levels and NF-κB-dependent genes tissue factor and IL-6 were increased compared with controls. Thus, endothelial GR is a critical regulator of NF-κB activation and nitric oxide synthesis in sepsis.
Collapse
|
37
|
Glucocorticoid-induced hypertension. Pediatr Nephrol 2012; 27:1059-66. [PMID: 21744056 DOI: 10.1007/s00467-011-1928-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
Glucocorticoid-induced hypertension is a common clinical problem that is poorly understood, thus rendering treatment strategies sub-optimal. This form of hypertension has been commonly thought to be mediated by excess sodium and water reabsorption by the renal mineralocorticoid receptor. However, experimental and clinical data in both humans and animal models suggest important roles for the glucocorticoid receptor as well, in both the pathogenesis and maintenance of this hypertension. The glucocorticoid receptor is widely expressed in a number of organ systems relevant to blood pressure regulation, including the kidney, the brain and the vasculature. In vitro studies in isolated kidney tissues as well as in vascular smooth muscle and vascular endothelial cells have attempted to elucidate the molecular physiology of glucocorticoid-induced hypertension, but have generally been limited by the inability to study signaling pathways in an intact organism. More recently, the power of mouse genetics has been employed to examine the tissue-specific contributions of vascular and extra-vascular tissues to this form of hypertension. Here we review recent developments in our understanding of the pathogenesis of glucocorticoid-induced hypertension.
Collapse
|
38
|
Abstract
Endocrine causes of hypertension are rare in children and screening for endocrine hypertension in children should be carried out only after ruling out renal and renovascular causes. Excess levels and/or action of mineralocorticoids associated with low renin levels lead to childhood hypertension and this can be caused by various conditions which are discussed in detail in the article. Childhood pheochromocytomas are being increasingly diagnosed because of the improved application of genetic testing for familial syndromes associated with pheochromocytomas. Adolescents with polycystic ovarian syndrome (PCOS) can also have hypertension associated with their obese phenotype.
Collapse
Affiliation(s)
- Nisha Bhavani
- Department of Endocrinology and Diabetes, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| |
Collapse
|